plant-derived substances and cardiovascular diseases

127
Linköping University Medical Dissertations No. 1097 Plant-Derived Substances and Cardiovascular Diseases -Effects of Flavonoids, Terpenes and Sterols on Angiotensin-Converting Enzyme and Nitric Oxide Ingrid A-L Persson Division of Drug Research / Pharmacology Department of Medical and Health Sciences Linköping University, Sweden Linköping 2009

Upload: others

Post on 02-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Plant-Derived Substances and Cardiovascular Diseases

Linköping University Medical Dissertations No. 1097

Plant-Derived Substances and Cardiovascular Diseases

-Effects of Flavonoids, Terpenes and Sterols on Angiotensin-Converting Enzyme and Nitric Oxide

Ingrid A-L Persson

Division of Drug Research / Pharmacology

Department of Medical and Health Sciences

Linköping University, Sweden

Linköping 2009

Page 2: Plant-Derived Substances and Cardiovascular Diseases

2

© Ingrid A-L Persson 2009

Photographs: Ginkgo biloba © Sven Persson, others © Ingrid A-L Persson

Published articles have been reprinted with the permission of the copyright

holder.

Printed in Sweden by Linköpings Tryckeri AB, 2009

ISBN 978-91-7393-706-1

ISSN 0345-0082

Page 3: Plant-Derived Substances and Cardiovascular Diseases

3

”Blommor är vilsamma att betrakta. De har

varken känslor eller konflikter”

Sigmund Freud

Page 4: Plant-Derived Substances and Cardiovascular Diseases

4

CONTENTS

ABSTRACT 7

LIST OF PAPERS 8

ABBREVIATIONS 9

INTRODUCTION 12

BACKGROUND 14

ANTIOXIDANTS 14

THE BIOSYNTHESIS OF SECONDARY METABOLITES IN PLANTS 15

Flavonoids 19

Purines 22

Terpenes 22

Sterols 25

HOMEOSTASIS OF THE VASCULAR WALL – REGARDING

ANGIOTENSIN II, NITRIC OXIDE AND REACTIVE OXYGEN SPECIES 26

THE ENDOTHELIUM 27

NITRIC OXIDE 29

THE RENIN-ANGIOTENSIN ALDOSTERONE SYSTEM 30

The Angiotensin-Converting Enzyme 35

AIMS 39

METHODS 41

INFUSIONS AND EXTRACTIONS 41

Tea Infusion, in vitro (Paper I) 41

Tea Infusion, in vivo (Paper V) 41

Coffee Infusion, in vitro 42

Cacao Extraction, in vitro 42

Bilberry 25E Extraction, in vitro (Paper IV) 42

Liquorice Extraction, in vitro 42

Page 5: Plant-Derived Substances and Cardiovascular Diseases

5

Ginkgo biloba Extraction, in vitro (Paper III) 43

Panax ginseng Extraction, in vitro (Paper II) 43

CULTURED ENDOTHELIAL CELLS FROM HUMAN UMBILICAL VEINS (HUVEC) 43

ANGIOTENSIN-CONVERTING ENZYME ACTIVITY IN HUVEC 44

ANGIOTENSIN-CONVERTING ENZYME RADIOENZYMATIC ASSAY 46

NITRITE/NITRATE ASSAY 46

LIQUID CHROMATOGRAPHY-MASS SPECTROMETRY-MASS MASS SPECTROMETRY (LC-MS-MS) 47

TENSION STUDIES 48

IN VIVO STUDY 49

ANGIOTENSIN-CONVERTING ENZYME GENOTYPE 50

CHEMICALS 52

CALCULATIONS 53

RESULTS AND DISCUSSION 54

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF BEVERAGES CONTAINING FLAVONOIDS AND XANTHINES 55

Green Tea, Black Tea and Rooibos Tea Infusions, in vitro (Paper I) 55

Coffee Infusion, in vitro 56

Cacao Extract, in vitro 56

Xanthines, in vitro (Paper I) 57

Green Tea, Black Tea and Rooibos Tea in Healthy Volunteers (Paper V) 57

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF DIETARY PRODUCTS CONTAINING ANTHOCYANINS OR ISOFLAVONOIDS 60

Bilberry Extract 25E, in vitro (Paper IV) 60

Liqourice Extract, in vitro 62

Genistein, in vitro 63

Page 6: Plant-Derived Substances and Cardiovascular Diseases

6

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF HERBAL MEDICINAL PLANT EXTRACTS 64

Panax Ginseng extract, in vitro (Paper II) 64

Ginkgo Biloba extract, in vitro (Paper III) 66

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF THE ANTIOXIDANTS ALPHA-TOCOPHEROL AND BETA-CAROTENE

Alpha-Tocopherol, in vitro 69

Beta-carotene, in vitro 70

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF STEROLS AND BLOOD LIPID-LOWERING DRUGS 72

Sterols, in vitro 72

Blood lipid –Lowering Drugs, in vitro 73

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF PRECURSOR MOLECULES 73

THE MECHANISM OF THE INHIBITORY EFFECT ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF THE FLAVONOIDS 74

EFFECTS ON NITRIC OXIDE 78

Nitric oxide, in vitro (Paper I, II, III) 78

Nitric oxide, in vivo (Paper V) 82

INTERACTIONS BETWEEN DIETARY PRODUCTS AND DRUGS 83

CLINICAL IMPORTANCE 84

SUMMARY 88

TABLE OF RESULTS 90

TACK 102

REFERENCES 105

Page 7: Plant-Derived Substances and Cardiovascular Diseases

7

ABSTRACT

Diet has for many years been known to play a key role in the development of chronic diseases. There are clear associations between consumption of vegetables, fruits and berries, and risk of cardiovascular diseases, the number one cause of death in the world. To maintain homeostasis of the vascular wall the balance between angiotensin II, nitric oxide and reactive oxygen species is of great importance in order to affect the development of cardiovascular diseases. Angiotensin II, a potent vasoconstrictor causing cell growth and nitric oxide, a signalling molecule influencing the vascular system as a vasodilatator, inhibiting cell proliferation and reactive oxygen species, are linked together in the renin-angiotensin aldosteron system. Angiotensin-converting enzyme will as a key enzyme in the renin-angiotensin aldosteron system convert angiotensin I to form angiotensin II and nitric oxide is known to inhibit angiotensin-converting enzyme and act as a scavenger of reactive oxygen species. Plant-derived substances as flavonoids, tocopherols and carotenoids are shown to have beneficial effects on the cardiovascular system due to their antioxidative effects. The aims of this study were to investigate beverages, dietary products, herbal medicinal plants, α- tocopherol, β-carotene, sterols and lipid-lowering drugs on angiotensin-converting enzyme activity and nitric oxide concentrations. This was done to investigate if the sole mechanism of plant-derived substances is their antioxidative properties and to investigate if there is any connection between effect and biosynthesis/structure of plant substances. The tested infusions and extracts containing high amounts of flavonoids, the flavonoids and β-carotene significantly inhibited angiotensin-converting enzyme activity in vitro. The other substances tested did not affect, or in some cases significantly increased, angiotensin-converting enzyme activity. The infusions and extracts containing high amounts of flavonoids, the flavonoids and β-carotene showed an increase on nitric oxide concentrations in vitro. Oral intake of a single dose of Rooibos tea significantly inhibited angiotensin-converting enzyme activity. A significant inhibition of angiotensin-converting enzyme activity was seen with the green tea for the angiotensin-converting enzyme genotypes II and ID. A significant inhibition of angiotensin-converting enzyme activity was also seen with the Rooibos tea for the angiotensin-converting enzyme genotype II.

Conclusion; flavonoids and β-carotene interact with the cardiovascular system in several ways, by reducing reactive oxygen species (as shown in several studies), increasing nitric oxide concentrations (as shown here and by others) and also by inhibiting angiotensin-converting enzyme activity (as shown here). Infusions and extracts as tea containing high amounts of flavonoids function as angiotensin-converting enzyme inhibitors. Angiotensin-converting enzyme contains two zink-dependent catalytic domains and angiotensin-converting enzyme inhibitors are designed to bind to the Zn2+ at the active site. If the inhibitory mechanism of flavonoids on angiotensin-converting enzyme activity is due to their ability to bind to Zn2+ ions then it would be possible for the flavonoids to also inhibit other zinc metallopeptidases, i.e. endothelin-converting enzyme, matrix metallopeptidases, neutral endopeptidase and maybe insulin-degrading enzyme, thereby exerting several additional positive effects on the cardiovascular system.

Page 8: Plant-Derived Substances and Cardiovascular Diseases

8

LIST OF PAPERS

This thesis is based on the following papers. Unpublished results are also

included in Results and Discussion. The roman numerals are used when

referring to the papers in the text.

I. Tea flavanols inhibit angiotensin-converting enzyme activity and increase nitric oxide production in human endothelial cells.

Persson, I.A-L., Josefsson, M., Persson, K. & Andersson, R.G.G. Journal of Pharmacy and Pharmacology 58: 1139-1144, 2006.

II. Effects of Panax ginseng extract (G115) on angiotensin-converting enzyme (ACE) activity and nitric oxide (NO) production.

Persson, I.A-L., Dong, L. & Persson, K.

Journal of Ethnopharmacology 105: 321-325, 2006.

III. Effects of Gingko biloba extract EGb 761 and its terpene-lactones on angiotensin converting enzyme activity and nitric oxide production in human endothelial cells.

Persson, I.A-L., Lindén, E., Andersson, M. & Persson, K.

Asian Journal of Traditional Medicines 3: 42-51, 2008.

IV. The Effect of Vaccinium myrtillus and its Polyphenols on Angiotensin-Converting Enzyme Activity in Human Endothelial Cells.

Persson, I.A-L., Persson, K. & Andersson, R.G.G. Submitted.

V. Effects of Green Tea, Black Tea and Rooibos Tea on Human Angiotensin-Converting Enzyme and Nitric Oxide in Healthy Volunteers.

Persson, I.A-L., Persson, K., Hägg, S. & Andersson, R.G.G. Submitted.

Page 9: Plant-Derived Substances and Cardiovascular Diseases

9

ABBREVIATIONS

ACC Acetyl-CoA carboxylase

ACE Angiotensin-converting enzyme

ADP Adenosine diphosphate

AMPA Aminopeptidase A

AMPN Aminopeptidase N

ANOVA One way analysis of variance

ANP Atrial natriuretic peptides

ANS Anthocyanidin synthase

AP Area postrema

APP Aminopeptidase P

ARPE-19 A retinal pigmented epithelium

AT1 Angiotensin receptor 1

AT2 Angiotensin receptor 2

AT3 Angiotensin receptor 3

AT4 Angiotensin receptor 4

ATP Adenosine triphosphate

BBB Blood-brain barrier

CAD Collision activated dissociation

cGMP Cyclic guanosine 3´, 5´monophosphate

CNS Central nervous system

CPM Carboxypeptidase M

CPN Carboxypeptidase N

CVO Circumventricular organs

D Deletion

DMAPP Dimethyl allyl diphosphate

DMSO Dimethylsulfoxide

Page 10: Plant-Derived Substances and Cardiovascular Diseases

10

ECE Endothelin-converting enzyme

ECGF Endothelial cell growth factor

EDRF Endothelium-derived relaxing factor

eNOS Endothelial nitric oxide synthase

ESI Electrospray ionisation

F3H Flavanone 3β-hydroxylase

FCS Fetal calf serum

FPP Farnesyl pyrophosphate

GGPP Geranylgeranyl pyrophosphate

GPP Geranyl pyrophosphate

HPLC High performance liquid chromatography

HUVEC Culured endothelial cells from human umbilical veins

I Insertion

IDE Insulin-degrading enzyme

iNOS Inducible nitric oxide synthase

IPP Isopentenyl diphosphate

IRAP Insulin-regulated aminopeptidase

LC Liquid chromatography

LDL Low-density lipoprotein

L-NMMA N G-monomethyl-L-arginine

MAP Mitogen-activated protein

MMP Matrix metallopeptidase

MS Mass spectrometry

NADPH Nicotinamide-adenine dinucleotide phosphate

NCEP National Cholesterol Education Program

NEP Neutral endopeptidase

nNOS Neuronal nitric oxide synthase

NO Nitric oxide

Page 11: Plant-Derived Substances and Cardiovascular Diseases

11

NOS Nitric oxide synthase

OVLT Organum vasculosum of the lamina terminalis

PAI-1 Plasminogen activator inhibitor-1

PBS Phosphate-buffered saline

PCR Polymerase chain reaction

PEP Prolyl endopeptidase

RAAS Renin-angiotensin aldosterone system

RHS Reactive halogen species

RNS Reactive nitrogen species

RSS Reactive sulphur species

ROS Reactive oxygen species

SCB Statistics Sweden

SFO Subfonical organs

WHO World Health Organization

WHOSIS World Health Organization Statistical Information System

1400-W N-3-(aminomethyl) benzylacetamidine

Page 12: Plant-Derived Substances and Cardiovascular Diseases

12

INTRODUCTION

Diet has for many years been known to play a key role in the development of

chronic diseases. During the latter half of the 2000 century there has been a

great change in diet habits almost all over the world. Food consumption has

changed from a traditionally plant-based diet containing complex carbohydrates

and dietary fibers present in vegetables, fruits and berries to a diet rich in

saturated fats and simple carbohydrates i.e. a diet consisting mainly of meat and

products with high energy content (WHO, 2003). Changes in diet patterns over

time are due to a complex interaction between many factors such as individual

preferences and beliefs, cultural traditions, geographical, environmental, social

and economic factors. As a result of these interactions, associations between

consumption of fibres (Truswell, 2002), unsaturated fats (Russo, 2008),

vegetables, fruits and berries, and risk of cardiovascular diseases have been

shown (Hertog et al., 1993; Keli et al., 1996; Joshipura et al., 1999; Bazzano et

al., 2002; Rissanen et al., 2003; Bruckdorfer, 2008).

Approximately 60% of total reported deaths in the world and approximately

46% of the global burden of diseases (WHO, 2003) are contributed to chronical

diseases. Almost half of the deaths related to chronical diseases are attributed to

cardiovascular events; i.e cardiovascular diseases are the number one cause of

death. As well as in the world, cardiovascular diseases is the number one cause

of death in Sweden; in 1996, 49% of all deaths were due to cardiovascular

diseases according to the Statistics Sweden (SCB) database.

WHO recommends an intake of 400-500 gram of vegetables (apart from

potatoes), fruits, berries and green leaves per day to reduce the risk of coronary

heart disease, stroke and high blood pressure (WHO, 2008). Only a very small

and negligible minority of the world population consumes this recommended

Page 13: Plant-Derived Substances and Cardiovascular Diseases

13

intake of vegetables, fruits and berries. However, in 2003 only 19 countries in

the WHO European Region had 600 grams of vegetables and fruits available per

capita and day, this to ensure the possible consumption of 400 g intake per day

and Sweden was not one of these 19 countries (WHO, 2008). In Sweden, the

recommended intake of vegetables and fruits is 500g per capita per day

(National Food Administration, Sweden).

Thus, lifestyle, i.e. diet and nutrition, is of great importance to human health

(Joshipura et al., 1999; Bazzano et al., 2002; Truswell, 2002; Rissanen et al.,

2003; Vanharanta et al., 2003; Jansen et al., 2004; Johnsen et al., 2004; Allen et

al., 2008; Chen et al., 2008; López et al., 2008; Patterson et al., 2008).

Vegetables, fruits and berries are well-known for their protection against and

prevention of non-communicable chronical diseases, such as cardiovascular

diseases, obesity, diabetes, cancer and osteoporosis (Steinmetz & Potter, 1991;

Hertog et al., 1995; Ness & Powles, 1997; WHO, 2003). These actions are

attributed to a diversity of effects such as antioxidative (Dragsted, 2003),

antithrombotic and anti-inflammatory properties, activation of endothelial nitric

oxide synthase (eNOS) and inhibition of low-density lipoprotein (LDL)

oxidation (Hwang et al., 2003; Ikizler et al., 2007; Kaliora & Dedoussis, 2007;

Aron et al., 2008; Boots et al., 2008;). Anticarcinogenic, antiatherogenic and

estrogenic effects have also been shown (Morton et al., 2000; Birt et al., 2001;

Nijveldt et al., 2001) as well as prevention of diabetes type 2 (Liese et al.,

2008). These proposed effects of vegetables, fruits and berries are

predominantly attributed to antioxidative mechanisms.

Page 14: Plant-Derived Substances and Cardiovascular Diseases

14

BACKGROUND

ANTIOXIDANTS

Antioxidants are chemical substances that delay or prevent oxidation and

oxidazible substrates include almost every molecule found in vivo. All living

organisms, except some anaerobic and aerotolerant species, require oxygen in

order to produce energy/ATP by electron transport chains that donate electrons

to dioxygen (O2) and reduce it to water as in the mitochondria of eukaryotic

cells. Under certain circumstances, e.g. certain diets, smoking, ultraviolet-light,

cold and heating, reactive oxygen species (ROS) are generated as side products

of this energy process. Apart from ROS, also called oxygen free radicals (a free

radical means any species capable of independent existence and containing one

or more unpaired electrons), ROS also include reactive nitrogen species (RNS),

reactive halogen species (RHS) and reactive sulphur species (RSS) (Halliwell &

Gutteridge, 2007). Thus, all ROS are not oxygen species, but all reactive species

are usually called ROS (Halliwell & Gutteridge, 2007).

Most of the damaging effects of oxygen are due to oxygen radicals (Muller et

al., 2007). As the oxygen content of the atmosphere increased many primitive

species i.e. anaerobs died out while other organisms began to evolve antioxidant

defence systems for protection against oxygen toxicity in order to survive.

Aerobic organisms survive in the presence of free radicals solely because they

have evolved antioxidant defences. There are two major mechanisms

contributed to antioxidative effects, free radical scavenging and metal chelating.

In healthy aerobs, the production of ROS is approximately in balance with the

antioxidant defence systems. If this balance fails, and the production of ROS

will outweigh the antioxidants defence, then oxidative damage will occur

(Halliwell & Whiteman, 2004). Thus, oxidative damage is the biomolecular

Page 15: Plant-Derived Substances and Cardiovascular Diseases

15

damage caused by ROS upon living organisms, as a result of diminished

antioxidants or increased production of ROS. This oxidative damage may induce

or become a result of endothelial cell damage, inflammation, atherosclerosis,

proliferation of vascular smooth muscle cells and cardiac remodelling associated

with hypertension and cardiovascular diseases (Grossman, 2008).

Antioxidants can either be syntesised in vivo e.g. glutathione, thiols, bilirubin,

transferin, lactoferin, erythrocytes, albumins or be administered via diet, e.g.

plant-derived substances like flavonoids (see below, section “Flavonoids”),

Vitamin E (see below, section “Terpenes”), carotenoids (see below, section

“Terpenes”) and Vitamin C. Plant-derived antioxidants as flavonoids,

tocopherols and carotenoids (Voutilainen et al., 2006; Kaliora et al., 2007;

Stocker, 2007) are shown to have beneficial effects on the cardiovascular system

(Devaraj et al., 2007; Svarcova et al., 2007; Aron et al., 2008; Choudhary et al.,

2008; Milman et al., 2008), and cancer prevention (Nandakumar et al., 2008).

Flavonoids are also shown to have effects on cognition and behaviour (DeKosky

et al., 2006; Williams et al., 2008). These beneficial properties of plant-derived

substances are proposed to be due to their antioxidative effects. From an

evolutionary point of view, oxygen and thereby antioxidative properties

appeared in significant amounts in the Earth atmosphere almost simultaneously

as the photosynthesis and biosynthesis of the secondary metabolites evolved in

cyanobacteria (Liang et al., 2006).

THE BIOSYNTHESIS OF SECONDARY METABOLITES IN PLANTS

The photosynthesis (Ingenhousz, 1779) can be divided into two major processes;

the energy-transduction reaction and the carbon-fixation reaction. In the energy-

transduction reaction, light energy is used to form adenosine triphosphate (ATP)

from adenosine diphosphate (ADP) and to reduce the oxidized electron-carrier

molecules nicotinamide-adenine dinucleotide phosphate (NADP+) to NADPH.

Page 16: Plant-Derived Substances and Cardiovascular Diseases

16

The received ATP and NADPH are used by the cells to provide energy for the

further biosynthetic pathways. The energy-transduction reaction is followed by

the second process, the carbon-fixation reaction. In this process the energy of

ATP is used to link carbondioxide, CO2 to an organic molecule and NADPH is

then used to reduce the newly fixed carbon atoms into a simple sugar (the Calvin

cycle after Melvin Calvin, NobelPrize winner in 1961). In the photosynthesis,

carbohydrates (sugars), carboxylic acids, α-amino acids, fats, proteins and

nucleic acids are produced; products involved in and essential for life processes

and suitable for storage as starch and the carbon skeleton from which all other

organic molecules can be built in further processes. Products derived from the

photosynthesis, so called primary metabolites, are also the precursors and

starting materials for the biosynthesis of the so called secondary metabolites. In

contrast to the primary metabolites, the secondary metabolites are not present in

all plants but are found in certain plants, species or families. Furthermore,

primary metabolites are essential to life, while the secondary metabolites are

not, but the secondary metabolites do contribute to the survival of the species

involved in e.g. the defense system and reproduction. Flavonoids, terpenes and

phytosterols are example of plant-derived secondary metabolites synthesised

from monosaccharides (glyceraldehyde-3-phosphate). All secondary metabolites

are linked to primary metabolites and most metabolites originate from a very

limited number of precursor molecules. There are three main biosynthesis

pathways of secondary metabolites: the shikimic, the polyketide and the

mevalonic pathways. As shown in figure 1, plant-sterols are derived from the

mevalonic pathway. The terpenes are an example of secondary metabolites

derived from two pathways, dependent on the type of terpene, from

glyceraldehyd-3-phosphate and pyruvic acid or from the mevalonic pathway

(figure 1). Several groups of metabolites have mixed origin, e.g the flavonoids

where one part of the flavonoid structure derives from the shikimic pathway and

the other part of the structure derives from the mevalonic pathway (figure 1).

Page 17: Plant-Derived Substances and Cardiovascular Diseases

Fig

ure

1. T

he

bios

ynth

esis

of f

lavo

noi

ds, t

erpe

nes

, st

erol

s an

d pu

rine

text

.

17

ds, t

erpe

nes

, st

erol

s an

d pu

rine

s/xa

nthin

es. S

econ

dary

met

abo

lites

test

ed in

this

stu

dy

are

prin

ted

in b

old

nes.

Sec

onda

ry m

etab

olit

es te

sted

in th

is s

tud

y a

re p

rinte

d in

bol

d

Page 18: Plant-Derived Substances and Cardiovascular Diseases

Figu

re 2. Basic chem

ical structures of flavonoids, stilbenes, aurones and ap

iforol.

18

hemical structures of flavonoids, stilbenes, aurone

s and apiforol.

Page 19: Plant-Derived Substances and Cardiovascular Diseases

19

Flavonoids

All dietary products originating from plants potentially contain flavonoids (Latin

flavus, yellow) and over 4000 compounds have been identified and the list is

still growing (Harborne & Williams, 2000). Flavonoids are water-soluble

pigments present in the cytosol and/or stored in the vacuole of the plant cell and

the flavonoids represent the largest group of phenolic compounds in plants.

Flavonoids are of mixed origin, biosynthesised by the shikimic acid pathway

and the mevalonic acid pathway (figure 1). The biosynthesis pathway of the

flavonoids is part of a larger phenylpropanoid pathway producing a range of

secondary metabolites e.g. phenolic acids, flavonoids, stilbenes, aurones and

apiforols (figure 1). The basic chemical structure of flavonoids is based on two

six-carbon rings linked by a three-carbon unit, the chalcone structure (figure 2).

The main classes/groups of flavonoids according to differences in the C-ring,

OH-substituents and double bondings are chalcones (the basic structure of the

flavonoids and unstable isomers of flavanones), flavanones (e.g.naringenin),

flavones (e.g. luteolin), flavonols (e.g. quercetin), flavanols (catechines),

isoflavones (e.g. genistein) and anthocyanidins (e.g. cyanidin, delphinidin and

malvidin) (figure 2). The difference between the various flavonoids in the

different groups is the number and position of substitution by hydroxylation,

hydrogenation, methylation, glycosylation, malonylation and sulphation

(Andersen & Markham, 2006). The most common forms of flavonoids found in

plants are the glycoside derivatives, except for the catechins which occur

without sugar molecules (Andersen & Markham, 2006). Flavonoid molecules

without sugar molecules are referred to as aglycones.

Flavonoids serve as communicators between the plant and the environment, and

are important for e.g. reproduction, resorption of mineral nutrients and as

antioxidants (Harborne & Williams, 2000); which make the functions of the

flavonoids critical for the survival of the plant. Concerning the antioxidative

Page 20: Plant-Derived Substances and Cardiovascular Diseases

20

effects of the flavonoids, yellow flavonoids and flavonoids with colours

invisible to the human eye (flavones, flavonols and isoflavonoids) seem to be

primarily involved in the protection against ultraviolet radiation (Stapleton,

1992) and plants often respond to ultraviolet light by activating the flavonoid

biosynthesis (Schmelzer, 1988). In humans, flavonoids are mostly associated

with the ability to donate hydrogen-ions and scavenge of reactive oxygen and

reactive nitrogen species, i.e. antioxidative effects (Sun et al., 2002; Chang et

al., 2007; Jiménez et al., 2007; Tomer et al., 2007).

Some of the flavonoids are more or less specific to particular dietary products

e.g. genistein found in soya-beans (Glycine max (L.) Merr.). Other flavonoids

are found in almost all plant-based dietary products e.g. the flavonol quercetin.

But, dietary products often contain mixtures of different flavonoids. The most

numerous reports on flavonoids and their antioxidative capacity concern the

flavonol quercetin (Rahman, 2006; Ratman et al., 2006) as well as studies on

ingestion of dietary products, mostly everyday beverages, containing large

amount of catechins, found in green tea and black tea, unfermented and

fermented leaves of Camellia sinensis L. (Theaceae) (Persson et al., 2006 (Paper

I); Basu & Lucas, 2007; Lambert et al., 2007). Rooibos or red tea, fermented

leaves and/or bark of Asparalathus linearis Dahlg. (Leguminosae) does not

contain catechins but dihydrochalcones, flavones and flavonols (Bramati et al.,

2002; Bramati et al., 2003). Antioxidative effects have also been reported

concerning procyanidins (polymer chain of catechins), found in beans of the

cacao tree, Theobroma cacao L. (Sterculiaceae) (Keen et al., 2005; Aron et al.,

2008).

In plants, anthocyanins (Greek, antos, flower and kyanos, blue), represent

pink/red/blue/violet to dark blue colours, and it is the anthocyanins that seems to

have the greatest antioxidative effect of all flavonoids and there is a strong

association between anthocyanin biosynthesis and plant stress (Andersen &

Page 21: Plant-Derived Substances and Cardiovascular Diseases

21

Markham, 2006). The anthocyanins consist of an anthocyanidin (an aglycon),

sugar(s), mostly glucose and galactose and in many cases, acyl group(s).

Anthocyanins are the only flavonoids able to form flavylium cations (figure 2)

and due to this ability the anthocyanins are very reactive towards ROS because

of their electron deficiency. The biosynthesis of the anthocyanins is shown in

figure 1. Anthocyanins may function as general antioxidants and are associated

with a broad range of stressors (Leshem et al., 1996). Anthocyanins are to be

found in dark-coloured foods like bilberries Vaccinium myrtillus L. (Ericaceae).

The anthocyanins are considered responsible for the main pharmacological

effects of Vaccinium myrtillus due to the antioxidative and free radical

scavenging properties (Kähkönen & Heinonen, 2003).

Isoflavonoids e.g. genistein are plant-derived non-steroidal secondary

metabolites also called phytooestrogens due to their structural relationship with

oestrogen and other sex hormones. Isoflavonoids exert both oestrogenic and

antioestrogenic activity by competing for receptor binding with oestrogen

(McCarty, 2006). Apart from acting on oestrogen receptors, genistein is shown

to induce apoptosis of cancer cells, exert antioxidative effect, inhibit cell

proliferation, modulate cell cycling, inhibit angiogenesis and suppress

lymphocyte functions (Polkowski & Mazurek, 2000). The isoflavonoids

originate from flavanons, and consequently the isoflavone genistein is

biosynthesised from the flavanone naringenin (figure 1 and 2). The structural

difference between isoflavonoids and other flavonoids is the linking of the B-

ring to the C-3 rather than to the C-2 position of the C-ring (figure 2).

Glycyrrhiza glabra L. (Fabaceae) contain a numerous of flavonoids, more than

300 species-specific phenolic compounds have been isolated from liquorice.

Glycyrrhiza species contain flavonoids, isoflavonoids, chalcones and bibenzyls

and about 70 of these are present in the root of the plant (Li et al., 2000). The

Page 22: Plant-Derived Substances and Cardiovascular Diseases

22

major flavonoid in Glycyrrhizin is the isoflavone glabridin (Belinky et al.,

1998).

Purines

Alkaloids (alkali-like) are a group of plant-derived secondary metabolites.

Alkaloids are basic compounds, containing one or more nitrogen atoms and they

are known to have marked physiological effects on humans e.g. caffeine,

morphine and nicotine. Alkaloids are based on amino acids and classified

according to chemical structure and one of the classes is purines. Purines are

heterocyclic aromatic organic compounds consisting of a pyrimidine ring fused

to an imidazole ring. In nature, the purines are widely distributed and their role

is not clear. The name purine (purum uricum) was given by Emil Fischer in

1884 who was the first to synthesise purines in 1899 (Nobel Prize winner in

1902 for his work on sugar and purine syntheses).

The purines are biosynthesised from ribose via pyruvic acid, amino acid and the

purine base xanthine. Purines are derivatives of xanthine, thereby also named

xanthines i.e. caffeine, theobromine and theophylline (figure 1) and present in

beverages as green tea, black tea, coffee and cacao. Pharmacological effects of

purines/ xanthines include bronchodilation, increased cardiac output, alertness

and dependence.

Terpenes

Terpenes (from turpentine, the fluid obtained by distillation of resin

(hydrocarbon secretion), originally from the Greek word terebinthine, Pistacia

terebinthus L., turpentine tree) are secondary metabolites built up from isoprene

units, C5H8 (figure 3), i.e. oligomers of isoprene units (C5)n, according to the

isoprene rule formed by Ruzicka and Wallach (Ruzicka, 1953).

Page 23: Plant-Derived Substances and Cardiovascular Diseases

23

Figure 3. Isoprene unit C5H8

Today, about 30000 terpenes have been identified, and most of these compounds

originate from plants. In nature, terpenes mainly exist as hydrocarbons, alcohols

and their glycosides, ethers, aldehydes, ketones, carboxylic acids and esters. The

differentiation of the chemical structure between the terpenes, hemi-(C5), mono-

(C5)2, sesqui-(C5)3, di-(C5)4, sester-(C5)5, tri-(C5)6, tetra-(C5)8 and polyterpenes

(C5)n , is dependent on the number of isoprene units. Isopentenyl diphosphate

(IPP) is the common precursor for all terpenes. Two biosynthetic pathways lead

to the formation of IPP, the formation of IPP in the plastid and the formation of

IPP in the cytoplasm. IPP and its isomer dimethyl allyl diphosphate (DMAPP)

are formed from glyceraldehyd-3-phosphate and pyruvic acid in the chloroplast

for further formation of mono-, di- and tetraterpenes (Lichtenthaler, 2007)

(figure 1). The terpenes derived from IPP formed in the cytoplasm via the

mevalonic pathway are sesqui- and triterpenes (Lichtenthaler, 2007) (figure 1).

Mevalonic acid consists of isoprene units, and the mevalonic acid pathway in

plants and mammals is similar from acetyl CoA to cholesterol except for the

synthesis of terpenes in plants. The biosynthesis of the terpenes is shown in

figure 1.

By combining isoprene units, the pathway produces geranyl pyrophosphate

(GPP) (C5)2, farnesyl pyrophosphate (FPP) (C5)3 and geranylgeranyl

pyrophosphate (GGPP) (C5)4, the precursor of the diterpenes (C5)4.

Tocopherols (Greek tocos, childbirth, phero, to bring forth), a group of

substances consisting of α-, β-, γ- and δ-tocopherols, and α-, β-, γ- and δ-

tocotrienols are cyclic diterpenes, prenylchromanols with a hydroxyl group that

can donate a hydrogen atom to reduce free radicals and a hydrophobic side

Page 24: Plant-Derived Substances and Cardiovascular Diseases

24

chain. In plants, tocopherols and tocotrienols serve as antioxidants by protecting

carotenoids (see below) and polyunsaturated fatty acids in the biomembranes

(Matringe et al., 2008). The most active of the tocopherols, α-tocopherol, (2-

prenyl-3,4-dihydro-2H-1-benzopyran-6-ol) is also known as Vitamin E. In

humans, α-tocopherol, as an antioxidant has been suggested to reduce

cardiovascular diseases (Meydani, 2004).

Ginkgo biloba L. (Ginkgoaceae) is considered to have cardioprotective effects

and these effects of Ginkgo biloba are often related to its unique terpene

compounds, the hexacyclic diterpenelactones ginkgolides A, B, C, J and the

tetracyclic sesquiterpene-lactone derivative bilobalide, and flavonol glycosides.

The biosynthesis pathway of the diterpenes and the sequiterpenes is shown in

figure 1.

The precursor of the triterpenes (C5)6 is squalene C30H50. The chemical structure

of the triterpenes is similar to the structure of the steroids (C27, C24, C21, C19,

C18). Triterpenes are also referred to as steroidal phytooestrogens. Effects of

Panax ginseng L. (Araliaceae) on the cardiovascular system are often attributed

to the tetracyclic triterpene saponins, the ginsenosides (Chen, 1996; He at al.,

2007). The biosynthesis pathway of the triterpenes is shown in figure 1.

Tetraterpenes, i.e carotenoids (the first to be isolated was from carrot, Daucus

carota L. in 1831), are based on a (C5)8 isoprene structure. Approximately 600

carotenoids have been identified, but only about 40 are present in ordinary

human diet. The carotenoids are present in all higher plants, in leaves, shoots

and roots. The chloroplast is the site of the photosynthesis containing

chlorophyll (a phytol, an acyclic diterpene alcohol containing a (C5)4 isoprene

side-chain and precursor of Vitamin E) and carotenoid pigments. The

carotenoids are prenol (3-methyl-2-buten-1-ol, an isoprene alcohol) lipids

representing yellow-orange and red colours in relation to the conjugated double

bonds in the carotenoid skeleton. Carotenoids may be acyclic (e.g.lycopene) or

Page 25: Plant-Derived Substances and Cardiovascular Diseases

25

having one or both ends modified into rings (e.g. β-carotene). Carotenoids

containing at least one oxygen atom are classified as xanthophylls (e.g.lutein)

and hydrocarbon carotenoids are classified as carotenes. In plants, the

carotenoids contributes to the photosynthetic processes, they have principal

functions as antioxidants by prevention of photo-oxidative damages to the

chlorophyll molecules and serve as communicators between the plant and the

environment. In humans, the effect of β-carotene is related to its antioxidative

activity, preventing cardiovascular diseases (Bjelakovic et al., 2008), cancer

(Bjelakovic et al., 2008)) and protection against light-induced skin damage

(Stahl & Sies, 2002). Beta-carotene is the carotenoid with the highest pro-

vitamin A activity and the most common of the carotenes. The biosynthesis of

the tetraterpenes is shown in figure 1.

Sterols

The terpenes, except the monoterpenes (see above, section “Terpenes”) and the

sterol lipids share a common biosynthetic pathway via IPP and FPP (figure 1),

but they have differences in structure and function. The sterol lipids can be

subdivided according to their biological functions; cholesterol and derivatives

e.g. plantsterols, steroids such as sex hormones and mineralcorticoids,

secosteroids i.e. Vitamin D2 and D3. Plant-derived sterols are also named

phytosterols, e.g. stigmasterol, sitosterol and campesterol. Stigmasterol is an

unsaturated phytosterol, the most common sterol present in plants. Animal-

derived sterols are named zoosterols e.g. cholesterol, the most common sterol of

animal origin also present in plants but not as common as in animals. The

precursor of cholesterol and animal steroids in general is lanosterol, while plant

sterols are formed via squalene, protosterol carbonium I and cycloartenol. The

sterol lipids and the prenol lipids share a common biosynthetic pathway via IPP,

GPP and FPP and are building blocks of cholesterol and thus of all steroids. The

Page 26: Plant-Derived Substances and Cardiovascular Diseases

26

biosynthesis of phytosterols and cholesterol is shown in figure 1. In 1976,

stigmasterol was used as starting material for approximately 28% of the world

production of steroids. Studies show that phytosterols have effect on the

cardiovascular system by lowering cholesterol levels (Klingberg et al., 2008;

Poli et al., 2008).

HOMEOSTASIS OF THE VASCULAR WALL

– REGARDING ANGIOTENSIN II, NITRIC OXIDE AND REACTIVE

OXYGEN SPECIES

To maintain homeostasis of the vascular wall, a balance between the

endogenous transmitters angiotensin II (see below, section “The renin-

angiotensin aldosterone system”), nitric oxide; (see below, section “Nitric

Oxide”) and ROS; (see above, section “Antioxidants”) is of great value.

Angiotensin II, nitric oxide and ROS are important participators in the

pathogenetic mechanisms of cardiovascular diseases. It has been shown that

hypertension caused by chronically elevated angiotensin II is mediated in part

by superoxide ions (O2-) and hypertension is a major risk factor for coronary

artery disease, congestive heart failure, cerebrovascular disease, peripheral

vascular disease and renal failure. This suggests that cardiovascular diseases

caused by chronically elevated angiotensin II levels are found to be mediated by

vasoconstriction and furthermore, partially mediated by ROS (Zhang et al.,

2007). Decreased vascular nitric oxide seems to promote angiotensin II

dependent cardiovascular diseases mediated by ROS (deGasparo, 2002).

Angiotensin II acting through angiotensin-1 receptors, AT1, mediates

vasoconstriction and stimulates membrane bound nicotineamide adenine

dinucleotide phosphate (NADPH) oxidase causing accumulation of ROS.

Angiotensin II acting on angiotensin-2 receptors, AT2, results in increased levels

Page 27: Plant-Derived Substances and Cardiovascular Diseases

27

of nitric oxide (and bradykinin and prostacyclin). Nitric oxide scavenges ROS

thereby consuming nitric oxide and blocking the beneficial properties of nitric

oxide (Doughan et al., 2008). Accumulation of ROS stimulates mitogen-

activated protein (MAP) kinases which promote cell growth and cell

proliferation (Zhang et al., 2007). The angiotensin receptors AT1 and AT2 are

with their physiologically antagonistic effects maintaining the balance between

nitric oxide and ROS. It is proposed that stimulation of AT1 receptors by

increased circulating or tissue levels of angiotensin II will stimulate cell growth,

cell proliferation, affect homeostasis of the vascular wall and give rise to

inflammation and cardiovascular diseases (deGasparo, 2002). Angiotensin-

converting enzyme (see below, section “The angiotensin-converting enzyme”),

is a key enzyme involved in the formation of the physiological antagonists

angiotensin II and nitric oxide.

THE ENDOTHELIUM

The entire vascular system is covered by endothelial cells, the endothelium, a

single layer of cells between the blood and the vascular smooth muscle cells.

The endothelium responds to neurotransmitters, e.g. acetylcholine, hormones

(e.g. angiotensin II), local mediators (e.g. bradykinin), platelet-derived

substances (e.g. thrombin), and mechanical force (shear stress) (Vanhoutte &

Mombouli, 1996; Lüscher & Barton, 1997). In concequence, endothelium-

derived substances are of a large variety and of importance in the regulation of

vascular tone, smooth muscle cell proliferation, vessel wall inflammation and

platelet function. The endothelium represents a complex interrelationsship

between physiological agonists and antagonists invaluable for homeostasis of

the vascular wall e.g. vasoconstriction versus vasodilatation, fibrinolysis versus

antifibrinolysis, thrombosis versus antithrombosis, growth promotion versus

Page 28: Plant-Derived Substances and Cardiovascular Diseases

growth inhibition and oxidation versus antioxidation (Lüscher & Barton, 1997;

de Gasparo, 2002) (figure 4).

Figure 4. Endothelial cell with endothelium

The communication between endothelial cells and smooth muscle cells is for

instance mediated by angiotensin

28

growth inhibition and oxidation versus antioxidation (Lüscher & Barton, 1997;

de Gasparo, 2002) (figure 4).

Figure 4. Endothelial cell with endothelium-derived substances.

The communication between endothelial cells and smooth muscle cells is for

instance mediated by angiotensin-converting enzyme and nitric oxide (Figure 4).

growth inhibition and oxidation versus antioxidation (Lüscher & Barton, 1997;

The communication between endothelial cells and smooth muscle cells is for

converting enzyme and nitric oxide (Figure 4).

Page 29: Plant-Derived Substances and Cardiovascular Diseases

29

NITRIC OXIDE

Nitric oxide (NO) was discovered in 1772 by Joseph Priestly as a toxic gas that

is produced by the combustion of air. The first discovered endothelium-derived

relaxing factor (EDRF) (Furchgott & Zawadski, 1980) produced and released by

the endothelium (see above, section “The endothelium”) was later shown to be

identical with NO (Ignarro et al., 1987; Palmer et al., 1987). NO function as a

signalling molecule (a hormone and a neurotransmitter) present throughout the

body. NO is highly reactive and diffuses freely across the membranes, with a

half life of a few seconds.

In humans, NO is biosynthesised from L-arginine (Arg), an α-amino acid

enzymatically transformed by nitric oxide synthase (NOS) forming NO and

citrulline (another α-amino acid, named after Citrullus lanatae, water melon,

from which citrulline was first isolated, and it is proposed that in plants citrulline

may serve as a nitrogen reserve) (Palmer et al., 1988; Sakuma et al., 1988).

Three isoforms of NOS exist: neuronal NOS (nNOS) found in a variety of cells

including endothelial cells, macrophages and neurons (Bredt & Snyder, 1990;

Tsutsui, 2004), inducible NOS (iNOS) found in macrophages and smooth

muscle cells (Hevel et al., 1991) and endothelial NOS (eNOS) found in

endothelial cells (Pollock et al., 1991).

The first known mechanism to produce NO in plants is by the enzyme nitrate

reductase reducing nitrite to NO, but further investigation has shown an

arginine-dependent NO synthesis in plants. This plant-derived NOS behaves like

the human eNOS but is also involved in defence responses as the human iNOS

(Crawford, 2006).

In humans, NO influences the vascular system as a vasodilator by relaxation of

smooth muscle cells (Furchgott & Zawadski, 1980; Rapoport & Murad, 1983)),

inhibition of smooth muscle cell proliferation (Garg & Hassid, 1989), reducing

Page 30: Plant-Derived Substances and Cardiovascular Diseases

30

platelet aggregation (Radomski et al., 1987), and platelet and monocyte

adhesion to the endothelium (Roberts et al., 2008). NO activates guanylyl

cyclase producing cyclic guanosine 3´, 5´ monophosphate (cGMP) (Rapoport &

Murad, 1983) which in turn activates proteinkinase G resulting in vasodilatation

and inhibited platelet aggregation. Furthermore, NO inhibits LDL oxidation

(Thomas et al., 2008) and expression of adhesion molecules and endothelin. NO

is also known to inhibit angiotensin-converting enzyme (ACE) (Ackermann et

al., 1998; Persson et al., 2005) (figure 5). NO also acts as a scavenger of ROS

(Doughan et al., 2008). NO together with the noradrenergic nervous system and

endothelin is tonically active in resistance vessels under basal conditions. A

decrease in NO e.g. by scavenging of ROS, thereby increases the risk of

developing atherosclerosis (Puddu et al., 2005) and renin-angiotensin

aldosterone (see below, section “The renin-angiotensin aldosterone system)

dependent diseases as coronary artery disease, diabetes, hypercholesterolemia

hypertension, migraine, peripheral vascular disease, vascular restenosis, stroke

and thrombosis.

THE RENIN-ANGIOTENSIN ALDOSTERONE SYSTEM

The renin-angiotensin aldosterone system (RAAS) (figure 5) is together with the

autonomic nervous system (Lohmeier, 2001), the most important mechanisms in

the body concerning regulation of blood pressure, fluid and electrolyte balance

(Reid, 1985; Ferrario, 1990). RAAS is a kidney-derived mechanism (Tigerstedt

& Bergman, 1898). Prorenin (Hobart et al., 1984), the inactive form of renin, is

released constituitively from the kidney and is found circulating in human

plasma in excess to renin. Prorenin can be activated proteolytically (in the

kidney) and nonproteolytically (in the plasma) (Danser & Deinum, 2005).

Decreased renal blood pressure, low salt content, sympathetic nerve stimulation,

β-adrenoreceptor agonists, circulating catecholamines and prostacyclin stimulate

Page 31: Plant-Derived Substances and Cardiovascular Diseases

31

the juxtaglomerular cells in the kidney to release the proteolytic enzyme renin

(Reid, 1985). Renin release is thus controlled by the macula densa cells,

vascular endothelial cells and smooth muscle cells (Davis, 1973). Renin is a

monospecific enzyme persisting in the circulation for 10 minutes to 1 hour.

During this time, renin continuously cleaves the α-2-globulin angiotensinogen

(Asp-Arg-Val-Tyr-Ile-His-Pro-Phe-His-Leu-Leu-Val-Tyr-Ser) at the N-terminal

end of the protein, to form the decaamino acid peptide angiotensin I (Asp-Arg-

Val-Tyr-Ile-His-Pro-Phe-His-Leu) (Oparil & Haber, 1974; Ferrario, 1990).

Angiotensinogen is produced constantly and released to the circulation mainly

by the liver and to some extent also by the kidneys (Kobori et al., 2002).

Angiotensin I is a very weak vasoconstrictor with virtually no activity of its

own. Angiotensin I is rapidly converted by angiotensin-converting enzyme

(ACE) (see below, section “The angiotensin-converting enzyme”) by the

removal of two amino acids to form the octaamino acid peptide angiotensin II

(Asp-Arg-Val-Tyr-Ile-His-Pro-Phe) (Skeggs et al., 1956a). Angiotensin II

causes feedback inhibition of renin release. Other enzymes generating

angiotensin II independently of ACE are tonin, catepsins, carboxypeptidase,

chymotrypsin and chymase (Peach, 1977; Roks et al., 1997). Angiotensin II is

formed both in the circulation and locally/tissue. The circulating form of

angiotensin II regulates systemic blood flow and pressure. The local/tissue

formation of angiotensin II ensures local control of blood flow independently of

blood-borne angiotensin II (Brunner et al., 1972) e.g. in the brain (Ganten et al.,

1971) and in the eye (Danser et al., 1994). An intracellular form of angiotensin

II has recently been discovered (Kumar et al., 2007) and this makes RAAS not

only an endocrine, but also a paracrine and intracrine system. Angiotensin II

functions as a potent vasoconstrictor and also causes cell growth (hypertrophy of

smooth muscle cells) and impairs learning and memory functions. Angiotensin

II regulates blood volume by release of aldosterone. Aldosterone is a

mineralcorticoid/steroid hormone produced by the adrenal cortex. A decrease in

Page 32: Plant-Derived Substances and Cardiovascular Diseases

32

sodium ion (Na+) concentration in the blood is sensed by the macula densa cells

in the kidney stimulating release of renin. Renin stimulates the formation of

angiotensin I and subsequently angiotensin II (see above) leading to the

stimulation of synthesis and release of aldosterone (Laragh et al., 1960; Sancho

et al., 1976) by the adrenal cortex (figure 5). Aldosterone increases the

reabsorption of Na+ and water, and decreases the reabsoption of potassium ions

(K+) from the renal collection duct (Brunner & Gavras, 1980). Apart from this

effect, aldosterone is involved in hypertension and cardiovascular diseases by

contributing to vascular inflammation, oxidative stress, fibrosis and vascular

injury (Brown, 2008). Concerning aldosterone and behaviour, amygdala, septum

pellucidum and hippocampus are the regions in the brain with the highest uptake

of aldosterone (Monder & White, 1993).

As the main effector of the RAAS, angiotensin II is involved in the development

of cardiovascular diseases. Angiotensin II binds to the angiotensin receptors

(Chiu et al., 1989) AT1, AT2 (Bumpus et al., 1991), AT3 and AT4 (Gulati, 1996;

Unger et al., 1996) which thereby are mediators of the actions of angiotensin II.

AT1 and AT2 receptors are specific membrane-bound G-protein coupled

receptors (Unger et al., 1996). AT1 receptors are expressed in blood vessels,

heart, kidneys, adrenal glands, liver, brain, lungs (Campbell, 1987; Chai et al.,

1993), in the endometrium (Ahmed et al., 1995) and in adipose tissue (Engeli et

al., 1999). Activation of AT1 receptors is associated with endothelial

dysfunction (Nishimura, 2000), vasoconstriction (Nishimura, 2000), cell

proliferation, cell growth in the heart and arteries (Benson et al., 2008), platelet

aggregation (Fogari & Zoppi, 2006), inhibition of nitric oxide synthase (Rush &

Aultman, 2008), increased aldosterone release from the adrenal glands and

increase in reactive oxygen species (ROS) (see above, section “Antioxidants”)

O2- by NADHoxidas (Reed et al., 2008). Activation of AT1 receptors is also

involved in hypertension, heart failure, salt and water retention (Weiss et al.,

Page 33: Plant-Derived Substances and Cardiovascular Diseases

33

2001), vasopressin release, thirst and cognitive and behavioural processes such

as depression, anxiety, decrease in learning and memory (Gard, 2002;

Birkenhäger & Staessen, 2006; McGuinness, 2006) and dementia (McGuiness,

2006). Several polymorphisms in the human AT1 receptor gene have been

discovered of which one (A1166C) is more frequent in hypertensive humans

(Bonnardeaux et al., 1994). It has been shown that AT2 receptors are expressed

in fetal life and in brain (Wright & Harding, 1997) and are supposed to be

involved in growth, differentiation and exploratory behavior (DeGasparo et al.,

2000). Effects of AT2 receptors on cardiovascular diseases e.g. hypertension,

cell proliferation and cell growth, seem to be relatively minor in contrast to the

effects of AT1. AT1 and AT2 receptors may act as physiological antagonists, i.e.

AT2 decrease while AT1 receptors increase blood pressure and cell proliferation

(Ichiki et al., 1995; Nakajima et al., 1995; Stoll et al., 1995). Stimulation of AT1

receptors generates ROS while AT2 stimulation generates the vasodilators

bradykinin and NO (see above, section “Nitric Oxide”). Plasma half life of

angiotensin II in the circulation is less than 2 minutes, then it is inactivated by

blood and tissue enzymes (Reid, 1985), i.e. aminopeptidase A and N, forming

peptide fragments, i.e. angiotensin III (angiotensin 2-8, Arg-Val-Tyr-Ile-His-

Pro-Phe), angiotensin IV (angiotensin 3-8, Val-Tyr-Ile-His-Pro-Phe) and

angiotensin 1-7 (Asp-Arg-Val-Tyr-Ile-His-Pro) (Peach, 1977; Schiavone et al.,

1990; Ferrario et al., 1991; Wright et al., 1995; Ferrario & Iyer, 1998; Ardaillou,

1999;). Effects of angiotensin III are mediated by AT1 and AT2 receptors; it

stimulates secretion of aldosterone and is involved in thirst (Fitzsimons, 1998).

Furthermore, angiotensin III acts as a major effector of RAAS in the brain (Zini

et al., 1996) by regulation of blood-pressure and vasopressin release (Reaux et

al., 2001; Bodineau et al., 2008). Apart from AT1 and AT2 receptors, AT3 and

AT4 receptors have been found (Gulati, 1996; Unger et al., 1996). The AT3

receptor has low affinity for angiotensin III and was initially described in mouse

neuroblastoma cells (Chaki & Inagami, 1992). The effects of the receptor AT3

Page 34: Plant-Derived Substances and Cardiovascular Diseases

34

are still unknown (Stanton, 2003). Angiotensin IV affects the endothelium to

release plasminogen activator inhibitor-1 (PAI-1) and this effect is mediated by

the AT4 receptor (DeGasparo et al., 2000; Stanton, 2003) present in human

prostate (DeGasparo et al., 2000) and in the brain e.g. in the amygdala,

hippocampus and thalamus (Wright et al., 2008) affecting learning and memory

(Bodineau et al., 2008). It has also been suggested that brain RAAS is involved

in stress responses (Ruiz-Ortega et al., 2001) and depression (Gard, 2004).

Even if angiotensin II is unable to penetrate the blood-brain barrier easily there

is a strong connection between brain structures outside and inside of the blood-

brain barrier. Furthermore, angiotensin II-sensitive structures inside the blood-

brain barrier may normally be stimulated by angiotensin II generated locally as

components of brain RAAS (Ganten et al., 1971). Many of these angiotensin II-

sensitive neurons in the central nervous system (Bickerton & Buckley, 1961) are

present in highly vascularized nervous structures, the circumventricular organs

(CVO) where the blood-brain barrier is deficient and these neurons are therefore

accessible to circulating molecules such as angiotensin II, although isolated

from the rest of the brain by other barriers. Blood-brain barrier is present in all

brain regions except the CVO including area postrema (AP) in the brainstem (a

part of the brain that controls vomiting), median eminence (connecting the

hypothalamus with the pituitary gland), posterior pituitary gland (neuronal

projections of the hypothalamus that secrete peptide-hormones vasopressin and

oxytocin into the circulation), pineal gland, subfonical organs (SFO) (involved

in vasopressin secretion) and organum vasculosum of the lamina terminalis

(OVLT) (controlling thirst, sodium excretion, blood volume regulation and

vasopressin secretion) (Lind & Johnson, 1982; Fitzsimons, 1998). These brain

structures are implicated in body fluid homeostasis and rich in angiotensin II

receptors and important concerning drinking behaviour, renal function and blood

pressure, and extremely sensitive to the action of angiotensin II. They also have

Page 35: Plant-Derived Substances and Cardiovascular Diseases

35

extensive connections with the hypothalamus and other limbic structures

translating information about the inner milieu into behaviour including

emotional-related behaviour, learning and memory (Mosimann et al., 1996). In

addition to thirst, sodium appetite and blood pressure control, angiotensin II in

the central nervous system (CNS) affects cell growth, membrane function,

protein synthesis, prostaglandin release, pituitary hormone synthesis and release

(Mosimann et al., 1996). The RAAS is shown in figure 5.

The Angiotensin-Converting Enzyme

Angiotensin-converting enzyme (ACE; EC 3.4.15.1) (Skeggs et al., 1956b),

initially known as kininase II (Yang & Erdös, 1967) a zink carboxypeptidase is

synthesized by the endothelium and present on the luminal surface of the

membrane of the endothelial cells (Baudin et al., 1997). ACE is bound with part

of the hydrophobic C-terminal in the cell membrane and with the active sites

protruding out into the vessel lumen (Ryan et al., 1975; Hooper et al., 1987; Wei

et al., 1991a) (figure 5). ACE may loose its C-terminal end and become

dissolved in plasma as circulating ACE (Hooper et al, 1987, Wei et al, 1991a,

Baudin et al, 1997). ACE is found mainly in the lungs due to their vast surface

of vascular endothelium (Ng &Vane, 1967). ACE is also present in other

vascular tissues than endothelium; such as smooth muscle cells (in tunica media

and tunica adventitia) (André et al., 1990; Arnal et al., 1994; Battle et al., 1994),

the heart (Lindpaintner et al., 1987), fibroblasts (Arnal et al., 1994; Battle et al.,

1994), the kidney, CNS, placenta and testis (Erdös & Skidgel, 1986; Erdös,

1990). ACE is a polyspecific enzyme metabolising angiotensin I (see above,

section “The renin-angiotensin aldosteron system”), angiotensin 1-7 (see above,

section “The renin-angiotensin aldosterone system”), and furthermore e.g.

enkephalins, substance P, luteinizing hormone-releasing hormone (LH-RH),

desArg9-bradykinin (Erdös & Skidgel, 1986; Berecek & Zhang, 1995,). In

Page 36: Plant-Derived Substances and Cardiovascular Diseases

36

insects, ACE is also involved in digestion (as a gastrointestinal hormone),

reproduction and immune defence (Macours & Hens, 2004). Furthermore, ACE

metabolise bradykinin, kallidin (that can be converted to bradykinin) and

kininogen (responsible for bradykinin generation) (Scharfstein et al., 2007).

Bradykinin is a 9-amino acid peptid (Arg-Pro-Pro-Gly-Phe-Ser-Pro-Phe-Arg),

an endothelial cell dependent vasodilator (Regoli & Barabé, 1980; Hall, 1992)

synthesised by the liver and present in plasma and tissues. Bradykinin is formed

as a result of increased vascular permeability (tissue injury) (Sharma et al.,

1996) i.e. inflammation, activated Hageman Factor (factor XII) and the effects

of bradykinin are mediated by the bradykinin receptors, B1, B2, B3 and B4.

Bradykinin activates eNOS and thereby generates NO (figure 5). Bradykinin is

involved in acute and chronic inflammation (Sharma et al., 1996), vasodilatation

(Regoli & Barabé, 1980; Hall, 1992), increases vascular permeability (Sharma et

al., 1996), stimulates and sensities sensory neurons (Whalley et al., 1987) and

increases airway secretion (Hall, 1992). Bradykinin is inactivated by ACE,

kininase I, aminopeptidase P (APP), neutral endopeptidase (NEP) and

carboxypeptidase M and N (CPM, CPN) (Kuoppala et al., 2000) generating des-

Arg-bradykinin a specific agonist to B1 receptor. Thus, ACE is not only

activating the vasoconstrictor angiotensin II but it also inactivates the

vasodilatator bradykinin (figure 5).

ACE contains two homologous sites (Soubrier et al., 1988; Bernstein et al.,

1989), catalytically and independently active (Wei et al., 1991b). Testicular

ACE contains a shorter amino acid sequence and only one active site (the C-

terminal site) (Erdös, 1990), suggesting a gene duplication during the evolution

(Corvol et al., 1995). Both active sites contain Zn2+ ion (Ehlers & Riordan,

1991, Wei et al, 1991b). ACE inhibitors are designed to bind to the Zn2+ ion at

the active sites of ACE (Sturrock et al., 2004).

Page 37: Plant-Derived Substances and Cardiovascular Diseases

37

The variation of plasma ACE activity between different individuals is very high

(Alhenc-Gelas et al., 1983), but there seems to be a familiar resemblance of

ACE activity levels (Cambien et al., 1988). The ACE gene is found on

chromosome 17 and the ACE gene polymorphic sites is an insertion/deletion

(I/D) type and explains 47% of the variations between individuals (Rigat et al.,

1990; Tiret et al., 1992). Individuals with the DD genotype have two-three fold

higher levels of ACE than those with the II genotype, and the ID genotype has

an intermediate level of ACE activity (Rigat et al., 1990; Beohar et al., 1995).

The D allele of the ACE gene is suggested to correlate with cardiovascular

diseases (Cambien et al., 1992; Samani et al., 1996). Hence, it seems that the

genotype DD could be a risk factor for cardiovascular diseases (Beohar et al.,

1995).

Page 38: Plant-Derived Substances and Cardiovascular Diseases

Figu

re 5. Th

e renin-angiotensin aldosterone syste

mA

minopeptidase A

(AM

PA

), Am

inopeptidase N (A

MP

N), Brad

ykinin (BK

), Brad

ykinin receptors (BK

R), Insulin

(IRA

P), M

as oncogen

e receptor (Mas), N

eutral endopeptidase (N

EP

), Nitric oxide (N

O),

endopeptidase (PE

P), R

enin/prorenin receptors (R

PR

38

angiotensin aldosterone syste

m. A

ngiotensin-converting enzyme (A

CE

), An

giotensin receptors (AT

Am

inopeptidase A (A

MP

A), A

minopeptidase N

(AM

PN

), Brad

ykinin (BK

), Brad

ykinin receptors (BK

R), Insulin

(IRA

P), M

as oncogen

e receptor (Mas), N

eutral endopeptidase (N

EP

), Nitric oxide (N

O), E

ndothelial nitric oxide synthase (eN

OS

),endopeptidase (P

EP

), Re

nin/prorenin receptors (RP

R).

enzyme (A

CE

), An

giotensin receptors (AT

1 , AT

2 , AT

3 , AT

4 ), A

minopeptidase A

(AM

PA

), Am

inopeptidase N (A

MP

N), Brad

ykinin (BK

), Brad

ykinin receptors (BK

R), Insulin-re

gulated aminopeptidase

Endothelial nitric oxide synth

ase (eNO

S),

Prolyl

Page 39: Plant-Derived Substances and Cardiovascular Diseases

39

AIMS

The effects of flavonoids are previously associated with their effects as

antioxidants. Epidemiological studies and meta-analyses strongly suggest that

long term consumption of a diet rich in vegetables and fruits will protect against

cardiovascular diseases (Hertog et al., 1993; Hertog et al., 1995; Ness &

Powles, 1997; Carlson et al., 2008) due to the involvement of reactive oxygen

species. Focus has been on the possible role of free radical scavenging and

radical suppression of nutrients in explaining the beneficial effect of diet

compounds. As for the beneficial protection from cardiovascular diseases, the

antioxidative effects of plant-derived substances as flavonoids are but one

explanation of several.

The antioxidative properties of flavonoids, tocopherols and carotenoids are of

importance, but to maintain homeostasis of the vascular wall in order to affect

the development of cardiovascular diseases the balance between angiotensin II,

nitric oxide (NO) and reactive oxygen species (ROS) is of great importance.

The aims of this study were to …..

…… investigate if the sole mechanism according to the benificial properties of

vegetables, fruits and berries on cardiovascular diseases, is their antioxidative

properties

…… investigate plant extracts with alleged effect on the cardiovascular system

on angiotensin-converting enzyme activity and nitric oxide concentration, in

vitro, and in vivo (after administration of green, black tea and Rooibos). Plant

extracts used as everyday beverage, dietary products, food supplements, herbal

medicinal products and traditional antioxidants

Page 40: Plant-Derived Substances and Cardiovascular Diseases

40

…… map the active substances with effect on angiotensin-converting enzyme

activity and nitric oxide concentration

…… investigate if there is any connection between effect and

biosynthesis/structure

Page 41: Plant-Derived Substances and Cardiovascular Diseases

41

METHODS

The studies on cultured endothelial cells from human umbilical veins (HUVEC)

(Paper I-IV) were approved by the regional ethics committee at the Faculty of

Health Sciences, Linköping, Sweden (Dnr 03-602).

The in vivo study (Paper V) was approved by the regional ethics committee at

the Faculty of Health Sciences, Linköping, Sweden (Dnr M56-07).

INFUSIONS AND EXTRACTIONS

Tea Infusion, in vitro (Paper I)

Tea infusions were prepared from green tea (Camellia sinensis, L. Theaceae;

Japanese Sencha), black tea (Camellia sinensis, L. Theaceae; Indian Assam

B.O.P.) and Rooibos (Aspalathus linearis, Dahlg. Leguminosae). Infusions were

made with 1 g tea in 20 ml sterile phosphate-buffered saline (PBS) for 5 min

(the green tea and the black tea) or 10 min (the Rooibos tea). The infusions were

filtered twice, first through a standard filter 0.45µm (Munktell, Grycksbo,

Sweden) and then through a sterile filter 0.2µm (Millipore). The obtained

filtrates were considered as 1:20 and were frozen at -20ºC in aliquots.

Tea Infusion, in vivo (Paper V)

The tea was prepared from green tea Japanese Sencha imported by Charabang,

Stockholm, Sweden, black tea Indian Assam B.O.P. imported by Norrköping

Kolonial, Sweden, and Rooibos tea imported by Norrköping Kolonial, Sweden.

The teas were bought at Tebladet, Linköping, Sweden. Infusions were made

with 10 g tea in 400 ml fresh-boiled water for 5 min with the green tea and the

black tea, and 10 min with the Rooibos tea, using a tea filter (Agatha´s Bester,

Germany).

Page 42: Plant-Derived Substances and Cardiovascular Diseases

42

Coffee Infusion, in vitro

Coffee infusion (Coffea arabica, L. Rubiaceae) was prepared from Santos Brazil

coffee. Infusion was made with 2 g coffee in 20 ml sterile PBS for 3 min. The

infusion was filtered twice, first through a standard filter 0.45µm (Munktell,

Grycksbo, Sweden) and then through a sterile filter 0.2µm (Millipore). The

obtained filtrate was considered 1:10 and was frozen at -20ºC in aliquots.

Cacao Extraction, in vitro

Cacao extract (Theobroma cacao, L. Sterculiaceae), containing approximately

10% cacao fat was used for preparation of solution. Solution was made of 1 g

cacao extract in 20 ml sterile PBS for 15 minutes in boiling waterbath. The

solution was filtered twice through a standard filter 0.45 µm (Munktell,

Grycksbo, Sweden). The obtained filtrate was considered as 50 mg/ml and was

frozen at -20ºC in aliquots.

Bilberry 25E Extraction, in vitro (Paper IV)

Bilberry extract 25E (Vaccinium myrtillus, L. Ericaceae) standardized for 25%

anthocyanidins (cyanidin, delphinidin and malvidin) was used for preparation of

solution. Solution was made of 1 g Bilberry 25E extract in 20 ml sterile PBS for

15 minutes in boiling waterbath. The solution was filtered twice through a

standard filter 0.45 µm (Munktell, Grycksbo, Sweden). The obtained filtrate was

considered as 50 mg/ml and was frozen at –20°C in aliquots.

Liquorice Extraction, in vitro

Liquorice (Glycyrrhiza glabra L. Fabaceae) dried powder was used for

preparation of solution. 2 g powder was dissolved in 20 ml sterile PBS and

submitted to boiling waterbath for 1 hour. The Glycyrrhiza suspension was then

Page 43: Plant-Derived Substances and Cardiovascular Diseases

43

filtered twice through a standard filter 0.45 µm (Munktell, Grycksbo, Sweden).

The obtained filtrate was considered as 100 mg/ml and was frozen at -20ºC in

aliquots (Persson et al., 2008).

Ginkgo biloba Extraction, in vitro (Paper III)

Ginkgo biloba, L. (Ginkgoaceae) standardized extract EGb 761, was used for

preparation of solution. One g EGb 761 was dissolved in 10 ml sterile PBS and

placed in 60ºC waterbath for 1 hour. The suspension was then filtered twice

through a standard filter 0.45µm (Munktell, Grycksbo, Sweden). The obtained

filtrate was considered as 100 mg/ml and was frozen at -20ºC in aliquots.

Panax ginseng Extraction, in vitro (Paper II)

Panax ginseng L. (Araliaceae) standardized extract G115, was used for

preparing the solution. 1 g G115 was dissolved in 10 ml sterile PBS and placed

in 60ºC waterbath for 1 hour as described previously (Friedl et al., 2001). The

suspension was filtered twice, first through a standard filter, then through a

sterile filter 0.2µm (Millipore). The obtained filtrate was considered as 100

mg/ml and frozen at -20ºC in aliqouts.

CULTURED ENDOTHELIAL CELLS FROM HUMAN UMBILICAL VEINS

(HUVEC) (PAPER I-IV)

Human umbilical cords were obtained after normal vaginal delivery (after

informed consent from the mothers), and kept in sterile bottles containing PBS

and antibiotics. Endothelial cells were isolated according to the method of

Nyhlén et al., (2000). In short, the veins were cannulated at each end, washed

with PBS, and then treated with collagenase in 37ºC for 25 minutes. The

collagenase+cell perfusate was washed twice, and then resuspended in cell

culture medium (Dulbecco´s modified Eagle´s medium, DMEM) supplemented

with nonessential amino acids (1:100), oxalacetic acid (1.2 mM), insulin (0.24

Page 44: Plant-Derived Substances and Cardiovascular Diseases

44

IE/ml), penicillin (5 U/ml), streptomycin (0.5 µg/ml), hepes (10 mM),

endothelial cell growth factor (ECGF, 30 µg/ml), heparin (20 U/ml) and 17 %

inactivated fetal calf serum (FCS). Resuspended HUVEC were seeded in 25

cm2 tissue culture flasks coated with 0.2 % gelatin, kept in an incubation

chamber, and medium was replaced every 48-72 hour. At confluence, cells were

harvested with trypsin-EDTA for 5-10 minutes, and then reseeded 1:2. Second

passage was seeded in a 96-well microtiter plate, and allowed to reach

confluence.

ANGIOTENSIN-CONVERTING ENZYME ACTIVITY IN HUVEC (PAPER

I-IV)

HUVEC cultured in 96-well microtiter plates (as described above; 2nd passage)

was used. Immediately prior to treating the cells with drugs or extract, the

medium was removed and replaced with serum free medium. This was done to

avoid discrepancies in results due to ACE present in the fetal calf serum

(Bramucci et al., 1999). Cells were treated with flavonoids, terpenes, sterols,

purines, precursor molecules, plant infusions and extracts, human steroids and

lipid-lowering drugs for 10 minutes. The flavonoids tested were the isoflavone

genistein (0.1, 0.5 and 1 mg/ml), the flavonol quercetin (0.1, 0.5 and 1 mg/ml),

the epi-flavan-3-ols (catechins) epicatechin (0.1, 0.5 and 1 mg/ml),

epicatechingallate (0.1, 0.5 and 1 mg/ml), epigallocatechin (0.1, 1 and 2 mg/ml)

and epigallocatechingallate (0.05, 0.1, 1 and 2 mg/ml), procyanidin (0.1, 0.5 and

1 mg/ml), the anthocyanidins cyanidin, delphinidin and malvidin (0.01, 0.025

and 0.05 mg/ml), myrthillin chloride (0.01, 0.025 and 0.05 mg/ml) containing

cyanidin, delphinidin and malvidin, and the biflavan sciadopitysin (0.1, 0.5 and

1 mg/ml). The terpenes tested were the diterpenes α-tocopherol (0.1, 0.5 and 1

mg/ml) and ginkgolides A, B, C, (0.1, 0.5 and 1 mg/ml), the sesquiterpene

bilobalide (0.1, 0.5 and 1 mg/ml), the triterpenes ginsenoside Rb1, Rb2, Rc, Rd,

Re, Rf, Rg1 (0.1, 0.5 and 1 mg/ml) and the tetraterpene β-carotene (0.1, 0.5 and

Page 45: Plant-Derived Substances and Cardiovascular Diseases

45

1 mg/ml). The sterols tested were stigmasterol (0.1, 0.5 and 1 mg/ml), lanosterol

(10-7, 10-6, 10-5 and 10-3 M) and cholesterol (10-5, 10-4 and 10-3 M). The purines

caffeine (0.1, 0,5 and 1 mg/ml), theobromine (0.01, 0.05 and 0.1 mg/ml) and

theophylline (0.1, 0.5 and 1 mg/ml) were also tested. The biosynthesis precursor

molecules tested were mevalonic acid, malonic acid, shikimic acid, chorismic

acid and the progenitor squalene (10-7, 10-6, 10-5 and 10-3 M). The phenol salicin

(0.1, 0.5 and 1 mg/ml) were tested. Bilberry extract 25E (0.00625, 0.0125,

0.025, 0.05 and 0.1 mg/ml) containing 25% anthocyanidins, cacao extract

(0.00625, 0.0125, 0.025, 0.05 and 0.1 mg/ml) containing procyanidin oligomers

derived from epicatechin were tested. Green tea, black tea and Rooibos tea

infusions, coffee infusion (1:3200, 1:1600, 1:800, 1:400 and 1:200), Ginkgo

biloba Egb761 extract (0.1, 0.5, 1, 5 and 10 mg/ml), Panax ginseng G115

extract (0.1, 0.5, 1, 5 and 10 mg/ml), liquorice extract (0.00625, 0.0125, 0.025,

0.05 and 0.1 mg/ml) were tested. The human-derived steroids aldosterone,

estradiol and testosterone (0.1, 0.5 and 1 mg/ml) were also tested. Furthermore,

the blood lipid-lowering drugs simvastatin and pravastatin were tested (10-8, 10-7

and 10-6 M). The bilberry extract, the cacao extract, the green, black and

Rooibos tea infusions, the coffee infusion, the Ginkgo biloba extract, the Panax

ginseng extract, the liquorice extract, salicin and cholesterol were dissolved in

PBS; all other drugs in dimethylsulfoxide (DMSO). Corresponding volumes of

PBS or DMSO were used as controls. Blank and standard serum, from the

commercial kit was added to wells with corresponding volumes of medium

without FCS. After 10 minutes incubation with drugs, ACE activity was

analysed as is described below.

Page 46: Plant-Derived Substances and Cardiovascular Diseases

46

ANGIOTENSIN-CONVERTING ENZYME RADIOENZYMATIC ASSAY

(PAPER I-V)

After incubating cells with drugs, extracts, infusions (Paper I-IV), or serum

(Paper V) ACE activity was analysed with a commercial radioenzymatic assay

(ACE-direct REA, Bühlmann Laboratories, Allschwil, Switzerland). In short,

the synthetic substrate 3H-hippuryl-glycyl-glycine was added to the samples and

cleaved by ACE to 3H-hippuric acid. After 2 hours for the in vitro experiments

and 1 hour for the in vivo study (Paper V) of incubation in 37ºC, the reaction

was stopped with 1 M HCl, scintillation liquid was added, and each sample was

counted in a scintillation counter.

NITRITE/NITRATE ASSAY (PAPER I-III, V)

After incubating the cells with drugs, extracts or infusions, medium was

removed and stored at -20ºC until nitrite/nitrate analysis, or for the in vivo study

(Paper V), serum was stored at -70ºC until analysis. Nitrite was analysed as a

marker of NO concentration as proposed by Lauer et al. (2001). Nitrite and

nitrate concentration in the tea infusions, cacao, Ginkgo biloba and Panax

ginseng extracts were also analysed. Nitrite/nitrate concentration was analysed

with a commercial nitrite/nitrate assay (Nitric oxide (NO2-/NO3

-) assay, R&D

Systems, UK). Nitrate was first reduced to nitrite which was then analysed. The

principle for the colorimetric detection of nitrite is the Griess reaction;

diazonium ions are produced when acidified nitrite reacts with sulfanilic acid.

Diazonium ions form chromophore agents when reacting with N-(1-

naphthyl)ethylenediamine. Optical density was determined with a Spectramax

reader at 540 nm.

Page 47: Plant-Derived Substances and Cardiovascular Diseases

47

LIQUID CHROMATOGRAPHY-MASS SPECTROMETRY-MASS

SPECTROMETRY (LC-MS-MS) (PAPER I)

For quantifications of epicatechin, epigallocatechin, epicatechingallate and

epigallocatechingallate, an ESI-LC-MS-MS system for gradient chromatography

was used (Paper I). The instrumentation consisted of a Perkin Elmer 200

chromatographic system equipped with two micro pumps, a solvent degasser

and an autosampler (Norwalk, CT, USA). Mass detection was performed on a

Sciex API 2000 triple quadrupole instrument equipped with a turbo ion-spray

interface (PE Sciex, Ontario, Canada) operating in positive ion mode. The

interface probe was set at 350ºC and the ion-spray needle was operated at -4500

V. Nitrogen was used as nebulizer-, auxiliary-, curtain- and CAD-gas and was

set at 25, 50, 30 psi and a value of 5 respectively. The high performance liquid

chromatography (HPLC) was carried out on a Hypersil Polar-RP 150 x 3.0 mm

(Phenomenex, Torrance, CA, USA) equipped with an Opti-Solv 2 µm column

inlet filter (Optimize, Portland, Oregon, USA). The mobile phases consisted of

acetonitrile, methanol and 20 mM ammonium formiate buffer, pH 3, in mixtures

of 5:5:90 (v/v/v) for phase A and 40:40:20 (v/v/v) for phase B. A linear gradient

chromatography from 0-100 % B-phase over seven minutes was run. Total

runtime including wash and reconditioning was 10 minutes. A flow-rate of 0.25

ml/min at ambient temperature was used. The detection and quantification of the

selected compounds were performed in the Multiple Reaction Monitoring mode

(MRM). MS-MS product ion spectra were produced by collision activated

dissociation (CAD) of the de-protonated molecule ion (M+1)-. The most intense

transitions; 289/245 (epicatechin), 305/179 (epigallocatechin) 441/169

(epicatechingallate), 457/169 (epigallocatechingallate) were used for

quantification. Linear calibration curves were performed ranging from 0.5 µg/ml

to 20 mg/ml. Tea extracts were diluted in methanol and buffer 1:10 and 1:100

Page 48: Plant-Derived Substances and Cardiovascular Diseases

48

respectively before analysis. Instrument control, integration and calculation were

performed with the PC based PE Sciex software, Analyst 1.4.

TENSION STUDIES (PAPER II)

Bovine mesenteric arteries were obtained at the local slaughterhouse and then

transported to the laboratory in Krebs´ buffer. The Krebs´ buffer had the

following composition (in mM): 137 Na+, 5.89 K+, 2.50 Ca2+, 1.20 Mg2+, 134 Cl-

, 18.0 HCO3-, 1.20 H2PO4

- and 5.60 glucose. The arteries were carefully cleaned

of adipose tissue, cut into rings of 3-5 mm and then cut open, forming arterial

strips. The strips were mounted between a fixed point in organ chambers

containing Krebs´ buffer, and a Grass FT 03 strain gauge transducer connected

to a Grass Polygraph (Grass Instrument, Quincy, MA, USA), allowing

continuous measurement of auxometric tension. The strips were stretched to 2-

2.5 g and allowed to equilibrate for 45 min, resulting in a passive tension of 0.5-

1.0 g (the optimal passive tension for these vessels). After wash, arteries were

incubated with or without ginseng extract for 10 min and then angiotensin I-

induced contractions were examined by submitting the strips to 10-5 M

angiotensin I. Then, the preparations were submitted to several washes until

tension returned to basal value. Maximal contraction of each strip was

determined by using a Krebs´ buffer with a high K+ concentration (128 mM K+,

15.4 mM Na+ and other ions as described above).

When endothelium-dependent relaxation was studied, two strips were used from

each cow. In one strip the endothelium was left intact, while on the other strip

the endothelium was removed by gently rubbing the luminal side of the strip

with a wooden stick as previously described (Persson and Andersson, 1998).

Then, the strips were precontracted with 10-5 M phenylephrine, and when a

stable contraction was obtained ginseng extract was added in cumulative doses.

Page 49: Plant-Derived Substances and Cardiovascular Diseases

49

IN VIVO STUDY (PAPER V)

Twenty healthy volunteers were randomised in a single-blind cross-over study to

receive either 400 ml green tea (Japanese Sencha), black tea (Indian Assam

B.O.P.) or Rooibos tea. The effect of single dose of the different teas was

compared with a 1-4 weeks washout period between the tea regimens. All

experiments started at 8 am and the subjects were not fasting. Three subjects

were excluded; two because of difficulties when collecting blood and one used

oral tobacco. The participants did not use any kind of drugs (medical or herbal)

2 weeks prior to and during the study. The volunteers did not use nicotine in any

form. Intake of beverages or food containing high amount of catechins, the

flavonol quercetin and anthocyanins, i.e. chokeberries, aubergine, blackberries,

bilberries, elderberries, raspberries, strawberries, coffee, cacao/chocolate,

cherries, onions, plums, pears, radishes, red and black currants, red cabbage,

black beans, cranberries, tea, wine, grapes and apples were not allowed 48 h

before the experiment. A list of food not allowed was distributed to the

participants when they registered to the study. The participants did also answer a

questionnaire concerning everyday intake of certain food i.e. vegetables, fruits

and berries containing the flavonoids catechins, quercetin and anthocyanins.

Data collected at each treatment were blood pressure, heart rate and venous

blood samples for analyses of ACE activity, NO concentration and ACE

genotype. The tea was newly made and cooled so the participants could drink it

within approximately 2 minutes. Blood pressure, heart rate and blood samples

for analyses of ACE activity and NO concentration were taken before drinking

tea and after 30 min, 60 minutes and 3 hours. Vaccutainer tubes without

anticoagulantia were used to collect blood for serum-ACE and serum-nitrite

analyses. After 2 hours the tubes were centrifugated 1000g for 20 minutes, at

4ºC. Serum was transferred to plastic tubes and ACE activity were analysed the

same day as described above. Remaining serum was frozen -70ºC until nitrite

Page 50: Plant-Derived Substances and Cardiovascular Diseases

50

analysis. Vaccutainer tubes with EDTA were used to collect blood for

determination of polymorphism concerning the ACE genotype. The blood was

transferred to sterile test tubes and frozen -70ºC until analysis (see above).

ANGIOTENSIN-CONVERTING ENZYME GENOTYPE (PAPER V)

Genomic DNA was isolated from venous blood using QIAmp DNA Mini Kit

(QIAGEN, Hilden, Germany).

The ACE gene contains a polymorphism based on the absence and/or presence

of a 287 base pair nonsense DNA domain in intron 16, which results in three

different genotypes: deletion/deletion (DD) homozygote, insertion/insertion (II)

homozygote and insertion/deletion (ID) heterozygote. Three primers 5´-CTG

CAG GTG TCT GCA GCA TGT GC-3´, 5´-GAT TAC AGG CGT GAT ACA

GTC ACT TTT-3´ and 5´-GCC ATC ACA TTC GTC AGA TCT GGT AG-3´

(Invitrogen Ltd., Paisley, UK) were used according to the method of Cheon et

al. (Cheon et al., 2000).

For the polymerase chain reaction (PCR) reaction a “ready-to-use” reaction

mixture (REDExtract-N-Amp Blood PCR ReadyMix, Sigma-Aldrich, St. Louis,

MO, USA) was used in a total volume of 20 µl. Each primer was used at a

concentration of 0.4 µM and for each reaction 2 µl DNA solution was added to

the reaction mixture.

The PCR was performed in a Perkin Elmer Cetus DNA Thermal Cycler and

started with 5 minutes of initial denaturation at 94ºC. Then the samples

underwent 30 cycles of amplification: 45 seconds of denaturation (94ºC), 45

seconds of annealing (65ºC) and 45 seconds of extension (72ºC). The reaction

was finished with 7 minutes of final extension (72ºC) and hold at 4ºC. The PCR

products were separated by electrophoresis on a 2% agarose gel, then stained

Page 51: Plant-Derived Substances and Cardiovascular Diseases

51

with ethidium bromide and visualized with ultraviolet light. The PCR products

detected with the primers are one 237 base pair fragment for the deleted allele,

and one 525 and one 155 base pair fragment for the inserted allele.

Page 52: Plant-Derived Substances and Cardiovascular Diseases

52

CHEMICALS

All chemicals for culturing cells were obtained from Life Technologies,

Scotland, UK, except Endothelial Cell Growth Factor that was bought at

Boehringer-Mannheim, Germany and heparin (Heparin LEO) from LEO

Pharma AB, Malmö, Sweden. Ginkgolides A, B, C, bilobalide, ginsenosides

Rb1, Rb2, Rc, Rd, Re, Rf and Rg1, cyanidin, delphinidin, malvidin and myrtillin

chloride were purchased from Extrasynthese Genay, France. Genistein,

quercetin, epicatechin, epicatechingallate, epigallocatechin,

epigallocatechingallate, procyanidin, sciadopitysin, α-tocopherol, β-carotene,

stigmasterol, lanosterol, cholesterol, mevalonic acid, malonic acid, shikimic

acid, chorismic acid, squalene, aldosterone, estradiol, testosterone, caffeine,

theobromine, theophylline, simvastatin, pravastatin, angiotensin I and

phenylephrine were bought from Sigma-Aldrich Chemical Co, MO, USA.

Enalaprilat (Renitec® 1 mg/ml) was bought from Merck Sharp and Dohme,

Haarlem, The Netherlands. NG-monomethyl-L-arginine (L-NMMA) and N-3-

(aminomethyl) benzylacetamidine (1400W) were bought from Alexis

Biochemicals, San Diego, USA. Salicin was a gift from Naturapoteket,

Norrköping, Sweden. The Bilberry extract 25E was a gift from Ferrosan,

Copenhagen, Denmark; the cacao extract and the liquorice powder was a gift

from Cloetta-Fazer, Ljungsbro, Sweden; the green tea, the black tea, the

Rooibos and the coffee was a gift from Tebladet, Linköping, Sweden; Ginkgo

biloba EGb 761 was a gift from Cederroth International AB, Upplands Väsby,

Sweden, and Dr Willmar Schwabe, Karlsruhe, Germany; Panax ginseng G115

was a gift from Pharmaton S.A. Switzerland.

Page 53: Plant-Derived Substances and Cardiovascular Diseases

53

CALCULATIONS

Results are presented as mean ±s.e.m. (standard error of the mean).

One unit (U) of ACE activity is defined as the amount of the enzyme required to

release 1 µmole of hippuric acid per minute and liter. Statistical calculations

were done with Graph Pad PrismTM 3.0 (Paper I and IV) Graph Pad PrismTM 4.0

(Paper II) and Graph Pad PrismTM 5.0 (Paper III and V). One way analysis of

variance (ANOVA) for repeated measures was performed followed by

Dunnett´s post test. Statistical significance is denoted as * p<0.05, **p<0.01 and

*** p<0.001.

Page 54: Plant-Derived Substances and Cardiovascular Diseases

54

RESULTS AND DISCUSSION

Results and discussion starts with effects seen on ACE activity followed by

effects on NO concentration, food-drug interactions and the results applied on

clinical importance. The results concerning ACE activity are divided in

beverages, other dietary products, herbal medicinal products, traditional

antioxidants, sterols, blood-lipid lowering drugs, precursor molecules and

proposed mechanism of effects of flavonoids on ACE activity.

Results on ACE activity and NO concentration in HUVEC and chemical

structure of the substances tested are summarised in Table 1, at the end of this

section.

NO concentrations was not measured with Bilberry extract 25E or its

anthocyanidins cyanidin, delphinidin and malvidin due to light absorption at 540

nm which is the same wavelength used in the nitrite/nitrate analysis. Neither was

NO concentration measured with genistein, ginsenosides, β-carotene, α-

tocopherol, stigmasterol, pravastatin and simvastatin.

Page 55: Plant-Derived Substances and Cardiovascular Diseases

55

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF

BEVERAGES CONTAINING FLAVONOIDS AND XANTHINES

Green Tea, Black Tea and Rooibos Tea, in vitro (Paper I)

Due to the climate, the tea tree/bush is cultivated all over the World, but mostly

in China, Japan, India and Sri Lanka, and tea is one of the most ordinary

beverages in the World. Green tea and black tea is produced from Camellia

sinensis L. (Theaceae); green tea from heated leaves to preserve the green

colour, and black tea from fermented leaves. Pharmacological effects on the

cardiovascular system of green and black tea have been attributed to

antioxidative effects of the catechin substances. Rooibos tea is a caffeine-free

alternative produced from fermented leaves and bark of a tree growing in South

Africa, Aspalathus linearis Dahlg. (Leguminosae). Rooibos tea do not contain

catechins but dihydrochalcones, flavones and flavonols.

In this study, 10 minutes incubation with green tea, black tea and Rooibos tea

showed a significant and dose-dependent inhibition of ACE activity in HUVEC

with the green tea 1:1600 *p<0.05, 1:800, 1:400 and 1:200 **p<0.01, with the

black tea 1:800 *p<0.05, 1:400 and 1:200 **p<0.01, while no significant effect

was seen with the Rooibos tea compared to the PBS control.

After ten minutes incubation a significant inhibition of ACE activity was seen

with the flavanols epicatechin 1 mg/ml *p<0.05, epicatechingallate 0.1mg/ml

*p<0.05, 0.5 mg/ml and 1 mg/ml **p<0.01, epigallocatechin 0.1 mg/ml

*p<0.05, 1 and 2 mg/ml **p<0.01 and epigallocatechingallate 0.1, 1 and 2

mg/ml ***p<0.001 compared to the DMSO control.

The green tea showed a stronger ACE inhibitory effect compared to the black

tea, probably because of the higher amount of catechins in green tea, especially

epigallocatechingallate. The tendency towards inhibition seen with the Rooibos

tea could be due to the content of flavonoids other than catechins. In contrast to

green and black tea, Rooibos tea do not contain any catechins, but the flavonol

Page 56: Plant-Derived Substances and Cardiovascular Diseases

56

quercetin is present in Rooibos tea (Bramati et al., 2003). The slight inhibition

of ACE activity in vitro by Rooibos tea might depend on its quercetin content,

since quercetin exhibits an ACE inhibitory action in vitro (Paper III).

The degree of ACE inhibition varied between the different flavanols/catechins

tested; lowest effect was seen with epicatechin, then epigallocatechin,

epicatechingallate, and the greatest effect was seen with epigallocatechingallate,

and consequently between extracts containing different amounts of catechins;

green tea and black tea. An increased number of hydroxyl groups and addition

of double-bound oxygen seemed to increase the inhibitory effect of the different

catechins on ACE activity (Table 1).

Coffee Infusion, in vitro

Coffee is produced by roasting the beans from the coffee bush, Coffea arabica

L. (Rubiaceae). Coffee is an everyday beverage mostly consumed due to its

amount of the psychostimulant caffeine. Studies investigating coffee in relation

to cardiovascular diseases are mostly focusing on its content of caffeine (George

et al., 2008). Besides caffeine, coffee contains catechin and epicatechin (Chen et

al., 2006).

After 10 minutes incubation with the coffee extract a significant inhibition on

ACE activity in HUVEC was seen with 5.0 mg/ml *p<0.05 compared to the

PBS control. This inhibition is proposed to be due to its content of catechins.

Cacao Extract, in vitro

Cacao is produced from the beans of Theobroma cacao L. (Sterculiaceae), a tree

cultivated in South-America, Africa and Asia. Cacao and chocolate is produced

from fermented, dried, roasted and crushed cacao-beans. Cacao contains

Page 57: Plant-Derived Substances and Cardiovascular Diseases

57

procyanidins which are oligomeric catechins linked together; dimers and trimers

are most common, e.g. epicatechin-epicatechin.

After 10 minutes incubation with cacao extract a significant and dose-dependent

inhibition of ACE activity in HUVEC was seen with 0.00625, 0.0125, 0.025,

0.05 and 0.1 mg/ml **p<0.01 compared to the PBS control. Compared to the tea

infusions, the cacao extract seems to have a stronger inhibitory effect on ACE

activity but to be noted; pure cacao extract was used not cacao-powder as we

usually drink or eat.

Xanthines, in vitro

The xanthines/purines caffeine, theobromine and theophylline are components

in coffee, tea and cacao and are known to have various pharmacological effects

on the cardiovascular system. Neither, caffeine, theobromine or theophylline did

show any significant effect on ACE activity in HUVEC after ten minutes

incubation compared to DMSO control, thus they are not responsible for the

effect on ACE activity of tea, coffee nor cacao.

Green Tea, Black Tea and Rooibos Tea in Healthy Volunteers (Paper V)

After oral intake of 400 ml green tea, black tea or Rooibos tea, a significant

**p<0.01 inhibition of serum ACE activity was seen with the Rooibos tea after

30 minutes and *p<0.05 after 60 minutes.

As previously mentioned, studies show that tea reduces mortality in

cardiovascular diseases (Mukamal et al., 2002; Hakim et al., 2003; Babu & Liu,

2008). The effects of tea on cardiovascular diseases are suggested to be due to

the high content of catechins i.e. epicatechin, epigallocatechin,

epicatechingallate and epigallocatechingallate (Paper I).

Page 58: Plant-Derived Substances and Cardiovascular Diseases

58

In HUVEC, green tea and black tea significantly inhibited ACE activity, in

contrast to Rooibos tea where no significant inhibition of ACE activity was seen

(Paper I). In the in vivo study, with healthy volunteers drinking tea, a significant

inhibition of serum ACE activity after intake of Rooibos tea was seen. This

discrepancy between the in vitro and in vivo studies may either be due to the

content of flavonoids, their absorption and/or metabolism or to a possible

different mechanism of action of the Rooibos tea compared to the green tea and

the black tea. The Rooibos tea did not show any significant effect on ACE

activity in vitro, but the flavonol quercetin found in Rooibos tea showed

significant inhibition of ACE activity in vitro.

To understand the mechanisms of action, and to be able to validate the results

from the in vitro experiments and to confirm the inhibiting effect on ACE

activity in vivo, it is necessary to gain information about the pharmacokinetic of

the flavonoids. During metabolism of flavonoids different metabolites are

formed (Kühnau, 1976; Williamson & Manach, 2005) which might be

responsible for the effects on ACE activity in vivo. An in vitro model of

degradation of quercetin by microorganisms formed the metabolites

dihydroquercetin (taxifolin), the auronol alphitonin and chalcones (Braune et al.,

2001) and, consequently, quercetin and its metabolites may affect ACE activity

in vivo. Tea catechins undergo phase II metabolism, methylation,

glucuronidation and sulphation. Epigallocatechingallate, the catechin with the

strongest effect on ACE activity in HUVEC, is hence degraded into methylated,

glucuronided and sulphated forms of epigallocatechin (Lambert et al., 2007)

which may explain the discrepancy of the results in vitro compared to in vivo of

green tea and black tea. Data available on flavonoids and their pharmacokinetics

consider unmetabolised flavonoids i.e. the ingested flavonoid or its conjugates.

Flavonoids are absorbed in the small intestine as glycosides or hydrolyzed to

their aglycones by microorganisms and then absorbed (Walle, 2004). The half-

Page 59: Plant-Derived Substances and Cardiovascular Diseases

59

life of catechins seems to be rather short, 1-3 hours, compared to genistein 6-10

hours and quercetin 11-28 hours (Manach et al., 2005). This discrepance in half-

life of the different flavonoids may also affect the results in vitro and in vivo

concerning tea.

When sub-grouping the participants, according to ACE genotype a significant

inhibition of serum ACE activity was seen with the green tea for the genotype II

*p<0.05, 30 minutes after intake, and for the genotype ID *p<0.05, 60 minutes

after intake. A significant inhibition of serum ACE activity was also seen with

the Rooibos tea for the genotype II *p<0.05, 60 minutes after intake. In

accordance with earlier studies (Rigat et al., 1990) the basal levels of serum

ACE activity was significantly higher ***p<0.001 for the participants with ID

and DD genotype compared to those with II genotype. The DD genotype is

proposed to be a risk factor for cardiovascular diseases, and in this study no

effect was seen on ACE activity for the DD genotype.

The difference on serum ACE activity between genotypes when drinking tea

may be a question of dosage or long term intake. ID and even more DD

genotype might need higher amount or long term intake of flavonoids to affect

serum ACE activity compared to II genotype. It is previously shown that the

effect of the ACE inhibitor drug enalaprilat is significantly greater among

individuals of II genotype (Ueda et al., 1998). Even so, in this study one cup of

tea was enough to significantly inhibit serum ACE activity in individuals with II

and ID genotypes. It is therefore tempting to suggest that “chronic” tea drinkers

have lower ACE activity no matter which genotype. The ACE gene

polymorphism explains 47% of the variations of serum ACE levels between

individuals (Rigat et al., 1990; Tiret et al., 1992). Although plasma levels of

ACE concentrations in one individual seems to be stable over time (Alhenc-

Gelas et al., 1991) environmental influence on gene expression can not be

excluded (Sing et al., 2003). Cardiovascular diseases have a complex

Page 60: Plant-Derived Substances and Cardiovascular Diseases

60

multifactorial etiology and can not be explained by nor genes neither

environmental factors alone. Over time environmental factors as e.g. culture and

diet may influence gene expression and consequently change gene expression

(Sing et al., 2003). The “evolution” of culture according to diet; from a

traditionally plant-based diet containing complex carbohydrates and dietary

fibers, vegetables, fruits and berries to a diet rich in saturated fats, and simple

carbohydrates i.e. a diet consisting mainly of meat and products with high

energy content (WHO, 2003), may have changed the expression of the ACE

genotypes. Changes of gene expressions due to long term intake of one-sided

food e.g. unsaturated fat and lack of vegetables, fruits and berries may be

underestimated.

In conclusion, the beneficial properties of tea besides antioxidative properties,

on cardiovascular diseases may be due to its inhibitory effect on ACE activity.

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME

ACTIVITY OF DIETARY PRODUCTS CONTAINING

ANTHOCYANINS OR ISOFLAVONOIDS

Bilberry Extract 25E, in vitro (Paper IV)

Bilberry, Vaccinium myrtillus L. (Ericaceae), grows in the north of Europe and

is one of the most common plants in Sweden and bilberry is known to contain

high amounts of anthocyanins.

The bilberry extract 25E, according to the manufacturer standardized for

containing 25% anthocyanins (the glucosides), showed a significantly inhibitory

effect on ACE activity in HUVEC. After incubation with Bilberry extract 25E

for 10 minutes, a significant and dose-dependent inhibition was seen with

Page 61: Plant-Derived Substances and Cardiovascular Diseases

61

0.00625, 0.0125, 0.025, 0.05 and 0.1 mg/ml ***p<0.001 compared to the PBS

control.

The anthocyanidins (the aglycones) cyanidin, delphinidin and malvidin did not

show any significant inhibition of ACE activity, while the combination of these

three anthocyanidins in myrtillin chloride 0.05 mg/ml *p<0.05 showed

significant inhibition of ACE activity compared to the DMSO control.

Previous studies show effect of anthocyanidins on blood circulation, especially

in vision disorders such as retinopathy caused by diabetes or hypertension and

venous insufficiency (Detre et al., 1986; Favot et al., 2003; Matsumoto et al.,

2005). Its mechanism of action is proposed to be due to its antioxidative and free

radical scavenging properties (Kähkönen & Heinonen, 2003).

The effect of bilberry and its anthocyanidins on ACE activity has not previously

been studied. The effect on ACE activity showed a more pronounced effect

using the bilberry extract compared to the respective anthocyanidins (combined

in myrtillin chloride or separately). The difference in inhibition of ACE activity

between the anthocyanidins and/or the bilberry extract may be due to other

compounds than anthocyanins present in the bilberry extract, e.g. the stilbene

resveratrol and the carotenoid lutein. The anthocyanidin substances are not

water soluble in contrast to anthocyanins (anthocyanidin with sugar) in the

Bilberry extract and this makes the anthocyanidins difficult to study as isolated

substances. In this study, the Bilberry extract 25E was dissolved in PBS while

myrtillin chloride and the anthocyanidins were dissolved in DMSO, and this

organic solvent is most likely of importance for the outcome of the results on

ACE activity. The ability of the anthocyanidins to form the natural electron

deficient flavylium cation makes them very sensitive to changes in pH, and

sensitive also to temperature, oxygen and light (Kim et al., 2003).

Supplementation based on bilberry extract is used for improvement of vision

(Canter & Ernst, 2004), macular degeneration and diabetic retinopathy (Zafra-

Page 62: Plant-Derived Substances and Cardiovascular Diseases

62

Stone et al., 2007). As vascular factors contribute to the development of various

ocular diseases such as diabetic retinophaty, macular degeneration and

glaucoma, the inhibition of ACE activity by Bilberry extract in HUVEC might

indicate that bilberry has positive effects on the eye. Supplementation based on

bilberry often also contain other substances as lutein (a carotenoid) and Vitamin

A, which also might be/probably is involved in the proposed effect on vision by

bilberry supplement (see below, section “Beta-carotene, in vitro”).

Liqourice Extract, in vitro (Persson et al., 2008)

Liqourice is produced from the roots of Glycyrrhiza glabra L. (Fabaceae) and

most frequently used as a flavouring/sweetening agent in drugs, herbal teas,

tobacco and as sweets. Glycyrrhiza species contain two types of main chemical

substances; the triterpene saponins glycyrrhizic acid and glycyrrhizinic acid, and

flavonoids. More than 300 different flavonoids have been isolated from

Glycyrrhiza species, numerous isoflavones and other flavonoids (Li et al.,

2000).

After 10 minutes incubation of HUVEC with Glycyrrhizin extract, a significant

and dose-dependent inhibition of ACE activity was seen with 0.00625, 0.0125,

0.025, 0.05 and 0.1 mg/ml ***p<0.001 compared to the PBS control (Persson et

al., 2008). This implies that liquorice can have positive effects on cardiovascular

diseases. However, liquorice is contraindicated in patients with hypertension due

to its content of the triterpene saponins glycyrrhizin, although it has been

reported that liquorice is valid against cardiovascular diseases due to its

antiatherosclerotic and antioxidative effects (Aviram & Fuhrman, 1998;

Fuhrman et al., 2002). Liquorice can induce hypertension (Russo et al., 2000)

by interaction with the hormonal system, i.e. aldosterone and cortisol, involved

in blood pressure homeostasis (Schambelan, 1994). Liquorice intake may

decrease the release of aldosterone and markedly increase cortisol levels thereby

Page 63: Plant-Derived Substances and Cardiovascular Diseases

63

inducing pseudohyperaldosteronism resulting in severe hypertension and

hypokalemia. Glycyrrhizin is believed to be responsible for the effects of

liqourice on the adrenal glands (Hammer & Stewart, 2006). The chemical

structure of glycyrrhizin is related to the structure of steroidal hormones in

humans and like these hormones, liquorice i.e. glycyrrhizin affects RAAS by

inhibition of renin release (Sigurjonsdottir et al., 2006). Fuhrman et al. have

shown that liquorice extract free of glycyrrhizinic acid reduces systolic blood

pressure (Fuhrman et al., 2002), and Quaschning et al. have shown that

glycyrrhizic acid increases systolic blood pressure (Quaschning et al., 2001).

Taken together, these two previous studies and our results imply that the

flavonoid/isoflavonoid fraction of the plant may act as an ACE inhibitor. It is

well known that angiotensin II increases the release of aldosterone from the

adrenal glands. The mechanism by which liquorice inhibits aldosterone release

is unknown, but may be explained by the inhibition of ACE activity in HUVEC

seen in this study. In this study aldosterone did not show any significant effect

on ACE activity in HUVEC (see below, section “Sterols, in vitro” and Table 1),

while other studies have shown significant increase of ACE activity by

aldosterone (Sugiyama et al., 2005).

In conclusion, even if liqourice is contraindicated in hypertension, this study

demonstrates that liquorice extract inhibits ACE activity in HUVEC. These are

benifical indicators on the cardiovascular system and on the blood pressure.

Substances in liquorice responsible for these effects are not clarified, but most

likely it is the flavonoids/isoflavonoids.

Genistein, in vitro

Genistein is an isoflavone found in soya-beans Glycine max (L.) Merr.

(Fabaceae). Isoflavonoids are so called phytooestrogens, according to their

structural resemblance with oestrogen and oestrogen-like effects in humans.

Page 64: Plant-Derived Substances and Cardiovascular Diseases

64

After ten minutes incubation of HUVEC, a significant and dose-dependent

inhibition of ACE activity was seen with genistein 0.1 mg/ml *p<0.05, 0.5

mg/ml ***p<0.001 and 1 mg/ml **p<0.01 compared to the DMSO control.

Prevention of cardiovascular diseases mediated by antioxidative effects and by

lowering of cholesterol and LDL has been reported by soy protein containing

high amounts of isoflavonoids as genistein (Sirtori & Lovati, 2001, Clarkson,

2002).

The inhibition of ACE activity by genistein seen in this study has also been seen

in rats by others (Xu et al., 2006). In contrast to genistein, oestrogen showed no

inhibition of ACE activity in HUVEC.

This study indicates that isoflavonoids as well as flavonoids inhibits ACE

activity in vitro.

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF

HERBAL MEDICINAL PLANTS EXTRACTS

Panax Ginseng extract, in vitro (Paper II)

Panax ginseng L. (Araliaceae) is a traditional herbal plant from Asia. Panax

ginseng extract is made from root and/or rhizome and used as a so called

“adaptogen”. Adaptogens are referred to as natural plant products that are

proposed to balance the homoestasis of the body i.e. by increase of mental and

physical performance (Keifer & Pantuso, 2003).

After 10 minutes incubation with Panax ginseng G115 extract, a significant and

dose-dependent inhibition of ACE activity in HUVEC was seen with 5 and 10

mg/ml **p<0.01 compared to the PBS control.

The effects of Panax ginseng are traditionally(?) suggested to be due to the

triterpene saponins, the ginsenosides.

Page 65: Plant-Derived Substances and Cardiovascular Diseases

65

After ten minutes incubation with the triterpenes ginsenosides Rb1, Rf and Rg1,

no significant inhibition of ACE activity was seen compared to the DMSO

control. The ginsenosides Rb2, Rc and Rd significantly increased ACE activity

(Rb2 0.5 mg/ml **p<0.01 and 1 mg/ml *p<0.05, Rc 0.05 mg/ml **p<0.01 and 1

mg/ml ***p<0.001, Rd 0.1, 0.5 mg/ml *p<0.05 and 1 mg/ml **p<0.01), while

the ginsenoside Re 1 mg/ml *p<0.05 significantly inhibited ACE activity in

HUVEC compared to the DMSO control (Table 1, unpublished data).

Beside the rather non-specific aphrodisiac and adaptogenic effects (Nocerino et

al., 2000), Panax ginseng is also believed to reduce blood pressure (Kim et al.,

1994; Han et al., 1998; Jeon et al., 2000; Sung et al., 2000), act as an

antioxidant (Kim et al., 1992) and increase nitric oxide concentration (Gillis,

1997). The cardiovascular effects of Panax ginseng, the antioxidative

properties, are proposed to be linked to NO (Gillis, 1997; Nocerino et al., 2000)

(see below, section “Nitric Oxide, in vitro” and Table 2).

The ginsenoside Re was the only ginsenoside to inhibit ACE activity, while

Rb2, Rc and Rd significantly increased ACE activity in HUVEC. This increase

of ACE activity seen in HUVEC could either be due to increased biosynthesis of

ACE in the endothelial cells or a direct increase of ACE activity. The

differentiation in effect of the ginsenosides on ACE activity in HUVEC might

be due to the location of the different glycosides attached to the aglycone

ginsenoside structures. There are divergent opinions about however ginseng is

appropriate as treatment of cardiovascular diseases (Coon & Ernst, 2002;

Messina, 2006) and a possible side effect reported for Panax ginseng is

hypertension (Coon & Ernst, 2002). The ginsenosides are triterpene saponins

and the chemical structure of the triterpenes is comparable to a steroid structure

and triterpenes are known to interact with the hormonal system (see above,

section “Liqourice Extract”). Hypertension due to Panax ginseng may be due to

increased ACE activity by the ginsenoside Rb2, Rc and Rd (seen in this study)

Page 66: Plant-Derived Substances and Cardiovascular Diseases

66

or the ginsenosides affecting the adrenal glands; a proposed possible mechanism

of the adaptogenic action of Panax ginseng is the augmentation of adrenal

steroidogenesis (Nocerino et al., 2000). Studies show that Panax ginseng may

slightly decrease blood pressure (Siegel, 1979) while other studies show

increase in blood pressure (Coon & Ernst, 2002). This discrepancy might

depend on the amount of ginsenosides and flavonoids in the extract tested.

Although, there is a standardized extract of Panax ginseng the amount of

respective compound may vary. Panax ginseng G115 extract is standardized

after the amount of ginsenosides (4%); however, the amount of and the

characterization of the flavonoids are usually not documented. The inhibitory

effect of Panax ginseng G115 on ACE activity in HUVEC may be associated

with the amount of flavonoids present in the extract. This conceivable inhibition

of ACE activity by the flavonoids in the Panax ginseng extract may interact

with the increase of ACE activity by the ginsenosides resulting in opposite

effects on ACE activity. Whether the inhibition of ACE activity by Panax

ginseng extract or the increase of ACE activity by the ginsenosides Rb2, Rc and

Rd seen in vitro also occur in vivo is dependent on the bioavailability of the

respective substances.

Ginkgo Biloba extract, in vitro (Paper III)

The standardized extract EGb 761 of Ginkgo biloba L. (Ginkgoaceae) is

prepared from leaves of the Ginkgo biloba tree growing in Asia. Ginkgo biloba

is the only living representative of the Ginkgoales.

After 10 minutes incubation with Ginkgo biloba EGb 761 extract, a significant

and dose-dependent inhibition of ACE activity in HUVEC was seen with 5 and

10 mg/ml ***p<0.001 compared to the PBS control.

Page 67: Plant-Derived Substances and Cardiovascular Diseases

67

The extract, EGb 761 is standardized concerning the content of the presumed or

proposed most active substances; 6% terpene lactones (ginkgolides A, B, C, J

and bilobalide), 24% flavonol glycosides (principally derivatives of quercetin,

kaempferol and isorhamnetin), 7% proanthocyanidins (e.g. procyanidin), 13%

carboxylic acids, 2% catechins and <0.005% ginkgotoxin (Van Beck, 2002;

Mesbath et al., 2005; Xie et al., 2006).

No significant effect on ACE activity in HUVEC was seen with the diterpenes

ginkgolides A, B, C or the sequiterpene bilobalide in contrast to the Gingko

biloba extract EGb 761.

The flavonol quercetin present in Gingko biloba extract was shown to affect

ACE activity. After ten minutes incubation with the flavonol quercetin a

significant inhibition of ACE activity in HUVEC was seen with 0.1 mg/ml

*p<0.05, 0.5 mg/ml ***p<0.01 and 1 mg/ml ***p<0.001 compared with the

DMSO control. This inhibition of ACE with quercetin is also described in other

studies (Häckl et al., 2002; Loizzo et al., 2007).

Procyanidin did not significantly affect ACE activity in HUVEC compared to

the DMSO control. However, there was a significant difference **p<0.01

between the concentrations 0.1 and 1 mg/ml of procyanidin.

Ginkgo leaves, but not the standardised extract EGb 761, also contain small

amounts of the biflavone sciadopitysin (Van Beck, 2002). Sciadopitysin did not

show any significant effect on ACE activity in HUVEC compared to the DMSO

control.

Ginkgo biloba is considered to have cardioprotective effects (Liebgott et al.,

2000; Pietri et al., 1997), antioxidative effects (Pietri et al., 1997), increase nitric

oxide concentration (Koltermann et al., 2007) and effects on cerebral blood flow

(Ahlemeyer & Krieglstein, 2003), dementia (DeKosky, 2006) and cognition

(Ward et al., 2002).

Page 68: Plant-Derived Substances and Cardiovascular Diseases

68

The inhibitory effect of Ginko biloba extract EGb 761 on ACE activity in

HUVEC is suggested to be associated with its content of flavonoids. This

inhibition of ACE activity may be one explanation of the pharmacological

effects of Ginkgo biloba on the cardiovascular system. Mechanisms related to

the effects of Ginkgo biloba on dementia and Alzheimer´s desease are related to

the increase of NO and to NO as a scavenger (Bastianetto & Quirion, 2002;

DeKosky et al., 2006) and antioxidant resulting in reduction of age-related loss

of cognitive functions (Ward et al., 2002). Studies have been performed with the

ginkgolides and the ginsenosides on cognitive functions like learning and

memory, showing that the ginkgolides may have effects in vitro and in animal

models (Birks & Grimley Evans, 2008) and that the ginsenosides are possibly

interacting with the steroid hormone receptors (Leung et al., 2007). However,

inhibition of ACE activity will increase local blood flow (in the CNS).

Furthermore, as ACE activity i.e. angiotensin II, III and IV, is involved in

cognitive functions (Chaki & Inagami, 1992; Ruiz-Ortega et al., 2001; Gard,

2004; Bodineau et al., 2008), the inhibition of ACE activity might also, at least

partly explain the proposed effects of Ginkgo biloba and Panax ginseng on

cognitive functions. Studies have been performed on flavonoids concerning

effect on behaviour and cognition showing a possible restoring beta-amyloid-

induced effect by epigallocatechingallate (found in tea) (Rasoolijazi et al.,

2007), and antioxidative effects altering stress signalling and neuronal

communication by vegetables and fruits containing polyphenols (Joseph et al.,

2007; Shukitt-Hale et al., 2008; Williams et al,. 2008). It has also been shown

that liqourice extract may improve learning and memory in mice (Dhingra et al.,

2004). There is shown to be a relationship between cardiovascular diseases and

emotional expressions like anger, hostility and depression (Sirois & Burg, 2003;

Davidson, 2008). A diversity of emotional expressions derives as a result of

cardiovascular disease, as part of a coping strategy. There is also a physiological

interaction between emotions like depression and cardiovascular diseases due to

Page 69: Plant-Derived Substances and Cardiovascular Diseases

69

e.g. the impact on RAAS, the importance of serotonin and the immune system

and dysfunction in the hypothalamic-pituitary-adrenocortal and

sympathoadrenal axis in response to stress (Bondy, 2007). Like cardiovascular

diseases, depression is a common disorder in developed countries and there is a

psychological as well as physiological connection between the brain, the

endrocrine system (including RAAS) and the immune system. Is there a

connection between cardiovascular diseases and mental disorders e.g. depression

and diet? An improvement of quality of life has been reported in patients using

ACE inhibitors and angiotensin receptor blockers (Maggioni, 2006) as well as

with a vegan diet (Link et al., 2008).

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF

THE ANTIOXIDANTS ALPHA-TOCOPHEROL AND BETA-CAROTENE

Alpha-Tocopherol, in vitro

The diterpene α-tocopherol showed no significant effect on ACE activity in

HUVEC.

Alfa-tocopherol is associated with antioxidative effects (Matringe et al., 2008).

However, α-tocopherol does not seem to lower cardiovascular mortality (Buijsse

et al., 2008). The fact that a substance is an antioxidant may not prove that the

same substance will prevent cardiovascular diseases. It could be speculated that

to have positive effects on cardiovascular diseases, an antioxidative effect is not

sufficient. Some additional mechanism of action may be required, perhaps ACE

inhibition.

Page 70: Plant-Derived Substances and Cardiovascular Diseases

70

Beta-Carotene, in vitro

The carotenoids are tetraterpenes and have some characteristics in common with

the flavonoids; they are plant-pigments present in the plastid. The carotenoids

derive from a different biosynthesis pathway compared to the flavonoids and in

contrast to the flavonoids, the carotenoids are lipid-soluble substances.

After ten minutes incubation with β-carotene, a significant and dose-dependent

inhibition of ACE activity in HUVEC was seen with 0.5 mg/ml **p<0.01 and 1

mg/ml ***p<0.001 compared to the DMSO control.

Carotenoids have principal functions as antioxidants in plants, and β-carotene is

the most common of the carotenes. In humans, the effect of β-carotene is related

to its antioxidant activity in prevention of a variety of diseases e.g.

cardiovascular diseases (Bjelakovic et al., 2008). The use of β-carotene as

antioxidative supplementation is debated as it has been shown that treatment

with β-carotene as well as α-tocopherol may increase mortality in various

diseases (Voutilainen et al., 2006; Bjelakovic et al., 2008).

Beta-carotene was the only non-flavonoid substance tested with a strong

inhibitory effect on ACE activity. Approximately 60 carotenoids (carotenes and

xanthophylls) have been found in human diet and few of these are present in

blood in measurable amounts e.g α-, β-carotene, lycopene, β-cryptoxanthin,

lutein and zeaxanthin. Do all carotenoids inhibit ACE activity or is it just β-

carotene? The difference in chemical structure between the carotenoids is;

acyclic or having one or both ends modified into rings, containing at least one

oxygen atom, or hydrocarbon atom (chemical structure of β-carotene is shown in

Table 1). The chemical similarities with flavonoids are the OH-groups and the

double bondings. From a speculative point of view probably all carotenoids

inhibit ACE activity, at least in vitro. To confirm this speculation further in

vitro/in vivo studies on ACE activity have to be performed.

Page 71: Plant-Derived Substances and Cardiovascular Diseases

71

The bioavailability of carotenoids is highly variable, dependent on e.g. species,

amount consumed in a meal, genetic factors and interactions. Food processing,

as cooking (β-carotene is absorbed 65% from cooked carrots and 41% from raw

carrots), and intake of fat improve the bioavailability of the carotenoids. The

bioavailability of carotenoids as supplements is usually higher than of

carotenoids in fruits and vegetables (Yonekura & Nagao, 2007). Beta-carotene is

the carotene best known for its function as a vitamin A precursor and vitamin A

is mainly studied for its role in eye function and vision.

After ten minutes incubation with β-carotene a significant inhibition of ACE

activity in vitro in the eye retinal pigmented epithelium cell line ARPE-19 was

seen with 0.5 mg/ml *p<0.05 and 1 mg/ml **p<0.01 compared with DMSO

control (figure 6; unpublished data).

Figure 6. ACE activity in ARPE-19 after incubation with β-carotene for 10 minutes. Statistics were calculated with one way ANOVA repeated measures and denoted as *p<0.05 and **p<0.01 compared with DMSO, n=3.

Beta-carotene

DMSO

0.1

mg/m

l

0.5

mg/m

l

1 m

g/ml

0.0

0.5

1.0

1.5

*

**

AC

E a

ctiv

ity (U

)

Page 72: Plant-Derived Substances and Cardiovascular Diseases

72

Carotenoids have been studied concerning effects on macular degeneration,

indicating a protective effect of carotenoid-rich food especially lutein and

zeaxanthin (Seddon, 2007). Decrease in ocular blood flow is one risk factor for

macular degeneration (Pemp & Schmetterer, 2008). Macular degeneration is a

common condition in the elderly and exists in two forms; as a cause of cellular

debris and angiogenesis of blood vessels from the choroid. Diabetic retinophaty

is described as changes in the retinal microvessels that can lead to macular

edema and vision loss. The major components of RAAS have been identified in

eye tissue and hypertension is a risk factor for diabetic retinopathy and local

formation of angiotensin II ensure local control of blood flow in e.g. the eye

(Ganten et al., 1971).

Activation of the angiotensin II receptors AT1 is proposed to be involved in

diabetic retinopathy (Clermont et al., 2006). ACE inhibiting drugs are shown to

inhibit capillary degeneration and to block the progress of diabetic retinopathy

(Clermont et al., 2006). The inhibition by β-carotene of ACE activity on ARPE-

19 in vitro is an indication of a possible mechanism of carotenoids on vision

disorders.

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF

STEROLS AND BLOOD LIPID-LOWERING DRUGS

Sterols, in vitro

So far, β-carotene, except for the ginsenoside Re, is the only lipid tested that

significantly inhibited ACE activity in HUVEC. Due to this we also tested the

sterol lipid, the phytosterol stigmasterol, cholesterol, lanosterol, the human

steroids oestradiol, testosterone and the mineralcorticoid aldosterone.

Page 73: Plant-Derived Substances and Cardiovascular Diseases

73

After ten minutes incubation with the sterols, no significant inhibition of ACE

activity in HUVEC was seen with stigmasterol, oestradiol, testosterone or

aldosterone compared to the DMSO control, nor with cholesterol compared to

the PBS control. Lanosterol significantly increased ACE activity in HUVEC 10-3

M **p<0.01 compared to the DMSO control. In this study, aldosterone did not

show any significant effect on ACE activity, while another study has shown a

significant increase of ACE activity by aldosterone (Sugiyama et al., 2005).

Blood lipid –Lowering Drugs, in vitro

In relation to the results on ACE activity in HUVEC in this study by the sterols

and their previously shown effects on the cardiovascular system by lowering

cholesterol levels (Klingberg et al., 2008; Poli et al., 2008), the blood lipid-

lowering drugs pravastatin and simvastatin were tested on ACE activity in

HUVEC.

After ten minutes incubation with the bloodlipid-lowering drugs pravastatin and

simvastatin, a significant increase of ACE activity was seen in HUVEC, 10-7 and

10-6 M *p<0.05, 10-7 M, **p<0.01 and 10-6 M *p<0.05, respectively.

These drugs are used as treatment of cardiovascular diseases by lowering

bloodlipid levels but might, as this study shows, increase ACE activity. This

might be a negative side effect of blood lipid-lowering drugs on the

cardiovascular system.

EFFECTS ON ANGIOTENSIN-CONVERTING ENZYME ACTIVITY OF

PRECURSOR MOLECULES

To investigate if the effect on ACE activity were associated to the biosynthesis

of plant substances (a biosynthesis-activity connection) the precursor molecules

involved in the biosynthesis of flavonoids, and the phenol salicin derived solely

from the shikimic pathway (figure 1) were studied.

Page 74: Plant-Derived Substances and Cardiovascular Diseases

74

After ten minutes incubation with the precursors, mevalonic acid, malonic acid,

shikimic acid, chorismic acid and the progenitor squalene, no significant effect

on ACE activity was seen in HUVEC compared to the DMSO control. Neither

did salicin show any significant effect on ACE activity after ten minutes

incubation of HUVEC compared to the PBS control. Studies show an inhibiting

effect on platelet aggregation/adhesion by flavonoids (Nardini et al., 2007; Babu

& Liu, 2008). This effect on platelets probably derives from the shikimic acid

pathway and coumaric acid, which is the connection between salicin (the

ancestor of acetylsalicylic acid) and the flavonoids (see figure 1).

THE MECHANISM OF THE INHIBITORY EFFECT ON ANGIOTENSIN-

CONVERTING ENZYME ACTIVITY OF THE FLAVONOIDS

Epidemiological studies have shown reduced cardiovascular morbidity among

tea drinkers (Mukamal et al., 2002; Hakim et al., 2003; Babu & Liu, 2008). It

has been proposed that this positive effect is due to the antioxidative effect of tea

catechins. Previously shown effects of green tea and black tea include

antioxidative (Locher et al., 2002; Kurihara et al., 2004), antiproliferative and

anti-angiogenic activity (Locher et al., 2002; Oak et al.,2005) and nitric oxide

synthase activation (Nakagawa & Yokozawa, 2002; Anter et al., 2004; Lorenz et

al., 2004). Pharmacological effects attributed to Rooibos tea are antioxidative

activity (Standley et al., 2001; Lee & Lang, 2004) and immunostimulatory

effects (Lambert & Yang, 2003).

Cacao contains catechin-related flavonoids as procyanidins and catechins as in

tea and coffee. Cacao is proposed to play a beneficial role on the cardiovascular

system as a anti-inflammatory modulator (Selmi et al., 2008) and procyanidins

found in cacao are shown to have antioxidative effects (Keen et al., 2005; Aron

et al., 2008).

Page 75: Plant-Derived Substances and Cardiovascular Diseases

75

The inhibition on ACE activity in HUVEC shown in this study may explain the

positive effects of green tea and black tea seen in habitual tea drinkers. The

results seen in this study on ACE activity in HUVEC using tea as well as cacao

and coffee add an additional and novel mechanism to the antioxidative effect;

flavonoids i.e. catechins as ACE inhibitors.

Do all flavonoids inhibit ACE activity and which is the possible molecular

mechanism of the ACE inhibitory action of the flavonoids and β-carotene?

Flavonoids are known to bind to enzymes and proteins (Andersen & Markham,

2006). Some proteins need additional components for their functions, So called

co-factors. In plants, as well as in humans, co-factors may be metal ions, e.g.

Fe2+ or Mg2+. Flavonoids as a group are known to have metal chelating capacity

to ions like Fe, Cu and Al (Raven, Evert and Eichhorn, 2003; Kostyuk et al.,

2001). The major flavonoid in liquorice, the isoflavone glabridin is known to

have metal chelating capacity (Belinky et al., 1998). ACE have two active sites

containing Zn2+ ion and the pharmacological mechanism of medicinal drugs

inhibiting ACE may be their ability to bind to the Zn2+ at the active site of ACE

and thereby decreasing its activity (Berecek & Zhang, 1995). Metal ions like

Mg2+ and Fe2+ are also involved in the biosynthesis of flavonoids. The

biosynthesis of the flavonoid precursor chalchone, the flavanols, the isoflavanols

and the anthocyanins are dependent on enzymes with metal chelating activity

(Andersen & Markham, 2006). A common feature of the biosynthesis of

flavanols (catechins), flavonols, isoflavanols and anthocyanins is that the

enzymes involved, flavanone 3β-hydroxylase (F3H), and anthocyanidin

synthase (ANS), bind and are dependent on Fe2+. Also Mg2+ functions as a

cofactor in the biosynthesis, for acetyl-CoA carboxylase (ACC) to form

malonyl-Co-A. It is possible that the flavonoid substrates bind to these enzymes

via the metal ions. The mechanism of this enzyme- and protein binding,

including the mechanism of the ACE inhibition, is so far speculative. However,

Page 76: Plant-Derived Substances and Cardiovascular Diseases

76

it seems that the flavonoids i.e. flavanols (catechins), flavonols (e.g.quercetin)

and anthocyanins do have the potential to bind Zn2+ at the active site of the

ACE. The flavonoids are relatively small chemical structures, with the ability to

form covalent bondings (Andersen & Markham, 2006). Furthermore, the

phenolic nucleus is a structural unit suitable for molecular, non-covalent

interactions with proteins related to the double bondings and OH-group(s) of the

flavonoids (Andersen &Markham 2006). This might be one explanation of the

ACE inhibitory effect of the flavonoids seen in this study.

In fact, flavonoids are known to be metal chelaters (Kostyuk & Potapovich,

1998; Kostyuk et al., 2001; Havsteen, 2002), to bind metal ions like Fe2+ and

Cu2+. It seems that flavonoids are involved in the resistance and protection of

plants against exposure to high levels of metals in soils (Havsteen, 2002;

Moreno et al., 2003). Furthermore, plants exposed to high levels of e.g.

aluminium have been found to synthesize high levels of flavonoids (Riesenberg

& Soltis, 1987). For instance, bilberries grow in soil contaminated with metal

ions like Zn2+ without being affected by this (Vallino et al., 2005). The metal-

chelating effect of the flavonoids might explain the ACE inhibiting effect of

these substances. Many enzymes, as well as ACE, contain Zn2+ due to the ability

of Zn2+ to easily react with other compounds. These enzymes are said to belong

to the zinc metallopeptidase family. Other zinc metallopeptidases except ACE

are e.g. aminopeptidase A (AMPA), aminopeptidase N (AMPN), insulin-

regulated aminopeptidase (IRAP), endothelin-converting enzyme (ECE), matrix

metallopeptidase (MMP), neutral endopeptidase (NEP) and insulin-degrading

enzyme (IDE). Peptidases are classified according to catalytic activity and

converged properties (Rawlings & Barrett, 1993). AMPA converts angiotensin

II to angiotensin III, AMPN converts angiotensin III to angiotensin IV, and

angiotensin IV activates IRAP which action may play a role in the development

of complications in type 2 diabetes (Keller, 2004). ECE catalyses the synthesis

Page 77: Plant-Derived Substances and Cardiovascular Diseases

77

of endothelin whith function as a vasoconstrictor, bronchoconstrictor and

stimulator of aldosterone secretion (Barton & Yanagisawa, 2008). MMPs are

involved in cell growth and tissue destruction in various diseases e.g myocardial

restenosis and macular degeneration (Marder & Greenwald, 2003; Dormán et

al., 2007). NEP catalyses the degradation of a variety of renal and CNS-active

peptides including substance P, bradykinin, enkephalins, atrial natriuretic

peptides (ANP), endothelin and angiotensin II (Aulakh et al., 2007). IDE,

degrading a range of substrates e.g. insulin, is a zinc metalloendopeptidase

(Authier et al., 1996), an evolutionary conserved neutral thiol-

metalloendopeptidase with structural similarities to NEP. If the inhibitory

mechanism of flavonoids on ACE activity is due to their ability to bind to Zn2+

ions then it would be possible for the flavonoids to also inhibit ECE, MMP, NEP

and maybe IDE, thereby exerting several additional positive effects on the

cardiovascular system. Kiss et al. have shown that Ligustrum vulgare L.

(Oleaceae) extract inhibit ACE and NEP in vitro (Kiss et al., 2008). Ligustrum

vulgare contain the flavonols kaempferol and quercetin (Romani et al., 2000).

No in vivo studies have been performed investigating the effects of flavonoids

on NEP, MMP, ECE or IDE.

In this study, β-carotene was the only non-flavonoid substance, except from the

ginsenoside Re, with a strong inhibitory effect on ACE activity.

Common features of the flavonoids and β-carotene are; they are plant pigments,

present in the plastid and antioxidants. It is unknown whether β-carotene has

metal-chelating properties and thereby exhibits ACE inhibition. The enzyme β-

carotene hydroxylase, like the enzymes involved in the biosynthesis of

catechins, flavonols, isoflavanols and anthocyanins, involved in the biosynthesis

of β-carotene is a Fe dependent enzyme. If β-carotene is a metal-chelator, the

metal-chelating mechanism could be the common feature of the plant-derived

Page 78: Plant-Derived Substances and Cardiovascular Diseases

78

substances affecting ACE activity, and thereby even all of the zinc

metallopeptidases.

Part of the aim of this study was to investigate if the sole mechanism according

to flavonoids and their protection against cardiovascular diseases is their

antioxidative effect. The results presented in this study show that ACE inhibition

is an additional mechanism.

Seen from an evolutionary perspective the flavonoids developed simultaneously

as vascular plants, approximately 440-430 million years ago (Raven et al.,

2003). Anthocyanins have been found in various orders of bryophytes (mosses),

the most primitive group of land plants, estimated 470 million years of evolution

(Gould, 2002). At this time the formation of soils was established. Between

approximately 470-430 million years ago plants went through a time of major

innovations: the colonization of land, development of a specialized system to

distribute water, solutes, photosynthetic products and the development of a

sexual reproductive system (Willis & McElwain, 2002). Seen from the

perspective of this study; it might be of importance that flavonoids developed

simultaneously as vascular plants, and are interacting with and affecting the

cardiovascular system as ACE inhibitors.

EFFECTS ON NITRIC OXIDE

Nitric oxide, in vitro (Paper I, II, III)

As the results on ACE activity showed, in some cases, a strong inhibition by the

flavonoids in vitro, it was interesting to investigate if there was a connection

between the effects on ACE activity, the antioxidative effects and the effects on

NO concentration.

Page 79: Plant-Derived Substances and Cardiovascular Diseases

79

After 24 hours incubation with the flavonoids and flavonoid-containing extracts,

a significant increase in NO concentration was seen in HUVEC with two of the

catechins, epigallocatechin 1 and 2 mg/ml **p<0.01 and epigallocatechingallate

0.5, 1 and 2 mg/ml ***p<0.001. Further, a significant increase in NO

concentration was seen with procyanidin 0.5 and 1 mg/ml **p<0.01, quercetin

0.1 mg/ml *p<0.05, 0.5 and 1 mg/ml ***p<0.001 and the biflavane

sciadopitysin 0.5 and 1 mg/ml ***p<0.001. Results were compared to DMSO

control.

After 24 hours incubation with plant-extracts containing high amounts of

flavonoids, a significant increase of NO concentration in HUVEC was seen with

the green tea 1:1600, 1:800, 1:400 and 1:200 ***p<0.001, black tea 1:800, 1:400

and 1:200 ***p<0.001, Rooibos tea 1:400 *p<0.05 and 1:200 ***p<0.001,

coffee extract 1:200 *p<0.05, cacao extract 100µg/ml ***p<0.001, liquorice

extract 50 and 100µg/ml ***p<0.001, Ginkgo biloba extract 5 and 10 mg/ml

***p<0.001 and Panax ginseng extract 0.5, 1, 5 and 10 mg/ml *p<0.05. Results

were compared to PBS control.

Other compounds significantly affecting NO concentration in HUVEC, apart

from the flavonoids, were caffeine increasing NO concentration, 1 mg/ml

*p<0.05, and the sesquiterpene bilobalide decreasing NO levels 0.5 mg/ml

*p<0.05 and 1 mg/ml ***p<0.001 compared to DMSO control.

Plants and plant-derived substances like green tea, black tea and various

catechins, cacao and procyanidins, bilberry and other anthocyanin-containing

berries, quercetin and genistein and their effect on NO concentration is a rather

well documented field of research (Squadrito et al., 2002; Hodgson, 2006;

Taubert et al., 2007; DalBó et al., 2008; Erlund et al., 2008; Faridi et al., 2008;

Grassi et al., 2008a; Hall et al., 2008; Loke et al., 2008; Nantz et al., 2008). NO

is known to influence the vascular system, and the increase in NO concentration

by flavonoids (Grassi et al., 2008b) will result in vasodilatation by relaxation of

Page 80: Plant-Derived Substances and Cardiovascular Diseases

80

smooth muscle cells (Squadrito et al., 2002; Hodgson, 2006; DalBó et al., 2008;

Grassi et al., 2008a; Hall et al., 2008), inhibition of smooth muscle cell

proliferation, reduced platelet aggregation (Erlund et al., 2008), reduced platelet

and monocyte adhesion to the endothelium, inhibition of LDL oxidation (Erlund

et al., 2008; Nantz et al., 2008,) and blood pressure reduction (Erlund et al.,

2008; Faridi et al., 2008; Grassi et al 2008; Nantz et al., 2008). An increase of

NO concentration by the flavonoids would thereby decrease the risk of

developing cardiovascular diseases (Napoli et al., 2006, Yetic-Anacak &

Catravas, 2006).

The results in this study showing an increase in NO concentration with the

flavonoids and flavonoid-containing extracts are in accordance with these

previous reports.

Is the increase in NO levels from HUVEC seen in this study related to an

increase of the cellular synthesis or to the nitrate present in the extracts

themselves? Studies have shown that inorganic nitrate found in vegetables and

fruits have the ability to convert to nitrite and NO in humans (Lundberg &

Govoni, 2004; Lundberg & Weitzberg, 2005; Lundberg et al., 2006). As shown

in Table 2, green tea, black tea, Rooibos, Ginkgo biloba and Panax ginseng

extracts themselves contain both nitrite and nitrate, and this could explain part of

the effect seen on NO concentration. However, the cacao extract did not contain

any nitrate but showed a significant increase in NO concentration from HUVEC

(Table 1). Furthermore, the Panax ginseng extract contains high amounts of

nitrate but did not show any significant increase on NO concentration after 10

minutes incubation (Paper I) and a significant but small increase, p<0.05 of NO

concentration after 24 hours incubation (Table 1). This indicates that the

increase in NO concentration seen with the flavonoids and extracts containing

high amounts of flavonoids is due to increases in cell-mediated NO

concentration and not nitrite/nitrate concentration of the extracts.

Page 81: Plant-Derived Substances and Cardiovascular Diseases

81

Table 2. The quantity of nitrite, nitrate and nitrite/nitrate in green tea, black tea, Rooibos, cacao extract, Ginkgo biloba extract and Panax ginseng extract compared to PBS.

Extract Nitrite µM Nitrate µM

Nitrite/Nitrate µM

PBS 0.32 0.53 0.85 Green tea 1:200 0.87 2.96 3.83 Green tea 1:800 0.74 2.73 3.47 Green tea 1:3200 0.42 2.57 2.99 Black tea 1:200 1.70 2.72 4.42 Black tea 1:800 0.42 3.29 3.71 Black tea 1:3200 0.32 3.15 3.47 Rooibos tea 1:200 1.93 2.37 4.30 Rooibos tea 1:800 0.64 1.76 2.40 Rooibos tea 1:3200 0.32 1.72 2.04 Cacao 0.1 mg/ml 0.32 0.53 0.85 Cacao 0.05 mg/ml 0.32 1.36 1.68 Cacao 0.025 mg/ml 0.32 1.36 1.68 Ginkgo biloba 10mg/ml 5.57 12.43 18.00 Ginkgo biloba 5mg/ml 3.21 13.36 16.57 Ginkgo biloba 1mg/ml 0.87 8.20 9.07 Gingko biloba 0.5mg/ml 0.32 4.93 5.25 Panax ginseng 10mg/ml 0.64 139.17 139.81 Panax ginseng 5mgml 0.42 66.04 66.46 Panax ginseng 1mg/ml 0.32 12.32 12.64

Page 82: Plant-Derived Substances and Cardiovascular Diseases

82

Nitric oxide, in vivo (Paper V)

After oral intake of 400 ml green tea, black tea or Rooibos tea, no significant

change in serum NO concentration was seen neither in the entire group of

volunteers nor in any of the genotype sub-groups.

Increased NO concentration may be the result of either an increased

synthesis/production due to increased NOS activity, or a prolonged half life of

NO. Increased NOS activity would result in a slow effect; the increase in NO

concentration in HUVEC was shown after incubation for 24 hours compared to

incubation for 10 minutes (Paper I, II and Table 1). Nicholson et al. has shown

that the stilbene resveratrol (figure 2) and quercetin affect the expression of the

gene encoding eNOS (Nicholson et al., 2008). Prolonged half life of NO may be

due to decreased metabolism of NO. NO is metabolised by reaction with O2-

forming peroxynitrit (Doughan et al., 2008). Flavonoids scavenging O2-, as

proposed in various studies (Sun et al., 2002; Chang et al., 2007; Jiménez et al.,

2007; Tomer et al., 2007), thereby inhibit the metabolism of NO and this

mechanism would result in rapid effect on NO concentration. Whether

flavonoids excert their antioxidative effects by scavenging free radicals or by

metal-chelating properties the effects will be the same. There are several known

connections between ACE and NO. NO inhibits ACE (Ackermann et al., 1998;

Persson et al., 2005) and downregulates AT1 receptors (Ichiki et al., 1998). ACE

degrades bradykinin, thereby inhibiting its stimulation of NO synthesis. Also,

angiotensin II activates NADH oxidase thereby increasing the synthesis of the

NO-scavenging radical O2-.

Page 83: Plant-Derived Substances and Cardiovascular Diseases

83

INTERACTIONS BETWEEN DIETARY PRODUCTS AND DRUGS

So far, interactions between herbal medicines and medicinal drugs are a rather

unexplored field. Results of this study showed a synergistic effect on ACE

activity in HUVEC by ginseng extract and the ACE inhibiting drug enalaprilat,

(Paper II) and by ginkgo biloba extract and enalaprilat (figure 7; unpublished

data). This indicates a possible interaction between ACE inhibiting drugs and

herbal medicines containing high amount of flavonoids with effect on ACE

activity.

Figure 7. ACE activity in HUVEC after incubation for 10 minutes with enalaprilat (E) alone

or in combination with Ginkgo biloba (GB) extract (ACE activity in PBS control 12.8±3.1 U),

n=3. Statistics calculated with one way ANOVA repeated measures and denoted as **p<0.01,

n=4 from different individuals.

-10 M

Enalap

rilat

10

M+G

B 0.1 m

g/ml

-10

E 10M

+GB 0.

5 mg/m

l

-10

E 10

GB 1.0 m

g/ml

M+

-10

E 10

M+G

B 5.0

mg/m

l

-10

E 10

M+G

B 10.0

mg/m

l

-10

E 10

0

5

10

15

****

AC

E a

ctiv

ity (U

)

Page 84: Plant-Derived Substances and Cardiovascular Diseases

84

As reported by Harris et al. (2003), a number of food-drug interactions in liver

metabolism have been reported (Harris et al., 2003). Food containing complex

mixtures of phytochemicals, e.g. vegetables, fruits and tea is more likely to

induce or inhibit the activity of drug-metabolising enzymes, and particularly

large interactions may occur due to intake of herbal dietary supplements (Harris

et al., 2003). Cytochrome P450 appears to be especially sensitive to dietary

effects (Cermak, 2008) and food-drug interactions may occur when food

substances use the same metabolizing enzymes as medicinal drugs. There are

indications on changes in gene expression of CYP 450 according to drug

metabolism (Ek et al., 2007) and this might also correspond to food intake.

Flavonoid-induced effects on drug bioavailability have also been reported

(Cermak, 2008).

CLINICAL IMPORTANCE

According to the WHO Statistical Information System (WHOSIS),

cardiovascular diseases is the number one cause of death and an estimated 20

million people will die from cardiovascular diseases in 2015; this will represent

30% of all global deaths (WHOSIS). The results of this investigation on plant-

derived substances inhibiting ACE activity and increasing NO concentration

could lead to new to ACE inhibiting drugs. Examining and mapping which

foods, i.e. vegetables, fruits and berries that contain these substances should lead

to specific diet recommendations concerning prevention and treatment of

cardiovascular diseases. ACE inhibiting drugs are used for treatment of

hypertension (Lim, 2007), congestive heart failure (Lim, 2007), ventricular

dysfunction (Lakhdar et al., 2008) and nephropathy in diabetes mellitus (Koitka

& Tikellis, 2008). Dependent on flavonoid(s) and/or carotenoid content,

vegetables, fruits and berries, containing plant-derived ACE inhibitors could be

an addition to medical drugs; food as medicine. Diet in the perspective of

Page 85: Plant-Derived Substances and Cardiovascular Diseases

85

lifestyle and cardiovascular diseases as a major cause of morbidity and mortality

is a combination of great interest, psychologically and physiologically. The

concomitant change in diet behavioural pattern and worldwide treatment of

cardiovascular diseases with medicinal drugs is probably not a mere accident.

Why do we eat; essentially for the same reason as we take medicinal drugs, to

survive and secondary, to stay healthy. Today, it seems that we eat to satisfy the

emotions and take medicinal drugs to survive and stay healthy.

Furthermore, flavonoids and β-carotene, due to inhibiting ACE activity, might

also increase blood flow and thereby positively affect cardiovascular diseases

and cognitive abilities.

Seen from a nutritional perspective, the role of the flavonoids in health and

disease is not clarified. As opposed to other well-known/recognized plant-

derived micronutrients e.g. Vitamins, lack of flavonoids does not induce obvious

deficiency syndroms. The initial classification of flavonoids as Vitamin P,

according to the action on vascular permeability and fragility (Rusznyak &

Szent-Györgyi, 1936) was withdrawn (Kühnau, 1976), and today flavonoids are

not classified as Vitamins, due to their lack of obvious deficiency syndroms.

Early analysis and identification of flavonoids in vegetables and fruits lead to an

estimated recommended dosage of a total of 1 g/day (Kühnau, 1976), distributed

between flavanones, flanonols and flavones (160-175 mg/day), anthocyanins

(180-215 mg/day), catechins (220 mg/day) and biflavans (460 mg/day)

(Kühnau, 1976). This estimated intake of flavonoids was applied until the 1990s,

when new results indicated this to be a highly overestimated dietary flavonoid

intake due to the accually flavonoid intake. An estimated daily intake of

flavonoids in Western population is about 100-200 mg (Manach et al., 2004).

However, considering the great number of flavonoids present in vegetables,

fruits and berries, an intake of 1 g per day could easily be acheived.

Page 86: Plant-Derived Substances and Cardiovascular Diseases

86

The Swedish National Food Administration recommends 500 g intake of

vegetables and fruits per capita and day, distributed over 5 times a day. This

recommendation is based on what is considered necessary for maintaining or

improving health and general well being, even if secondary metabolites from

plants i.e. flavonoids is not considered as essential nutritients. Essential (Latin,

unknown) nutrients are often described as necessary to maintain normal life

or/and normal health and must be administered to the body via food (Swedish

National Food Administration; WHO). So far, vitamins and minerals are the

only minor food components derived from plant biosynthesis classified as

necessary/essential for maintaining and/or improving health and general well

being. However, do flavonoids have to be administered to the human body via

food and necessary for health? When it comes to essential nutritients as vitamins

and minerals, there is a recommended average intake to minimize the risk of

inadequate intake and a lowest recommended intake to avoid the risk of

developing deficiency symptom. According to the result of the questionnaire

(Paper V), average intake of vegetables and fruits was 3.16 times a day for the

participants of this study. A population based questionnaire in Sweden, showed

an average intake of vegetables and fruits of 3.0 times a day; less than 10% ate

vegetables and fruits 5 times a day, and 25% ate vegetables and fruits 3-4 times

a day (Sepp et al., 2004). According to this, there is an inadequate intake of

vegetables and fruits. Long term inadequate intake increases the risk of

developing defiency symptom e.g. inadequate intake of flavonoids will increase

the risk of cardiovascular diseases. Appropriate recommended average intake

values to regulate the intake of flavonoids might give substance to their

importance and involvement in maintaining and/or improving health and general

well being. It might also prevent intake of too high concentrations of flavonoids

in e.g. supplements, as high intake may give rise to side effects as kidney and/or

liver toxicity.

Page 87: Plant-Derived Substances and Cardiovascular Diseases

87

Statins are used as lipid-lowering drugs and according to guidelines of the

National Cholesterol Education Program (NCEP) statins play an important role

in prevention of coronary heart disease, myocardial infarction, stroke and

peripheral artery disease. Studies also show that statins appear to have effect on

inflammation (Jones & Farmer, 2008), dementia (Orr, 2008), cancer (Xie &

Itkowitz, 2008), nuclear cataracts (Bariciak & Nichols, 1995) and hypertension

(Cernes et al., 2008). The fact that the statins i.e. pravastatin and simvastatin

increase ACE activity in vitro, as shown in this study is to be considered as a

negative effect of these drugs. After ten minutes incubation with simvastatin and

pravastatin, a significantly increased ACE activity was seen in HUVEC.

Simvastatin 10-8, 10-6 M *p<0.05 and 10-7 M **p<0.01, pravastatin 10-7 and 10-6

M *p<0.05 compared to the DMSO control (Table 1). The pharmacological

mechanism of the effect of statins on ACE activity is unknown. Intake of

flavonoid-rich food might also as prevention or complement lipid-lowering

drugs since studies show that flavonoids lower lipids (Hwang et al., 2003;

Ikizler et al., 2007; Kaliora et al., 2007; Aron et al., 2008; Boots et al., 2008) as

well as is shown in this study inhibit ACE activity, while lipid-lowering drugs as

this study shows increase ACE activity in human endothelial cells. Is it possible

to replace lipid-lowering drugs with vegetables, fruits and berries containing

high enough doses of flavonoids, at least in patients with low to modest elevated

blood lipids?

Page 88: Plant-Derived Substances and Cardiovascular Diseases

88

SUMMARY

Flavonoids interact with the cardiovascular system in several ways, by reducing

ROS (as shown in several studies), increasing NO concentration (as shown here

and by others) and also by inhibiting ACE activity (as shown here).

In plants, flavonoids have antioxidative effects and metal chelating properties

and the antioxidative effects affect NO. There are two major mechanisms

contributed to antioxidative effects; free radical scavenging and metal chelating.

The two active sites of ACE contain Zn2+ and ACE inhibitors are designed to

bind to the Zn2+ at the active sites. The effects seen in this study on ACE activity

could be due to the metal chelating properties of the flavonoids. Substances

affecting ACE activity, NO concentration and thereby the cardiovascular system

will affect the individual as a whole; ACE as a part of the endocrine system and

NO as a part of different systems in the human body. Physiologically as well as

psychologically the endocrine system, the immune system and the

cardiovascular system are linked together.

Green tea and black tea infusions, cacao extract, Bilberry extract 25E, liquorice

extract, Ginkgo biloba extract EGb 761 and Panax ginseng extract G115 showed

significant inhibition on ACE activity in vitro.

The most potent ACE inhibitors in vitro of the plant-derived substances tested

were the flavonoids, i.e. epicatechingallate, epigallocatechin.

epigallocatechingallate, quercetin, genistein and the tetraterpene β-carotene.

In vivo, Rooibos tea significantly inhibited ACE activity.

In vivo, Green tea significantly inhibited ACE activity for the genotype II and

ID.

In vivo, Rooibos tea significantly inhibited ACE activity for the genotype II.

Page 89: Plant-Derived Substances and Cardiovascular Diseases

89

Among the terpenes tested, the tetraterpene β-carotene was the only one

showing a strong ACE inhibiting effect, i.e β-carotene was the only non-

flavonoid with a strong inhibitory effect on ACE activity.

Bilobalide, ginsenoside Rb2, Rc, Rd and the statins, i.e. pravastatin and

simvastatin increased ACE activity, in vitro.

Green tea, black tea and Rooibos tea infusion, cacao extract, liqourice extract,

Ginkgo biloba extract EGb 761 and Panax ginseng extract G115 significantly

increased NO concentration in vitro. The most potent plant-derived substances

increasing NO concentration in vitro were epigallocatechingallate, procyanidin,

quercetin and sciadopitysin. Bilobalide significantly decreased NO

concentration in vitro.

It has been proposed that the positive effects of flavonoids are due to the

antioxidative effects and NO effect. However, a substance with antioxidative

effects, do not automatically have to prevent cardiovascular diseases, e.g. α-

tocopherol impliying an additional mechanism. Our results on ACE activity

show an additional important mechanism; extracts containing high amounts of

flavonoids and flavonoids as ACE inhibitors.

Page 90: Plant-Derived Substances and Cardiovascular Diseases

90

Table 1. A

CE

activity and N

O concentration in H

UV

EC

after incubation

with different con

centration of flavonoids, purine

s, terpenes, sterols,

salicin, biosynthesis precursor m

olecules, human steroids, blood lipid-low

ering drugs and plant extrac

t for ten minutes resp

ective 24 hours

compared to control (P

BS

or DM

SO

as stated). Statis

tics were calculated w

ith one wa

y AN

OV

A repeated m

easures, *p<

0.05, **p<0.01 and

***p<0.001. n=

5-7 from different individuals. a

ap<0.01 betw

een 0.1 and 1 m

g/ml. A

CE

activity is exp

ressed in Units (U

) and N

O concentration

in µM

.

Flavonoids

Epicatechin

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 11.9±1.6

11.2±2.2 9.4±1.9

7.9±2.1*

N

O concentration

4.3±1.8

5.2±1.7 6.2±1.9

5.9±2.2

Epicatechingallate

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 11.9±1.6

7.7±1.9* 3.8±1.7**

1.2±0.6**

N

O concentration

4.3±1.8

5.7±2.0 11.1±4.1

17.0±6.0

OO

H

OH

OH

OH

OH

O

O

OH

OH

OH

OH

OO

H

OH

OH

Page 91: Plant-Derived Substances and Cardiovascular Diseases

91

Epi

gallo

cate

chin

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

1mg/

ml

2mg/

ml

AC

E a

ctiv

ity

11.9

±1.6

7.

5±2.

0*

3.1±

0.8*

* 0*

*

N

O c

once

ntra

tion

4.

3±1.

8 6.

7±2.

3 21

.0±7

.8*

23.6

±6.5

*

Epi

gallo

cate

chin

galla

te

Con

cent

ratio

n

DM

SO

50

µg/

ml

0.1m

g/m

l 0.

5mg/

ml

1mg/

ml

2m

g/m

l A

CE

act

ivity

9.

1±1.

5 5.

5±1.

7 5.

3±2.

0 1.

1±0.

8***

0.

1±0

.1**

* 0*

**

NO

con

cent

ratio

n

4.3±

1.8

25.7

±3.7

***

31.8

±2.9

***

40.4

±3.0

***

Gre

en te

a ex

trac

t Jap

anes

e S

ench

a

Con

cent

ratio

n

PB

S

1:32

00

1:16

00

1:80

0 1:

400

1:20

0 A

CE

act

ivity

14

.9±3

.9

10.2

±3.2

7.

7±2.

0*

5.7±

1.8*

* 2

.2±0

.9**

0.

8±0.

6**

NO

con

cent

ratio

n

2.2±

0.9

2.5±

0.9

3.3±

1.0*

**

4.0±

1.1

***

5.0±

1.1*

**

7.1±

1.2*

**

Bla

ck te

a ex

trac

t Ind

ian

Ass

am B

.O.P

.

Con

cent

ratio

n

PB

S

1:32

00

1:16

00

1:80

0 1:

400

1:20

0 A

CE

act

ivity

14

.9±3

.9

11.8

±2.8

9.

4±2.

5 6.

4±1.

4*

4.7

±0.5

**

2.3±

0.6*

* N

O c

once

ntra

tion

2.

2±0.

9 2.

4±0.

9 2.

6±1.

0 3.

6±1.

0***

4.

8±1.

1***

7.

0±1.

1***

R

ooib

os te

a ex

trac

t

Con

cent

ratio

n

PB

S

1:32

00

1:16

00

1:80

0 1:

400

1:20

0 A

CE

act

ivity

14

.9±3

.9

15.4

±5.1

13

.3±3

.8

13.4

±3.7

11

.9±3

.2

8.5±

3.9

NO

con

cent

ratio

n

2.2±

0.9

2.3±

1.0

2.3±

1.0

2.5±

1.1

2.8±

1.0*

4.

5±1.

3***

O

OH

OH

OH

OH

OH

OH

O

O

OH

OH

OH

OH

OH

OO

H

OH

OH

Page 92: Plant-Derived Substances and Cardiovascular Diseases

92

Coffee extract S

antos Brasil

Concentration

P

BS

1:3200

1:1600 1:800

1:400 1:200

AC

E activity

19.2±3.7 18.4±4.2

17.0±3.8 15.0±3.2

9.5±2.6

4.0±1.7* N

O concentration

3.7±0.8

4.9±0.9 5.5±1.0

4.9±0.9 6.

1±0.9 7.0±0.6*

Procyanidin E

picatechin-epicatechin

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 4.8±0.5

7.8±0.9 6.8±0.5

4.7±0.7a

a

N

O concentration

2.0±0.5

2.0±0.5 3.7±1.0**

8.2±0.6**

Cacao extract

Concentration

P

BS

6.25µ

g/ml

12.5µg/m

l 25µ

g/ml

50µg/

ml

100µg/m

l A

CE

activity 30.8±4.1

18.1±2.9** 11.7±2.9**

5.9±2.2** 1.4±1.2**

0** N

O concentration

3.6±1.4

3.4±1.4 3.5±1.4

3.8±1.2 4.

3±1.2 5.9±1.2***

Genistein

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 7.7±1.3

4.7±1.2* 2.7±0.6***

3.6±1.1**

Quercetin

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 4.8±0.5

3.0±0.9* 1.7±0.4***

1.5±0.4***

N

O concentration

2.0±0.5

6.1±1.3* 9.1±0.6***

9.8±0.7***

OO

OH

OH

OH

OO

OH

OH

OH

OH

Page 93: Plant-Derived Substances and Cardiovascular Diseases

93

Sci

adop

itysi

n

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

4.

8±0.

5 5.

4±0.

7 5.

3±0.

6 6.

1±0.

6

N

O c

once

ntra

tion

2.

0±0.

5 5.

8±0.

6 19

.2±1

.6**

* 46

.1±2.5**

*

Cya

nidi

n

R1=

H

R2=

OH

Con

cent

ratio

n

DM

SO

0.

01m

g/m

l 0.

025m

g/m

l 0.

05m

g/m

l

A

CE

act

ivity

8.

4±2.

0 13

.5±3

.2

4.1±

1.4

2.6±

1.1

Del

phin

idin

R

1=O

H

R2=

OH

Con

cent

ratio

n

DM

SO

0.

01m

g/m

l 0.

025m

g/m

l 0.

05m

g/m

l

A

CE

act

ivity

8.

4±2.

0 8.

4±2.

4 5.

7±1.

7 2.

9±1.

3

M

alvi

din

R1=

OC

H 3 R

2=O

CH 3

Con

cent

ratio

n

DM

SO

0.

01m

g/m

l 0.

025m

g/m

l 0.

05m

g/m

l

A

CE

act

ivity

8.

4±2.

0 5.

0±1.

8 5.

3±2.

1 3.

3±1.

3

M

yrtil

lin c

hlor

id

Con

cent

ratio

n

DM

SO

0.

01m

g/m

l 0.

025m

g/m

l 0.

05m

g/m

l

A

CE

act

ivity

5.

5±1.

9 2.

5±0.

9 0.

9±0.

5*

0.9±

0.6*

B

ilber

ry e

xtra

ct 2

5E

Con

cent

ratio

n

PB

S

6.25

µg/

ml

12.5

µg/

ml

25µ

g/m

l 50

µg/

ml

100µ

g/m

l A

CE

act

ivity

25

.0±5

.4

5.7±

3.2*

**

2.7±

1.8*

**

1.0±

0.7**

* 0.

2±0.

2***

0*

**

O

O OH

OH

O O

OC

H3

OC

H3

OH

O

CH

3

O+

OH

OH

OH

R1

OH

R2

Page 94: Plant-Derived Substances and Cardiovascular Diseases

94

Liquorice extract

Concentration

P

BS

6.25µ

g/ml

12.5µg/m

l 25µ

g/ml

50µg/

ml

100µg/m

l A

CE

activity 29.4±4.0

13.4±4.5*** 11.3±3.5***

5.8±2.3*** 2.4±1.4***

0.7±0.5*** N

O concentration

2.8±1.5

3.4±1.6 4.2±1.6

5.9±1.7 9.

1±2.1*** 22.8±3.2***

Purines

Caffeine

R1=C

H3 R

2=C

H3 R

3=C

H3

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 11.9±1.6

15.8±3.3 18.5±3.1

18.8±3.4

N

O concentration

4.3±1.8

4.5±1.5 4.2±1.5

5.2±1.7*

T

heobromine R

1=H

R2=

CH3 R

3=C

H3

Concentration

D

MS

O

0.01mg/m

l 0.05m

g/ml

0.1mg/m

l

A

CE

activity 11.9±1.6

13.7±2.4 14.2±2.2

11.1±2.1

N

O concentration

4.3±1.8

4.4±1.6 4.5±1.6

8.6±3.7

T

heophylline R1=

CH

3 R2=

CH

3 R3=

H

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 11.9±1.6

11.3±1.6 12.6±1.7

9.7±2.2

N

O concentration

4.3±1.8

4.5±1.8 5.7±1.9

4.7±1.7

N

NN N

O

O

R1

R3

R2

Page 95: Plant-Derived Substances and Cardiovascular Diseases

95

Ter

pene

s

Gin

kgol

ide

A

R1=

OH

R

2=H

R

3=H

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

15

.4±1

.7

12.9

±2.5

13

.7±3

.6

12.8

±2.6

N

O c

once

ntra

tion

3.

2±0.

6 2.

9±0.

4 2.

8±0.

4 2.

9±0.

4

G

inkg

olid

e B

R

1=O

H

R2=

OH

R

3=

H

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

15

.4±1

.7

15.1

±3.4

15

.5±4

.4

14.4

±2.7

N

O c

once

ntra

tion

3.

2±0.

6 2.

8±0.

4 2.

7±0.

4 2.

7±0.

4

G

inkg

olid

e C

R

1=O

H

R2=

OH

R

3=

OH

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

15

.4±1

.7

17.0

±5.1

18

.1±5

.8

23.3

±6.9

N

O c

once

ntra

tion

3.

2±0.

6 3.

2±0.

5 3.

1±0.

5 3.

0±0.

5

Bilo

balid

e

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

15

.4±1

.7

13.4

±2.5

12

.3±2

.0

13.5

±2.3

N

O c

once

ntra

tion

3.

2±0.

6 2.

8±0.

4 2.

6±0.

4*

2.4±

0.4*

**

O O

OH

OO

H

R3

OH

R2

O

R1

OO

OH

OH

OO

OH

OO

OH

Page 96: Plant-Derived Substances and Cardiovascular Diseases

96

Ginkgo biloba extract E

Gb 761

Concentration

P

BS

0.1m

g/ml

0.5mg/m

l 1m

g/ml

5mg/m

l 1

0mg/m

l A

CE

activity 33.0±1.9

29.8±4.8 23.4±4.6

15.6±4.5***

2.7±1.2*** 0.6±0.3***

NO

concentration

3.5±0.6 3.1±0.4

3.4±0.4 4.2±0.5

11.0±1.3***

19.2±2.4***

Ginsenoside R

b1 R1=

Glc- 2G

lc R2=

Glc- 2G

lc- R3=

Glc- 6G

lc

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 24.7±6.3

25.0±6.6 25.9±5.8

27.4±5.1

G

insenoside Rb2 R1=

Glc- 2G

lc R2=

Glc- 2G

lc- R3=

Ara(p)- 6G

lc

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 24.7±6.3

29.8±6.2 33.3±8.5**

32.4±7.7*

Ginsenoside R

c R1=G

lc- 2Glc R

2=G

lc- 2Glc- R

3=A

ra(f)- 6G

lc-

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 24.7±6.3

26.4±6.3 32.4±8.0**

34.7±9.0***

Ginsenoside R

d R1=G

lc- 2Glc R

2=H

R3=

Glc-

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 24.7±6.3

33.2±8.6* 33.2±7.8*

35.4±10.2**

Ginsenoside R

e R1=H

R2=

Rha- 2G

lc-O- R

3=G

lc-

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 24.7±6.3

26.3±7.1 27.0±6.8

19.3±4.4*

R2O

R3

R1O

OH

Page 97: Plant-Derived Substances and Cardiovascular Diseases

97

Gin

seno

side

Rf

R1=

H

R2=

Glc

-2 Glc

-O

R3=

H

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

24

.7±6

.3

18.2

±3.4

18

.4±2

.8

20.3

±3.1

G

inse

nosi

de R

g1

R

1=H

R

2=G

lc-O

- R

3=G

lc-

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

24

.7±6

.3

23.3

±5.2

20

.9±6

.6

21.2

±5.6

P

anax

gin

seng

ext

ract

G11

5

Con

cent

ratio

n

PB

S

0.1m

g/m

l 0.

5mg/

ml

1mg/

ml

5mg/

ml

10m

g/m

l A

CE

act

ivity

16

.5±3

.6

16.8

±3.0

15

.9±3

.7

13.7

±2.7

8.

1±2.

8**

5.4±

2.0*

* N

O c

once

ntra

tion

6.

5±1.

4 6.

1±1.

3 5.

1±1.

1*

5.1±

0.8*

5.

0±1.

0*

4.8±

0.9*

β-c

arot

ene

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

7.

7±1.

2 4.

7±1.

3 3.

5±1.

4**

1.9±

0.8*

**

α-t

ocop

hero

l

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

7.

7±1.

3 7.

3±0.

9 7.

4±0.

8 8.

6±1.

3

H

H

O

CH

3

CH

3

OH

CH

3

Page 98: Plant-Derived Substances and Cardiovascular Diseases

98

Sterols

Cholesterol

Concentration

P

BS

10 -5M

10

-4M

10-3M

A

CE

activity 20.7±8.0

19.5±6.8 20.5±7.4

20.1±7.2

Lanosterol

Concentration

D

MS

O

10 -7M

10-6M

10

-5M

10-3M

AC

E activity

5.1±1.9 6.8±0.9

7.1±0.9 7.2±0.7

8.0±2.1**

Stigm

asterol

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 7.7±1.3

6.1±0.9 7.2±1.5

6.4±0.9

OH

HH

H

H

HO

H

OH

H

H

H

Page 99: Plant-Derived Substances and Cardiovascular Diseases

99

Phe

nol

Sal

icin

Con

cent

ratio

n

PB

S

0.1m

g/m

l 0.

5mg/

ml

1mg/

ml

AC

E a

ctiv

ity

28.1

±2.9

29

.4±2

.9

26.8

±3.0

27

.4±3

.0

NO

con

cent

ratio

n

4.5±

0.3

4.4±

0.3

4.7±

0.1

4.5±

0.3

Pre

curs

or m

olec

ules

Cho

rism

ic a

cid

Con

cent

ratio

n

PB

S

10-6M

10

-5M

10

-4M

A

CE

act

ivity

26

.2±5

.9

16.7

±4.6

17

.3±4

.5

18.0

±4.6

Mal

onic

aci

d

Con

cent

ratio

n

PB

S

10-6M

10

-5M

10

-4M

A

CE

act

ivity

24

.1±6

.8

19.0

±5.7

22

.2±7

.0

20.6

±7.1

O

OHO

OH

OH

OH

O

OH

CO

OH

CO

OH

CH

2

OH

OH

OO

Page 100: Plant-Derived Substances and Cardiovascular Diseases

10

0

Mevalonic acid

Concentration

P

BS

10 -6M

10

-5M

10-4M

A

CE

activity 29.6±5.3

29.0±6.2 27.2±6.7

28.1±6.9

N

O concentration

4.5±0.3

4.9±0.2 4.8±0.2

5.2±0.2

Shikim

ic acid

Concentration

P

BS

10 -6M

10

-5M

10-4M

A

CE

activity 26.3±5.8

23.4±6.1 23.8±6.2

22.6±5.3

N

O concentration

4.4±0.3

4.4±0.2 5.0±0.3

4.9±0.5

Squalene

Concentration

D

MS

O

10 -7M

10-6M

10

-5M

10-3M

AC

E activity

5.1±1.9 7.5±3.4

7.2±2.4 6.4±1.9

7.4±1.7

H

uman steroids

Aldosterone

Concentration

D

MS

O

0.1mg/m

l 0.5m

g/ml

1mg/m

l

A

CE

activity 7.7±1.3

7.1±1.3 9.0±1.9

9.9±2.3

OH

OH

OO

H

OH

O

OH

OH

OH

O

H

H

OO

OH

Page 101: Plant-Derived Substances and Cardiovascular Diseases

10

1

Oes

trad

iol

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

7.

7±1.

3 6.

3±1.

0 6.

1±1.

2 6.

5±1.

6

Tes

tost

eron

e

Con

cent

ratio

n

DM

SO

0.

1mg/

ml

0.5m

g/m

l 1m

g/m

l

A

CE

act

ivity

7.

7±1.

3 8.

5±2.

9 8.

1±2.

0 6.

7±2.

2

B

lood

lipid

-low

erin

g dr

ugs

Pra

vast

atin

Con

cent

ratio

n

PB

S

10-8M

10

-7M

10

-6M

A

CE

act

ivity

32

.1±9

.0

34.6

±9.8

35

.7±9

.6*

35.6

±9.3

*

Sim

vast

atin

Con

cent

ratio

n

DM

SO

10-8

M

10-7

M

10-6

M

AC

E a

ctiv

ity

1.6±

1.0

7.9±

2.7*

9.

5±3.

4**

7.2±

2.0*

OH

H

OH

HH

O

H

OH

HH

CH

3

O

CO

OH

CH

3

O

HH

3C

HO

OH

CH

3

CH

3

O

CH

3

CH

3C

H3

OO

O

HHO

Page 102: Plant-Derived Substances and Cardiovascular Diseases

102

TACK……..

Forska…Forska???....Tänk om jag skall forska..????

Det var många funderingar och känslor som dök upp i mitt huvud.

Jo, jag vill, men??…kan jag???? Typiskt mig, jag vill (jag vill det mesta), men

vad är det jag skall göra och kan jag ????? Jag förstod att det måste handla om

att tänka, systematik och vetenskap. För mig är det jobbigt att tänka, men jag är

bra på att känna, och systematik… vad är det??

Jag började med att slå upp ordet forskning, och läste: forskning är en aktiv (jag

är aktiv, så det klarar jag) och systematisk process (här får jag problem), jag

läser vidare i ordboken, forskningen drivs av forskarens nyfikenhet (nyfikenhet

har jag inbyggt i generna, det ligger i släkten, så det klarar jag) och intresse ???

(i ordboken står det: intresse t.ex. fågelskådning. Fågelskådning är roligt och jag

har fågelskådat i många år, så då klarar jag intressedelen också).

Mitt liv som forskare kunde börja!

Det behövs visst en handledare också…….

För mig var det självklart, det måste bli mannen som erbjudit mig att bli

forskare, Rolf Andersson. Jag fick, fast det var nog snarare så att vi kom överens

om att han skulle bli min handledare, Rolf Andersson alltså………..

Professor Rolf G.G. Andersson….. undrar vad G.G. betyder? Great Guru

kanske, (Guru är ”den som skingrar mörkret”). Det är en verkligt bra sak när

handledaren tror att det skall gå bra, för det händer att självförtroendet sviktar

ibland, det hände i alla fall mig. Handledare är en person som tar dig i handen

och leder dig längs vägen fram till målet. Fast nu är det ju så att jag har alltid

gått mina egna vägar, och lycka på jorden, du tillät mig att bana min egen väg i

Page 103: Plant-Derived Substances and Cardiovascular Diseases

103

forskningssnåret. Jag tror, om jag får bli lite djup, att det är detta som jag är mest

tacksam över, att jag fick bana min egen väg, att det har varit min forskning. Jag

är också tacksam för din entusiasm och din alltid så positiva inställning till min

forskning.

Anita……när jag arbetade med min ”dricka-te studie” kom jag till jobbet tidigt

på morgonen, men hur tidigt jag än kom så fanns Anita där. Det är bra när någon

alltid finns där, det är tryggt. Anita har många bra egenskaper och en är hennes

goda kakor, onsdags-kakorna har varit en mitt-i-veckan ljuspunkt i den mörka

forskningstunneln. ”De som vandrar i mörker skall se ett stort ljus”, jag såg

Anita.

Tack till co-forskare, co-kollegor och alla som har visat intresse för min

forskning och bidragit med en hjälpande hand när jag har behövt och bett om

hjäälp!

Familjen Ranidae, Temporaria, Arvalis och Dalmatina. Era alltid glada hopp och

vackra sång, ni är mina prinsar.

Fresse…… nu undrar ni förståss vad det finns för anledning att tacka en katt?

Men alla ni som efter en lång dag och en ännu längre vecka fullproppad av

forskning och för att inte tala om allt tänkande, har fått vila ert trötta huvud på

en katt ni förstår precis varför jag tackar Fresse. Med en katt som huvudkudde

kan Tempur och allt vad ni nu heter slänga er i väggen. Nu sitter Fresse i

katthimlen och jag saknar min mjuka, sköna och trogne huvudkudde.

Karin, min lillasyster. I ordboken, under ordet lillasyster står det: ”Lillasyster är

ett svenskt rockband som bildades 2006, bandets debutalbum heter Hjärndöd

musik för en Hjärndöd generation”. Det är något som inte stämmer; när Karin

spelar Julkväll på Hawaii på orgeln låter det visserligen som hjärndöd musik för

en hjärndöd generation, men Lillasyster, rockbandet alltså bildades 2006 och jag

är säker på att lillasyster, Karin alltså är äldre än 3 år. Det är bra att du har lagt

Page 104: Plant-Derived Substances and Cardiovascular Diseases

104

orgeln på hyllan. Karin har också intresse, fågelskådning alltså och vi utövar

intresse tillsammans. Det har varit många roliga och vilsamma dagar med fika,

fika och fågelskådning för att inte tala om alla steg vi har gått (ca 100 miljoner).

Vi har nött ut många par vandringskängor och trampat ner i många hål i marken

(Karin i alla fall). Under våra vandringar har havsörn, skäggmes, majvivor,

naturguider, ACE och kväveoxid blandats i en salig röra.

Det har varit roligt!

I.P.

Page 105: Plant-Derived Substances and Cardiovascular Diseases

105

REFERENCES

Ackermann, A., Fernández-Alfonso, M.S., Sánchez de Rojas, R., Ortega, T., Paul, M. & González, C. Modulation of angiotensin-converting enzyme by nitric oxide. Br J Pharmacol 1998; 124: 291-298.

Ahlemeyer, B. & Krieglstein, J. (2003). Neuroprotective effects of Ginkgo biloba extract. Cell Mol Life Sci, 60, 1779-1792.

Ahmed, A., Li, F.X., Shams, M., Gregory, J., Rollason, T., Barnes, N.M. & Newton, J.R. (1995). Localization of the angiotensin II and its receptor subtype expression in human endometrium and identification of a novel high-affinity angiotensin II binding site. J Clin Invest, 96, 848-857. 848-857.

Alhenc-Gelas, F., Richard, J., Courbon, D., Warnet, J.M. & Corvol, P. (1991). Distribution of plasma angiotensin I-converting enzyme levels in healthy men: relationship to environmental and hormonal parameters. J Lab Clin Med, 117, 33-39.

Alhenc-Gelas, F., Weare, J.A., Johnson, R.L. & Erdös, E.G. (1983). Measurement of human converting enzyme level by direct radioimmunoassay. J Lab Clin Med, 101, 83-96.

Allen, R.R., Carson, L., Kwik-Uribe, C., Evans, E.M. & Erdman Jr, J.W. (2008). Daily consumption of a dark chocolate containing flavanols and added sterol esters affect cardiovascular risk factors in a normotensive population with elevated cholesterol. Nutr, 138, 725-731.

Andersen, Ö.M. & Markham, K.R. (2006). Flavonoids, Chemistry, Biochemistry and applications. Taylor and Francis Group, Boca Raton FL. USA.

André, P., Schott, C., Nehlig, H. & Stoclet, J.-C. (1990). Aortic smooth muscle cells are able to convert angiotensin I to angiotensin II. Biochem Biophys Res Commun, 173, 1137-1142.

Anter, E., Thomas, S.R., Schulz, E., Shapiro, O.M., Vita, J.A. & Keaney, J.F.Jr. (2004). Activation of endothelial nitric-oxide synthase by the p38 MAPK in response to black tea polyphenols. J Biol Chem, 279, 46637-46643.

Ardaillou, R. (1999). Angiotensin II receptors. J Am Soc Nephrol, 10, S30-S39.

Arnal, J.F., Battle, T., Rasetti, C., Challah, M., Costerousse, O., Vicaut, E., Michel, J.B. & Alhenc-Gelas, F. (1994). ACE in three tunicae of rat aorta: expression in smooth muscle and effect of renovascular hypertension. Am J Physiol, 267, H1777-H1784.

Aron, P.M. & Kennedy, J.A. (2008). Flavan-3-ols: Nature, occurrence and biological activity. Mol Nutr Food Res, 52, 79-104.

Aulakh, G.K., Sodhi, R.K. & Singh, M. (2007). An update on non-peptide angiotensin receptor antagonists and related RAAS modulaters. Life Sciences, 81, 615-639.

Authier, F., Posner, B.I. & Bergeron, J.J. (1996). Insulin-degrading enzyme. Clin Invest Med, 19, 149-160.

Page 106: Plant-Derived Substances and Cardiovascular Diseases

106

Aviram, M. & Fuhrman, B. (1998). Polyphenolic flavonoids inhibit macrophage-mediated oxidation of LDL and attenuate atherogenesis, Atherosclerosis, 137, 45-50S

Babu, P.V. & Liu, D. (2008). Green tea catechins and cardiovascular health: an update. Curr Med Chem, 15, 1840-1850.

Bariciak, M.D. & Nichols, B.D. (1995). Cataracts in systemic diseases and syndromes. Curr Opin Ophthalmol, 6, 3-8.

Barton, M. & Yanagisawa, M. (2008). Endothelin: 20 years from discovery to therapy. Can J Physiol Pharmacol, 86, 485-498.

Bastianetto, S. & Quirion, R. (2002). Natural extracts as possible protective agents of brain aging. Neurobiol Aging, 23, 891-897.

Basu, A. & Lucas, E.A. (2007). Mechanisms and effects of green tea on cardiovascular health. Nutr Rev, 65, 361-375.

Battle, T., Arnal, J.F., Challah, M. & Michel, J.B. (1994). Selective isolation of rat aortic wall layers and their cell types in culture-application to converting enzyme activity measurement. Tissue Cell, 26, 943-955.

Baudin, B., Berard, M., Carrier, J.L., Legrand, Y. & Drouet, L. (1997). Vascular origin determines angiotensin I-converting enzyme expression in endothelial cells. Endothelium, 5, 73-84.

Bazzano, L.A., He, J., Ogden, L.G., Loria, C.M., Vupputuri, S., Myers, L. & Whelton, P.K. (2002). Fruit and vegetable intake and risk of cardiovascular disease in US adults: the first national health and nutrition examination survey epidemiologic follow-up study. Am J Clin Nutr, 76, 93-99.

Belinky, P.A., Aviram, M., Fuhrman, B., Rosenblat, M. & Vaya, J. (1998). The antioxidative effects of the isoflavan glabridin on endogenous constituents of LDL during its oxidation. Atherosc,137, 49-61.

Benson, S.C., Iguchi, R., Ho, C.I., Yamamoto, K. & Kurtz, T.W. (2008). Inhibition of cardiovascular cell proliferation by angiotensin receptor blockers: are all molecules the same? J Hypertens, 26, 973-980.

Beohar, N., Damaraju, S., Prather, A., Yu, Q.T., Raizner, A., Kleiman, N.S., Roberts, R. & Marian, A.J. (1995). Angiotensin-I converting enzyme genotype DD is a risk factor for coronary artery disease. J Investig Med, 43, 275-280.

Berecek, K. & Zhang, L. (1995). Biochemistry and cell biology of angiotensin-converting enzyme and converting enzyme inhibitors. Adv Exp Med Biol, 377, 141-168.

Bernstein, K.E., Martin, B.M., Edwards, A.S. & Bernstein, E.A. (1989). Mouse angiotensin-converting enzyme is a protein composed of two homologous domains. J Biol Chem, 264, 11945-11951.

Bickerton, R.K. & Buckley, J.P. (1961). Evidence for a central mechanism in angiotensin induced hypertension. Proc Soc Exp Biol Med, 106, 834-836.

Page 107: Plant-Derived Substances and Cardiovascular Diseases

107

Birkenhäger, W.H & Staessen, J.A. (2006). Progress in cardiovascular diseases: cognitive function in essential hypertension. Prog Cardiovasc Dis, 49, 1-10.

Birks, J. & Grimley Evans, J. (2008). Ginkgo biloba for cognitive impairment and dementia (Review). The Cochrane Library, Issue 4.

Birt, D.F., Hendrich, S. & Wang, W. (2001). Dierary agents in cancer prevention: flavonoids and isoflavonoids. Pharmacol Therapeutics, 90, 157-177.

Bjelakovic, G., Nikolova, D., Gluud, L.I., Simonetti, R.G. & Gluud, C. (2008). Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. The Cochrane Library, Issue 4.

Bodineau, L., Frugière, A., Marc, Y., Claperon, C & Llorens-Cortes, C. (2008). Aminopeptidase A inhibitors as centrally acting antihypertensive agents. Heart Fail Rev, 13, 311-319.

Bondy, B. (2007). Common genetic factors for depression and cardiovascular disease. Dialogues Clin Neurosci, 9, 19-28.

Bonnardeaux, A., Davies, E., Jeunemaitre, X., Féry, I., Charru, A., Clauser, E., Tiret, L., Cambien, F., Corvol, P. & Soubrier,F. (1994). Angiotensin II type 1 receptor gene polymorphisms in human essential hypertension. Hypertension, 24, 63-69.

Boots, A.W., Haenen, G.R.M.M. & Bast, A. (2008). Health effects of quercetin: from antioxidant to nutraceutical. Eur J Pharmacol, 585, 325-337.

Bramati, L., Minoggio, M., Gardana, C., Simonetti, P., Mauri, P. & Pietta, P. (2002). Quantitative characterization of flavonoids by HPLC-UV and determination of the total antioxidant activity. J Agr food Chem, 50, 5513-5519.

Bramati, L., Aquiland, F. & Pietta, P. (2003). Unfermented Rooibos tea. Quantitative characterization of flavonoids by HPLC-UV and determination of the total antioxidant activity. J Agr Food Chem, 51, 7472-7474.

Bramucci, M., Miano, A., Quassinti, L., Maccari, E., Murri, O. & Amici, D. (1999). Presence and comparison of angiotensin converting enzyme in commercial cell culture sera. Biochem Mol Biol Int, 47, 107-115.

Braune, A., Gütschow, M., Engst, W. & Blaut, M. (2001) Degradation of quercetin and luteolin by Eubacterium ramulus. Applied Environment Microbiol, 67, 5558-5567.

Bredt, D.S. & Snyder, S.H. (1990). Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci USA, 87, 682-685.

Brown, N.J. (2008). Aldosterone and vascular inflammation. Hypertens, 51, 161-167.

Bruckdorfer, K.R. (2008). Antioxidants and CVD. Proceedings of the Nutr Soc, 67, 214-222.

Brunner, H.R. & Gavras, H. (1980). Is the renin system necessary? Am J Med, 69, 739-745.

Brunner, H.R., Laragh, J.H., Baer, L., Newton, M.A., Goodwin, F.T., Krakoff, L.R., Barnd, R.H. & Bühler, F.R. (1972). Essential hypertension: renin and aldosterone, heart attack and stroke. N Eng J Med, 286, 441-449.

Page 108: Plant-Derived Substances and Cardiovascular Diseases

108

Buijsse, B., Feskens, E.J.M., Kwape, L., Kok, F.J. & Kronhout, D. (2008). Both α- and β-carotene, but not tocopherols and vitamin C, are inversely related to 15-year cardiovascular mortality in Dutch elderly men. J Nutr, 138, 344-350.

Bumpus, F.M., Catt, K.J., Chiu, A.T., DeGasparo, M., Goodfriend, T., Husain, A., Peach, M.J., Taylor, D.G. & Timmermans, P.B.M.W.M. (1991). Nomenclature for angiotensin receptors. A report of the nomenclature committee of the council for high blood pressure research. Hypertension, 17, 270-271.

Cambien, F., Alhenc-Gelas, F., Herbeth, B., Andre, J.L., Rakotovao, R., Gonzales, M.F., Allegrini, J. & Bloch, C. (1988). Familial resemblance of plasma angiotensin-converting enzyme level: The Nancy study. Am J Hum Genet, 43, 774-780.

Cambien, F., Poirier, O., Lecerf, L., Evans, A., Cambou, J.P., Arveiler, D., Luc, G., Bard, J.M., Bara, L., Ricard, S et al. (1992). Deletion polymorphism in the gene for angiotensin-converting enzyme is a potent risk factor for myocardial infarction. Nature, 15, 641-644.

Campbell, D.J. (1987). Circulating and tissue angiotensin systems. J Clin Invest, 79, 1-6.

Canter, P.H. & Ernst, E. (2004). Anthocyanidines of Vaccinium myrtillus (bilberry) for night vision – a systematic review of placebo-controlled trials. Survey Ophthalmol, 49, 38-50.

Carlson, S., Peng, N., Prasain, J.K. & Wyss, J.M. (2008). Effects of botanical dietary supplements on cardiovascular, cognitive, and metabolic function in males and females. Gender Med, 5, S76-S90.

Cermak, R. (2008). Effect of dietary flavonoids on pathways involved in drug metabolism. Expert Opin Drug Metab Toxicol, 4, 17-35.

Cernes, R., Zimlichman, R. & Shargorodsky, M. (2008). Arterial elasticity in cardiovascular diseases: focus on hypertension, metabolic syndrome and diabetes. Adv Cardiol, 45, 65-81.

Chai, S.Y., Zhuo,J & Mendelsohn, F.A.O. (1993). Localization of components of the renin-angiotensin system and site of action of inhibitors. Arzneim-Forsch, 43, 214-221.

Chaki, S. & Inagami, T. (1992). Identification and characterization of a new binding site for angiotensin II in mouse neuroblastoma neuro-2A cells. Biochem Biophys Res Commun, 182, 388-394.

Chang, W-T., Shao, Z-H., Yin, J-J., Mehendale, S., Wang, C-Z., Qin, Y., Li, J., Chen, W-J., Chien, C-T., Becker, L.B. Hoek, T.L.V. & Yuan, C-S. (2007). Comparative effects of flavonoids on oxidant scavenging and ischemia-reperfusion injury in cardiomyocytes. Eur J Pharmacol, 566, 58-66.

Chen, X. (1996). Cardiovascular protection by ginsenosides and their nitric oxide releasing action. Clin Exp Pharmacol Physiol, 23, 728-732.

Chen, C.M., Zhao, W., Yang, Z., Wu, Y. & Kong, L. (2008). The role of dietary factors in chronic disease control in China. Obesity Rev, 9, 100-103.

Chen, Z., Liang, J., Zhang, C. & Rodrigues, C.J.Jr. (2006). Epicatechin and catechin may prevent coffee berry disease by inhibition of appressorial melanization of Colletotrichum kahawae. Biotechnol Lett, 28, 1637-1640.

Page 109: Plant-Derived Substances and Cardiovascular Diseases

109

Cheon, K.T., Choi, K.H., Lee, H.B., Park, S.K., Rhee, Y.K. & Lee, Y.C. (2000). Gene polymorphisms of endothelial nitric oxide synthase and angiotensin-converting enzyme in patients with lung cancer. Lung, 178, 351-360.

Chiu, A., Herblin, W.F., McCall, D.E., Ardecky, R.J., Carini, D.J., Duncia, J.V., Pease, L.J., Wong, P.C., Wexler, R.R., Johnson, A.L. & Timmermans, P.B. (1989). Indentifications of angiotensin II receptors subtypes. Biochem Biophys Res Commun, 165, 196-203.

Choudhary, R., Palm-Leis, A., Scott III, R.C., Guleria, R.S., Rachut, E., Baker, K.M. & Pan, J. (2008). All-trans retinoic acid prevents development of cardiac remodeling in aortic banded rats inhibiting the renin-angiotensin system. Am J Physiol Heart Circ Physiol, 294, H633-H644.

Clarkson, T.B. (2002). Soy, soy phytoestrogens and cardiovascular disease. J Nutr, 132, 566S-569S.

Clermont, A., Bursell, S-E. & Feener, E.P. (2006). Role of the angiotensin II type 1 receptor in the pathogenesis of diabetic retinopathy: effects of blood pressure control and beyond. J Hypertens, 24, S73-S80.

Coon, J.T. & Ernst, E. (2002). Panax ginseng: a systematic review of adverse effects and drug interactions. Drug Saf, 25, 323-344.

Corvol, P., Michaud, A., Soubrier, F. & Williams, T.A. (1995). Recent advances in knowledge of the structure and function of the angiotensin I converting enzyme. J Hypertens, 13, S3-S10.

Crawford, N.M. (2006). Mechanisms for nitric oxide synthesis in plants. J Exp Botany, 57, 471-478.

DalBó, S., Goulart, S., Horst, H., Pizzolatti, M.G. & Ribeiro-do-Valle, R.M. (2008). Activation of endothelial nitric oxide synthase by proanthocyanidin-rich fraction from Croton celtidifolius (Euphorbiaceae): Involvement of extracellular calcium influx in rat thoracic aorta. J Pharmacol Sci, 107, 181-189.

Danser, A.H.J. & Deinum, J. (2005). Renin, prorenin and the putative (pro)renin receptor. Hypertens, 46, 1069-1076.

Danser, A.H.J., Derkx, F.H.M., Admiraal, P.J.J., Deinum, J., deJong, P.T.V.M. & Schalekamp, M.A.D.H. (1994). Angiotensin levels in the eye. Invest Ophthalmol Vis Sci, 35, 1008-1018.

Davidson, K.W. 2008. Emotional predictors and behavioral triggers of acute coronary syndrome. Cleve Clin J Med, 75, S15-S19.

Davis, J.O. (1973). The control of renin release. Am J Med, 55, 333-350.

DeGasparo, M. (2002). Angiotensin II and Nitric Oxide Interaction. Heart Failure Rev, 7, 347-358.

DeGasparo, M., Catt, K.J., Inagami, T., Wright, J.W. & Unger, T.H. (2000). International Union of Pharmacology. XXIII. The angiotensin II receptors. Pharmacol Rev, 52, 415-472.

Page 110: Plant-Derived Substances and Cardiovascular Diseases

110

DeKosky, S.T., Fitzpatrick, A., Ives, D.G., Saxton, J., Williamson, J., Lopez, O.L., Burke, G., Fried, L., Kuller, L.H., Robbins, J., Tracy, R., Woolard, N., Dunn, L., Kronmal, R., Nahin, R. & Furberg, C.; GEMS Investigators, (2006). The Ginkgo evaluation of memory (GEM) study: Design and baseline data of a randomized trial of Ginkgo biloba extract in prevention of dementia. Cont Clin Trials, 27, 238-253.

Detre, Z., Jellinek, H., Misulin, M. & Robert, A.M. (1986). Studies on vascular permeability in hypertension: action of anthocyanosides. Clin Physiol & Biochem, 4, 143-149.

Devaraj, S., Tang, R., Adams-Huet, B., Harris, A., Seenivasan, T de Lemos, J.A. & Jialal, I. (2007). Effect of high-dose α-tocopherol supplementation on biomarkers of oxidative stress and inflammation and carotid atherosclerosis in patients with coronary artery disease. Am J Clin Nutr, 86, 1392-1398.

Dhingra, D., Parle, M. & Kulkarni, S.K. (2004). Memory enhancing activity of Glycyrrhiza glabra in mice. J Ethnopharmacol, 91, 361-365.

Dormán, G., Kocsis-Szommer, K., Spadoni, C. & Ferdinandy, P. (2007). MMP inhibitors in cardiac diseases: an update. Recent Patents Cardiovasc Drug Discov, 2, 186-194.

Doughan, A.K., Harrison, D.G. & Dikalov, S.I. (2008). Molecular mechanisms of angiotensin II-mediated mitochondrial dysfunction, linking mitochondrial oxidative damage and vascular endothelial dysfunction. Cisc Res, 102, 488-496.

Dragsted, L.O. (2003). Antioxidant actions of polyphenols in humans. Int J Vit Nutr Res, 73, 112-119.

Ehlers, M.R.W. & Riordan, J.F. (1991). Angiotensin-converting enzyme: zinc- and inhibitor-binding stoichiometries of the somatic and testis isoenzymes. Biochemistry, 30, 7118-7126.

Ek, M., Söderdahl, T., Küppers-Munther, B., Edsbagge, J., Andersson, T.B., Björquist, P., Cotgreave, I., Jernström, B., Ingelman-Sundberg, M. & Johansson, I. (2007). Expression of drug metabolizing enzymes in hepatocyte-like cells derived from human embryonic stem cell. Biochem Pharmacol, 74, 496-503.

Engeli, S., Gorzelniak, K., Kreutz, R., Runkel, N., Distler, A & Sharma, A.M. (1999). Co-expression of renin-angiotensin system genes in human adipose tissue. J Hypertens, 17, 555-560.

Erdös, E.G. (1990). Angiotensin I converting enzyme and the changes in our concepts through the years. Hypertension, 16, 363-370.

Erdös, E.G. & Skidgel, R.A. (1986). The unusual substrate specificity and the distribution of human angiotensin I converting enzyme. Hypertension, 8, I-34-I-37.

Erlund, I., Koli, R., Alfthan, G., Marniemi, J., Puukka, P., Mustonen, P., Mattila, P. & Jula, A. (2008). Favorable effects of berry consumption on platelet function, blood pressure, and HDL cholesterol. Am J Clin Nutr, 87, 323-331.

Faridi, Z., Njike, V.Y., Dutta, S, Ali, A. & Katz, D.L. (2008). Acute dark chocolate and cocoa ingestion and endothelial function: a randomized controlled crossover trial. Am J Clin Nutr, 88, 58-63.

Page 111: Plant-Derived Substances and Cardiovascular Diseases

111

Favot, L., Martin, S., Keravis, T., Andriantsitohaina, R. & Lugnier, C. (2003). Involvment od cyclin-dependent pathway in the inhibition effect of delphinidin on angiogenesis. Cardivasc Res, 59, 479-487.

Ferrario, C.M. (1990). The renin-angiotensin system: importance in physiology and pathology. J Cardiovasc Pharmacol, 15, S1-S5.

Ferrario, C.M., Brosnihan, K.B., Diz, D.I., Jaiswal, N., Khosla, M.C., Milsted, A. & Tallant, E.A. (1991). Angiotensin-(1-7): a new hormone of the angiotensin system. Hypertens, 18, 126-133.

Ferrario, C.M. & Iyer, S.N. (1998). Angiotensin-(1-7): a bioactive fragment of the rennin-angiotensin system. Regul Pept, 78, 13-18.

Fitzsimons, J.T. (1998). Angiotensin, thirst, and sodium appetite. Physiol Rev, 78, 583-686.

Fogari, R. & Zoppi, A. (2006). Antihypertensive drugs and fibrinolytic function. Am J Hypertens, 19, 1293-1299.

Friedl, R., Moeslinger, T., Kopp, B. & Spieckermann, P.G. (2001). Stimulation of nitric oxide synthesis by the aqueous extract of Panax ginseng root in RAW 264.7 cells. Br J Pharmacol, 134, 1663-1670.

Fuhrman, B., Volkova, N., Kaplan, M., Presser, D., Attias, J., Hayek, T. & Avriram, M. (2002). Antiatherosclerotic effects of licorice extract supplementation on hypercholesterolemic patients: Increased resistance of LDL to atherogenic modifications, reduced plasma lipid levels, and decreased systolic blood pressure. Nutrition, 18, 268-273.

Furchgott, R.F. & Zawadski, J.V. (1980). The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature, 288, 373-376.

Ganten, D., Minnich, J.L., Granger, P., Hayduk, K., Brecht, H.M., Barbeau, A., Boucher, R. & Genest, J. (1971). Angiotensin-forming enzyme in brain tissue. Science, 173, 64-65.

Gard, K.S. (2002). The role of angiotensin II in cognition and behavior. Eur J Pharmacol, 438, 1-14.

Gard, P.R. (2004). Angiotensin as a target for the treatment of Alzheimer´s disease, anxiety and depression. Expert Opin Ther Targets, 8, 7-14.

Garg, U.C. & Hassid, A. (1989). Nitric oxide-generating vasodilators and 8-bromo-cyclic guanosine monophosphate inhibit mitogenesis and proliferation of cultured rat vascular smooth muscle cells. J Clin Invest, 83, 1774-1777.

George, S.E., Ramalakshmi, K. & Mohan Rao, L.J. (2008). A perception on health benefits of coffee. Crit Rev Food Sci Nutr, 48, 464-486.

Gillis, C.N. (1997). Panax ginseng pharmacology: a nitric oxide link? Biocem Pharmacol, 54, 1-8.

Gould, P.R. (2002). Nature´s Swiss army knife: the diverse protective roles of anthocyanins in leaves. J Biomed Biotechnol, 5, 314-320.

Page 112: Plant-Derived Substances and Cardiovascular Diseases

112

Grassi, D., Aggio, A., Onori, L., Croce, G., Tiberti, S., Ferri, C., Ferri, L. & Desideri, G. (2008b). Tea, flavonoids, and nitric oxide-mediated vascular reactivity. J Nutr, 138, 1554S-1560S.

Grassi, D., Desideri, G., Necozione, S., Lippi, C., Casale, R., Properzi, G., Blumberg, J.B. & Ferri, C. (2008a). Blood pressure is reduced and insulin sensitivity increased in glucose-intolerant, hypertensive subjects after 15 days of consuming high-polyphenol dark chocolate. J Nutr, 138, 1671-1676.

Grossman, E. (2008). Does increased oxidative stress cause hypertension? Diabetes Care, 31, S185-S189.

Gulati, K. (1996). Angiotensin II-receptor subtypes characterization and pathophysiological implications. Indian J Exp Biol, 34(2), 91-97.

Hakim, I.A., Alsaif, M.A., Alduwaihy, M., Al-Rubeaan, K., Al-Nuaim, A.R. & Al-Attas, O.S. (2003). Tea comsumption and the prevalence of coronary heart disease in Saudi adults: results from a Saudi national study. Prev Med, 36, 64-70.

Hall, J.M. (1992). Bradykinin receptors: pharmacological properties and biological roles. Pharmacol Ther, 56, 131-190.

Hall, W.L., Formanuik, N.L., Harnpanich, D., Cheung, M., Talbot, D., Chowienczyk, P.J. & Sanders, T.A.B. (2008). A meal enriched with soy isoflavones increases nitric oxide-mediated vasodilation in healthy postmenopausal woman. J Nutr, 138, 1288-1292.

Halliwell, B. & Gutteridge, J.M.C. (2007). Free Radicals in Biology and Medicine. Oxford University Press Inc., N.Y.

Halliwell, B. & Whiteman, M. (2004). Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol, 142, 231-255.

Hammer, F. & Stewart, P.M. (2006). Cortisol metabolism in hypertension. Endocrin Hypertens, 20, 337-353.

Han, K.H., Choe, S.C., Kim, H.S., Sohn, D.W., Nam, K.Y., Oh, B.H., Lee, M.M., Park, Y.B., Choi, Y.S., Seo, J.D. & Lee, Y.W. (1998). Effect of red ginseng on blood pressure in patients with essentioal hypertension and white coat hypertension. Am J Chinese Med, 26, 199-209.

Harborne, J.B. & Williams, C.A. (2000). Advances in flavonoid research since 1992. Phytochemistry, 55, 481-504.

Harris, R.Z., Jang, G.R. & Tsunoda, S. (2003). Dietary effects on drug metabolism and transport. Clin Pharmacokinet, 42, 1071-1088.

Havsteen, B. (2002). The biochemistry and medical significance of the flavonoids. Pharmacol & Ther, 96, 67-202.

He, F., Guo, R., Wu, S.L. & Li, M. (2007). Protective effects of ginsenoside Rb1 on human umbilical vein endothelial cells in vitro. J Cardiovasc Pharmacol, 50, 314-320.

Page 113: Plant-Derived Substances and Cardiovascular Diseases

113

Hertog, M.G., Feskens, E.J., Hollman, P.C., Katan, M.B. & Kromhout, D. (1993). Dietary antioxidant flavonoids and risk of coronary heart disease: the Zutphen elderly study. Lancet, 23;342(8878), 1007-1011.

Hertog, M.G., Kromhout, D., Aravanis, C., Blackburn, H., Buzina, R., Fidanza, F., Giampaoli, S., Jansen, A., Menotti, A., Nedeljkovic, S., et al. (1995). Flavonoid intake and long-term risk of coronary heart disease and cancer in the seven countries study. Arch Intern Med, 155, 381-386.

Hevel, J.M., White, K.A. & Marletta, M.A. (1991). Purification of the inducible murine macrophage nitric oxide synthase. J Biol Chem, 266, 22789-22791.

Hobart, P.M., Fogliano, M., O´Connor, B.A., Schaefer, I.M. & Chirgwin, J.M. (1984). Human renin gene: Structure and sequence analysis. Proc Natl Acad Sci USA, 81, 5026-5030.

Hodgson, J.M. (2006). Effects of tea and tea flavonoids on endothelial function and blood pressure: a brief review. Clin Exp Pharmacol Physiol, 33, 838-841.

Hopper, N.M., Keen, J., Pappin, D.J.C. & Turner, A.J. (1987). Pig kidney angiotensin converting enzyme. Purification and characterization of amphipathic and hydrophilic forms of the enzyme establishes C-terminal anchorage to the plasma membrane. Biochem J, 247, 85-93.

Hwang, J., Wang, J., Morazzoni, P., Hodis, H.N. & Sevanian, A. (2003). The phytoestrogen equol increases nitric oxide availability by inhibiting superoxide production: an antioxidant mechanism for cell-mediated LDL modification. Free Radical Biol & Med, 34, 1271-1282.

Häckl, L.P.N., Cuttle, G., Sanches Dovichi, S., Lima-Landman, M.T. & Nicolau, M. (2002). Inhibition of angiotensin-converting enzyme by quercetin alters the vascular response to bradykinin and angiotensin I. Pharmacol, 65, 182-186.

Ichiki, T., Labosky, P.A., Shiota, C., Okuyama, S., Imagawa, Y., Fogo, A., Nimura, F., Ichikawa, I., Hogman, B.L.M. & Inagami, T. (1995). Effects on blood pressure and exploratory behavior of mice lacking angiotensin II type-2receptor. Nature, 377, 748-750.

Ignarro, L.J., Buga, G.M., Wood, K.S., Byrns, R.E. & Chaudhuri, G. (1987). Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc Natl Acad Sci USA, 84, 9265-9269.

Ikizler, M., Erkasap, N., Dernek, S., Kural, T. & Kaygisiz, Z. (2007). Dietary polyphenol quercetin protects rat hearts during reperfusion: enhanced antioxidant capacity with chronic treatment. Anatolian J Cardiol, 7, 404-410.

Ingenhousz, J. (1779). Experiments upon vegetables, discovering their great power of purifying the common air in the sun-shine, and of injuring it in the shade and at night. To which is joined, a new method of examining the accurate degree of salubrity of the atmosphere. From Leicester, H.M. & Klickstein, H.S. A source book in chemistry 1400-1900, New York: McGraw Hill, 1952.

Jansen, M.C., Bueno-de-Mesquita, H.B., Feskens, E.J., Streppel, M.T., Kok, F.J. & Kromhout, D. (2004). Quantity and variety of fruit and vegetable consumption and cancer risk. Nutr Cancer, 48, 142.148.

Page 114: Plant-Derived Substances and Cardiovascular Diseases

114

Jeon, B.H., Kim, C.S., Park, K.S., Lee, J.W., Park, J.B., Kim, K-J., Kim, S.H., Chang, S.J. & Nam, K.Y. (2000). Effects of Korea red ginseng on the blood pressure in conscious hypertensive rats. Gen Pharmacol, 35, 135-141.

Jiménez, R., López-Sepúlveda, R., Kadmiri, M., Romero, M., Vera, R., Sánchez, M., Vargas, F., O´Valle, F., Zarzuelo, A., Duenas, M., Santos-Buelga, C. & Duarte, J. (2007). Polyphenols restore endothelial function in DOCA-salt hypertension: Role of andothelin-1 and NADPH oxidase. Free Rad Biol Med, 43, 462-473.

Johnsen, N., Hausner, H., Olsen, A., Tetens, I., Christensen, J., Bach Knudsen, K.E., Overvad, K. & Tjønneland, A. (2004). Intake of whole grains and vegetables determines the plasma enterolactone concentration of Danish women. J Nutr, 134, 2691-2697.

Jones, P.H. & Farmer, J.A. (2008). Adjunctive interventions in myocardial infarction: the role of statin therapy. Curr Atheroscler Rep, 10, 142-148.

Joseph, J.A., Shukitt-Hale, B. & Lau, F.C. (2007). Fruit polyphenols and their effects on neuronal signaling and behavior in senescence. Ann NY Acad Sci, 1100, 470-485.

Joshipura, K.J., Ascherio, A., Manson, J.E., Stampfer, M.J., Rimm, E.B., Speizer, F.E., Hennekens, C.H., Spiegelman, D. & Willett, W.C. (1999). Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA, 282, 1233-1239.

Joubert, E., Gelderblom, W.C.A., Louw, A. & de Beer, D. (2008). South African herbal teas: Aspalathus linearis, Cyclopia spp. And Athrixia phylicoides-a review. J Ethnopharmacol, 119, 376-412.

Kaliora, A.C. & Dedoussis, G.V.Z. (2007). Natural antioxidant compounds in risk factors for

CVD. Pharmacol Res, 56, 99-109.

Keen, C.L., Holt, R.R., Oteiza, P.I., Fraga, C.G. & Schmitz, H.H. (2005). Cocoa antioxidants and cardiovascular health. Am J Clin Nutr, 81, 298S-303S.

Keifer, D. & Pantuso, T. (2003). Panax ginseng. Am Family Physician, 68, 1539-1542.

Keli, S.O., Hertog, M.G., Feskens, E.J. & Kromhourt, D. (1996). Dietary flavonoids, antioxidant vitamins, and incidence of stroke: the Zutphen study. Arch Intern Med, 25;156(6):, 637-642.

Keller, S.R. (2004). Role of the insulin-regulated aminopeptidase IRAP in insulin action and diabetes. Biol Pharmaceutical Bull, 27, 761-764.

Kim, H., Chen, X. & Gillis, C.N. (1992). Ginsenosides protect pulmonary vascular endothelium against free radical-induced injury. Biochem Biophys Res Comm, 189, 670-676.

Kim, N.D., Kang, S.Y. & Schini, V.B. (1994). Ginsenosides evoke endothelium-dependent vascular relaxation in rat aorta. Gen Pharmacol, 25, 1071-1077.

Kim, S.J., Cho, Y.H., Park, W., Han, D., Chai, C.H. & Imm, J.Y. (2003). Solubilization of water soluble anthocyanins in apolar medium using recerse micelle. J Agric Food Chem, 51, 7805-7809.

Page 115: Plant-Derived Substances and Cardiovascular Diseases

115

Kiss, A.K., Ma´nk, M. & Melzig, M.F. (2008). Dual inhibition of metollopeptidases ACE and NEP by extracts, and iridoids from Ligustrum vulgare L. J Ethnopharmacol, in press.

Klingberg, S., Ellegård, L., Johansson, I., Hallmans, G., Weinehall, L., Andersson, H. & Winkvist, A. (2008). Inverse relation between dietary intake of naturally occuring plant sterols and serum cholesterol in northern Sweden. Am J Clin Nutr, 87, 993-1001.

Kobori, H., Harrison-Bernard, L.M. & Navar, L.G. (2002). Urinary excretion of angiotensinogen reflects intrarenal angiotensinogen production. Kidney International, 61, 579-585.

Koitka, A. & Tikellis, C. (2008). Advances in the rennin-angiotensin-aldosterol system: relevance to diabetic nephropathy. Sci World J, 28, 434-445.

Koltermann, A., Hartkorn, A., Koch, E., Fürst, R., Vollmar, A.M. & Zahler, S. (2007). Ginkgo biloba extract EGb 761 increases endothelial nitric oxide production in vitro and in vivo. Cell Mol Life Sci, 64, 1715-1722.

Kostyuk, V. & Potapovich, A.I. (1998). Antiradical and chelating effects in flavonoid protection against silica-induced cell injury. Arch Biochem Biophys, 355, 43-48.

Kostyuk, V., Potapovich, A.I., Vladykovskaya, E.N., Korkina, L.G. & Afanasév, I.B.A. (2001). Influence of metal ions on flavonoid protection against asbestos-induced cell injury. Arch Biochem Biophys, 385, 129-137.

Kumar, R., Singh, V.P. & Baker K.M. (2007). The intracellular renin-angiotensin system: a new paradigm. Trends Endocrinol Metab, 18, 208-214.

Kuoppala, A., Lindstedt, K.A., Saarinen, J., Kovanen, P.T. & Kokkonen, J.O. (2000). Inactivation of bradykinin by angiotensin-converting enzyme and by carboxypeptidase N in human plasma. Am J Physiol Heart Circ Physiol, 278, H1069-H1074.

Kurihara, H., Fukami, H., Asami, S., Toyoda, Y., Makai, M., Shibata, H. & Yao, X-S. (2004). Effects of Oolong tea on plasma antioxidative capacity in mice loaded with restraint stress assessed using the oxygen radical absorbance capacity (ORAC) assay. Biol Pharm Bull, 27, 1093-1098.

Kühnau, J. (1976). The flavonoids. A class of semi-essential food components: their role in human nutrition. Wld Rev Nutr Diet, 24, 117-191.

Kähkönen, M. & Heinonen, M. (2003). Antioxidant activity of anthocyanins and their aglycons. J Agr Food Chem, 51, 628-633.

Lakhdar, R., Al-Mallah, M.H. & Lanfear, D.E. (2008). Safety and tolerability of angiotensin-converting enzyme inhibitors versus the combination angiotensin-converting enzyme inhibitor and angiotensin receptor blocker in patients with left ventricular dysfunction: a systematic review and meta-analysis of randomized controlled trials. J Cardiac Failure, 14, 181-188.

Lambert, J.D., Sang, S. & Yang, C.S. (2007). Biotransformation of green tea polyphenols and the biological activities of those metabolites. Mol Pharmaceut, 4, 819-825.

Lambert, J.D. & Yang, C.S. (2003). Mechanisms of cancer prevention by tea constituents. Am Soc Nutr Sci, 133, 3262S-3267S.

Page 116: Plant-Derived Substances and Cardiovascular Diseases

116

Laragh, J.H., Angers, M., Kelly, W.G. & Lieberman, S. (1960). Hypotensive agents and pressor substances. The effect of epinephrine, norepinephrine, angiotensin II, and others on the secretory rate of aldosterone in man. JAMA, 174, 234-240.

Lauer, T., Preik, M., Rassaf, T., Strauer, B.E., Deussen, A., Feelisch, M. & Kelm, M. (2001). Plasma nitrite rather than nitrate reflects regional endothelial nitric oxide synthase activity but lacks intrinsic vasodilator action. Proc Natl Acad Sci USA, 98, 12814-12819.

Lee, E.J. & Lang, H.D. (2004). Antioxidant activity and protective effects on DNA strand scission of Rooibos tea (Aspalathus linearis). Biofactors, 21, 285-292.

Leshem, Y.Y. & Kniper, P.J.C. (1996). Is there a GAS (general adaption syndrome) response to various types of environmental stress? Biol Plant, 38, 1-9.

Leung, K.W., Yung, K.K., Mak, N.K., Yue, P.Y., Luo, H.B., Cheng, Y.K., Fan, T.P., Yeung, H.W., Ng, T.B. & Womg, R.N. (2007). Angiomodulatory and neurological effects of ginsenosides. Curr Med Chem, 14, 1371-1380.

Li, W., Asada, Y. & Toshikawa, T. (2000). Flavonoid constituents from Glycyrrhiza glabra hairy root cultures. Phytochem, 55, 447-456.

Liang, C., Zhao, F., Wei, W., Wen, Z. & Qin, S. (2006). Carotenoid biosynthesis in cyanobacteria: structural and evolutionary scenarios based on comparative genomics. Int J Biol Sci, 2, 197-207.

Lichtenthaler, H.K. (2007). Biosynthesis accumulation and emission of carotenoids, α-tocopherol, plastoquinone, and isoprene in leaves under high photosynthetic irradiance. Photosynth Res, 92, 163-179.

Liebgott, T., Miollan, M., Berchadsky, Y., Drieu, K., Calcasi, M. & Pietri, S. (2000). Complementary cardioprotective effects of flavonoids metabolites and terpenoid constituents of Ginkgo biloba extract (EGb 761) during ischemia and reperfusion. Bais Res Cardiol, 95, 368-377.

Liese, A.D., Weis, K.E., Schulz, M. & Tooze, J.A. (2008). Food intake patterns associated with incident type 2 diabetes: The insulin resistance atherosclerosis study. Diabetes Care, In press.

Lim, S. (2007). Recent update in the management of hypertension. Acta Med Indones, 39, 186-191.

Lind, R.W. & Johnson, A.K. (1982). Subfornical organ-median preoptic connections and drinking and pressor responses to angiotensin II. J Neurosc, 2, 1043-1051.

Lindpainter, K., Wilhelm, M.J., Jin, M., Unger, T., Lang, R.E., Schoelkens, B.A. & Ganten, D. (1987). Tissue renin-angiotensin systems: focus on the heart. J Hypertension, 5, S33-S38.

Link, L.B., Hussaini, N.S. & Jacobson, J.S. (2008). Change in quality of life and immune markers after a stay at a raw vegan institute: A pilot study. Complementary Therapies in Medicine, 16, 124-130.

Locher, R., Emmanuele, L., Suter, P.M., Vetter, W. & Barton, M. (2002). Green tea polyphenols inhibit human vascular smooth muscle cell proliferation stimulated by native low-density lipoprotein. Eur J Pharmacol, 434, 1-7.

Page 117: Plant-Derived Substances and Cardiovascular Diseases

117

Lohmeier, T.E. (2001). The sympathetic nervous system and long-term blood pressure regulation. Am J Hypertens,14, 147S-154S.

Loizzo, M.R., Said, A., Tundis, R., Rashed, K., Statti, G.A., Hufner, A. & Menichini, F. (2007). Inhibition of angiotensin converting enzyme (ACE) by flavonoids isolated from Ailanthus Excelsa (Roxb) (Simaroubaceae). Phytother Res, 21, 32-36.

Loke, W.M., Hodgson, J.M., Proudfoot, J.M., McKinley, A.J., Puddey, I.B. & Croft, K.D. (2008). Pure dietary flavonoids quercetin and (-)-epicatechin augment nitric oxide products and reduce endothelin-1 acutely in healthy men. Am J Clin Nutr, 88, 1018-1025.

López, E.P., Rice, C., Weddle, D.O. & Rahill, G.J. (2008). The relationship among cardiovascular risk factors, diet patterns, alcohol consumption, and ethnicity among women aged 50 years and older. J Am Diet Ass, 108, 248-256.

Lorenz, M., Wessler, S., Follmann, E., Michaelis, W., Düsterhöft, T., Baumann, G., Stangl, K. & Stangl, V. (2004). A constituent of green tea, epigallocatechin-3-gallate, activates endothelial nitric oxide synthase by a phosphatidylinositol-3-OH-kinase-, cAMP-dependent protein kinase-, and Akt-dependent pathway and leads to endothelial-dependent vasorelaxation. J Biol Chem, 279, 6190-6195.

Lundberg, J.O. & Govoni, M. (2004). Inorganic nitrate is a possible source for systemic generation of nitric oxide. Free Radic Biol Med, 37, 395-400.

Lundberg, J.O., Feelisch, M., Björne, H., Jansson, E.Å. & Weitzberg, E. (2006). Cardioprotective effects of vegetables: is nitrate the answer? Nitric oxide, 15, 359-362.

Lundberg, J.O. & Weitzberg, E. (2005). NO generation from nitrate and its role in vascular control. Arter Thromb Vasc Biol, 25, 915-922.

Lüscher, T.F. & Barton, M. (1997). Biology of the endothelium. Clin Cardiol, 20, II-3-II-10.

Macours, N. & Hens, K. (2004). Zinc-metalloproteases in insects: ACE and ECE. Insect Biocem Mol Biol, 34, 501-510.

Maggioni, A.P. (2006). Efficacy of angiotensin receptor blockers in cardiovascular disease. Cardiovasc Drugs Ther, 20, 295-308.

Manach, C., Williamson, G., Morand, C., Scalbert, A. & Rémésy, C. (2005). Bioavailability and bioeffecacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr, 81, 230S-242S.

Manach, C., Scalbert, A., Morand, C., Rémésy, C. & Jiménez, L. (2004). Polyphenols: food sources and bioavailability. Am J Clin Nutr, 79, 727-747.

Marder, G. & Greenwald, R.A. (2003). Potential applications of matrix metalloproteinase inhibitors in geriatric practice. Isr Med Assoc J, 5, 361-364.

Matringe, M., Ksas, B., Rey, P. & Havaux, M. (2008). Tocotrienols, the unsaturated forms of vitamin E, can function as antioxidants and lipid protectors in tobbaco leaves. Plant Physiol, 147, 764-768.

Page 118: Plant-Derived Substances and Cardiovascular Diseases

118

Matsumoto, H., Kamm, K.E., Stull, J.T. & Azuma, H. (2005). Delphinidin-3-rutinoside relaxes the bovine ciliary smooth muscle through activation of ETB receptor and NO/cGMP pathway. Exp Eye Res, 80, 313-322.

McCarty, M.F. (2006). Isoflavones made simple – Genistein´s agonist activity for the beta-type estrogen receptor mediates their health benefits. Med Hypothes, 66, 1093-1114.

McGuiness, B. Todd, S., Passmore, P. & Bullock. R. (2006). Blood pressure lowering in patients without prior cerebrovascular disease for comparative pharmacological interruption of the rennin system. Hypertens, 32, 387-392.

Mesbah, M.K., Khalifa, S.I., El-Gindy, A. & Tawfik, K.A. (2005). HPLC determination of certain flavonoids and terpene-lactones in selected Ginkgo biloba L. phytopharmaceuticals. Il Farmaco, 60, 583-590.

Messina, B.A.M. (2006). Herbal supplements: facts and myths-talking to your patients about herbal supplements. J PeriAnesthes Nurs, 4, 268-278.

Meydani, M. (2004). Vitamin E modulation of cardiovascular disease. Ann N.Y. Acad Sci,1031, 271-279.

Milman, U., Blum, S., Shaoira, C., Aronson, D., Miller-lotan, R., Anbinder, Y., Alshiek, J., Bennett, L., Kostenko, M., Landau, M., Keidar, S., Levy, Y., Khemlin, A., Radan, A. & Levy, A.P. (2008). Vitamin E supplementation reduces cardiovascular events in a subgroup of middle-aged individuals with both type 2 diabetes mellitus and the haptoglobin 2-2 genotype, a perspective double-blinded clinical trial. Arterioscler Thromb Vasc Biol, 28, 341-347.????

Monder, C. & White, P.C. (1993). 11 beta-hydroxysteroid dehydrogenase. Vitam Horm, 47, 187-271.

Moreno, D.A., Víllora, G., Ruiz, J.M. & Romero, L. (2003). Growth conditions, elemental accumulation and induced physiological changes in Chinese cabbage. Chemosphere, 52, 1031-1040.

Morton, L.W., Abu-Amsha Caccetta, R., Puddey, I.B. & Croft, K.D. (2000). Chemistry and biological effects of dietary phenolic compounds: relevance to cardiovascular disease. Clin Exp Pharmacol Physiol, 27, 152-159.

Mosimann, R., Imboden, H. & Felix, D. (1996). The neuronal role of angiotensin II in thirst, sodium appetite, cognition and memory. Biol Rev Camb Philos Soc, 71, 545-549.

Mukamal, K.J., Maclure, M., Muller, J.E., Sherwood, J.B. & Mittleman, M.A. (2002). Tea comsuption and mortality after acute myocardial infarction. Circulation, 105, 2476-2481.

Muller, F.L., Lustgarten, M.S., Jang, Y., Richardsson, A. & Van Remmen, H. (2007). Trends in oxidative aging theories. Free Rad Biol Med, 43, 477-503.

Nandakumar, V., Singh, T. & Katiyar, S.K. (2008). Multi-targeted prevention anf therapy of cancer by proanthocyanidins. Cancer Lett doi: 10.1016/j.canlet.2008.03.049.S

Nakagawa, T. & Yokozawa, T. (2002). Direct scavenging of nitric oxide and superoxide by green tea. Food Chem Toxicol, 40, 1745-1750.

Page 119: Plant-Derived Substances and Cardiovascular Diseases

119

Nakajima, M., Hutchinson, H.G., Fujinaga, M., Hayashida, W., Morishita, R., Zhang, L., Horiuchi, M., Pratt, R.E. & Dzau, V.J. (1995). The angiotensin II type 2 (AT2) receptor antagonizes the growth effects of the AT1 receptor: gain-of-function study using gene transfer. Proc Natl Acad Sci USA, 92, 10663-10667.

Nantz, M.P., Rowe, C.A., Bukowski, J.F. & Percival, S.S. (2008). Standardized capsule of Camellia sinensis lowers cardiovascular risk factors in a randomized, double-blind, placebo-controlled study. Nutrition, in press.

Napoli, C., deNigris, F., Williams-Ignarro, S. & Pignalosa, O. (2006). Nitric oxide and atherosclerosis: An update. Nitric Oxide, 15, 265-279.

Nardini, M., Natella, F. % Scaccini, C. (2007). Role of dietary polyphenols in platelet aggregation. A review of the supplementation studies. Platelets, 18, 224-243.

Ness, A.R. & Powles, J.W. (1997). Fruit and vegetables, and cardiovascular disease: a review. Intern J Epidemiol, 26, 1-13.

Ng, K.F. & Vane, J.R. (1967). Conversion of angiotensin I to angiotensin II. Nature, 216, 762-766.

Nicholson, S.K., Tucker, G.A. & Brameld, J.M. (2008). Effects of dietary polyphenols on gene expression in human vascular endothelial cells. Proceedings Nutr Soc, 67, 42-47.

Nijveldt, R.J., van Nood, E., van Hoorn, E.C., Boelens, P.G., van Norren, K. & van Leeuwen, P.A.M. (2001). Flavonoids: a rewiew of probable mechanisms of action and potential applications. Am J Clin Nutr, 74, 418-425.

Nishimura, H. (2000). Angiotensin receptors- evolutionary overview and perspectives. Comp Biochem Physiol, 128, 11-30.

Nocerino, E., Amato, M. & Izzo, A.A. (2000). The aphrodisiac and adaptogenic properties of ginseng. Filoterapia, 71, S1-S5.

Nyhlén, K., Linden, M., Andersson, R.G.G. & Uppugunduri, S. (2000). Corticosteroids and interferons inhibit cytokine induced production of IL-8 by human endothelial cells. Cytokine, 12, 355-360.

Oak, M-H., El Bedoui, J. & Schini-Kerth, V.B. (2005). Antiangiogenic properties of natural polyphenols from red wine and green tea. J Nutr Biochem, 16, 1-8.

Oparil, S. & Haber, E. (1974). The renin-angiotensin system (first of two parts). N Eng J Med, 291, 389-401.

Orr, J.D. (2008). Statins in the spectrum of neurologic disease. Curr Atheroscler Rep, 10, 11-18.

Palmer, R.M.J., Ferrige, A.G. & Moncada, S. (1987). Nitric oxide release accounts for the biological activity of endothelium-relaxing factor. Nature, 327, 524-526.

Palmer, R.M.J., Ashton, D.S. & Moncada, S. (1988). Vascular endothelial cells synthesize nitric oxide from L-arginine. Nature, 333, 664-666.

Page 120: Plant-Derived Substances and Cardiovascular Diseases

120

Patterson, C., Feightner, J.W., Garcia, A., Hsiung, G-Y.R., MacKnight, C. & Sasovnick, A.D. (2008). Diagnosis and treatment of dementia: Risk assessment and primary prevention of Alzheimer disease. Can Med Ass J, 178, 548-556.

Peach, M.J. (1977). Renin-angiotensin system: Biochemistry and mechanisms of action. Physiol Rev, 57, 313-370.

Pemp, B. & Schmetterer, L. (2008). Ocular blood flow in diabetes and age-related macular degeneration. Can J Ophthalmol, 43, 295-301.

Persson, I.A-L. & Persson, K. (2008). Liquorice, Glycyrrhiza glabra inhibits Angiotensin-converting enzyme activity and increases Nitric oxide production in human endothelial cells. Special Edition on Standardization of Herbal / Ayurvedic Formulation Vol 23 of the Serial Publication “Recent Progress in Medicinal Plants”.

Persson, K. & Andersson, R.G.G. (1998). Biphasic response to bradykinin in isolated porcine iliac arteries is mediated by bradykinin B1 and B2 receptors. J Cardivasc Pharmacol, 31, 306-313.

Persson, K., Säfholm, A.C.E., Andersson, R.G.G. & Ahlner, J. (2005). Glyceryl trinitrate-induced angiotensin-converting enzyme (ACE) inhibition in healthy volunteers is dependent on ACE genotype. Can J Physiol Pharmacol, 83, 1117-1122.

Pietri, S.M., Drieu, K. & Calcasi, M. (1997). Cardioprotective and anti-oxidant effects of the terpenoid constituents of Ginkgo biloba extract (EGb 761). J Cell Cardiol, 29, 733-742.

Poli, A., Marangoni, F., Paoletti, R., Mannarino, E., Lupattelli, G., Notarbartolo, A., Aureli, P., Bernini, F., Cicero, A., Gaddi, A., Catapano, A., Cricelli, C., Gattone, M., Marrocco, W., Porrini, M., Stella, R., Vanotti, A., Volpe, M., Cannella, C., Pinto, A., Del Toma, E., La Vecchia, C., Tavani, A., Manzato, E., Riccardi, G., Sirtori, C & Zambon, A. (2008). Non-pharmacological control of plasma cholesterol levels. Nutr Metab Cardiovasc Dis, 18, 1-16.

Polkowski, K. & Mazurek, A.P. (2000). Biological properties of genistein. A review of in vitro and in vivo data. Acta Poloniae Pharmaceutica, 57, 135-155.

Pollock, J.S., Förstermann, U., Mitchell, J.A., Warner, T.D., Schmidt, H.H.H.W., Nakane, N. & Murad, F. (1991). Purification and characterization of particulate endothelium-derived relaxing factor synthase from cultured and native bovine aortic endothelial cells. Proc Natl Acad Sci USA, 88, 10480-10484.

Puddu, P., Cravero, E., Puddu, G.M. & Muscari, A. (2005). Genes and atherosclerosis: at the origin of the predisposition. Int J Clin Pract, 59, 462-472.

Quaschning, T., Ruschitzka, F., Niggli, B., Lunt, C.M.B., Shaw, S., Christ, M., Wehling, M. & Luscher, T.F. (2001). Influence of aldosterone vs endothelin receptor antagonism on renovascular function in liquorice-induced hypertension. Nephrol Dial Transplant, 16, 2146-2151.

Quaschning, T., Ruschitzka, F., Shaw, S. & Luscher, T. (2001). Aldosterone receptor antagonism normalizes vascular function in liquorice-induced hypertension. Hypertension, 37, 801-805.

Page 121: Plant-Derived Substances and Cardiovascular Diseases

121

Radomski, M.W., Palmer, R.M.j. & Moncada, S. (1987). The anti-aggregating properties of vascular endothelium: interactions between prostacyclin and nitric oxide. Br J Pharmacol, 92, 639-646.

Rahman, I. (2006). Antioxidant therapies in COPD. Int J Chon Obstruc Pulm Dis, 1, 15-29.

Rapoport, R.M. & Murad, F. (1983). Agonist-induced endothelium-dependent relaxation in rat thoracic aorta may be mediated through cGMP. Circ Res, 52, 352-357.

Rasoolijazi, H., Joghataie, M.T., Roghani, M. & Nobakht, M. (2007). The beneficial effect of (-)-epigallocatechin-3-gallate in an experimental model of Alzheimer´s disease in rat: a behavioral analysis. Iran Biomed J, 11, 237-243.

Ratman, D.V., Ankola, D.D., Bhardwaj, V., Sahana, D.K. & Kumar, M.N.V.R. (2006). Role of antioxidants in prophylaxis and therapy: A pharmacological perspective. SJ Controll Release, 113, 189-207.

Raven, P.H., Evert, R.F. & Eichhorn, S.E. (2003). Biology of Plants. W.H. Freeman and Company, New York, USA:

Rawlings, N.D. & Barrett, A.J. (1993). Evolutionary families of peptidases. Biochem J, 290, 205-218.

Reaux, A., Fournie-Zaluski, M.C. & Llorens-Cortes, C. (2001). Angiotensin III: a central regulator of vasopressin release and blood pressure. TRENDS Endocrinol Metabol, 12, 157-162.

Reed, R., Kolz, C., Potter, B. & Rocic, P. (2008). The mechanistic basis for the disparate effects of angiotensin II on coronary collateral growth. Arterioscler Thromb Vasc Biol, 28, 61-67.

Regoli, D. & Barabé, J. (1980). Pharmacology of bradykinin and related kinins. Pharmacol Rev, 32, 1-46.

Reid, I.A. (1985). The renin-angiotensin system and body function. Arch Int Med, 145, 1475-1479.

Riesenberg, L.H. & Soltis, D. (1987). Flavonoids of fossil Miocene Platanus and it sextant relatives. Biochem System Ecol, 15, 109-112.

Rigat, B., Hubert, C., Alhenc-Gelas, F., Cambien, F., Corvol, P. & Soubrier, F. (1990). An Insertion/Deletion polymorphism in the angiotensin I-converting enzyme gene accouting for half the variance of serum enzyme levels. J Clin Invest,86, 1343-1346.

Rissanen, T.H., Voutilainen, S., Virtanen, J.K., Venho, B., Vanharanta, M., Mursu, J. & Salonen, J.T. (2003). Low intake of fruits, berries and vegetables is associated with excess mortality in men: the Kuopio ischaemic heart disease risk factor (KIHD) study. J Nutr, 133, 199-204.

Roberts, W., Riba, R., Homer-Vanniasinkam, S., Farndale, R.W. & Naseem, K.M. (2008). Nitric oxide specifically inhibits integrin-mediated platelet adhesion and spreading on collagen. J Thromb Haemost, in press.

Page 122: Plant-Derived Substances and Cardiovascular Diseases

122

Roks, A., Buikema, H., Pinta, Y.M. & van Gilst, W.H. (1997). The renin-angiotensin system and vascular function. The role of angiotensin II, angiotensin-converting enzyme, and alternative conversion of angiotensin I. Heart Vessels, Suppl 12, 119-124.

Romani, A., Pinelli, P., Mulinacci, N., Vincieri, F.F., Gravano, E. & Tattini, M. (2000). HPLC analysis of flavonoids and secoiridoids in leaves of Ligustrum vulgare L. (Oleaceae). J Agric Food Chem, 48, 4091-4096.

Ruiz-Ortega, M., Lorenzo, O., Rupérez, M., Esteban, Y., Suzuki, Y., Mezzano, S., Plaza, J.J. & Egido, J. (2001). Role of the rennin-angiotensin system in vascular diseases: expanding the field. Hypertens, 38, 1382-1387.

Rush, J.W. & Aultman, C.D. (2008). Vascular biology of angiotensin and the impact of physical activity. Appl Physiol Nutr Metab, 33, 162-172.

Russo, G.L. (2008). Dietary n-6 and n-3 polyunsaturated fatty acids: From biochemistry to clinical implications in cardiovascular prevention. Biochemistry Pharmacol, In press.

Russo, S., Mastropasqua, M. Mosetti, M.A., Persegani, C. & Paggi, A. (2000). Low doses of liquorice can induce hypertension encephalopathy. Am J Nephrol, 20, 145-148.

Rusznyák, St. & Szent-Györgui, A. (1936). Vitamin P: Flavonols as Vitamins. Nature, 138, 27.

Ruzicka, L. (1953). The isoprene Rule and the biogenesis of terpenic compounds. Experientia, IX, 357-396.

Ryan, J.W., Ryan, U.S., Schultz, D.R., Whitaker, C. & Chung, A. (1975). Subcellular localization of pulmonary angiotensin-converting enzyme (kininase II). Biochem J, 146, 497-499.

Sakuma, I., Stuehr, D.J., Gross, S.S. Nathan, C. & Levi, R. (1988). Identification of arginine as a precursor of endothelium-derived relaxing factor. Proc Natl Acad Sci USA, 85, 8664-8667.

Samani, N.J., Thompson, J.R., O´Toole, L., Channer, K. & Woods, K.L. (1996). A meta-analysis of the association of the deletion allele of the angiotensin-converting enzyme gene with myocardial infarction. Circulation, 94, 708-712.

Sancho, J., Re, R., Burton, J., Barger, A.C. & Haber, E. (1976). The role of the renin-angiotensin-aldosterine system in cardiovascular homeostasis in normal human subjects. Circulation, 53, 400-405.

Schambelan, M. (1994). Licorice ingestion and blood pressure regulating hormones. Steroids, 59, 127-130.

Scharfstein, J., Schmitz, V., Svensjö, E., Granato, A. & Monteiro, A.C. (2007). Kininogens coordinate adaptive immunity through the proteolytic release of bradykinin, an endogenous danger signal driving dendrite cell maturation. Scand J Immunol, 66, 128-136.

Schiavone, M.T., Khosla, M.C. & Ferrario, C.M. (1990). Angiotensin-(1-7): evidence for novel actions in the brain. J Cardiovasc Pharmacol, 4, S19-24.

Page 123: Plant-Derived Substances and Cardiovascular Diseases

123

Schmelzer, E., Jahnen, W. & Hahlbrock, K. (1988). In situ localization of light-induced chalcone sunthase mRNA, chalcone synthase, and flavonoid endproducts in epidermal cells of parsley leaves. Proc Nat Acad Sci USA, 85, 2989-2996.

Seddon, J.M. (2007). Multivitamin-multimineral supplements and eye disease: age-related macular degeneration and cataract. Am J Clin Nutr, 85, 304S-307S.

Selmi, C., Cocchi, C.A., Lanfredini, M., Keen, C.L. & Gershwin, M.E. (2008). Chocolate at heart: The anti-inflammatory impact of cocoa flavanols. Mol Nutr Food Res, 52, 1340-1348.

Sepp, H., Ekelund, U. & Becker, W. (2004). Enkätfrågor om kost och fysisk aktivitet bland vuxna. National Food Administration, Sweden, Rapport 21.

Sharma, J.N., Uma, K., Noor, A.R. & Rahman, A.R.A. (1996). Blood pressure regulation by the kallikrein-kinin system. Gen Pharmacol, 27, 55-63.

Shukitt-Hale, B., Lau, F.C. & Joseph, J.A. (2008). Berry fruit supplementation and the aging brain. J Agric Food Chem, 56, 636-641.

Siegel, R.K. (1979). Ginseng abuse syndrome Problems with the panacea, JAMA, 241, 1614-1615.

Sigurjonsdottir, H., Axelson, M., Johannsson, G., Manhem, K., Nyström, E. & Wallerstedt, S. (2006). The liquorice effect on the RAAS differs between the genders. Blood Pressure, 15, 169-172.

Sing, C.F., Stengård, J.H. & Kardia, S.L.R. (2003). Genes, environment, and cardiovascular disease. Arterioscler Thromb Vasc Biol, 23, 1190-1196.

Sirois, B.C. & Burg, M.M. 2003. Negative emotion and coronary heart disease. Behav Modif, 27, 83-102.

Sirtori, C.R. & Lovati, M.R. (2001). Soy Proteins and cardiovascular disease. Curr Atheroscler Rep, 3, 47-53.

Skeggs, L.T., Kahn, J.R. & Shumway, N.P. (1956b). The preparation and function of the hypertension-converting enzyme. J Exp Med, 103, 295-299. Skeggs, L.T., Lentz, K.E., Kahn, J.R., Shumway, N.P. & Woods, K.R. (1956a). The amino acid sequence of hypertension II. J Exp Med, 104, 193-197. Soubrier, F., Alhenc-Gelas, F., Hubert, C., Allegrini, J., John, M., Tregear, G. & Corvol, P. (1988). Two putative active centers in human angiotensin I-converting enzyme revealed by molecular cloning. Proc Natl Acad Sci USA, 85, 9386-9390. Stahl, W. & Sies, H. (2002). Carotenoids and protection against solar UV radiation. Skin Pharmacol Appl Skin Physiol, 15, 291-296. Standley, L., Wintertom, P., Marnewick, J.L., Gelderblom, W.C.A., Joubert, E. & Britz, T.J. (2001). Influence of processing stages on antimutagenic and antioxidant potentials of Rooibos tea. J Agr Food Chem, 49, 114-117. Stanton, A. (2003). Potential of renin inhibition in cardiovascular disease. J RAAS, 4, 6-10.

Page 124: Plant-Derived Substances and Cardiovascular Diseases

124

Stapleton, A.E. (1992). Ultraviolet radiation and plants: burning questions. Plant Cell, 4, 1353-1358. Steinmetz, K.A. & Potter, J.D. (1991). Vegetables, fruit, and cancer. I. Epidemiology. Cancer Causes Control, 2, 325-357. Stocker, R. (2007). Vitamin E. Diet Supplem Health, 282, 77-92. Stoll, M., Steckelings, U.M., Paul, M., Bottare, S.P., Metzger, R. & Unger, T. (1995). The angiotensin AT2-receptor mediates inhibition of cell proliferation in coronary endothelial cells. J Clin Invest, 95, 651-657. Sturrock, E.D., Natesh, R., van Rooyen, J.M. & Acharva, K.R. (2004). Structure of angiotensin I-converting enzyme. Cell Mol Life Scien, 61, 2677-2686.

Sugiyama, T., Yoshimoto, T., Tsuchiya, K., Gochou, N., Hirono, Y., Tateno, T., Fukai, N., Shichiri, M. & Hirata, Y. (2005). Aldosterone induces angiotensin converting enzyme gene expression via a JAK2-dependent pathway in rat endothelial cell. Endocrinol, 146, 3900-3906. Sun, J., Chu, Y-F., Wu, X. & Liu, R.H. (2002). Antioxidant and antiproliferative activities of common fruits. J Agric Food Chem, 50, 7449-7454. Sung, J., Han, K.H., Zo, J.H., Park, H.J., Kim, C.H. & Oh, B.H. (2000). Effects of red ginseng upon vascular endothelial function in patients with essential hypertension. Am J Chinese Med, 28, 205-216. Svarcova, I., Heinrich, J. & Valentova, K. (2007). Berry fruits as a source of biologically active compounds: The case of Lonicera Caerulea. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub, 151, 163-174. Squadrito, F., Altavilla, D., Morabito, N., Crisafulli, A., D́anna, R., Corrado, F., Ruggeri, P., Campo, G.M., Calapai, G., Caputi, A.P. & Squadrito, G. (2002). The effect of the phytoestrogen genistein on plasma nitric oxide concentrations, endothelin-1 levels and endothelium dependent vasodilatation in postmenopausal women. Atherosclerosis, 163, 339-347. Taubert, D., Roesen, R., Lehmann, C., Jung, N. & Schömig, E. (2007). Effects of low habitual cocoa intake on blood pressure and bioactive nitric oxide. JAMA, 298, 49-60. Thomas, S.R., Witting, P.K. & Drummond, G.R. (2008). Redox control of endothelial function and dysfunction: molecular mechanisms and therapeutic opportunities. Antioxid Redox Signal, 10, 1713-1765. Tigerstedt, R. & Bergman, P.G. (1898). Niere, und Kreislauf. Skand Archiv Physiol, 8, 223-271.

Page 125: Plant-Derived Substances and Cardiovascular Diseases

125

Tiret, L., Rigat, B., Visvikis, S., Breda, C., Corvol, P., Cambien, F. & Soubrier, F. (1992). Evidence, from combined segregation and linkage analysis, that a variant of the angiotensin I-converting enzyme (ACE) gene controls plasma ACE levels. J Hum Genet, 51, 197-205. Tomer, D.P., McLeman, L.D., Ohmine, S., Scherer, P.M., Murray, B.K. & O´Neill, K.L. (2007). Comparison of the total oxyradical scavenging capacity and oxygen radical absorbance capacity antioxidant assays. J Med Food, 10, 337-344. Truswell, A.S. (2002). Cereal grains and coronary heart disease. Eur J Clin Nutr, 56, 1-14. Tsutsui, M. (2004). Neuronal nitric oxide synthase as a novel anti-atherogenic factor. J Atheroscler Thromb, 11, 41-48. Ueda, S., Meredith, P.A., Morton, J.J., Connell, J.M.C. & Elliott, H.L. (1998). ACE (I/D) genotype as a predictor of the magnitude and duration of the response to an ACE inhibitor drug (enalaprilat) in humans. Circ, 98, 2148-2153. Unger, T., Chung, O., Csikos, T., Culman, J., Gallinat, S., Gohlke, P., Höhle, S., Meffert, S., Stroll, M., Stroth, U. & Zhu, Y.-Z. (1996). Angiotensin receptors. J Hypertens, 14, S95-S103. Vallino, M., Drogo, V., Abba, S. & Perotto, S. (2005). Gene expression of the ericoid mycorrhizal fungus Oidiodendron maius in the presence of high zinc concentrarion. Mycorrhiza, 15, 333-344. Van Beck, T.A. (2002). Chemical analysis of Ginkgo biloba leaves and extracts. J Chromat, 967, 21-55. Vanharanta, M., Voutilainen, S., Rissanen, T.H., Adlercreutz, H. & Salonen, J.T. (2003). Risk of cardiovascular disease-related and all-cause death according to serum concentration of enterolactone. Arch Intern Med, 163, 1099-1104. Vanhoutte, P.M. & Mombouli, J-V. (1996). Vacular endothelium: vasoactive mediators. Prog Cardivasc Dis, 34, 229-238. Voutilainen, S., Nurmi, T., Mursu, J., Rissanen, T.H. (2006). Carotenoids and cardiovascular health. Am J Clin Nutr, 83, 1265-1271. Walle, T. (2004). Absorption and metabolism of flavonoids. Free Rad Biol Med, 36, 829-837. Ward, C.P., Redd, K., Williams, B.M., Caler, J.R., Luo, Y. & McCoy, J.G. (2002). Ginkgo biloba extract: Cognitive enhancer or antistress buffer. Pharmacol Biocem Behav, 72, 913-922. Whalley, E.T., Clegg, S., Stewart, J.M. & Vavrek, R.J. (1987). The effect of kinin agonists and antagonists on the pain response of the human blister base. Naunyn-Schmiedelberg´s Arch Pharmacol, 336, 652-655. Williams, C.M., El Mohsen, M.A., Vauzour, D., Rendeiro, C., Butler, L.T., Ellis, J.A., Whiteman, M. & Spencer, J.P.E. (2008). Blueberry-induced changes in spatial working

Page 126: Plant-Derived Substances and Cardiovascular Diseases

126

memory correlated with changes in hippocampal CREB phosporylation and brain-derived neurotropic factor (BDNF) levels. Free Radic Biol Med, 45, 295-305. Williamson, G. & Manach, C. (2005). Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am J Clin Nutr, 81, 243S-255S. Willis, K.J. & McElwain, J.C. (2002). The evolution of plants. Oxford University Press New York, USA. Wei, L., Alhenc-Gelas, F., Corvol, P. & Clauser, E. (1991b). The two homologous domains of human angiotensin I-converting enzyme are both catalytically active. J Biol Chem, 266, 9002-9008. Wei, L., Alhenc-Gelas, F., Soubrier, F., Michaud, A., Corvol, P. & Clauser, E. (1991a). Expression and characterization of recombinant human angiotensin I-converting enzyme. J Biol Chem, 266, 5540-5546. Weiss, D., Sorescu, D. & Taylor, W.R. (2001). Angiotensin II and atherosclerosis. Am J Cardiol, 87, 25C-32C. WHO, (2003). Diet, nutrition and the prevention of chronic diseases. World Health Org, Technical Report Series 916. WHO, (2008). WHO European action plan for food and nutrition policy 2007-2012. World Health Org, WHO Regional Office for Europe, Copenhagen, Denmark. Wright, J.W. & Harding, J.W. (1995). Brain angiotensin receptor subtypes AT1, AT2 and AT4 and their functions. Regul Pept, 59, 269-295. Wright, J.W. & Harding, J.W. (1997). Important roles for angiotensin III and IV in the brain renin-angiotensin system. Brain Res Rev, 25, 96-124. Wright, J.W., Krebs, L.T., Stobb, J.W. & Harding, J.W. (1995). The angiotensin IV system: functional implications. Front Neuroendocrinol, 16, 23-52. Wright, J.W., Yamamoto, B.J. & Harding, J.W. (2008). Angiotensin receptor subtype mediated physiologies and behaviors: new discoveries and clinical targets. Progress Neurobiol, 84, 157-181. Xie, J. & Itzkowitz, S. (2008). Cancer in inflammatory bowel disease. World J Gastroenterol, 14, 378-389. Xie, P., Chen, S., Liang, Y., Wang, X., Tian, R. & Upton, R. (2006). Chromatographic fingerprint analysis- a rational approach for quality assessment of traditional Chinese herbal medicine. J Chromat, 1112, 171-180. Xu, Y-Y., Yang, C. & Li, S-N. (2006). Effects of genistein on angiotensin-converting enzyme in rats. Life Sciences, 79, 828-837.

Page 127: Plant-Derived Substances and Cardiovascular Diseases

127

Yang, H.Y.T. & Erdös, E.G. (1967). Second kininase in human blood plasma. Nature, 215, 1402-1403. Yetik-Anacak, G. & Catravas, J.D. (2006). Nitric oxide and the endothelium: History and impact on cardiovascular disease. Vasc Pharmacol, 45, 268-276. Yonekura, L. & Nagao, A. (2007). Intestinal absorption of dietary carotenoids. Mol Nutr Food Res, 51, 107-115. Zafra-Stone, S., Yasmin, T., Bagchi M., Chatterjee, A., Vinson, J.A. & Bagchi, D. (2007). Berry anthocyanins as novel antioxidants in human health and disease prevention. Mol Nutr Food Res, 51, 675-683. Zang, G-X., Lu, X-M., Kimura, S. & Nishiyama, A. (2007). Role of mitochondria in angiotensin II-induced reactive oxygen species and mitogen-activated protein kinase activation. Cardiovasc Res, 76, 204-212. Zini, S., Fournie-Zaluski, M-C., Chauvel, E., Roques, B.P., Corvol, P. & Llorens-Cortes, C. (1996). Identification of metabolic pathways of brain angiotensin II and III using specific aminopeptidase inhibitors: Predominant role of angiotensin III in the control of vasopressin release. Proc Natl Acad Sci USA, 93, 11968-11973.