Open access

Dietary Antioxidant Properties of Vegetable Oils and Nuts – The Race Against Cardiovascular Disease Progression

Written By

Wongama Given Pantsi, Dirk Jacobus Bester, Adriaan Johannes Esterhuyse and Guillaume Aboua

Submitted: 16 September 2013 Published: 05 February 2014

DOI: 10.5772/57184

From the Edited Volume

Antioxidant-Antidiabetic Agents and Human Health

Edited by Oluwafemi Oguntibeju

Chapter metrics overview

3,492 Chapter Downloads

View Full Metrics

1. Introduction

Cardiovascular disease (CVD) is a substantial and growing problem in most of the developing regions of the world. Evidence from experimental, clinical and epidemiological studies has unequivocally pointed to oxidative stress as the key culprit in the pathogenesis of CVD [1, 2]. CVD continues to remain a concern in developed countries and is a growing health concern worldwide. Although death rates from CVD have decreased in many countries due to advances in the field of medicine, the prevalence of CVD risk factors continues to increase. Diet is a centrally important, modifiable risk factor in the prevention of CVD [221-224].

The protection offered by foods is probably mediated through multiple beneficial nutrients contained in these foods, including mono- and polyunsaturated fatty acids, antioxidant vitamins, minerals, phytochemicals, fibre and plant protein. In dietary practice, healthy plant-based diets do not necessarily have to be low in fat. Instead, these diets may include unsaturated fats as the predominant form of dietary fat (e.g., fats from natural vegetable oils and nuts).

Consistent evidence suggests that diets rich in fruit and vegetables and other plant foods are associated with moderately lower overall mortality rates and lower death rates from chronic diseases including CVD [3- 6]. The ‘antioxidant hypothesis’ proposes that vitamin C, vitamin E, carotenoids and other antioxidant nutrients offer protection against CVD by decreasing oxidative damage [7-9]. As evidence began to mount from animal studies and human epidemiological studies on the potential protective effects of antioxidants, excitement in both the lay and medical communities also began to increase.

There has been a global increase in the use of medicinal plants that contain significant amounts of antioxidant-rich oils, offering multiple health benefits with fewer side effects compared to their synthetic counterparts. The idea is that natural compounds, if taken in supplement form, may offer a broad and inexpensive means of decreasing the risk for CVD. Natural products, such as vegetable oils and nuts, may be viewed as a cocktail of active ingredients that often have a synergistic effect on health. The (n-3) PUFAs have been shown in epidemiological and clinical trials to reduce the incidence of CVD. Large-scale epidemiological studies suggest that individuals at risk of coronary heart disease (CHD) benefit from the consumption of plant and marine derived (n-3) PUFAs, although the ideal intake is presently unclear. Overall, in view of the prevalence of CHD, consumption of (n-3) PUFA oils should be considered as a useful complementary option for the amelioration of CVD. Several researchers have shown encouraging findings on the protective effects of some vegetable oils and nuts. However, more research needs to be done with regards to the nutrients in these vegetable oils and nuts to elucidate the protective effects against CVD progression. This chapter focuses on the beneficial roles of antioxidant-rich vegetable oils and nuts in the management of CVD, their mechanisms of action and future prospects.

The term “cardiovascular disease (CVD)” encompasses the major clinical end-points related to the heart and vascular system, including ischaemic myocardium (heart failure and angina), myocardial infarction (heart attack), cerebrovascular disease (stroke), high blood pressure (hypertension), peripheral arterial disease (ischaemia of the limbs), arrhythmias, congenital heart disease and rheumatic heart disease. The facts are unequivocal and disturbing; CVD is the leading cause of death worldwide [10-12].

Chronic diseases are disorders with a long duration and generally slow progression. They comprise four major non-communicable diseases (NCDs) as listed by the World Health Organization (WHO), namely CVD, cancer, chronic respiratory disease and diabetes [13], which are now reaching epidemic proportions in low- and middle-income countries (LIMIC) of the world [14-18]. NCDs constitute the major global health burden of the 21st century [19-20] without discriminating among age groups [21]. Chronic diseases are implicated in 35 million deaths annually worldwide and a large portion of these deaths occurs due to CVD in LIMIC [22].

There is a rising epidemic of NCDs in sub-Saharan Africa (SSA). However, as in other LIMICs, individuals in SSA suffer from the dual burdens of infectious disease and NCDs [22, 23]. Walker and colleagues [24] reported that SSA continues to suffer under the weight of infectious diseases such as HIV and malaria, as well as high rates of undernutrition. Facing these issues in conjunction with the chronic diseases that accompany high rates of overnutrition is a daunting task [25] for the health burden in Africa. SSA has a disproportionate burden of both infectious and chronic diseases compared with other parts of the world [26]. South Africa (SA) is a country of great diversity extending from highly industrialized cities with an urban advanced-economy lifestyle to remote rural areas with more traditional lifestyles. SA, like many SSA countries, is not immune to the NCD epidemic accompanied by the continued burden of undernutrition. In SA, approximately 28% of deaths annually are attributed to infectious diseases, while NCDs account for 25% of the lives lost [27]. The burden of diseases related to NCDs is predicted to rise substantially in SA over the next decade if necessary measures are not in place to combat the trend [28]. WHO estimates the burden from NCDs in SA to be two to three times higher than that in developed countries [13].

Approximately 35-65% of all deaths worldwide occur due to CVDs and death rates exceed these estimated figures owing to malnutrition and infections [29, 30]. CVDs and their risk factors are increasing in SSA [17, 31] with a high prevalence of ischaemic heart disease among men in their sixties followed by women of the same age group [17]. The common potential risk factors for NCDs are tobacco use, physical inactivity and an unhealthy diet, which all lead to CVD, diabetes and cancer [32, 33]. This burgeoning epidemic of NCDs has many root causes. Additional perpetuators of these epidemics are globalization and urbanization [34-37] with abdominal obesity contributing significantly to CVD in the SSA region [38]. Compelling evidence demonstrates a rise in mortality and morbidity from the NCDs in all strata of South African society. Leeder and colleagues [39] estimated that even without changes in the risk factor profile or the mortality rates from CVD, the demographic changes will result in a doubling of the number of cardiovascular deaths in SA by 2040. Chronic diseases such as CVD, obesity and diabetes have therefore become at least as important as infectious disease.

In summary, CVD is a substantial and growing problem in most of the developing regions of the world. The burden of NCD on the African continent and in SA in particular continues to demonstrate the potential for a sustained rise. A significant investment in the health care system and in particular the primary health care system is therefore justified. Further innovative strategies and plans are needed to address the determinants of this disease burden. However, indications still point to the paucity of community-based studies aimed at investigating NCD prevalence, incidence and risk factors. Consistent evidence suggests that diets rich in fruit and vegetables and other plant foods are associated with moderately lower overall mortality rates and lower death rates from chronic diseases including CVD [3-6]. The ‘antioxidant hypothesis’ proposes that carotenoids, polyphenols, vitamin C, vitamin E and other antioxidant nutrients afford protection against CVD by decreasing oxidative damage [7-9]. As the evidence began to mount from animal studies and human epidemiologic studies on potential protective effects of antioxidants, excitement in both the lay and medical communities also began to increase. The idea that natural compounds, if taken in supplement form, may offer a broad and inexpensive means of decreasing the risk for CVD and other age-related diseases is a very attractive hypothesis. Enthusiasm has grown to the point where people around the globe have become aware of the need to consume a diet with a high content of fruit and vegetables.

Indeed, evidence from experimental, clinical and epidemiologic studies has unequivocally pointed to oxidative stress as the key culprit in the pathogenesis of CVD [1, 2, 40, 41]. CVD continues to remain a significant problem in developed countries and is a growing health concern worldwide. Although death rates from CVD have decreased in many countries, due to advances in the field of medicine, the prevalence of CVD risk factors continues to increase. Diet is a centrally important, modifiable risk factor in the prevention of CVD [221-224].

Advertisement

2. Can vegetable oils and nuts be the natural solution for CVD?

Oxidative stress is common in many clinically important cardiac disorders, including ischaemia/reperfusion (I/R) injury, diabetes and hypertensive heart disease [42-46]. Several animal models suggest that when endogenous anti-oxidant systems are compromised, as is the case under oxidative stress conditions, exogenous antioxidant supplementation can be used for preventive and/or therapeutic intervention of CVD [42, 43, 47-49].

2.1. Composition and health benefits of vegetable oils

2.1.1. Dietary fats

Fats are the most concentrated form of energy for the body. They also aid in the absorption of fat-soluble vitamins (A, D, E and K) and other fat-soluble biologically active components [50]. Chemically, most of the fats in foods are triglycerides, made up of a unit of glycerol combined with free fatty acids, each of which may be the same or different. Other dietary fats include phospholipids, phytosterols and lipoproteins associated with cholesterol [50-52]. A balanced diet, including oils and fats that supply energy and essential fatty acids is needed for good health.

The different types of fatty acids are the most important characteristics of dietary fats. According to the degree of unsaturation (double bonds and hydrogen content), fatty acids are largely classified into three major types: saturated fatty acids, monounsaturated fatty acids (MUFA) and polyunsaturated fatty acids (PUFA). A fourth form, the trans fatty acids, are mainly produced by partial hydrogenation of polyunsaturated oils in food processing but also occur naturally in animal foods in small amounts [53].

Fatty acids consist of a hydrocarbon chain with a hydrophobic methyl group at one end and a hydrophilic carboxyl group at the other end. Greek letters (α, β, γ, ω) have been used to identify the location of the double bonds in fatty acids. The “alpha” carbon is the carbon closest to the carboxyl group. The methyl group of the molecule is also referred to as the omega end and the terminal carboxyl group is located at the delta end. Current chemical numerical terms number the carbon chain form one to “n”, with n being the last carbon at the methyl end. The terms “n” and “omega” are synonymous [54].

2.1.2. Saturated fat

Saturated fatty acids contain no double bond; they are fully saturated with hydrogen. The main saturated fatty acids are lauric acid (C12:0), myristic acid (C14:0), palmitic acid (C16:0) and stearic acid (C18:0). Saturated fats are found in animal-based products, such as milk, cream, butter and cheese, meat from most land animals, palm oil and coconut oil, as well as manufactured products made from these, such as pies, biscuits, cakes and pastries [55].

2.1.3. Monounsaturated fat (MUFA)

MUFAs are predominant in vegetable oils, such as olive oil, canola oil and peanut oil and are also found in high proportions in animal fats [56]. Much of the interest in the role of MUFA in the prevention of coronary heart disease (CHD) stems from the observed beneficial effects of the Mediterranean diet [57], which includes high consumption of olive oil. MUFAs are less susceptible to oxidation when compared to PUFAs. This in turn leads to increased availability of antioxidants in the active form and better stability of olive oil [58-61]. Olive oil also contains some antioxidant micronutrients, namely polyphenols and squalene [58, 62-64]. The main MUFA in the human diet is oleic acid (C18:1n-9), which has one double bond. MUFA intake has been associated with a slight cardioprotective effect [65]. MUFAs are known to have a beneficial effect on the serum lipid profile and thus decrease the risk of CVD [66-68]. Furthermore, these fatty acids are stable in oxidative stress conditions and are less likely to react with reactive oxygen species (ROS) when compared with PUFA [58-59]. However, studies reporting associations between dietary intake of MUFAs and CHD risk have been inconclusive [69-71].

2.1.4. Polyunsaturated fat (PUFA)

PUFAs are naturally occurring endogenous substances, present in almost all tissues and are essential components of all mammalian cells. They are essential for survival and cannot be synthesized in the body. Hence, they have to be obtained in our diet and are therefore essential [54, 72]. There are two types of naturally occurring PUFAs in the body, the (n-6) PUFAs derived from linoleic acid (LA, C18:2) and the (n-3) PUFAs derived from α-linolenic acid (ALA, C18:3). They are categorized depending on the location of their first double bond: (n-3) PUFAs have their first double bond located at the third carbon molecule and (n-6) PUFAs at the sixth. Both of these two forms of PUFAs are metabolized by the same set of enzymes as their respective long-chain metabolites [73]. The differences between (n-3) and (n-6) PUFAs are shown in Table 1 below.

Vegetable oils are the predominant sources of alpha linolenic acid (ALA). ALA is found in legumes, flax seeds, walnuts, pinto beans, soybeans and spinach [74]. Dietary intake of ALA among Western adults is typically in the range of 0.5–2g/d [75]. The (n-6) PUFA is the main PUFA in most Western diets and is typically consumed in greater amounts than ALA [75, 76]. The evidence for a beneficial role of dietary (n-6) PUFAs is less convincing and for the purpose of this chapter we will focus on the (n-3) PUFA. The three main forms of (n-3) PUFAs are ALA, eicosapentaenoic acid (EPA, C20:5 n-3) and docosahexaenoic acid (DHA, C22:6 n-3) [77], with ALA being the simplest form. The (n-3) PUFAs are a family of biologically active fatty acids. The simplest member of this family, ALA, can be converted to the more biologically active and very long-chain (n-3) PUFAs; EPA and DHA. This process, as shown in Figure 1, occurs by a series of desaturation and elongation reactions, with stearidonic acid being an intermediate in the pathway [54, 75, 78].

Research has shown that long-chain (n-3) PUFAs protect against CVD [77, 79-82]. The cardioprotective effects of (n-3) PUFAs have long been recognized. Epidemiologic data suggest that (n-3) PUFAs derived from fish oil reduce CVD. Fish oil is a rich source of EPA (C20:5 n-3) and DHA (C22:6 n-3) (Table 1) [67, 83, 84]. The cardioprotective roles of these two forms of (n-3) PUFA are extensively reviewed by Bester and co-workers [48]. Fish oil may also reduce mortality after a cardiovascular incident, as it plays a role in reducing potentially fatal arrhythmias ([85-87]. There are several prospective studies relating the use of fish or the intake of long-chain (n-3) PUFAs to lower risk of CVD [88, 89]. Long chain (n-3) PUFAs have several beneficial cardiovascular properties, including antiatherothrombotic, antiarrhythmic, anti-inflammatory, antihypertensive and triglyceride lowering [81, 90, 91]. In summary, studies investigating the dietary roles of fatty acids demonstrate that dietary supplementation with (n-3) PUFAs decreases cardiac deaths, nonfatal cardiovascular events and all-cause mortality. These benefits are most apparent in high-risk patients. (n-3) PUFA supplementation appears to confer additional benefits in patients eating a Mediterranean diet.

The original observation is from almost 57 years ago, when Hugh M. Sinclair [92] published his observations on the negative effects of essential fatty acid deficiency on CVD. He strengthened his hypothesis by noting the low mortality rate from CHD (coronary heart disease) in Greenland Eskimos, a population consuming a high fat diet, but rich in (n-3) PUFAs [92]. Clinical studies suggest that (n-3) PUFAs reduce mortality from coronary heart disease and the rate of sudden cardiac death [92-95]. Significant antiarrhythmic effects of (n-3) PUFAs were observed in some but not all human studies on atrial fibrillation [96, 97]. In addition, animal studies show strong antiarrhythmic effects of (n-3) PUFAs [98-102].

  (n-3) PUFA (n-6) PUFA
Molecular structure First double-bond on the third carbon counting from the methyl end (the “nth” carbon) First double-bond on the sixth carbon counting from the methyl end (the “nth” carbon)
Types α-Linolenic acid (ALA) [C18:3]
Eicosapentaenoic acid (EPA) [C20:5]
Docosahexaenoic acid (DHA) [C22:6]
Linoleic acids (LA) [C18:2]
Arachidonic acid (AA) [C20:4]
Food sources Flaxseed oil (ALA)
Canola oil (ALA)
Soybean oil (ALA)
Oily fish (EPA/DHA)
Fish oil capsules (EPA/DHA)
Corn oil(LA)
Soybean oil (LA)
Sunflower oil (LA)
Poultry (AA)
Meats (AA)

Table 1.

Molecular structure, types and food sources of (n-3) and (n-6) PUFAs.

Figure 1.

The biosynthesis of (n-3) PUFA.

Long-chain (n-3) PUFAs are important constituents of all cell membranes and confer on membranes properties of fluidity and thus, determine and influence the behaviour of membrane-bound enzymes and receptors [103-107]. These PUFAs are found in abundance in the myocardium, retina, brain and spermatozoa, and are essential for the proper functioning of these tissues and growth, being important modulators of many physiological processes. The fact that these tissues have developed the cellular machinery to preferentially incorporate these minor dietary components into their membranes suggests that these PUFAs play a role in the proper function of the cell [108-110].

The fatty acid composition of myocardial membrane phospholipids, in particular, is sensitive to the type of fatty acid consumed in the diet. Studies show that indeed the myocardium and myocardial membrane phospholipids are rich in (n-3) PUFAs after fish oil consumption [111,112]. Diet-induced changes in the PUFA composition of a cell membrane have an impact on the cell’s function, partly because these fatty acids represent a reservoir of molecules that perform important signalling roles within and between cells. In particular, dietary (n-3) PUFAs compete with dietary (n-6) PUFAs for incorporation into all cell membranes [113,114]. (n-3) PUFAs modulate the expression of adhesion proteins such as selectins [115] and exert an effect by modulating the intracellular signalling pathways associated with the control of transcription factors (e.g., nuclear factor-κB) and gene transcription [116,117]. Research has shown that enrichment of monocyte membranes with (n-3) PUFAs results in the synthesis and secretion of reduced quantities of cytokines (e.g., tumour necrosis factor-α, interleukin-1β) that are involved in the amplification of the inflammatory response [117,118]. Therefore, at a cellular level, (n-3) PUFAs from fish oils can directly or indirectly modulate a number of cellular activities associated with inflammation.

2.1.5. Polyphenols

Polyphenols constitute one of the most numerous and ubiquitously distributed groups of plant secondary metabolites, with more than 8000 phenolic structures currently known. Natural polyphenols can range from simple molecules (phenolic acids, phenylpropanoids, and flavonoids) to highly polymerised compounds (lignins, melanins, tannins), with flavonoids representing the most common and widely distributed sub-group [119]. These secondary plant metabolites are known to have potential antioxidant activity and radical scavenging capacity [120-124]. Polyphenols are gaining increased importance due to their beneficial effects on health. Flavonoids are the most abundant polyphenols in our diets. They can be divided into several classes according to the degree of oxidation of the oxygen heterocycle: flavones, flavonols, isoflavones, anthocyanins, flavanols, proanthocyanidins and flavanones [125]. A complication of the epidemiological observations regarding members of the flavonoid family is that subtle differences in their chemical structures can translate into marked differences in their absorption, metabolism and bioactivities [126]. South African herbal teas, rooibos (Aspalathus linearis) and honeybush (Cyclopia ssp.) are currently gaining popularity worldwide [127, 128], owing to their anti-oxidant, anti-cancer and anti-mutagenic properties [129-131]. Rooibos is a herbal tea made from the leaves and stems of the indigenous South African plant, Aspalathus linearis (Brum.f) Dahlg. (family Fabaceae; tribe Crotalarieae) [132,133]. Research has demonstrated that this herbal tea is rich in flavonoids [127, 134]. Animal studies that have investigated the cardioprotective effects of natural or synthetic flavonoids have focused mainly on the acute pharmacological activity of these compounds. For example, in vivo studies using animal models have reported acute cardioprotection obtained from intravenous injections of natural or synthetic flavonoids [135,136].

2.1.6. Vitamin E

Natural vitamin E is composed of eight chemical compounds: α-, β-, γ- and δ-tocopherols and their corresponding tocotrienols. α-Tocopherol is the most active form of vitamin E in vitro. The tocopherols are saturated forms of vitamin E, whereas the tocotrienols are unsaturated and have an isoprenoid side chain. Tocopherols possess a chromanol ring and a 15-carbon tail. The presence of three trans double bonds in the tail distinguishes tocopherols from tocotrienols [137-139]. This may account for the differences in their efficacy and potency in vitro and in vivo [140,141].

Red palm oil (RPO) is a rich source of vitamin E. It contains 560–1000 parts per million of vitamin E, of which approximately 18–22% are tocopherols and 78–82% tocotrienols [142-144]. RPO has been shown to offer protection against I/R injury [42, 43, 47, 48] leading to a reduction in oxidative stress [145]. It has also been suggested that palm oil may have some anti-arrhythmogenic effects, which may reduce sudden death after ischaemic incidents [146].

Of all the vegetable oils, RPO has the highest content of tocotrienols with γ-tocotrienol the most abundant. This form of vitamin E has been demonstrated to reduce cholesterol production and platelet aggregation [147-151]. RPO may also exert a neutral or positive effect on the serum lipid profile through the effects of its fatty acid composition and tocotrienols [152-155]. Investigations into vitamin E showed that tocotrienols are more potent than tocopherols as antioxidants. The tocotrienols present in palm oil have been shown to offer protection from myocardial I/R injury in an isolated perfused rat heart model [156, 157]. Animal studies with tocopherols and tocotrienols that investigate these compounds’ potential against chronic diseases are extensively reviewed by Aggarwal and co-workers [158]. These authors argue that the evidence overwhelmingly suggests that tocotrienols may be superior in their biological properties than tocopherols and that their anti-inflammatory and antioxidant activities could prevent CVD among other chronic diseases.

2.1.7. Carotenoids

Carotenoids are nature’s most widespread pigments, well known for their orange-red to yellow colours, which they impart to many fruits and vegetables. These fat-soluble phytochemicals have also received substantial attention because of their provitamin A and antioxidant roles [159]. Carotenoids are polyenoic terpenoids with conjugated trans double bonds. They include carotenes (β-carotene and lycopene), which are polyene hydrocarbons and xanthophylls (lutein, zeaxanthin, capsanthin, canthaxanthin, astaxanthin and violaxanthin) that have oxygen in the form of hydroxy, oxo, or epoxy groups [160]. The majority of the 600 carotenoids found in nature are 40 carbons in length and may be pure hydrocarbons, called carotenes, or possess oxygenated functional groups, in which case they are called xanthophylls [161]. The long-chain conjugated polyene structure accounts for the ability of these compounds to absorb visible light, but also makes them quite susceptible to oxidation. This latter property is closely related to their ability to act as antioxidants [162].

The properties and therefore functions of a carotenoid molecule are primarily dependent upon its structure and hence its chemistry [163]. In particular, the conjugated C = C double bond system is associated with energy transfer reactions, such as those found in photosynthesis [164]. In human plasma and tissues, several carotenoids have been well characterized including cyclic (such as β-carotene and α-carotene) and acyclic carotenes (such as lycopene and phytoene), together with a number of xanthophylls (such as zeaxanthin, lutein and beta-cryptoxanthin), all of which can be directly derived from dietary sources [165]. Carotenoids have generated considerable interest as several studies have suggested an inverse association between the dietary intake of carotenoids and the risk for CVD [166, 167]. Conversely prooxidant roles of these phytochemicals have also been reported [168-170].

2.1.8. Possible mechanism(s) of action

As mentioned earlier, RPO supplementation does offer protection against myocardial I/R injury via several suggested mechanisms. Amongst the proposed mechanisms are the NO–cyclic GMP pathway, phosphorylation of mitogen-activated protein kinases and scavenging of deleterious reactive oxygen species by RPO [42, 43, 47, 48].

Investigations concerning (n-3) PUFAs show that these forms of essential fatty acids reduce the risk of sudden cardiac death as well as fatal and nonfatal myocardial infarction [171-173]. A number of mechanisms have been implicated in the protective effects of (n-3) PUFAs [174, 175]. The (n-3) PUFAs have been demonstrated as altering the transcription of specific genes. These effects are mediated by a variety of mechanisms that involve indirect (i.e., by eicosanoids, hormones) and direct nuclear effects on genes. The PUFAs (i.e., both (n-3) and (n-6) PUFAs) modulate the expression of genes involved in lipogenesis, glycolysis, production of glucose transporters, inflammatory mediators, early response genes and genes for cell adhesion molecules [176, 177].

The primary source of MUFA that lowers cholesterol levels is olive oil [178, 179]. It is evident that olive oil, due to its micronutrient content and fatty acid composition, can play a vital role in maintaining beneficial serum lipid profiles. Together with its ability to reduce systemic oxidative stress, blood pressure and inflammation, it has become an appropriate dietary supplement for lowering the risk of CHD.

2.2. Composition and health benefits of nuts

Nuts are highly nutritious and of prime importance for people in several regions in Asia and Africa. Most nuts contain a great deal of fat (e.g., pecan 70%, macadamia nut 66%, Brazil nut 65%, walnut 60%, almonds 55% and peanut butter 55%). Most have a good protein content (in the 10–30% range) and only a few have a very high starch content [180]. Many nuts have also been identified as especially rich in antioxidants [181, 182]. Nuts therefore constitute one of the most nutritionally concentrated kinds of food available. Most nuts, left in their shell, have a remarkably long shelf life and can conveniently be stored for winter use [183]. Nuts are foods rich in fat, ranging from 46% in cashews and pistachios to 76% in macadamia nuts and provide 20–30 kJ/g per nut. Despite their high fat content, they are not harmful because they contain a low proportion (4–16%) of saturated fatty acids. Nearly one half of the fat content of nuts consists of unsaturated fatty acids, including both mono- (oleic acid) and poly- (linoleic and α-linolenic acid) unsaturated fatty acids (MUFA and PUFA respectively). The fatty fraction of nuts also contains plant sterols with anti-oxidants [184] and cholesterol-lowering effects [185]. Nuts are also rich sources of other bioactive macronutrients, such as protein (25% of energy) and dietary fibre, which ranges from 4 to 11g/100 g and in standard servings provide 5–10% of daily fibre requirements. They also contain significant micronutrients (Table 2), among them folate [185] antioxidant vitamins (e.g., tocopherols) and phenolic compounds [183].

Table 2.

Composition of Nuts (data from the US department of agriculture nutrient database)

By virtue of their unique composition, nuts are likely to benefit modern cardiovascular risk biomarkers, such as LDL oxidizability, soluble inflammatory molecules and endothelial dysfunction. The complex pathophysiology of atherosclerotic disease has evolved beyond the accumulation of cholesterol in the arterial wall. A series of circulating, functional, structural and genomic biological markers that reflect arterial vulnerability have been proposed as potential novel risk factors for the development of CVD (Vasan, 2006). Among them, biomarkers for oxidation [186], inflammation [187] and endothelial dysfunction [188] have received increasing attention.

Studies had shown that whole, unprocessed and unpeeled nuts have a unique composition that consists of important macro- and micronutrients, which give nuts their multiple beneficial effects on cardiovascular outcomes [189-192]. Most nut constituents have shown beneficial effects when clinically tested, in isolation or as part of enriched foods, for effects on diverse cardiovascular outcomes, including novel risk markers [189-192].

2.2.1. Antioxidant effects

Nuts are important sources of tocopherols and phenolic antioxidants, which protect against LDL oxidation [183]. Walnuts have been shown to contain substantial amounts of melatonin, which contributed to a significant antioxidant effect in an experimental rat model [193]. In addition, a substantial fraction of nut fat comes from MUFAs, which are not susceptible to oxidation. The PUFAs are contained mainly in walnuts and are more susceptible to oxidation. However, nuts are a rich source of many antioxidants, which protect the PUFA in vivo against oxidative modification [194].

2.2.2. Anti-inflammatory effects

Plasma high-sensitivity CRP, an accepted measure for systemic low-grade inflammation, was a secondary outcome in several controlled nut feeding trials conducted in hypercholesterolemic subjects with almonds [195-198] or walnuts [197, 199]. Some of them have demonstrated a CRP-lowering effect [196, 197, 198]. Zhao et al., who used walnuts and walnut oil to enrich the diet in PUFA and especially ALA, showed a decrease in inflammatory markers [197] and proinflammatory cytokine production by mononuclear cells [197].

2.2.3. Effects on endothelial function

Endothelial dysfunction is a critical event in atherogenesis and is implicated both in early disease and in advanced atherosclerosis [201]. Short-term feeding studies have shown consistently that diets rich in saturated fatty acids impair endothelial function [181, 202, 203] and that even a single fatty meal rich in saturated fatty acids is followed by transient endothelial dysfunction [204, 205]. These detrimental effects can be counteracted by the administration of PUFA and other nutrients contained in nuts, such as antioxidant vitamins and arginine [179]. Another feeding trial showed that, compared with an isoenergetic Mediterranean diet with similar saturated fatty acid content, a walnut diet attenuated the endothelial dysfunction associated with hypercholesterolemia [199]. Moreover, changes in circulating levels of cellular adhesion molecules critical to leukocyte recruitment on the arterial wall also reflect endothelial dysfunction [201]. Several studies have shown that diets enriched with ALA from walnuts [197, 199, 206] reduce endothelial activation as assessed by decreased plasma cellular adhesion molecules. Walnut feeding also reduced the expression of endothelin-1, a potent endothelial activator in an animal model of accelerated atherosclerosis [207].

2.2.4. Effects on body weight changes

As the interest in incorporating nuts into the diet grows, it is important that consumers understand how to include them in a healthy diet without promoting weight gain. They are high-fat, energy dense foods and are therefore a potential threat for contributing to positive energy balance. Numerous epidemiological and clinical studies have shown that nuts are not associated with higher body weight [208, 209] or weight gain [210-215]. This could be attributed along with other potential mechanisms for the high satiety properties of nuts [216]. The enhanced satiety, which is also achieved via other mechanisms such as the decreased eating rate [217], leads to reduced energy consumption and therefore a decreased risk of weight gain and obesity.

Blomhoff et al. [190] argued that the inverse association between nut intake and cardiovascular and coronary heart diseases in epidemiological studies may, or may not, be associated with antioxidants. According to these authors, epidemiologic studies are not ideally suited for studying the role of specific nuts or biological mechanisms. Nevertheless, they are in agreement with findings supporting the theory that a complex and rich mix of nut constituents is able to offer protection against CVD and perhaps other chronic diseases [183].

2.2.5. Possible mechanism(s) of action

Epidemiologic and clinical trial evidence has demonstrated the beneficial effect of nut consumption on coronary heart disease and its associated risk factors. The cardioprotective properties of nuts, due partially to their favourable lipid fatty acid profile (rich in unsaturated fatty acids), exceed the LDL-C lowering. Nuts, especially walnuts, contain (n-3) PUFAs, which have been shown to have a favourable impact on multiple factors related to CVD, such as inflammation, platelet function, arrhythmias, hypertriglyceridemia and nitric oxide-induced endothelial relaxation [218]. Nuts are also excellent sources of other bioactive compounds such as vegetable protein, dietary fibre, potassium, calcium, magnesium, tocopherols, phytosterols, phenolic compounds, resveratrol and arginine [179]. This unique nutrient composition explains the benefits of nut consumption for the prevention of CVD through mechanisms of oxidation, inflammation and vascular reactivity.

Advertisement

3. Conclusions and future directions

Investigation of the mechanisms underlying CVD showed that the disease has a complex cause beyond the accumulation of cholesterol on the arterial wall, with enhanced oxidative stress and a prominent inflammatory response. Diet has been shown to be associated with cardiovascular events. PUFAs are essential in our diet because we cannot synthesize them. They are also essential nutrients for optimal health of the cardiovascular, nervous and undoubtedly other organ systems. Dietary (n-3) PUFAs are incorporated into the cellular membranes of all tissues. The extent of incorporation into tissue membranes is dependent on dietary intake. The enrichment of membranes with (n-3) PUFAs can modulate cellular signalling events, membrane protein function and gene expression.

Interest in the possible health benefits of flavonoids has increased owing to their potent antioxidant and free-radical scavenging activities observed in vitro. There is growing evidence from human feeding studies that the absorption and bioavailability of specific flavonoids is much higher than originally believed. However, epidemiologic studies exploring the role of flavonoids in human health have been inconclusive. Some studies support a protective effect of flavonoid consumption in CVD and cancer; other studies demonstrate no effect and a few studies suggest potential harm. More recently, results from human studies provide evidence that rooibos can offer protection against oxidative stress conditions such as CVD [131,219]. In a study by Pantsi et al., the beneficial effects of dietary rooibos flavonoids were observed ex vivo in isolated perfused rat hearts. Epidemiological studies suggest that the beneficial cardiovascular health effects of diets rich in fruit and vegetables are in part mediated by their flavonoid content, with particular benefits provided by one member of this family, the flavonols [49].

Polyphenols are abundant micronutrients in our diet and evidence for their role in the prevention of degenerative diseases is emerging. Bioavailability differs greatly from one polyphenol to another, so the most abundant polyphenols in our diet are not necessarily those leading to the highest concentrations of active metabolites in target tissues. Because there are many biological activities attributed to the flavonoids, some of which could be beneficial or detrimental depending on specific circumstances, further studies in both the laboratory and with populations are warranted.

However, the fatty acid components of nuts may differently influence oxidation processes and this needs to be considered for the synergy or opposition to the effects of constituent antioxidants. There is growing evidence that dietary polyphenols in nuts, tea and wine may have anti-inflammatory effects, mediated by both their antioxidant action and modulation of signal transduction pathways, such as the nuclear transcription factor kB, with ensuing down-regulation of inflammatory genes in endothelial cells and macrophages [220]. The increased diversity and availability of sources of dietary fatty acids will likely allow the continued expansion of food products fortified with these fatty acids, a trend that may result in the attainment of the recommended dietary intake of these nutrients.

Future studies in oils should be carried out in order to elucidate the effects of oils in various models in which effects remain unknown. Little is known about the effects of nuts on a diseased heart. Studies should be performed to test whether nuts may offer protection against the severity or progression of various models of CVD.

References

  1. 1. Ceconi C, Boraso A, Cargnoni A, Ferrari R. Oxidative stress in cardiovascular disease: myth or fact? Biochemistry and Biophysics 2003; 420: 217-221.
  2. 2. Dalle-Donne I, Rossi R, Colombo R, Giustarini D, Milzani A. Biomarkers of oxidative damage in human disease. Clin Chem 2006; 52: 601–623.
  3. 3. He FJ, Nowson CA, Lucas M, MacGregor GA. Increased consumption of fruit and vegetables is related to a reduced risk of coronary heart disease: meta-analysis of cohort studies. J Hum Hypertens 2007; 21: 717–728.
  4. 4. Dembitsky VM, Poovarodom S, Leontowicz H, Leontowicz M, Vearasilp S, Traktenberg S. The multiple nutrition properties of some exotic fruits: Biological activity and active metabolites. Food Research International 2011; 44: 1671–1701.
  5. 5. Devalaraja S, Jain S, Yadav H. Exotic fruits as therapeutic complements for diabetes, obesity and metabolic syndrome. Food Research International 2011; 44: 1856–1865.
  6. 6. Sant'Ana AS. Special issue on exotic fruits. Food Research International 2011; 44: 1657.
  7. 7. Rufino MSM, Ricardo E, Alves RE, Brito ES, Jiménez JP, Calixto FS et al. Bioactive compounds and antioxidant capacities of 18 non-traditional tropical fruits from Brazil. Food Chemistry 2010; 121: 996–1002.
  8. 8. Borges GSC, Vieira FGK, Copetti C, Valdemiro GL, Zambiazi RC, Mancini Filho J. Chemical characterization, bioactive compounds, and antioxidant capacity of Jussara (Euterpe edulis) fruit from the Atlantic Forest in southern Brazil. Food Research International 2011; 44: 2128–2133.
  9. 9. Coimbra MC, Jorge N. Proximate composition of guariroba (Syagrus oleracea), jerivá (Syagrus romanzoffiana) and macaúba (Acrocomia aculeata) palm fruits. Food Research International 2011; 44: 2139–2142.
  10. 10. Yach D, Hawkes C, Gould CL, Hofman KJ. The global burden of chronic diseases: overcoming impediments to prevention and control. JAMA 2004; 291: 2616 –2622.
  11. 11. Fuster V, Voute J. MDGs: chronic diseases are not on the agenda. Lancet 2005; 366: 1512-1514.
  12. 12. Goyal A, Yusuf S. The burden of cardiovascular disease in the Indian subcontinent. Indian J Med Res 2006; 124: 235-244.
  13. 13. World Health Organization. The Global Burden of Disease: 2004 Update. Geneva: WHO, 2008.
  14. 14. Ezzati M, Vander Hoorn S, Lawes CM et al. Rethinking the ‘diseases of affluence’ paradigm: global patterns of nutritional risks in relation to economic development. PLoS Med 2005; 2: e133.
  15. 15. Connor MD, Walker R, Modi G, Warlow CP. Burden of stroke in black populations in sub-Saharan Africa. Lancet Neurol 2007; 6: 269–278.
  16. 16. Daar AS, Singer PA, Persad, et al. Grand challenges in chronic non-communicable diseases. Nature 2007; 450: 494-496.
  17. 17. Mensah GA. Ischaemic heart disease in Africa. Heart 2008; 94: 836–843.
  18. 18. Parkin DM, Sitas F, Chirenje M et al. Cancer in indigenous Africans—burden, distribution and trends. Lancet Oncol 2008; 9: 683–692.
  19. 19. Alwan A, Maclean DR, Riley LM, d’Espaignet ET, Mathers CD, Stevens GA, Bettcher D, Narayan KM, Ali MK, Koplan JP. Global noncommunicable disease - where worlds meet. N Engl J Med 2010; 363: 1196-1198.
  20. 20. Beaglehole R, Horton R. Chronic diseases: global action must match global evidence. Lancet 2010; 376: 1619-1621.
  21. 21. Narayan KM, Ali MK, Koplan JP. Global noncommunicable diseases - where worlds meet. N Engl J Med 2010;363:1196-1198.
  22. 22. Lopez AD, Mathers CD, Ezzati M, Jamison DT, Murray CJ. Global and regional burden of disease and risk factors, 2011: systematic analysis of population health data. Lancet 2006; 367: 1747-1757.
  23. 23. Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. PLoS Med 2006; 3: e442.
  24. 24. Walker A, Walker B, Segal I. Some puzzling situations in the onset, occurrence and future of coronary heart disease in developed and developing populations, particularly such in sub-Saharan Africa. Journal of the Royal Society for the Promotion of Health 2004; 124: 40–46.
  25. 25. Salman Z, Kirk G, DeBoer M. High rate of obesity-associated hypertension among primary schoolchildren in Sudan. International Journal of Hypertension, Volume 2011, Article ID 629492, 5 pp.
  26. 26. Dalal S, Beunza JJ, Volmink J, Adebamowo C, Bajunirwe F, Njelekela M, Mozaffarian D, Fawzi W, Willet W, Adami H, Holmes MD. Non-communicable disease in sub-Saharan Africa: what we know. International Journal of Epidemiology 2011; 40: 885-901.
  27. 27. Steyn K, Bradshaw D, Norman R, Laubscher R. Determinants and Treatment of Hypertension in South Africa/Determinants of Hypertension and Its Treatment in South Africa During 1998: The First Demographic and Health Survey. Tygerburg, South Africa: South Africa Medical Research Council; 2003.
  28. 28. Abegunde DO, Mathers CD, Adam T, Ortegon M, Strong K. The burden and costs of chronic diseases in low-income and middle-income countries. Lancet 2007; 370: 1929–1938.
  29. 29. Critchley J, Liu J, Zhao D, Wei W, Capewell S. Explaining the increase in coronary heart disease mortality in Beijing between 1984 and 1999. Circulation 2004; 110: 1236–1244.
  30. 30. Gaziano T. Global burden of cardiovascular disease. Braunwald’s Heart Disease: A Textbook of Cardiovascular Medicine, 8th ed. Saunders; 2008: 1–22.
  31. 31. Vorster HH. The emergence of cardiovascular disease during urbanisation of Africans. Public Health Nutr 2002; 5: 239–243.
  32. 32. Medical Research Council. South Africa Demographic and Health Survey 2003. Pretoria: Department of Health, 2007.
  33. 33. Norman R, Bradshaw D, Schneider M, et al. and the SA CRA Collaborating Group. A Comparative risk assessment for South Africa in 2000: towards promoting health and preventing disease. S Afr Med J 2007; 97: 637–641.
  34. 34. Misra A, Khurana L. Obesity and the metabolic syndrome in developing countries. Journal of Clinical Endocrinology and Metabolism 2008; 93: S9–S30.
  35. 35. Abdulai A. Socio-economic characteristics and obesity in underdeveloped economies: does income really matter? Applied Economics 2010; 42: 157–169.
  36. 36. Maher D, Smeeth L, Sekajugo J. Health transition in Africa: practical policy proposals for primary care. Bulletin of the World Health Organization 2010; 88: 943–948.
  37. 37. Greenberg H, Raymond S, Leeder S. The prevention of global chronic disease: academic health’s new frontier. American Journal of Public Health 2011; 101: 1386–1391.
  38. 38. Steyn K, Sliwa K, Hawken S et al. Risk factors associated with myocardial infarction in Africa: the INTERHEART Africa study. Circulation 2005; 112: 3554–3561.
  39. 39. Leeder SR, Raymond SU, Greenberg H. A race against time: the challenge of cardiovascular diseases in developing economies. New York: Columbia University, 2004.
  40. 40. Manikandan P, Sumitra M, Aishwarya S, Manohar BM, Lokanadam B, Puvanakrishnan R. Curcumin modulates free radical quenching in myocardial ischaemia in rats. Int J Biochem Cell Biol 2004; 36: 1967–1980.
  41. 41. Ungvari Z, Gupte SA, Recchia FA, Batkai S, Pacher P. Role of oxidative-nitrosative stress and downstream pathways in various forms of cardiomyopathy and heart failure. Curr Vasc Pharmacol 2005; 3: 221–229.
  42. 42. Esterhuyse AJ, du Toit EF, Benade AJS, van Rooyen J. Dietary red palm oil improves reperfusion cardiac function in the isolated perfused rat heart of animals fed a high cholesterol diet. Prostaglandins, Leukotrienes and Essential Fatty Acids 2005; 72: 153 161.
  43. 43. Engelbrecht AM, Esterhuyse J, du Toit EF, Lochner A, van Rooyen J. p38-MAPK and PKB/Akt, possible role players in red palm oil-induced protection of the isolated perfused rat heart? Journal of Nutritional Biochemistry 2006; 17: 265-271.
  44. 44. Qin CX, Williams SJ, Woodman OL. Antioxidant activity contributes to flavonol cardioprotection during reperfusion of rat hearts. Free Radical Biology and Medicine 2011; 51: 7-1437.
  45. 45. Yu E, Mercer J, Bennett M. Mitochondria in vascular disease, Cardiovascular Research 2012; 95: 2-173.
  46. 46. Morsy MD, Bashir SO. Alpha-tocopherol ameliorates oxidative renal insult associated with spinal cord reperfusion injury. Journal of Physiology and Biochemistry 2013; 69, 3-487.
  47. 47. Kruger M, Engelbrecht AM, Esterhuyse J, du Toit EF, van Rooyen J. Dietary Red Palm Oil (RPO) reduces ischaemia/reperfusion injury in a hypercholesterolemic diet. Br J Nutr 2007; 97: 653-660.
  48. 48. Bester D, Esterhuyse AJ, Truter EJ, van Rooyen J. Cardiovascular effects of edible oils: a comparison between four popular edible oils. Nutrition Research Reviews 2010; 23: 334–348.
  49. 49. Pantsi WG, van Rooyen J, Marnewick JL, Esterhuyse AJ, Rautenbach F. Rooibos (Aspalathus linearis) offers cardiac protection against ischaemia/reperfusion in the isolated perfused rat heart. Phytomedicine 2011; 18: 1220–1228.
  50. 50. Fahy E, Subramaniam S, Murphy RC, et al. Update of the LIPID MAPS comprehensive classification system for lipids. J Lipid Res 2009; 50(suppl): S9–S14.
  51. 51. Quehenberger O, Armando AM, Brown AH, et al. Lipidomics reveals a remarkable diversity of lipids in human plasma. J Lipid Res. 2010; 51: 3299-3305.
  52. 52. Quehenberger O, Dennis EA. The human plasma lipidome. N Engl J Med. 2011; 365: 1812-1823.
  53. 53. Innis SM. Dietary triacylglycerol structure and its role in infant nutrition. Adv Nutr. 2011; 2: 275-283.
  54. 54. Das UN. Essential fatty acids: Biochemistry, physiology, and pathology. Biotechnology J 2006; 1: 420-439.
  55. 55. Hu FB, Stampfer MJ, Manson JE, Ascherio A, Colditz GA, Speizer FE, et al. Dietary saturated fats and their food sources in relation to the risk of coronary heart disease in women. Am J Clin Nutr 1999; 70: 1001–1008.
  56. 56. Guthrie HA, Pcciano MF. Human nutrition. Boston, MA7 WCB/ McGraw Hill; 1995, pp. 419.
  57. 57. Keys A. Mediterranean diet and public health: personal reflections. Am J Clin Nutr 1995; 61: 1321S–1323S.
  58. 58. Owen RW, Mier W, Giacosa A, et al. Phenolic compounds and squalene in olive oils: the concentration and antioxidant potential of total phenols, simple phenols, secoiridoids, lignans and squalene. Food Chem Toxicol 2000; 38: 647–659.
  59. 59. Diniz YSA, Cicogna AC, Padovani CR, et al. Diets rich in saturated and polyunsaturated fatty acids: metabolic shifting and cardiac health. Nutrition 2004; 20: 230–234.
  60. 60. Wahle KW, Caruso D, Ochoa JJ, et al. Olive oil and modulation of cell signaling in disease prevention. Lipids 2004; 39: 1223–1231.
  61. 61. Eder E, Wacker M, Lutz U, et al. Oxidative stress related DNA adducts in the liver of female rats fed with sunflower-, rapeseed-, olive- or coconut oil supplemented diets. Chem Biol Interact 2006; 159: 81–89.
  62. 62. Newmark HL. Squalene, olive oil, and cancer risk: a review and hypothesis. Cancer Epidemiol Biomarkers Prev 1997; 6: 1101–1103.
  63. 63. Quiles JL, Ochoa JJ, Ramirez-Tortosa C, et al. Dietary fat type (virgin olive vs. sunflower oils) affects age-related changes in DNA double-strand-breaks, antioxidant capacity and blood lipids in rats. Exp Gerontol 2004; 39: 1189–1198.
  64. 64. Caravaca AMG, Pancorbo AC, Diaz BC, et al. Electrophoretic identification and quantitation of compounds in the polyphenolic fraction of extra-virgin olive oil. Electrophoresis 2005; 26: 3538–3551.
  65. 65. Hu FB, Stampfer MJ, Manson JE, Rimm E, Colditz GA, Rosner BA, et al. Dietary fat intake and the risk of coronary heart disease in women. New Engl J Med 1997; 337: 1491–149.
  66. 66. Heyden S. Polyunsaturated and monounsaturated fatty acids in the diet to prevent coronary heart disease via cholesterol reduction. Ann Nutr Metab 1994; 38: 117–122.
  67. 67. Demonty I, Chan YM, Pelled D, et al. Fish-oil esters of plant sterols improve the lipid profile of dyslipidemic subjects more than do fish-oil or sunflower oil esters of plant sterols. Am J Clin Nutr 2006; 84: 1534–1542.
  68. 68. Metcalf RG, James MJ, Gibson RA, et al. Effects of fish-oil supplementation on myocardial fatty acids in humans. Am J Clin Nutr 2007; 85: 1222–1228.
  69. 69. McGee DL, Reed DM, Yano K, Kagan A, Tillotson J. Ten-year incidence of coronary heart disease in the Honolulu Heart Program. Relationship to nutrient intake. Am J Epidemiol 1984; 119: 667–676.
  70. 70. Pietinen P, Ascherio A, Korhonen P, Hartman AM, Willett WC, Albanes D, et al. Intake of fatty acids and risk of coronary heart disease in a cohort of Finnish men. The Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study. Am J Epidemiol 1997; 145: 876–87.
  71. 71. Jakobsen MU, Overvad K, Dyerberg J, Schroll M, Heitmann BL. Dietary fat and risk of coronary heart disease: possible effect modification by gender and age. Am J Epidemiol 2004; 160: 141–149.
  72. 72. Lerman RH. Essential fatty acids. Altern Ther Health Med 2006;12: 20-29.
  73. 73. Das UN. Essential fatty acids and their metabolites could function as endogenous HMG-CoA reductase and ACE enzyme inhibitors, anti-arrhythmic, anti-hypertensive, antiatherosclerotic, anti-inflammatory, cytoprotective, and cardioprotective molecules. Lipids Health Dis 2008; 7: 37.
  74. 74. Simopoulos AP. Summary of the NATO advanced research workshop on {omega} 3 and {omega} 6 fatty acids: Biological effects and nutritional essentiality. J Nutr 1989; 119: 521-528.
  75. 75. Burdge GC, Calder PC. Dietary α-linolenic acid and health-related outcomes: a metabolic perspective. Nutr Res Rev 2006; 19: 26–52.
  76. 76. Blasbalg TL, Hibbeln JR, Ramsden CE, Majchrzak SF, Rawlings RR. Changes in consumption of omega-3 and omega-6 fatty acids in the United States during the 20th century. Am J Clin Nutr. 2011; 93: 950–962.
  77. 77. De Caterina R, Massaro M. Omega-3 fatty acids and the regulation of expression of endothelial pro-atherogenic and pro-inflammatory genes. J Membr Biol 2005; 206: 103 -116.
  78. 78. Calder PC. Mechanisms of Action of (n-3) Fatty Acids. J. Nutr 2012; 142: 592S–599S.
  79. 79. Kris-Etherton PM, Harris WS, Appel LJ. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease. Circulation 2002; 106: 2747–2757.
  80. 80. Lands WE. Dietary fat and health: the evidence and the politics of prevention: careful use of dietary fats can improve life and prevent disease. Ann N Y Acad Sci 2005; 1055: 179–192.
  81. 81. Lee JH, O’Keefe JH, Lavie CJ, Marchioli R, Harris WS. Omega-3 fatty acids for cardioprotection. Mayo Clin Proc 2008; 83: 324–332.
  82. 82. Lavie CJ, Milani RV, Mehra MR, Ventura HO. Omega-3 polyunsaturated fatty acids and cardiovascular diseases. J Am Coll Cardiol 2009; 54: 585–594.
  83. 83. Dahl L, Bjorkkjaer T, Graff IE, et al. Fish – more than just omega 3. Tidsskr Nor Laegeforen 2006; 126: 309–311.
  84. 84. Conde CMS, Cyrino FZGA, Bottino DA, et al. Longchain n-3 polyunsaturated fatty acids and microvascular reactivity: observation in the hamster cheek pouch. Microvasc Res 2007; 3: 237–247.
  85. 85. McLennan PL. Relative effects of dietary saturated, monounsaturated, and polyunsaturated fatty acids on cardiac arrhythmias in rats. Am J Clin Nutr 1993; 57: 207-212.
  86. 86. Mclennan PL, Abeywardena MY. Membrane basis for fish oil effects on the heart: linking natural hibernators to prevention of human sudden cardiac death. Membrane Biol 2005; 206: 85–102.
  87. 87. Hlavackova M, Neckar J, Jezkova J, et al. Dietary polyunsaturated fatty acids alter myocardial protein kinase-C expression and affect cardioprotection induced by chronic hypoxia. Exp Biol Med 2007; 232: 823–832.
  88. 88. Mozaffarian D, Lemaitre RN, Kuller LH, Burke GL, Tracy RP, Siscovick DS. Cardiac benefits of fish consumption may depend on the type of fish meal consumed: the cardiovascular health study. Circulation 2003; 107: 1372–1377.
  89. 89. He K, Song Y, Daviglus ML, Liu K, Van Horn L, Dyer AR, et al. Accumulated evidence on fish consumption and coronary heart disease mortality: a meta-analysis of cohort studies. Circulation 2004; 109: 2705–11.
  90. 90. Anand RG, Alkadri M, Lavie CJ, et al. The role of fish oil in arrhythmia prevention. J Cardiopulm Rehabil Preven. 2008; 28: 92–98.
  91. 91. Jung UJ, Torrejon C, Tighe AP, et al. n-3 Fatty acids and cardiovascular disease: mechanisms underlying beneficial effects. Am J Clin Nutr. 2008; 87: S2003–2009.
  92. 92. Sinclair HM. Deficiency of essential fatty acids and atherosclerosis, etcetera. Lancet 1956; 270: 381–383.
  93. 93. Leaf A, Kang JX, Xiao YF, Billman GE. Clinical prevention of sudden cardiac death by n-3 polyunsaturated fatty acids and mechanism of prevention of arrhythmias by n-3 fish oils. Circulation 2003; 107: 2646–2652.
  94. 94. Mozaffarian D. Fish and n-3 fatty acids for the prevention of fatal coronary heart disease and sudden cardiac death. Am J Clin Nutr 2008; 87: 1991S–1996S.
  95. 95. Mozaffarian D, Wu JH. Omega-3 fatty acids and cardiovascular disease: effects on risk factors, molecular pathways, and clinical events. J Am Coll Cardiol. 2011; 58: 2047-2067.
  96. 96. Calo L, Bianconi L, Colivicchi F, et al. N-3 fatty acids for the prevention of atrial fibrillation after coronary artery bypass surgery: a randomized, controlled trial. J Am Coll Cardiol 2005; 45: 1723–1728.
  97. 97. Virtanen JK, Mursu J, Voutilainen S, Tuomainen TP. Serum long-chain n-3 polyunsaturated fatty acids and risk of hospital diagnosis of atrial fibrillation in men. Circulation 2009; 120: 2315–2321.
  98. 98. Matthan NR, Jordan H, Chung M, Lichtenstein AH, Lathrop DA, Lau J. Asystematic review and meta-analysis of the impact of omega-3 fatty acids on selected arrhythmia outcomes in animal models. Metabolism 2005; 54: 1557–1565.
  99. 99. Sarrazin JF, Comeau G, Daleau P, et al. Reduced incidence of vagally induced atrial fibrillation and expression levels of connexins by n-3 polyunsaturated fatty acids in dogs. J Am Coll Cardiol 2007; 50: 1505–1512.
  100. 100. Fischer R, Dechend R, Qadri F, et al. Dietary n-3 polyunsaturated fatty acids and direct renin inhibition improve electrical remodeling in a model of high human renin hypertension. Hypertension 2008; 51: 540–546.
  101. 101. Gao JY, Yasuda S, Tsuburaya R, et al. Long-term treatment with eicosapentaenoic acid ameliorates myocardial ischemia–reperfusion injury in pigs in vivo. Circ J 2011; 75: 1843–1851.
  102. 102. Kitamura K, Shibata R, Tsuji Y, Shimano M, Inden Y, Murohara T. Eicosapentaenoic acid prevents atrial fibrillation associated with heart failure in a rabbit model. Am J Physiol Heart Circ Physiol 2011; 300: H1814–H1821.
  103. 103. Stubbs CD, Smith AD. The modification of mammalian membrane polyunsaturated fatty acid composition in relation to membrane fluidity and function. Biochim Biophys Acta. 1984; 779: 89–137.
  104. 104. Murphy MG. Dietary fatty acids and membrane protein function. J Nutr Biochem. 1990; 1: 68–79.
  105. 105. Yaqoob P. The nutritional significance of lipid rafts. Annu Rev Nutr. 2009; 29: 257–282.
  106. 106. Das UN. Metabolic Syndrome Pathophysiology: The Role of essential fatty acids and their metabolites. Ames, IA, USA: Wiley-Blackwell Publishers; 2010.
  107. 107. Das UN. Molecular Basis of Health and Disease. New York: Springer; 2011.
  108. 108. Zuijdgeest-van Leeuwen SD, Dagnelie PC, Rietveld T, et al. Incorporation and washout of orally administered n-3 fatty acid ethyl esters in different plasma lipid fractions. Br J Nutr 1999; 82: 481-488.
  109. 109. Connor WE. Importance of n-3 fatty acids in health and disease. Am J Clin Nutr 2000; 71: 171–175S.
  110. 110. Surette ME, Koumenis IL, Edens MB, et al. Inhibition of leukotriene synthesis, pharmacokinetics, and tolerability of a novel dietary fatty acid formulation in healthy adult subjects. Clin Ther 2003; 25: 948-971.
  111. 111. Nair SS, Leitch J, Falconer J, Garg ML. Cardiac (n-3) nonesterified fatty acids are selectively increased in fish oil-fed pigs following myocardial ischemia. J Nutr 1999; 129: 1518–1523.
  112. 112. McLennan PL. Myocardial membrane fatty acids and the antiarrhythmic actions of dietary fish oil in animal models. Lipids 2001; 36: S111–114S.
  113. 113. Healy DA, Wallace FA, Miles EA, et al. Effect of low-to-moderate amounts of dietary fish oil on neutrophil lipid composition and function. Lipids 2000; 35: 763-768.
  114. 114. Calder PC. n-3 polyunsaturated fatty acids, inflammation and inflammatory diseases. Am J Clin Nutr 2006; 83: 1505S-1519S.
  115. 115. De Caterina R, Massaro M. Omega-3 fatty acids and the regulation of expression of endothelial pro-atherogenic and pro-inflammatory genes. J Membr Biol 2005; 206: 103 -116.
  116. 116. Weber C, Erl W, Pietsch A, et al. Docosahexaenoic acid selectively attenuates induction of vascular cell adhesion molecule-1 and subsequent monocytic cell adhesion to human endothelial cells stimulated by tumor necrosis factor-alpha. Arterioscler Thromb Vasc Biol 1995; 15: 622-628.
  117. 117. Novak TE, Babcock TA, Jho DH, et al. NF-kappa B inhibition by omega-3 fatty acids modulates LPS-stimulated macrophage TNF-alpha transcription. Am J Physiol Lung Cell Mol Physiol 2003; 284: L84-L89.
  118. 118. Caughey GE, Mantzioris E, Gibson RA, et al. The effect on human tumor necrosis factor alpha and interleukin 1 beta production of diets enriched in n-3 fatty acids from vegetable oil or fish oil. Am J Clin Nutr 1996; 63: 116-122.
  119. 119. Bravo L. Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance, Nutr. Rev. 1998; 56: 317–333.
  120. 120. Higdon JV, Frei B. Tea catechins and polyphenols: health effects, metabolism, and antioxidant functions. Critical Reviews in Food Science and Nutrition 2003; 43: 89–143.
  121. 121. Yilmaz Y. Novel uses of catechins in foods. Trends in Food Science and Technology 2006; 17: 64–71.
  122. 122. Lobo V, Patil A, Phatak A, Chandra N. Free radicals, antioxidants and functional foods: impact on human health. Pharmacognosy Review 2010; 4: 118–126.
  123. 123. Wootton-Beard PC, Ryan L. Improving public health? The role of antioxidant-rich fruit and vegetable beverages. Food Research and Technology 2011; 44: 3135–3148.
  124. 124. Ignat I, Volf I, Popa VI. A critical review of methods for characterisation of polyphenolic compounds in fruits and vegetables. Food Chemistry 2012; 126: 1821–1835.
  125. 125. Heiss C, Keen CL, Kelm M. Flavanols and cardiovascular disease prevention. European Heart Journal 2010; 31: 2583–2592.
  126. 126. Erdman JW Jr, Balentine D, Arab L, Beecher G, Dwyer JT, Folts J, Harnly J, Hollman P, Keen CL, Mazza G, Messina M, Scalbert A, Vita J, Williamson G, Burrowes J. Flavonoids and heart health: proceedings of the ILSI North America Flavonoids Workshop, May 31–June 1, 2005, Washington, DC. J Nutr 2007; 137: 718S–737S.
  127. 127. Joubert E, Joubert ME, Bester C, De Beer D, De Lange JH. Honeybush(Cyclopia spp.): From local cottage industry to global markets – the catalytic and supporting role of research. South African Journal of Botany 2011; 77: 887–907.
  128. 128. Joubert E, De Beer D. Rooibos (Aspalathus linearis) beyond farm gate: from herbal tea to potential phytopharmaceutical. South African Journal of Botany 2011; 77: 869–886.
  129. 129. Joubert E,Gelderblom WC, Louw A, De Beer D. South African herbal teas: Aspalathus linearis, Cyclopia spp. and Athrixia phylicoides–a review. Journal of Ethnopharmacology 2008a; 119: 376–412.
  130. 130. Joubert E, Gelderblom WC, De Beer D. Phenolic contribution of South African herbal teas to a healthy diet. Natural Product Communications 2009; 4: 701–718.
  131. 131. Marnewick JL, Rautenbach F, Venter I, Neethling H, Blackhurst DM, Wolmarans P, Macharia M. Effects of rooibos (Aspalathus linearis) on oxidative stress and biochemical parameters in adults at risk for cardiovascular disease. J. Ethnopharmacol. 2011; 133: 46–52.
  132. 132. McKay DL, Bloemberg JB. A review of the bioactivity of South African herbal teas: Rooibos (Aspaluthus Linearis) and Honeybush (Cyclopia intermedia). Physiother. Res 2007; 21: 1–16.
  133. 133. Marnewick JL. Rooibos and honeybush: recent advances in chemistry, biological activity and pharmacological activity and pharmacogynosy. In: Juliana HR, Simon JE, Ho CT. (Eds.), African Natural Plant Products: New Discoveries and Challenges in Chemistry and Quality. ACS Symposium Series, 1021. Oxford University Press, 2009; pp. 277–294.
  134. 134. Bramati L, Minoggio M, Gardana C, Simonetti P, Mauri P, Pietta P. Quantitative characterization of flavonoid compounds in rooibos tea (Aspalathus linearis) by LC–UV/DAD. J. Agric. Food Chem. 2002; 50: 5513–5519.
  135. 135. Ji ES, Yue H, Wu YM, He RR. Effects of phytooestrogen genistein on myocardial ischaemia reperfusion and apoptosis in rabbits. Acta Pharmacol. Sin 2004; 25; 306–312.
  136. 136. Wang S, Dusting GJ, May CN, Woodman OL. 3_,4_-Dihydroxyflavonol reduces infarct size and injury associated with myocardial ischaemia and reperfusion in sheep. Br. J. Pharmacol. 2004; 42: 352–443.
  137. 137. Qureshi AA, Sami SA, Salser WA, Khan FA. Synergistic effect of tocotrienol-rich fraction (TRF(25)) of rice bran and lovastatin on lipid parameters in hypercholesterolemic humans. J Nutr Biochem 2001; 12: 318–329.
  138. 138. Qureshi AA, Sami SA, Salser WA, Khan FA. Dose-dependent suppression of serum cholesterol by tocotrienol-rich fraction (TRF25) of rice bran in hypercholesterolemic humans. Atherosclerosis 2002; 161: 199–207.
  139. 139. Sen CK, Khanna S, Rink C, Roy S. Tocotrienols: the emerging face of natural vitamin E. Vitam Horm 2007; 76: 203–261.
  140. 140. Serbinova E, Kagan V, Han D, Packer L. Free radical recycling and intramembrane mobility in the antioxidant properties of alpha-tocopherol and alpha-tocotrienol. Free Radic Biol Med 1991; 10: 263–275.
  141. 141. Yoshida Y, Niki E, Noguchi N. Comparative study on the action of tocopherols and tocotrienols as antioxidant: chemical and physical effects. Chem Phys Lipids 2003; 123: 63–75.
  142. 142. Nagendran B, Unnithan UR, Choo YM. Characteristics of red palm oil, a-carotene- and vitamin E-rich refined oil for food uses. Food Nutr Bull 2000; 2: 189–194.
  143. 143. Sundram K, Sambanthamurthi R, Tan YA. Palm fruit chemistry. Asia Pac J Clin Nutr 2003; 12: 355–362.
  144. 144. Schroeder MT, Becker EM, Skibsted LH. Molecular mechanism of antioxidant synergism of tocotrienols and carotenoids in palm oil. J Agric Food Chem 2006; 54: 3445–3453.
  145. 145. Narang D, Sood S, Thomas MK, et al. Effect of dietary palm oil on oxidative stress associated with ischaemic–reperfusion injury in isolated rat heart. BMC Pharmacol 2004; 4: 29.
  146. 146. Charnock JS, Sundram K, Abeywardena MY et al. Dietary fats and oils in cardiac arrhythmia in rats. Am J Clin Nutr 1991; 53: 1047S–1049S.
  147. 147. Steiner M, Anatasi J. Vitamin E and platelet aggregation. J Clin Invest 1975; 57: 732–737.
  148. 148. Chan AC, Leith MK. Decreased prostacyclin synthesis in vitamin E deficient rabbit aorta. Am J Clin Nutr 1981; 34: 2341–2347.
  149. 149. Helub BJ, Sicilia I, Mahadevappa VG. Effect of tocotrienol derivatives on collagen and ADP-induced plasma platelet aggregation. Abstracts, PORUM International Palm Oil Development Conference, 1989; 5–9 September, Kuala Lumpur, N 9.
  150. 150. Qureshi AA, Qureshi N, Weight JJK, et al. Lowering of serum cholesterol in hypercholesterolaemic humans by tocotrienols (palmvitee). Am J Clin Nutr 1991; 53: 1021S–1026S.
  151. 151. Pearce BC, Parker RA, Deason ME, et al. Hypocholesterolaemic activity of synthetic and natural tocotrienols. J Med Chem 1992; 35: 3595–3606.
  152. 152. Hornstra G. Dietary lipids and cardiovascular disease: effects of palm oil. Oleagineux 1988; 43: 75–81.
  153. 153. Qureshi AA, Bradlow BA, Brace L, et al. Response of hypercholesterolaemic subjects to administration of tocotrienols. Lipids 1995; 30: 1171–1177.
  154. 154. Sundram K, Anisah I, Hayes KC, et al. Trans (elaidic) fatty acids adversely impact lipoprotein profiles relative to specific saturated fatty acids in humans. J Nutr 1997; 127: 514S–520S.
  155. 155. Kritchevsky D. Impact of red palm oil on human nutrition and health. Food Nutr Bull 2000; 2: 182–188.
  156. 156. Simopoulos AP. Omega-3 fatty acids in health and disease and in growth and development. Am J Clin Nutr 1991; 54: 438–463.
  157. 157. Serbinova E, Khavaja S, Catudioc J, et al. Palm oil vitamin E protects against ischaemia/reperfusion injury in the isolated perfused Langendorff heart. Nutr Res 1992; 12: 989S–1009S.
  158. 158. Aggarwal BB, Sundaram C, Prasad S, Kannappan R. Tocotrienols, the vitamin E of the 21st century: Its potential against cancer and other chronic diseases. Biochemical Pharmacology 2010; 80: 1613-1631.
  159. 159. Ribayamercado JD, Solon SF, Tang G, Cabal-Borza M, Perfecto SC, Russel RM. Bionconversion of plant carotenoids to vit-A in Filipino school-aged children varies inversely with vit-A status. Am J Clin Nutr 2000; 72: 455–465.
  160. 160. Choe E, Min DB. Mechanisms of antioxidants in the oxidation of foods. Comp Rev Food Sci Food Safe 2009; 8: 345–358.
  161. 161. Krinsky NI. The antioxidant and biological properties of carotenoids. Ann NY Acad Sci 1998; 854: 443–447.
  162. 162. Reboul E, Thap S, Tourniaire F, Andre M, Juhel C, Morange S, Amiot MJ, Lairon D, Borel P. Differential effect of dietary antioxidant classes (carotenoids, polyphenols, vitamins C and E) on lutein absorption. Br J Nutr 2007; 97: 440–446.
  163. 163. Young AJ, Lowe GM. Antioxidant and prooxidant properties of carotenoids. Arch Biochem Biophys 2001; 385: 20–27.
  164. 164. Christensen RL. The photochemistry of carotenoids. In: Frank HA, Young AJ, Britton G, Cogdell RJ, (Eds.), Dordrecht, The Netherlands: Kluwer Academic 1999; 137–157.
  165. 165. Khachik F, Spangler CJ, Smith, JC, Canfield LM, Steck A, Pfander H. Identification, quantification, and relative concentrations of carotenoids, and their metabolites in human milk and serum. Anal Chem 1997; 69: 1873–1881.
  166. 166. Chen J, Jarvi M, Lo PC, Stefflova K, Wilson BC, Zheng G. Using the singlet oxygen scavenging property of carotenoid in photodynamic molecular beacons to minimize photodamage to non-targeted cells. Photochem Photobiol Sci 2007; 6: 1311–1317.
  167. 167. McNulty H, Jacob RF, Mason RP. Biologic activity of carotenoids related to distinct membrane physicochemical interactions. Am J Cardiol 2008; 101: 20–29.
  168. 168. Burton GW, Ingold KU. beta-Carotene: an unusual type of lipid antioxidant. Science. 1984; 224: 569–73.
  169. 169. Sies H, Stahl W. Vitamins E and C, β-carotene, and other carotenoids as antioxidants. Am J Clin Nutr 1995; 62: 1315S–1321S.
  170. 170. Amengual J, Lobo GP, Golczak M, Li HN, Klimova T, Hoppel CL, Wyss A, Palczewski K, von Lintig J. A mitochondrial enzyme degrades carotenoids and protects against oxidative stress. FASEB J 2011; 25: 948–959.
  171. 171. Daviglus ML, Stamler J, Orencia AJ, et al. Fish consumption and the 30-year risk of fatal myocardial infarction. N Engl J Med.1997; 336: 1046–1053.
  172. 172. Albert CM, Hennekens CH, O’Donnell CJ, et al. Fish consumption and risk of sudden cardiac death. JAMA. 1998; 279: 23–28.
  173. 173. Leaf A, Albert CM, Josephson M, et al. Prevention of fatal arrhythmias in high-risk subjects by fish oil n-3 fatty acid intake. Circulation. 2005; 112: 2762–2768.
  174. 174. Zaloga GP. Dietary lipids: ancestral ligands and regulators of cell signaling pathways. Crit Care Med. 1999; 27: 1646–1648.
  175. 175. Siddiqui RA, Shaikh SR, Sech LA, et al. Omega 3-fatty acids: health benefits and cellular mechanisms of action. Mini Rev Med Chem.2004; 4: 859–871.
  176. 176. Clarke SD, Jump DB. Regulation of gene transcription by polyunsaturated fatty acids. Prog Lipid Res. 1993; 32: 139–149.
  177. 177. Abia R, Perona JS, Pacheco YM, Montero E, Muriana FJ, Ruiz-Gutierrez V. Postprandial triacylglycerols from dietary virgin olive oil are selectively cleared in humans. J Nutr 1999; 129: 2184–2191.
  178. 178. Kris-Etherton PM, Yu-Poth S, Sabaté J, Ratcliffe HE, Zhao G, Etherton TD. Nuts and their bioactive constituents: effects on serum lipids and other factors that affect disease risk. Am J Clin Nutr 1999; 70: 504–511.
  179. 179. Davidson A. The Oxford Companion to Food. Oxford, UK: Oxford University Press 1999.
  180. 180. Halvorsen BL, Holte K, Myhrstad MC, et al. A systematic screening of total antioxidants in dietary plants. J Nutr 2002; 132: 461–471.
  181. 181. Wu X, Beecher GR, Holden JM, Haytowitz DB, Gebhardt SE, Prior RL. Lipophilic and hydrophilic antioxidant capacities of common foods in the United States. J Agric Food Chem 2004; 52: 4026–4037.
  182. 182. Blomhoff R, Carlsen MH, Andersen LF, Jacobs DR J. Health benefits of nuts: potential role of antioxidants. Br J Nutr 2006; 96: 52–60.
  183. 183. Vivancos M, Moreno JJ. beta-Sitosterol modulates antioxidant enzyme response in RAW 264.7 macrophages. Free Radic Biol Med 2005; 39: 91–97.
  184. 184. Segura R, Javierre C, Lizarraga MA, Ros E. Other relevant components of nuts: phytosterols, folate and minerals. Br J Nutr 2006; 96: 36–44.
  185. 185. Fraley AE, Tsimikas S. Clinical applications of circulating oxidized low-density lipoprotein biomarkers in cardiovascular disease. Curr Opin Lipidol 2006; 17: 502–509.
  186. 186. Ferri N, Paoletti R, Corsini A. Biomarkers for atherosclerosis: pathophysiological role and pharmacological modulation. Curr Opin Lipidol 2006; 17: 495–501.
  187. 187. Deanfield JE, Halcox JP, Rabelink TJ. Endothelial function and dysfunction. Testing and clinical relevance. Circulation 2007; 115: 1285–95.
  188. 188. Brown AA, Hu FB. Dietary modulation of endothelial function: implications for cardiovascular disease. Am J Clin Nutr 2001; 73: 673–686.
  189. 189. Blomhoff R. Dietary antioxidants and cardiovascular disease. Curr Opin Lipidol 2005; 16: 47–54.
  190. 190. Basu A, Devaraj S, Jialal I. Dietary factors that promote or retard inflammation. Arterioscler Thromb Vasc Biol 2006; 26: 995–1001.
  191. 191. Kay CD, Kris-Etherton PM, West SG. Effects of antioxidant-rich foods on vascular reactivity: review of the clinical evidence. Curr Atheroscler Rep 2006; 8: 510–522.
  192. 192. Reiter RJ, Manchester LC, Tan DX. Melatonin in walnuts: influence on levels of melatonin and total antioxidant capacity of blood. Nutrition 2005; 21: 920–924.
  193. 193. Alexiadou K, Katsilambros N. Nuts: Anti-atherogenic food? European Journal of Internal Medicine 2011; 22: 141–146.
  194. 194. Abbey M, Noakes M, Belling GB, Nestel PJ. Partial replacement of saturated fatty acids with almonds or walnuts lowers total plasma cholesterol and low-densitylipoprotein cholesterol. Am J Clin Nutr 1994; 59: 995–999.
  195. 195. Jenkins DJ, Kendall CW, Marchie A, Faulkner DA, Wong JM, de Souza R, et al. Effects of a dietary portfolio of cholesterol-lowering foods vs lovastatin on serum lipids and C-reactive protein. JAMA 2003; 290: 502–510.
  196. 196. Zhao G, Etherton TD, Martin KR, West SG, Gillies PJ, Kris-Etherton PM. Dietary alpha-linolenic acid reduces inflammatory and lipid cardiovascular risk factors in hypercholesterolemic men and women. J Nutr 2004; 134: 2991–2997.
  197. 197. Jenkins DJ, Kendall CW, Marchie A, Faulkner DA, Josse AR, Wong JM, et al. Direct comparison of dietary portfolio vs statin on C-reactive protein. Eur J Clin Nutr 2005; 59: 851–860.
  198. 198. Ros E, Núñez I, Pérez-Heras A, Serra M, Gilabert R, Casals E, et al. A walnut diet improves endothelial function in hypercholesterolemic subjects: a randomized crossover trial. Circulation 2004; 109: 1609–1614.
  199. 199. Zhao G, Etherton TD, Martin KR, Gillies PJ, West SG, Kris-Etherton PM. Dietary alpha-linolenic acid inhibits proinflammatory cytokine production by peripheral blood mononuclear cells in hypercholesterolemic subjects. Am J Clin Nutr 2007; 85: 385–391.
  200. 200. Bonetti PO, Lerman LO, Lerman A. Endothelial dysfunction: a marker of atherosclerotic risk. Arterioscler Thromb Vasc Biol 2003; 23: 168–175.
  201. 201. West SG. Effect of diet on vascular reactivity: an emerging marker for vascular risk. Curr Atheroscler Rep 2001; 3: 446–455.
  202. 202. Sanderson P, Sattar N, Olthof M, Grimble RF, Calder PC, Griffin BA, et al. Dietary lipids and vascular function: UK Food Standards Agency workshop report. Br J Nutr 2004; 91: 491–500.
  203. 203. de Koning EJ, Rabelink TJ. Endothelial function in the post-prandial state. Atheroscler 2002; 3: 11–16.
  204. 204. Tentolouris N, Arapostathi C, Perrea D, Kyriaki D, Revenas C, Katsilambros N. Differential effects of two isoenergetic meals rich in saturated or monounsaturated fat on endothelial function in subjects with type 2 diabetes. Diab Care 2008; 31: 2276–2278.
  205. 205. Estruch R, Martínez-González MA, Corella D, Salas-Salvadó J, Ruiz-Gutiérrez V, Covas MI, et al. PREDIMED study investigators. Effects of a Mediterranean-style diet on cardiovascular risk factors: a randomized trial. Ann Intern Med 2006; 145: 1–11.
  206. 206. Davis P, Valacchi G, Pagnin E, Shao Q, Gross HB, Calo L, et al. Walnuts reduce aortic ET-1 mRNA levels in hamsters fed a high-fat, atherogenic diet. J Nutr 2006; 136: 428-432.
  207. 207. Sabaté J. Nut consumption and body weight. Am J Clin Nutr 2003; 78: 647–650.
  208. 208. Griel AE, Eissenstat B, Juturu V, Hsieh G, Kris-Etherton PMJ. Improved diet quality with peanut consumption. Am Coll Nutr 2004; 23: 660–668.
  209. 209. Alper CM, Mattes RD. Effects of chronic peanut consumption on energy balance and hedonics. Int J Obes Relat Metab Disord 2002; 26: 1129–1137.
  210. 210. Fraser GE, Bennett HW, Jaceldo KB, Sabaté J. Effect on body weight of a free 76 kilojoule (320 calorie) daily supplement of almonds for six months. J Am Coll Nutr 2002; 21: 275–83.
  211. 211. Sabaté J, Cordero-Macintyre Z, Siapco G, Torabian S, Haddad E. Does regular walnut consumption lead to weight gain? Br J Nutr 2005; 94: 859–64.
  212. 212. Rajaram S, Sabaté J. Nuts, body weight and insulin resistance. Br J Nutr 2006; 96: 79-86.
  213. 213. Hollis J, Mattes R. Effect of chronic consumption of almonds on body weight in healthy humans. Br J Nutr 2007; 98: 651–656.
  214. 214. Bes-Rastrollo M, Wedick NM, Martinez-Gonzalez MA, Li TY, Sampson L, Hu FB. Prospective study of nut consumption, long-term weight change, and obesity risk in women. Am J Clin Nutr 2009; 89: 1913–1919.
  215. 215. Mattes RD, Kris-Etherton PM, Foster GD. Impact of peanuts and tree nuts on body weight and healthy weight loss in adults. J Nutr 2008; 138: 1741–1745.
  216. 216. Kokkinos A, le Roux CW, Alexiadou K, Tentolouris N, Vincent RP, Kyriaki D, et al. Eating slowly increases the postprandial response of the anorexigenic gut hormones, peptide YY and glucagon-like peptide-1. J Clin Endocrinol Metab 2010; 95: 333.
  217. 217. Defilippis AP, Blaha MJ, Jacobson TA. Omega-3 Fatty acids for cardiovascular disease prevention. Curr Treat Options Cardiovasc Med 2010; 12: 365–380.
  218. 218. Villano D, Pecorari M, Testa MF, Raguzzini A, Stalmach A, Crozier A, Tubili C, Serafini M. Unfermented and fermented rooibos teas (Aspalathus linearis) increase plasma total antioxidant capacity in healthy humans. Food Chem. 2010; 123: 679–683.
  219. 219. Rahman I, Biswas SK, Kirkham PA. Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol 2006; 72: 1439–1452.
  220. 220. Stephens NG, Parsons A, Schofield PM, Kelly F, Cheeseman K, Mitchinson MJ. Randomised controlled trial of Vitamin E in patients with coronary disease: Cambridge Heart Antioxidant Study (CHAOS). Lancet1996; 347: 781–786.
  221. 221. Pryor WA. Vitamin E and heart disease: basic science to clinical intervention trials. Free Radic Biol Med 2000; 28: 141–164.
  222. 222. Guidelines for the prevention of hypertension and associated cardiovascular disease. Joint World Health Organization/International Society of Hypertension Meeting. J Hypertens 1992; 10: 97-99.
  223. 223. Goyal A, Yusuf S. The burden of cardiovascular disease in the Indian subcontinent. Indian J Med Res 2006; 124: 235-244.

Written By

Wongama Given Pantsi, Dirk Jacobus Bester, Adriaan Johannes Esterhuyse and Guillaume Aboua

Submitted: 16 September 2013 Published: 05 February 2014