Hostname: page-component-7c8c6479df-5xszh Total loading time: 0 Render date: 2024-03-17T05:44:05.050Z Has data issue: false hasContentIssue false

New hypotheses for the health-protective mechanisms of whole-grain cereals: what is beyond fibre?

Published online by Cambridge University Press:  22 June 2010

Anthony Fardet*
Affiliation:
INRA, UMR 1019 Nutrition Humaine, F-63122Saint-Genès-Champanelle, France Clermont Université, UFR Médecine, UMR 1019 Nutrition Humaine, F-63000Clermont-Ferrand, France
Rights & Permissions [Opens in a new window]

Abstract

Epidemiological studies have clearly shown that whole-grain cereals can protect against obesity, diabetes, CVD and cancers. The specific effects of food structure (increased satiety, reduced transit time and glycaemic response), fibre (improved faecal bulking and satiety, viscosity and SCFA production, and/or reduced glycaemic response) and Mg (better glycaemic homeostasis through increased insulin secretion), together with the antioxidant and anti-carcinogenic properties of numerous bioactive compounds, especially those in the bran and germ (minerals, trace elements, vitamins, carotenoids, polyphenols and alkylresorcinols), are today well-recognised mechanisms in this protection. Recent findings, the exhaustive listing of bioactive compounds found in whole-grain wheat, their content in whole-grain, bran and germ fractions and their estimated bioavailability, have led to new hypotheses. The involvement of polyphenols in cell signalling and gene regulation, and of sulfur compounds, lignin and phytic acid should be considered in antioxidant protection. Whole-grain wheat is also a rich source of methyl donors and lipotropes (methionine, betaine, choline, inositol and folates) that may be involved in cardiovascular and/or hepatic protection, lipid metabolism and DNA methylation. Potential protective effects of bound phenolic acids within the colon, of the B-complex vitamins on the nervous system and mental health, of oligosaccharides as prebiotics, of compounds associated with skeleton health, and of other compounds such as α-linolenic acid, policosanol, melatonin, phytosterols and para-aminobenzoic acid also deserve to be studied in more depth. Finally, benefits of nutrigenomics to study complex physiological effects of the ‘whole-grain package’, and the most promising ways for improving the nutritional quality of cereal products are discussed.

Type
Review Article
Copyright
Copyright © The Author 2010

Introduction

There is growing evidence that whole-grain cereal products protect against the development of chronic diseases. The most important of these in terms of public health are obesity(Reference Koh-Banerjee and Rimm1, Reference van de Vijver, van den Bosch and van den Brandt2), the metabolic syndrome(Reference Esmaillzadeh, Mirmiran and Azizi3, Reference Sahyoun, Jacques and Zhang4), type 2 diabetes(Reference de Munter, Hu and Spiegelman5, Reference Murtaugh, Jacobs and Jacob6), CVD(Reference Mellen, Walsh and Herrington7) and cancers(Reference Chan, Wang and Holly8Reference Schatzkin, Park and Leitzmann12). Whole-grain cereal consumption has also been shown to be protective against mortality, as was shown with inflammation-related death (i.e. non-cardiovascular and non-cancer inflammatory diseases such as, for example, respiratory system diseases)(Reference Jacobs, Andersen and Blomhoff13) and with cancer and CVD(Reference Sahyoun, Jacques and Zhang4, Reference Adom, Sorrells and Liu14, Reference Jacobs, Meyer and Solvoll15). These conclusions are supported by the effects of consuming refined cereal products (bread, pasta and rice), as these have been associated with an increased risk of digestive tract, pharynx, larynx and thyroid cancers in northern Italians(Reference Chatenoud, La Vecchia and Franceschi16). However, an association between a lower risk of developing a chronic disease and a high whole-grain cereal consumption does not mean a direct causal relationship and provides no information about the physiological mechanisms involved.

These metabolic diseases are related to our daily lifestyle, notably an unbalanced energy-rich diet lacking fibre and protective bioactive compounds such as micronutrients and phytochemicals. Today, it is agreed to advance that this is the synergistic action of the compounds, mainly contained in the bran and germ fractions of cereals, which is protective(Reference Jensen, Koh-Banerjee and Franz17, Reference Liu18). Some specific mechanisms are today well recognised. For example, food structure influences satiety and the slow release of sugars recommended for type 2 diabetes. Dietary fibre improves gut health, and the antioxidant and anti-inflammatory properties of most phytochemicals can help prevent cancer and CVD. However, the precise physiological mechanisms involved are far from being elucidated.

The main whole-grain cereals consumed worldwide are wheat, rice and maize, followed by oats, rye, barley, triticale, millet and sorghum. Whole-grain wheat, which is the focus of the present review, is composed of 10–14 % bran, 2·5–3·0 % germ and 80–85 % endosperm, depending on the intensity of the milling process. The bioactive compounds are unevenly distributed within these parts (Fig. 1), and this distribution also varies according to the type of cereal considered. Whole-grain cereals are a rich source of fibre and bioactive compounds. For example, whole-grain wheat contains about 13 % dietary fibre and at least 2 % bioactive compounds other than fibre (Table 1), which accounts for at least 15 % of the whole grain. In the bran and germ fractions, still higher proportions are reached: about 45 and 18 % of dietary fibre, and about 7 % and at least 6 % of bioactive compounds, respectively; which represents about 52 % and at least 24 % of these fractions. These proportions obviously depend on the cereal type. It is therefore easy to understand that refined cereal products that lack the bran and germ fractions have lost most of their protective compounds. For example, refining whole-grain wheat may lead to the loss of about 58 % of fibre, 83 % of Mg, 79 % of Zn, 92 % of Se, 70 % of nicotinic acid, 61 % of folates and 79 % of vitamin E(Reference Truswell19).

Fig. 1 The three wheat fraction (bran, germ and endosperm) with their main bioactive compounds as obtained from Tables 1 and 2. Whole-grain wheat has an heterogeneous struture with bioactive compounds unevenly distributed within its different parts (with permission from Surget & Barron for original image(Reference Surget and Barron476), and adapted from the brochure ‘Progress in HEALTHGRAIN 2008’, HealthGrain Project, European Community's Sixth Framework Programme, FOOD-CT-2005-514008, 2005–2010). * No published data on the precise locations of policosanol and phytosterols in a specific layer of the wheat bran fraction.

Table 1 Average content of the major bioactive compounds in whole-grain wheat and wheat bran and germ fractions (%)*

* Mean percentages of bioactive compounds found in wheat bran, whole-grain wheat and wheat germ are calculated from Table 2 as follows: % = (minimum value+maximum value)/2.

Expressed as g/100 g food.

No data found.

§ Total free glutathione is given as glutathione equivalents = reduced glutathione+(oxidised glutathione × 2).

Dietary fibre content is measured according to the AOAC method as such or modified (for details, see American Association of Cereal Chemists(53)).

Oligosaccharides include fructans, raffinose and stachyose.

However, the exact nature of the positive physiological effects exerted by whole-grain cereal products remains unresolved because of the huge number of phytochemicals and biological effects involved (Tables 2 and 3). The most significant of them in wheat, besides fibre, are n-3 fatty acids, sulfur amino acids, oligosaccharides (stachyose, raffinose and fructans), lignin, minerals, trace elements, vitamins B and E, carotenoids, polyphenols (especially phenolic acids such as ferulic acid and smaller amounts of flavonoids and lignans), alkylresorcinols, phytic acid, betaine, total choline-containing compounds, inositols, phytosterols, policosanol and melatonin. Each one of these compounds has numerous physiological functions and recognised health benefits (Tables 3 and 4). While studying each compound separately, the main approach used to date, may well be unavoidable, it also involves considerable risk. This is because it ignores two important factors. One is the importance of synergy between the actions of compounds which is poorly characterised and more difficult to assess than the biological action of an isolated compound. The second is the importance of the cereal matrix and its influence on the accessibility of compounds in the digestive tract and hence on their availability within the organism. Indeed, little is often known of the bioavailability of many bioactive compounds derived from complex cereal products (Table 2). Thus, the amount of a particular compound in whole-grain cereals is rarely the same as the amount that is available to exert a given physiological action, in contrast to the result of consuming the free compound.

Table 2 Content, apparent absorption and fermentability of bioactive compounds and fibre from whole-grain wheat and wheat bran and germ fractions*

nd, Not detected.

* All data are based on international references unless specified (see references in Appendices); for bioavailability data, methods used for determining percentage apparent absorption, the subject status and the model used (animals v. humans) differ from one study to another which may explain the sometimes very large range of values given: data remain therefore indicative and should be taken cautiously.

When expressed on a DM basis in references, results were converted on a wet matter basis considering that whole grain, bran and germ contain 13, 10 and 11·4 g water/100 g food, respectively.

No data found as regard with whole-grain wheat, and wheat bran and germ.

§ Total glutathione equivalents = reduced glutathione+(oxidised glutathione × 2).

Degree of fermentation.

Small-intestinal phytases (high activity in rats and very much lower in humans and pigs) are able to hydrolyse phytic acid.

** High ranges are likely to result from the different types of extraction procedure used.

†† Expressed in gallic acid equivalents/100 g.

‡‡ Expressed in catechin equivalents.

§§ Expressed as rutin equivalents.

∥∥ Sum of genistein and daidzein (whole-wheat flour type not specified).

¶¶ Total choline refers to the sum of free choline, glycerophosphocholine, phosphatidylcholine and sphingomyelin.

*** Toasted wheat germ(Reference Zeisel, Mar and Howe477).

††† Chiro-inositol refers to the sum of free d-chiro-inositol and chiro-inositol moieties mainly derived from pinitol (i.e. methyl chiro-inositol) and glycosylated pinitol.

‡‡‡ Evaluation based on the fact that about 95 % of total myo-inositol would come almost exclusively from phytic acid(Reference Matheson and Strother250).

Table 3 Main physiological functions, potential protective mechanisms and health benefits of isolated bioactive compounds found in whole-grain wheat, rice and oat*

* All data concerning physiological mechanisms and health effects are based on international references (in vitro studies on culture cells and in vivo studies in animals and human subjects; see references in Appendices).

For these compounds, the intensity of the symbol in brackets (+,++ or +++) refers to the importance of the compound as supplied by a predominantly cereal-based diet, based on British data collected by Truswell(Reference Truswell19); for other compounds, the intensity of the symbol in brackets was estimated based on the compound content in whole-grain wheat compared with other food sources.

Mechanisms and health outcomes are associated with plant saponins in general, not exclusively cereal saponins.

Table 4 Whole-grain cereal bioactive compounds potentially involved in the prevention of major health outcomes and in antioxidant protection*

* Prepared from data in Table 3.

There may be many protective physiological mechanisms associated with consuming whole-grain cereal because of the high number of protective compounds. They may be mechanical within the digestive tract (insoluble fibre can increase transit time and faecal bulking), hormonal (Zn, Se and nicotinic acid participating in hormone activation and synthesis), antioxidative (almost all micronutrients), anti-inflammatory (for example, n-3 α-linolenic acid, Cu and ferulic acid), anti-carcinogenic (almost all micronutrients), or linked to gene regulation (for example, flavonoids), cell signalling (for example, polyphenols and redox status), energy metabolism (for example, the B-complex vitamins) and effects on enzymes (for example, some minerals and trace elements) (Table 3).

The main objective of the present paper is to propose new hypotheses for exploring the mechanisms behind the protective actions of whole-grain cereals using wheat as the main example. I have therefore exhaustively itemised all the bioactive compounds in whole-grain wheat and in the two fractions that are usually removed during refining: bran and germ. I have also listed their contents (range) in wheat, their bioavailability when obtained from complex whole-grain wheat products, their potential physiological effect(s) and the resulting health outcomes, with particular attention to some compounds that are specific to cereals other than wheat. The proposed new hypotheses are based on the action of compounds that are all bioactive when tested alone in their free form, such as the B vitamins, lignin, phytic acid, betaine, choline-containing compounds, inositols, policosanol, melatonin, para-aminobenzoic acid, sulfur amino acids, α-linolenic acid, phytosterols and some oligosaccharides.

First, I define the term ‘whole-grain cereal products’ and then examine the presently accepted mechanisms for explaining the role played by whole-grain cereals in preventing chronic diseases, as identified by studies on human subjects (for example, the importance of food structure and antioxidants), on rats (for example, the anti-carcinogenic property of many phytochemicals) and in vitro (cell-associated mechanisms). I then discuss my new hypotheses that are based on recent findings and on the potential physiological effects of whole-grain cereal compounds. I develop a broader view of the well-known antioxidant hypothesis that takes into account the actions of polyphenols on cell signalling and gene regulation in relation to the redox status. I review recent publications that have also revealed the great potential of the nutrigenomic approach for extending our knowledge of the protective mechanisms associated with complex foods. Finally, I briefly review the ways by which the nutritional quality of cereal products can be improved so as to optimally preserve the protective properties of whole-grain cereals.

What are whole-grain cereal products?

Definition

The American Association of Cereal Chemists (AACC) gave the following scientific and botanical definition in 1999: ‘Whole grains shall consist of the intact, ground, cracked or flaked caryopsis, whose principal anatomical components – the starchy endosperm, germ and bran – are present in the same relative proportions as they exist in the intact caryopsis’(20). The definition given by the Whole Grains Council in May 2004 includes processed food products: ‘Whole grains or foods made from them contain all the essential parts and naturally-occurring nutrients of the entire grain seed. If the grain has been processed (e.g. cracked, crushed, rolled, extruded, and/or cooked), the food product should deliver approximately the same rich balance of nutrients that are found in the original grain seed’(21). The US Food and Drug Administration published a Draft Guidance on Whole-grain Label Statements in 2006 that adopted the international AACC definition and included amaranth, barley, buckwheat, bulgur, maize (including popcorn), millet, quinoa, rice, rye, oats, sorghum, teff, triticale, wheat and wild rice; pearled barley was not included because some outer layers of the bran fraction are removed(22). Pseudocereals such as amaranth, buckwheat and quinoa have similar macronutrient compositions (carbohydrates, proteins and lipids), and are used in the same traditional ways as cereals(23, Reference Jones24). The response to the US Food and Drug Administration Draft Guidance by the AACC International recommended that some traditional cereals such as ‘lightly pearled barley, grano (lightly pearled wheat), nixtimalized corn and bulgur that has been minimally processed be also classified as whole grains’(23), making allowance for small losses of components that occur through traditional processing. The Whole Grain Task Force stated in 2008 that it ‘supports the use of the term whole-grain for products of milling operations that divide the grain into germ, bran and endosperm, but then recombine the parts into their original proportions before the flour leaves the mill’(Reference Jones24). However, as I will explain later, most of the products defined as whole-grain foods in studies showing the health benefits of whole-grain cereals are made of recombined whole-grain flours(Reference Jones24), which rarely contain the same proportions of bran, germ and endosperm as the intact grain before milling. Thus, the germ fraction is almost always removed because its high lipid content (about 9 %) may go rancid upon storage(Reference Srivastava, Sudha and Baskaran25). Processing whole-grain cereals also leads to losses of bioactive compounds so they cannot really deliver ‘approximately the same rich balance of nutrients that are found in the original grain seed’(21). Thus, if researchers had referred strictly to the definitions given above, few studies could have concluded that whole-grain cereal foods protect human health. Alternative definitions have therefore been proposed by the Whole Grain Task Force in which ‘as they exist in the intact caryopsis’ in the AACC definition is replaced by ‘as found in the least-processed, traditional forms of the edible grain kernels’ or completed by adding ‘as they exist in the intact caryopsis to the extent feasible by the best modern milling technology’(Reference Jones24). This last definition is probably the best adapted to our Western country technologies. But none of these alternative definitions has been adopted to date and there is still no official international definition of whole-grain cereal products in Europe.

What proportions?

Finally, the proportion of whole grains that must be present in a cereal product needs to be defined for it to be considered a whole-grain product. The issue is still debated. The definition given by the American Food and Drug Administration(26) in 1999 was: ‘For purposes of bearing the prospective claim, the notification defined ‘whole grain foods’ as foods that contain 51 percent of total weight or more whole grain ingredient(s) by weight’ (extract). This definition was debated and contested by the European Whole Grain Task Force in 2008. They explained that: ‘Using total weight gives advantage to products sold by dry weight such as crackers and ready-to-eat cereal. Because foods like breads have a proportionally high water content, even some breads made with all whole grain flours but containing significant amounts of nuts, seeds and fruit would fail to meet the 51 % by weight rule’(Reference Jones24). Apparently, there is still no international consensus as to the right proportion of whole grain by dry weight (DW) in a product in order for it to be called a whole-grain product. Each country has its own definition and standards(21). However, most research and observational studies, particularly those on breakfast cereals, estimate the whole-grain intake from products containing at least 25 % whole grains or bran by weight(Reference de Munter, Hu and Spiegelman5, Reference Adom, Sorrells and Liu14, Reference Jacobs, Meyer and Kushi27, Reference Liu, Manson and Stampfer28). Thus, a study on young individuals aged 4–18 years found that using a 51 %-based definition underestimated the whole-grain intake by 28 %, breakfast cereals (56 %) and bread (25 %) being the major sources of whole-grain cereals(Reference Thane, Jones and Stephen29). In another study on adiposity among two cohorts of British adults, the same research team assumed that whole-grain foods contained ≥ 10 % whole grains and found little or no association between the whole-grain intake and anthropometric indices(Reference Thane, Stephen and Jebb30). This suggests that the threshold of 10 % is probably too low and emphasises the need to harmonise how the whole-grain cereal food intake is calculated. In these studies, generally carried out in Western countries, whole-grain cereal foods considered are, for the most cited, whole-grain breads (for example, dark, brown, wholemeal and rye bread), whole-grain breakfast cereals (for example, muesli), popcorn, cooked porridges (oatmeal or whole wheat), wheat germ, brown rice, bran, cooked grains (for example, wheat, millet and roasted buckwheat) and other grain-based foods such as bulgur and couscous. A complete list of food ingredients classified as whole grains in the US Department of Agriculture (USDA) pyramid servings database is reported by Cleveland et al. (Reference Cleveland, Moshfegh and Albertson31). Refined grain foods generally include white breads (for example, French baguette), sweet rolls, noodles, pasta, cakes, biscuits, viennoiseries, muffins, refined grain breakfast cereals, white rice, pancakes, waffles and pizza.

The importance of whole-grain cereal product consumption

There are far fewer whole-grain cereal products on the market than there are refined products, at least in Western countries. The major sources of whole-grain cereals are breads, breakfast cereals and whole-grain cereals consumed as such (for example, brown rice or quick-cooking whole-grain barley and wheat). Epidemiological data show that the consumption of two to three servings of whole-grain cereal per d is sufficient to get beneficial health effects(Reference Lang and Jebb32). The recommended consumption of whole-grain cereal products differs from one country to another, but most recommend increased whole-grain cereal product consumption(21, Reference Lang and Jebb32). For example, at least three servings daily are recommended in the USA, that is, about 48 g of whole-grain cereals(Reference Welsh, Shaw and Davis33); between six and twelve servings daily are recommended in Australia and four servings daily in Denmark(21). Other countries such as Canada, UK, Greece, Germany, Austria and Switzerland are not so precise and generally recommend an increase in cereal consumption with emphasis on whole-grain products(21). Surveys carried out in the USA and the UK showed that most individuals consume less than one serving per d and about 30 % any, and that only 0·8 to 8 % of those surveyed in the USA consumed the recommended three servings per d(Reference Cleveland, Moshfegh and Albertson31, Reference Lang and Jebb32, Reference Albertson and Tobelmann34). The situation is quite different in Scandinavian countries, where individuals consume more whole-grain cereal products, particularly rye-based(Reference Lang and Jebb32). For example, Norwegians consume an estimated four times more whole-grain products than do Americans(35), but less than the Finns, 40 % of whom may consume four or more slices of dark bread per d(Reference Prättälä, Helasoja and Mykkänen36). Why is consumption so low in other Western countries? There are probably several reasons. First, unlike fruits and vegetables, individuals do not know about the benefits of whole-grain cereal products. Second, individuals tend to think that whole-grain cereal products are not very tasty. And third, whole-grain cereal products are less common and many are difficult to identify as being whole-grain (problem of labelling). Last, time and money have been cited as obstacles to eating more nutritiously(Reference Adams and Engstrom37).

Whole-grain and wholemeal

The terms ‘whole-grain’ and ‘wholemeal’ are mostly used synonymously. It is generally believed that whole-grain products are made with wholemeal flour, and that they may secondarily also contain intact grains. But the form in which grain is incorporated into food, intact or milled, is nutritionally significant. Thus ‘wholemeal’ (made of milled whole-grain flour) and ‘whole-grain’ (made with intact cereal grains) breads have different effects on postprandial glycaemia. The whole-grain breads produce a significantly lower glycaemic response than the wholemeal breads(Reference Jenkins, Wesson and Wolever38). This underlines the importance of food structure on physiology. Thus, for clarity, the term ‘whole-grain’ should be used for cereal products containing more or less intact cereal kernels, and ‘wholemeal’ for cereal products made of more or less refined flour, in which bran, germ and endosperm are first separated, and then reassembled, in proportions that rarely correspond to those of intact grains, as the germ fraction is generally removed.

Current hypotheses and mechanisms for the protective action of whole-grain cereals

The mechanisms underlying the health benefits of whole-grain cereals are undoubtedly multi-factorial. A recent cross-sectional study on 938 healthy men and women showed that a higher consumption of whole grains, bran and germ was associated with a significant decrease in plasma homocysteine (hyperhomocysteinaemia is a risk factor for CVD) and of some markers of blood glucose control, inflammation and lipid status(Reference Jensen, Koh-Banerjee and Franz17). Other studies have linked the consumption of high-whole-grain diets with improved BMI and insulin sensitivity, lower concentrations of serum TAG, total and LDL-cholesterol and inflammation markers, and higher plasma or serum enterolactone(Reference van de Vijver, van den Bosch and van den Brandt2, Reference Jacobs, Pereira and Stumpf39Reference Newby, Maras and Bakun42). Except for enterolactone, for which high serum levels are associated with reduced risk of CVD(Reference Vanharanta, Voutilainen and Lakka43), all of the other biomarkers, when outside a normal healthy range, are all risk factors associated with the development of diabetes and CVD. There is the same kind of significant negative association between whole-grain consumption and the risk of digestive cancer(Reference Levi, Pasche and Lucchini44, Reference Slavin45). Other mechanisms are involved in this, including the capacity of several whole-grain compounds to suppress tumour growth(Reference Slavin, Martini and Jacobs46). The next section describes the main known mechanisms by which whole-grain cereals help protect the gut and prevent the development of obesity, diabetes, CVD and cancers.

Food structure

The structure of food has long been recognised as an important parameter governing the health benefit of whole-grain cereal products. The first study was performed in 1977 by Haber et al. on the influence of apple structure (intact apples v. apple purée v. fibre-free apple juice) on satiety, plasma glucose and serum insulin. The removal of fibre and/or the disruption of the physical food structure was accompanied by reduced satiety, disturbed glucose homeostasis and an inappropriate insulin response(Reference Haber, Heaton and Murphy47). Almost 10 years later, it was shown that simply swallowing carbohydrate-rich foods (rice, apple, potato and sweetcorn) without chewing was sufficient to significantly decrease postprandial glycaemia(Reference Read, Welch and Austen48). This was the simplest way to emphasise the importance of food structure (chewing v. no chewing) on digestion. Then, Jenkins et al. studied the effects of wholemeal and wholegrain breads and showed that the glycaemic index (GI) of wholemeal breads (wheat or barley flour-based) without intact grains was the same as that of white bread made of refined flour (>90), and that increasing the intact barley kernel or cracked wheat grain content of the bread (50 and 75 %) resulted in a significantly large decrease in the GI from 92–96 to 39(Reference Jenkins, Wesson and Wolever38). Thus, an intact botanical food structure is more important than the composition of the food (the presence of fibre in wholemeal bread and absence from white bread) for influencing physiological responses like those related to satiety and glucose metabolism. Many later studies have confirmed these results, emphasising the importance of preserving the natural initial fibrous network, particularly in more or less intact wheat, barley, rye and oat kernels(Reference Fardet, Leenhardt and Lioger49Reference Nilsson, Ostman and Granfeldt52).

Whole-grain cereals as a rich source of fibre

Dietary fibre is defined by the AACC as ‘the edible parts of plants or analogous carbohydrates that are resistant to digestion and absorption in the human small intestine with complete or partial fermentation in the large intestine. Dietary fibre includes polysaccharides, oligosaccharides, lignin and associated plant substances. It promotes beneficial physiological effects including laxation and/or blood cholesterol attenuation and/or blood glucose attenuation’(53). This definition includes that fraction of starch not digested in the small intestine, resistant starch (RS). Whole-grain wheat may contain from 9 to 17 g total fibre per 100 g edible portion (Table 2), which is more than in most vegetables (generally < 6 g/100 g edible portion). Thus, consuming whole-grain cereal products is undoubtedly a good way of increasing the fibre intake from the 10–15 g/d eaten by most Western populations to the recommended level of about 30–35 g/d.

Wheat is relatively poor in soluble fibre. It has been found that the soluble:insoluble fibre ratio is about 1:5 for whole-grain wheat, 1:10 for wheat bran and 1:3 for wheat germ (Table 2). Whole-grain wheat therefore provides large quantities of insoluble fibre (up to 11 g/100 g) and RS (up to 22 % for certain high-amylose barley varieties(Reference Nilsson, Ostman and Holst54)). Cereal fibre is now recognised to be beneficial for bowel health. Wheat has a great diversity of fermentable carbohydrates. Except for lignin, whose nutritional benefits are not really known, all the types of fibre compounds, including soluble and insoluble fibre, oligosaccharides and RS, have important physiological properties and provide significant health benefits(Reference Liu18, Reference Topping55). For example, soluble fibre increases viscosity, which delays gastric emptying and limits glucose diffusion towards the enterocytes for absorption. This leads to a lower glucose response when sufficient quantities are ingested(Reference Wood56).

Cereal fibres also increase satiety and help control body weight(Reference Koh-Banerjee, Franz and Sampson57). The mechanisms by which dietary fibre positively affect body weight have been previously described: briefly, they involve hormonal effects via reduction of the insulin secretion, metabolic effects via increased fat oxidation and decreased fat storage due to greater satiety, and colonic effects via SCFA production(Reference Slavin58). Thus, the consumption of highly viscous fibre such as β-glucans, found mainly in barley and oats, is now recommended for the management of glucose homeostasis in type 2 diabetic subjects(Reference Jenkins, Jenkins and Zdravkovic59). Soluble fibre has also been shown to reduce cholesterolaemia in ileostomy subjects(Reference Zhang, Hallmans and Andersson60) by probably favouring an increase in bile acid excretion as shown in ileostomates following oat β-glucans consumption(Reference Lia, Hallmans and Sandberg61). Increased bile acid excretion stimulates bile acid synthesis from serum cholesterol, so reducing cholesterolaemia(Reference Lia, Hallmans and Sandberg61).

The fermentation of fibre and RS within the colon produces SCFA that are associated with a lower risk of cancer(Reference Scheppach, Bartram and Richter62, Reference Slavin63), favouring the development of a healthy colonic microbiota (i.e. prebiotic effect)(Reference Costabile, Klinder and Fava64). These SCFA also reduce the proliferation of human colon cancer cell lines in vitro (Reference Scheppach, Bartram and Richter62, Reference Slavin63). RS is known to produce large quantities of butyrate(Reference Brouns, Kettlitz and Arrigoni65). The increased butyrate production by rats fed wheat bran is negatively associated with the proliferation of colon crypt cells that are involved in the development of colorectal cancer(Reference Boffa, Lupton and Mariani66). RS also significantly increases fat oxidation in humans, probably by increased SCFA production that inhibits glycolysis in the liver, so rendering it more dependent on fat-derived acetyl CoA as fuel, this effect being associated with a concomitant decrease in carbohydrate oxidation and fat storage(Reference Higgins, Higbee and Donahoo67).

In contrast, insoluble fibre, which is poorly fermented in the colon, favours an increased transit time and greater faecal bulking(Reference McIntosh, Noakes and Royle68), two parameters that probably prevent colon cancer by diluting carcinogens and reducing their time in contact with epithelial cells(Reference Ferguson and Harris69). The fermentation of some fibre also increases mineral absorption in rats, mainly by increasing the surface area available for absorption (epithelial cell hypertrophy) and/or by favouring better hydrolysis of phytic acid via enhanced fermentation, as was shown with RS(Reference Lopez, Coudray and Bellanger70, Reference Lopez, Levrat-Verny and Coudray71) and inulin (a fructan-type compound)(Reference Coudray, Bellanger and Castiglia-Delavaud72, Reference Lopez, Coudray and Levrat-Verny73).

Whole-grain cereals and butyrate production

Whole-grain cereal products are an important indirect source of butyrate, produced notably through RS fermentation(Reference Brouns, Kettlitz and Arrigoni65). Butyrate has cancer-preventing properties in rats by inducing apoptosis(Reference Reddy, Hamid and Rao74) or reducing tumour mass(Reference McIntyre, Gibson and Young75). But its positive physiological action may not be restricted to these two effects. The precise mechanisms involved in the anti-colon cancer effect of butyrate have been reviewed from in vitro, animal and human studies and they mainly include a combination of several physiological modifications in relation to abnormal cell growth inhibition, immune system stimulation and modulation of DNA repair and synthesis(Reference Brouns, Kettlitz and Arrigoni65). Butyrate might also protect against breast and prostate cancers, as shown by in vitro studies on mammary(Reference Heerdt, Houston and Anthony76) and prostate(Reference Ellerhorst, Nguyen and Cooper77) cancer cell lines(Reference Brouns, Kettlitz and Arrigoni65). The RS content of whole-grain cereal products depends on the proportion of the different types of RS: RS1 which is physically inaccessible to α-amylase, RS2 which is raw starch granules, and RS3 which is recrystallised/retrograded amylose that is formed when cooked food cools. It is therefore difficult to obtain precise data on the RS content of whole-grain cereal products, but some products are enriched in RS by selecting high-amylose varieties of cereal. Nevertheless, products containing whole grains or made from high-amylose cereal varieties will have proportionally higher RS contents and produce more butyrate, as was shown in human subjects fed various breads, breakfast cereals and crackers(Reference Bird, Vuaran and King78, Reference Liljeberg and Bjorck79). Whole-grain cereal products with an intact botanical structure, that is with intact kernels, will have a higher RS1 content, since it is inaccessible to α-amylase, and butyrate production. The relationship between the consumption of whole-grain cereals and/or their bran and germ fractions, butyrate production and long-term health effects deserve to be studied more thoroughly in human subjects, particularly because of the effects in rats of butyrate on fat oxidation and of total SCFA production on cholesterol synthesis reduction(Reference Hara, Haga and Aoyama80).

The ‘second-meal effect’

The ‘second-meal effect’ is characterised by an improved carbohydrate tolerance at a meal (either lunch or breakfast, called the ‘second meal’) about 4–5 or 10–12 h after the consumption of a low-GI meal (i.e. the ‘first meal’), an effect which may contribute to the long-term metabolic benefits of low-GI diets. It was first described by Jenkins et al. who used viscous guar gum(Reference Jenkins, Wolever and Nineham81), and thereafter for low-GI carbohydrate foods such as lentils(Reference Jenkins, Wolever and Taylor82). Recently, mechanisms have been proposed to explain the sustained positive effect of low-GI whole-grain products composed of intact barley or rye kernels consumed at diner or breakfast on the glycaemic response at the following meal, breakfast or lunch(Reference Nilsson, Ostman and Granfeldt52, Reference Nilsson, Ostman and Holst54, Reference Nilsson, Granfeldt and Ostman83).

The physiological mechanisms involved appear to differ according to the interval between the two meals, dinner to breakfast (about 10–12 h) or breakfast to lunch (about 4–5 h). The shorter period seems to be sufficient for the low-GI feature of the cereal product consumed at breakfast to reduce the glucose response at lunch, probably by improving blood sugar regulation and insulin sensitivity(Reference Nilsson, Ostman and Holst54). The longer interval between dinner and breakfast involved the fermentation of indigestible carbohydrates in the colon, reduced plasma NEFA and modified glucose metabolism. This indicates that the presence of specific dietary fibre (soluble or insoluble or RS) in boiled barley kernels is more significant in this ‘second-meal effect’ than is its low GI.

SCFA produced during the fermentation of fibre in the colon might be particularly involved(Reference Nilsson, Granfeldt and Ostman83) through at least three potential processes: a possible decrease of the gastric emptying rate by SCFA as reviewed in rats and humans(Reference Cherbut84), notably through an increased level of the polypeptide YY in blood by SCFA, that may lead to a reduced rate of glucose entry into the bloodstream; the ability of propionate and acetate to reduce serum NEFA in humans(Reference Wolever, Spadafora and Eshuis85), circulating fatty acids being able to induce peripheral and hepatic insulin resistance in humans(Reference Homko, Cheung and Boden86); and, finally, the possible specific action of propionate on glucose metabolism by increasing hepatic glycolysis and decreasing hepatic glucose production as shown in isolated rat hepatocytes(Reference Anderson and Bridges87). A later study on healthy subjects(Reference Nilsson, Ostman and Holst54) confirmed that the low-GI feature of the products consumed in the evening meal was not per se involved in the improved glucose response at breakfast, and that the lower plasma NEFA concentration combined with the high plasma propionate content (from fermentation in the colon) contributed to the overnight benefits in terms of glucose tolerance(Reference Nilsson, Granfeldt and Ostman83). The quantity and quality of the indigestible carbohydrates (for example, barley fibre and RS) are most important. There is also an important relationship between gut microbial metabolism and insulin resistance(Reference Nilsson, Ostman and Holst54).

These results suggest that the influence of carbohydrates on glucose tolerance over a longer time (semi-acute) is optimal when the food structure is preserved (i.e. a low-GI feature) and content of RS and/or fibre is high (i.e. production of specific SCFA). Eating barley or rye kernels for breakfast resulted in lower cumulative postprandial increases in blood glucose after breakfast, lunch and dinner (a total of 9·5 h) than did a breakfast of white-wheat bread(Reference Nilsson, Ostman and Granfeldt52). From a technological point of view, the quantity and quality of the indigestible carbohydrates is therefore particularly important, in addition to preserving a more or less intact botanical food structure, for a better control of glucose metabolism, especially to prevent type 2 diabetes.

Whole-grain cereals as rich sources of anti-carcinogenic compounds

A survey of 61 433 women found that a high consumption of whole grains (hard whole-grain rye bread, soft whole-grain bread, porridge, and cold breakfast cereals) was associated with a lower risk of colon cancer(Reference Larsson, Giovannucci and Bergkvist11). An inverse association between cereal fibre and whole-grain cereal consumption and small-intestinal cancer incidence has also been reported(Reference Schatzkin, Park and Leitzmann12). The roles played by dietary fibre and phytochemicals in preventing intestinal cancer in humans and animals have been reviewed and discussed for both human intervention and animal studies(Reference Slavin45, Reference Ferguson and Harris69, Reference Liu88). The positive action of the wheat bran oil on colon tumour incidence in rats (azoxymethane-induced cancer)(Reference Reddy, Hirose and Cohen89) and mice (Min cancer model)(Reference Sang, Ju and Lambert90) has also been demonstrated. This anti-carcinogenic effect is mainly attributed to the antioxidant and anti-inflammatory properties of several bioactive compounds, as increased oxidative stress and inflammation are involved in cancer aetiology(Reference Bartsch and Nair91). Phenolic acids, flavonoids, carotenoids, vitamin E, n-3 fatty acids, lignan phyto-oestrogens, steroid saponins (found mainly in oats), phytic acid and Se are all potential suppressors of tumour growth, but human, animal and/or in vitro cell studies indicate that their mechanisms of action may differ (Tables 3 and 4)(Reference Slavin, Martini and Jacobs46, Reference Ferguson and Harris69, Reference Graf and Eaton92Reference Shamsuddin95). For example, cereal lignans are converted by fermentation into mammalian lignans or phyto-oestrogens (enterodiol and enterolactone). These may have a weak oestrogenic activity, and may protect against hormone-dependent cancers (prostate and breast cancers) and/or colon cancer(Reference Adlercreutz96). Studies on postmenopausal women, ovariectomised rats and liver and breast cancer cell cultures indicate that phyto-oestrogens inhibit cell proliferation by competing with oestradiol for type II oestrogen binding sites(Reference Adlercreutz, Mousavi and Clark97, Reference Markaverich, Webb and Densmore98). Phytic acid would help reduce the rate of cell proliferation during the initiation and post-initiation stages (for example, decreased incidence of aberrant colon crypt foci) by complex mechanisms that involve its antioxidant properties, signal transduction pathways, gene regulation and immune response through enhancing the activity of natural killer cells(Reference Reddy99), and its anti-carcinogenic effect seems to be dose-dependent(Reference Ullah and Shamsuddin100). The high phytic acid content of whole-grain cereals (up to 6 % in wheat bran) has led to questions about whether the anti-cancer activity of wheat bran should be attributed more to phytic acid than to dietary fibre(Reference Ferguson and Harris69, Reference Graf and Eaton92). Indeed, pure phytic acid is more efficient at reducing the incidence and multiplicity of mammary tumours in rats than is the bran fraction (All Bran; Kellogg®)(Reference Vucenik, Yang and Shamsuddin101). The many anti-carcinogenic actions of flavonoids include their ability to inhibit various stages of tumour development in animals(Reference Hollman and Katan102) and to reduce the mutagenicity of several dietary carcinogens in Salmonella typhimurium TA98NR(Reference Edenharder, Rauscher and Platt103). The anti-carcinogenic activity of ferulic acid is mainly attributed to its antioxidant capacity; it scavenges the free oxidative radicals that are involved in the aetiology of cancer, and to its ability to stimulate cytoprotective enzymes(Reference Barone, Calabrese and Mancuso104, Reference Kawabata, Yamamoto and Hara105). Studies on azoxymethane-treated rats indicate that vitamin E and β-carotene inhibit the progression of aberrant crypt foci to colon cancer, especially the later stages of carcinogenesis, while wheat bran is better at inhibiting earlier stages(Reference Alabaster, Tang and Shivapurkar106). Lignins, by hydrophobically binding bile salts, might reduce the formation of carcinogens from them(Reference Eastwood and Girdwood107, Reference Eastwood and Hamilton108). Their adsorptive ability would increase with increased methylation of the hydroxyl moieties on the phenyl-propane units(Reference Eastwood and Girdwood107, Reference Eastwood and Hamilton108). Lignins also reduce DNA lesions in rat testicular cells and lymphocytes both in vitro and ex vivo (Reference Labaj, Slamenova and Lazarova109). Se inhibits the occurrence of neoplasia in rats and mice, suggesting that an Se-poor diet is associated with an increased prevalence of neoplasia in specific human populations(Reference Wattenberg110). This probably depends on the activity of the selenoprotein glutathione peroxidase, which is involved in the development of cancers(Reference Jablonska, Gromadzinska and Sobala111). Cereal bioactive compounds act via several other anti-mutagenic and anti-carcinogenic mechanisms(Reference Stavric112). Important ones are the adsorption and dilution of carcinogens by insoluble dietary fibre and lignins(Reference Ferguson and Harris69, Reference Alabaster, Tang and Shivapurkar106, Reference Harris and Ferguson113, Reference Harris, Roberton and Watson114), and the action of SCFA produced by fibre fermentation(Reference Morita, Tanabe and Sugiyama115). Butyrate is a major factor, as more is produced in the presence of RS, and favours apoptosis in human cancer cell lines(Reference Scheppach, Bartram and Richter62) and DNA repair in rats(Reference Toden, Bird and Topping116). Interestingly, contrary to what was believed since the works of Burkitt emphasising the preponderant role of fibre in the prevention of Western diseases, notably colon cancer observed in Western countries and not in African rural population consuming high levels of dietary fibre(Reference Story and Kritchevsky117), it is more and more believed today that the effect against colon cancer development might be before all attributed to RS(Reference Bauer-Marinovic, Florian and Muller-Schmehl118), since a lower risk of colon cancer was recently observed in populations with a low level of fibre consumption but with a high intake of RS(Reference Bingham119, Reference O'Keefe, Kidd and Espitalier-Noel120). This reinforces the idea that specific products of RS fermentation within the colon, such as butyric acid, are the active components. Betaine(Reference Cho, Willett and Colditz121) may be added to the list of anti-carcinogenic compounds, as its concentration can reach 0·3 % in whole-grain wheat and 1·5 % in wheat bran (Table 2).

To summarise, the anti-carcinogenic effects of insoluble fibre (including lignin), phytochemicals and wheat bran oil can be distinguished. Insoluble fibre may act directly by adsorbing or diluting carcinogens (through increased faecal bulk by water absorption), or indirectly by decreasing colon pH (through SCFA production) and increasing butyrate production. The role of phytochemicals is complex and multi-factorial, and notably involves their antioxidant properties since increased oxidative stress is a major factor in the aetiology of cancers(Reference Bartsch and Nair91, Reference Klaunig, Xu and Isenberg122). The exact components of wheat bran oil that reduce the development of colon tumours are still to be identified(Reference Reddy, Hirose and Cohen89, Reference Sang, Ju and Lambert90). However, animal experiments indicate that dietary fibre, particularly soluble fibre, may not protect against or even enhance carcinogenesis. This may be due to the abrasive property of insoluble fibre, a too low pH ( < 6·5) reached within the colon following soluble fibre and RS fermentation, the enhanced colon glucuronidase activity (that converts conjugated carcinogens to free carcinogens) and the increased production of secondary bile acids (tumour promoters) within the colon due to the increased viscosity of some soluble fibre which reduces the reabsorption of bile salt in the small intestine(Reference Harris and Ferguson123).

Whole-grain cereals as a rich source of antioxidants

Whole-grain cereals can protect the body against the increased oxidative stress that is involved and/or associated with all the major chronic diseases: metabolic syndrome(Reference Ford, Mokdad and Giles124), obesity(Reference Higdon and Frei125, Reference Keaney, Larson and Vasan126), diabetes(Reference Evans, Goldfine and Maddux127, Reference Maiese, Morhan and Chong128), cancers(Reference Bartsch and Nair91) and CVD(Reference Cai and Harrison129, Reference Castelao and Gago-Dominguez130). Whole-grain cereals are good sources of antioxidants (thirty-one compounds or groups of compounds are listed in Table 4), as shown by measurements made in vitro of the antioxidant capacity of whole-grain, bran and germ fractions(Reference Martinez-Tome, Murcia and Frega131Reference Zielinski and Kozlowska135). However, this may not be the same in vivo (Reference Fardet, Rock and Rémésy136), and up to today, to my knowledge, the number of studies exploring the in vivo antioxidant effect of whole-grain cereals and/or their fractions in human subjects does not exceed eleven(Reference Andersson, Tengblad and Karlstrom137Reference Wang, Han and Zhang147). The antioxidants in cereals differ in their structure and mode of action(Reference Slavin, Martini and Jacobs46, Reference Fardet, Rock and Rémésy136). There are indirect antioxidants, such as Fe, Zn, Cu and Se, which act as cofactors of antioxidant enzymes, and direct radical scavengers such as ferulic acid, other polyphenols (lignans, anthocyanins and alkylresorcinols), carotenoids, vitamin E and compounds specific to cereals other than wheat, such as γ-oryzanol in rice and avenanthramides in oats. These can neutralise free radicals and/or stop the chain reactions that lead to the production of oxidative radical compounds (for example, the lipid chain peroxidation stopped by vitamin E within cell membranes). Another antioxidant mechanism involves phytic acid, which can chelate Fe and thus stop the Fenton reaction producing the highly oxidative and damaging free radical OH, ultimately reducing lipid peroxidation(Reference Graf, Empson and Eaton148). Lignins are also considered to be antioxidants in vitro (radical-scavenging activity)(Reference Dizhbite, Telysheva and Jurkjane149), but precisely how they act in vivo is not known: they may adsorb oxidative damaging compounds within the digestive tract in a way similar to bile salts adsorption(Reference Eastwood and Girdwood107, Reference Eastwood and Hamilton108). While the action of cereal antioxidants is not well characterised once the epithelial barrier has been crossed, there is a growing belief that cereal antioxidants protect the intestinal epithelium cells from oxygen-derived free radicals(Reference Fardet, Rock and Rémésy136, Reference Vitaglione, Napolitano and Fogliano150), particularly those produced by bacteria that may help form active carcinogens by oxidising procarcinogens or those that may result from increased stool Fe content (Fenton reaction) due to a diet high in red meat(Reference Babbs151). The concept of ‘dietary fibre-bound phytochemicals/phenolic compounds’ was proposed recently(Reference Liu18, Reference Vitaglione, Napolitano and Fogliano150). The authors suggest that the antioxidant polyphenols survive digestion in the small intestine because most of them are bound to fibre (for example, esterification of phenolic acids to arabinoxylans) in the cereal food matrix. They reach the colon where the fibre is fermented and some of the antioxidants are released(Reference Vitaglione, Napolitano and Fogliano150). Vitaglione et al. hypothesised ‘the slow and continuous release in the gut of the dietary fibre bound antioxidants’, such as that of ferulic acid, which will determine the effects of these antioxidants, and considered dietary fibre to be a ‘natural functional ingredient to deliver phenolic compounds into the gut’(Reference Vitaglione, Napolitano and Fogliano150). For example, only 0·5–5 % of the ferulic acid is absorbed within the small intestine, mainly the soluble free fraction(Reference Adam, Crespy and Levrat-Verny152Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154), and this typical whole-grain wheat phenolic acid (about 90 % of total phenolic acids) would probably exert a major action in the protection of the colon from cancer. Thus, bound antioxidant phenolic acids might act along the whole length of the digestive tract by trapping oxidative compounds. This fraction of bound polyphenols has often led to an important underestimation of the real antioxidant capacity of whole-grain cereals – and of their fractions – as measured in vitro and generally based on the measurement of the easily extractable polyphenol fraction(Reference Perez-Jimenez and Saura-Calixto133, Reference Pellegrini, Serafini and Salvatore155). In vivo studies are now needed to examine this hypothesis, and to characterise and quantify this potential antioxidant effect within the digestive tract.

The antioxidants in whole-grain cereals act via different, complex, and synergetic mechanisms in vivo. However, the antioxidant action of whole-grain cereals has not yet been convincingly validated in human subjects and requires further exploration.

Whole-grain cereals as rich sources of magnesium

Among plant-based foods, whole-grain cereals, together with legumes, nuts and seeds, are one of the best sources of Mg: whole-grain wheat contains 104 mg Mg/100 g, wheat bran 515 mg, and wheat germ 245 mg (Table 2). The high Mg content of whole-grain cereals may explain its favourable impact on insulin sensitivity and diabetes risk (Fig. 2)(Reference McCarty156), diabetes being otherwise frequently associated with Mg deficiency(Reference Durlach and Collery157). Mg can increase insulin secretion and the rate of glucose clearance from the blood in humans(Reference Paolisso, Sgambato and Gambardella158, Reference Paolisso, Sgambato and Pizza159). This was also proposed to explain the lower insulin response in obese and overweight adults following the consumption of a whole-grain-based diet as compared with those on a refined cereal-based diet(Reference Pereira, Jacobs and Pins160). High-Mg diets reduce insulin resistance in rats fed a high-fructose diet(Reference Balon, Jasman and Scott161); they also reduce the development of spontaneous diabetes in obese Zucker rats, a model of non-insulin-dependent diabetes mellitus, but these rats had to be given Mg before the onset of diabetes to obtain protection(Reference Balon, Gu and Tokuyama162). Most explanations of the prevention of type 2 diabetes by Mg are based on the finding that Mg stimulates insulin-dependent glucose uptake in elderly subjects(Reference Paolisso, Sgambato and Gambardella158, Reference Gould and Chaudry163). It also protects Mg-deficient animals from the production of reactive oxygen species(Reference Weglicki, Mak and Kramer164). Reactive oxygen species are partly responsible for the increased hyperglycaemia-mediated oxidative stress in diabetic subjects(Reference Ceriello, Bortolotti and Crescentini165, Reference Pereira, Ferderbar and Bertolami166). Mg also acts as a mild physiological Ca antagonist(Reference Iseri and French167). Obese and diabetic patients with insulin resistance have excess free intracellular Ca and these two clinical conditions are associated with hypertension(Reference Resnick168). In addition, Mg helps keep the concentration of intracellular Ca optimal through various complex cellular mechanisms involving Ca channels, Ca sequestration/extrusion by the endoplasmic reticulum and Ca binding sites on proteins and membranes(Reference McCarty156). Finally, low serum plasma Mg has been positively associated with a higher risk of coronary atherosclerosis or acute thrombosis(Reference Liao, Folsom and Brancati169), suggesting that whole-grain cereal Mg might also contribute to the prevention of CVD. This may also involve the inhibition of platelet-dependent thrombosis by Mg supplementation in patients with coronary artery disease(Reference Shechter, Merz and Paul-Labrador170) and the positive effect of Mg upon blood pressure regulation in hypertensive patients(Reference Kawano, Matsuoka and Takishita171). The capacity of a regular prolonged consumption of whole-grain cereals to sustain a high plasma Mg concentration therefore deserves to be investigated in the context of type 2 diabetes prevention.

Fig. 2 Current accepted mechanisms for how whole grain protects against major chronic diseases (modified with permission from Professor I. Björck (University of Lund, Sweden); see the HealthGrain brochure for original diagram: ‘Progress in HEALTHGRAIN 2008’, a project from the European Community's Sixth Framework Programme, FOOD-CT-2005-514008, 2005–2010; see Poutanen et al. (Reference Poutanen, Shepherd and Shewry478) for more details about the Project). GI, glycaemic index; II, insulinaemic index.

The action of some anti-nutrients on starch hydrolysis and glycaemia

Whole-grain cereals are also a source of antinutrients with both adverse and positive health effects. The most important are phytic acid, lectins, tannins, saponins and inhibitors of enzymes such as proteases and α-amylases. Their main negative effect is their ability to reduce the bioavailability and the absorption of some nutrients (for example, the chelation of minerals by phytic acid and tannins), the binding of lectins to epithelial cells that damages the intestinal microvillae, and inhibition of digestive enzymes by tannins, which inhibits growth in animals(Reference Al-Mamary, Al-Habori and Al-Aghbari172, Reference Thompson173). Cereal products in the human diet are cooked; this leads to losses of antinutrients such as lectins and enzyme inhibitors, and the major health outcome appears to be the low dietary Fe bioavailability in African populations that consume sorghum or finger millet-based beverages, gruels and porridges, both cereals containing phytic acid and a high tannin content(Reference Gillooly, Bothwell and Charlton174, Reference Tatala, Svanberg and Mduma175). For example, the phytate and Fe-binding phenolic compounds in whole-grain millet flour may reach 0·6 g/100 g (DW)(Reference Lestienne, Besancon and Caporiccio176). This is one of the key factors responsible for Fe-deficiency anaemia in developing countries(Reference Tatala, Svanberg and Mduma175). On the other hand, the use of traditional processing such as germination, soaking, pre-fermentation and cooking may help to decrease the tannin and phytic acid contents, so improving Fe bioavailability(Reference Hassan and El Tinay177Reference Towo, Matuschek and Svanberg180).

However, phytic acid, lectins, protease inhibitors and tannins also contribute to the low-GI property of whole-grain foods(Reference Thompson181, Reference Yoon, Thompson and Jenkins182). In wheat and derived whole-grain food products, since lectins and enzyme inhibitors are inactivated by cooking processes, this is primarily phytic acid which would reduce glycaemia through several potential mechanisms: thus, binding with proteins closely associated with starch, association with digestive enzymes, chelation of Ca required for α-amylase activity, direct binding with starch, effect on starch gelatinisation during cooking processes and slowing of gastric emptying rate might be involved(Reference Thompson181).

Conclusion

The proposed mechanisms by which whole-grain cereals may protect the body are shown in Fig. 2. The most important ones are the preservation of food structure, fibre fermentation in the colon, the hypoglycaemic and hypoinsulinaemic, antioxidant, anti-inflammatory and anti-carcinogenic properties of several bioactive compounds, improved insulin sensitivity by Mg and reduced hyperhomocysteinaemia by betaine, a significant CVD risk factor (for details about betaine, see the ‘New hypotheses’ section below). However, an extensive list of all the bioactive compounds in whole-grain wheat and its fractions (Table 2), the ways they act and their health effects as isolated free compounds (Tables 3 and 4) makes it possible to formulate new hypotheses to explain the protective role of whole-grain cereals. Whole-grain cereals, particularly wheat and/or wheat bran and germ, are also a source of n-3 fatty acids (especially α-linolenic acid), sulfur compounds (reduced glutathione (GSH), oxidised glutathione (GSSG), methionine and cystine), oligosaccharides (fructans, raffinose and stachyose), P, Ca, Na, K, B vitamins, flavonoids (for example, anthocyanins and isoflavonoids), alkylresorcinols, betaine, choline, phytosterols, inositols, policosanol and melatonin. The actions of these compounds will be described in the next ‘New hypotheses’ section. The antioxidant hypothesis will be discussed with a broader perspective, as well as the health benefits of active compounds from whole-grain cereals that are less often studied, such as B vitamins, sulfur compounds, methyl donors and lipotropes, α-linolenic acid, lignins, oligosaccharides, policosanol and melatonin.

New hypotheses: a broader perspective for the protective action of whole-grain cereals

The antioxidant hypothesis must not be reduced to free radical scavenging and antioxidant enzyme activation

There is more and more evidence that the primary effect of antioxidants from whole-grain cereals is in the digestive tract, where they protect intestinal epithelial cells from attack by free radicals(Reference Fardet, Rock and Rémésy136, Reference Vitaglione, Napolitano and Fogliano150). However, the mechanisms by which antioxidants that cross the intestinal barrier protect the body remain uncertain. Published studies on animals and human subjects fed the free compounds give rise to new explanations of the antioxidant protection by whole-grain cereals. The antioxidant action of whole-grain cereals might be multi-factorial and much more complex than it first appears. There are at least four new mechanisms to be studied in the context of whole-grain cereals: the action of polyphenols on cell signalling and gene regulation modifying the redox status of tissues and cells, the action of sulfur amino acids on glutathione synthesis, the possible stimulation of endogenous antioxidants by whole-grain cereal bioactive compounds, and the underestimated antioxidant properties of phytic acid and lignin.

Whole-grain cereals as a source of polyphenols involved in cell signalling

The polyphenols in complex foods are generally not readily absorbed in the small intestine: 2–5 % for whole-grain cereal phenolic acids (Table 2), and 30–40 % for flavonoids from vegetables, beverages and fruits, depending on the food(Reference Manach, Williamson and Morand183). The resulting plasma concentrations of these absorbed compounds are generally in the nanomolar (nm) or micromolar (μm) range, lower than that of endogenous antioxidant compounds such as GSH and vitamin C (millimolar). However, this does not mean that they have no antioxidant action. Some quite recent studies on isolated compounds have shown that flavonoids(Reference Choi, Choi and Shin184, Reference Crespo, García-Mediavilla and Gutiérrez185) and phenolic acids(Reference Maggi-Capeyron, Ceballos and Cristol186, Reference Yun, Koh and Kim187) act on cell signalling pathways, so modifying gene regulation and/or cell redox status, as has been discussed in several recent reviews(Reference Moskaug, Carlsen and Myhrstad188Reference Williams, Spencer and Rice-Evans191). However, most of the studies were performed with flavonoids, not phenolic acids which are more abundant in whole-grain wheat (up to 100 mg/100 g) than are flavonoids (30–43 mg/100 g) (Table 2). Results obtained with isolated flavonoids, mainly in in vitro cell cultures, may be extrapolated to flavonoids found in whole-grain wheat once they have entered the bloodstream and then reached cells. Little work has been done to precisely identify wheat flavonoids. Nevertheless, some of them are catechin and proanthocyanidins(Reference McCallum and Walker192), tricine(Reference Ferguson and Harris69), apigenin glycosides(Reference Feng and McDonald193), and vicenin and schaftosides(Reference Gallardo, Jiménez and García-Conesa194). These flavonoids may act as signals within cells. The main mechanisms probably involve the redox status and antioxidant and pro-inflammatory genes activated by increased oxidative stress, i.e. a modified redox state of the cell, through signalling pathways that may be up- and down-regulated by polyphenols via activation or inactivation of transcription factors such as NF-κB(Reference Rahman, Biswas and Kirkham189, Reference Yun, Koh and Kim187) or activator protein-1 (AP-1)(Reference Maggi-Capeyron, Ceballos and Cristol186). Thus, flavonoids can increase GSH synthesis through the transcription factor Nrf2 (nuclear factor-erythroid 2-related factor 2) which binds to specific antioxidant/electrophile response element (AREs/EpRE)-containing gene promoters(Reference Moskaug, Carlsen and Myhrstad188). For example, oxidised quercetin (quinone) can react with thiols in the Keap1 protein (Kelch-like ECH-associated protein 1 bound to the cytoskeleton), releasing Nrf2 and then activating specific genes via ARE/EpRE involved in GSH synthesis(Reference Moskaug, Carlsen and Myhrstad188). Here, more than the antioxidant property of the flavonoids, it is its activated or metabolised form which would be active within cells. Kaempferol and quercetin, two flavonoids, also modulate the production of γ-glutamylcysteine synthetase(Reference Myhrstad, Carlsen and Nordstrom195), an important enzyme in the synthesis of GSH. The authors conclude that flavonoids are important for regulating the intracellular concentration of GSH(Reference Myhrstad, Carlsen and Nordstrom195). There is therefore a strong link between the intra- and/or extra-cellular actions of polyphenols, redox cell status and gene regulation, broadening the notion of antioxidant polyphenols to activities other than just free radical scavenging. However, most studies have used higher polyphenol concentrations (>10 μm) than those found in vivo. For example, the postprandial plasma ferulic acid concentrations following wheat bran consumption in rats were about 1 μm(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154) and about 0·2 μm in human subjects(Reference Kern, Bennett and Mellon196). However, a study conducted in vitro on cell cultures with six wine phenolic acids in the 20 nm–20 μm range showed that ferulic, sinapic, p-coumaric and caffeic acids (all found in whole-grain wheat) are able to inhibit the action of pro-inflammatory transcription factor AP-1 as low as 20 nm in a range of 5–15 %(Reference Maggi-Capeyron, Ceballos and Cristol186). Besides, it may reasonably be supposed that the true plasma polyphenol concentration is higher than the 0·2–1 μm reached with ferulic acid due to the presence of other polyphenols such as sinapic acid and, to a lesser extent flavonoids, as recently reported in human subjects where a+5 μm increase in plasma total polyphenols has been observed 1 h after boiled wheat bran consumption(Reference Price, Welch and Lee-Manion146). Most of the sinapic acid in whole-grain wheat is free or in a soluble conjugated form (approximately equal to 70 %), and may reach a total concentration of 4–18 mg/100 g whole-grain wheat(Reference Li, Shewry and Ward197). However, whether the low plasma polyphenol concentrations obtained following a whole-grain cereal meal are compatible with cell signalling activity remains to be explored.

Whole-grain cereals are a rich source of sulfur compounds

The sulfur amino acid contents (methionine and cystine) of whole-grain wheat, wheat bran and germ are 0·5, 0·6 and 1·0 % (Table 2), and may be higher in some cereal varieties (see ranges in Table 2). Methionine and cystine are both precursors of GSH, an intracellular antioxidant, and as such contribute to the control of the cell oxidative status by participating in gene expression through modification of the thiol redox status, as has been recently reviewed(Reference Métayer, Seiliez and Collin198, Reference Tesseraud, Métayer Coustard and Collin199). Thus, rats fed a 0·6 % free methionine diet had a higher hepatic GSH content than rats fed a control 10 % casein-based diet without methionine supplementation(Reference Morand, Rios and Moundras200). It has also been shown in rat gut mucosa and plasma that an inadequate intake of sulfur amino acids leads to the oxidation of the thiol/disulfide redox status (expressed by the ratios cysteine:cystine and GSH:GSSG), i.e. a less reductive potential, that in the end increases oxidative stress(Reference Nkabyo, Gu and Jones201). Methionine also generates cysteine via the cystathionine pathway(Reference Tateishi, Hirasawa and Higashi202), cysteine being oxidised to cystine (two cysteine moieties linked by a disulfide bond).

For humans, average daily intakes of 305–2770 mg methionine and 197–1561 mg cystine have been reported for a usual diet(Reference Flagg, Coates and Eley203). The estimated daily requirements of methionine+cysteine are 910–2100 mg/d for a 70 kg adult(Reference Martin204). Based on the methionine and cystine content of commercially prepared whole-wheat bread (USDA database, 155 and 214 mg/100 g)(205) and on a daily consumption of one serving of whole-grain cereal products (i.e. about 30 g for a slice of bread)(Reference Smith, Kuznesof and Richardson206), whole-grain cereals provide an average 47 mg methionine and 64 mg cystine per d. This suggests that whole-grain cereals contribute little to methionine and cystine intakes, at least for low consumers. However, quite significant amounts of at least 280 mg methionine and 380 mg cystine per d can be obtained by following the USDA food guide pyramid that recommends between six and eleven daily servings of whole-grain cereal products. This would significantly contribute either to the average daily intakes as previously reported(Reference Flagg, Coates and Eley203) or to the daily recommendations(Reference Martin204). However, it is not known how a regular daily consumption of between six and eleven servings of whole-grain cereal products would contribute to GSH synthesis and/or an improved antioxidant status in humans.

GSH can be hydrolysed in the small intestine by γ-glutamyltransferase and/or absorbed intact, mainly in the upper jejunum(Reference Hagen, Wierzbicka and Bowman207). It is therefore available to cells where it may exert its physiological effects as an antioxidant, anti-carcinogenic and/or immunostimulating(Reference Gmünder, Roth and Eck208) agent and also as detoxifier of xenobiotics. Human subjects given a solution of 46 mg GSH/kg body weight (a single oral dose of 3 g) showed no significant increase in postprandial plasma GSH(Reference Witschi, Reddy and Stofer209). Dietary GSH, but also its dietary precursors methionine and cystine, are therefore not major determinants of circulating GSH(Reference Flagg, Coates and Eley203), probably because GSH is rapidly hydrolysed in the small intestine(Reference Witschi, Reddy and Stofer209); however, it might help detoxify reactive electrophiles in the diet within the intestinal lumen(Reference Hagen, Wierzbicka and Bowman207) or protect epithelial cells against attack by free radicals. The human daily total GSH consumption is 13–110 mg (mean 35 mg)(Reference Flagg, Coates and Eley203). Using the GSH highest content in whole-grain wheat (Table 2), that is about 5·7 mg/100 g, and eating 30 g whole-grain cereal per d as bread (about 38 % water), it may be calculated that whole-grain bread provides less than 1·3 mg GSH per d. Increasing the consumption of whole-grain cereal products to between six and eleven servings daily as recommended by the USDA food pyramid (epidemiological data show that an average 2·7 servings of whole-grain foods have beneficial health effects), especially servings containing wheat germ since this fraction may have 246 mg GSH/100 g – and probably more if total glutathione equivalents (GSH+(2 × GSSG)+protein-bound glutathione) are considered – might therefore provide a substantial supply of GSH. Thus, the total GSH content of high-grade extraction wheat flours (1·44–1·73 g ash/100 g) is 11·6–17·6 mg/100 g (with a water content for whole-grain wheat flour of 13·0 %), which is about three times the total GSH content of low-grade extraction wheat flours (0·54–0·59 g ash/100 g and 4·7–5·0 mg total GSH/100 g flour with an 11·9 % water content for white wheat flour), clearly showing that GSH is mainly in the bran(Reference Sarwin, Walther and Laskawy210). However, a higher total glutathione content of 15·8 mg/100 g (thirty-six wheat varieties) was evaluated from data by Li et al. for white wheat flours(Reference Li, Bollecker and Schofield211, Reference Weber and Grosch212). The contribution of total whole-grain wheat GSH to the antioxidant defence, either within the gut lumen or as a substrate supplying cysteine for endogenous GSH synthesis in the liver, might be explored by comparing low-methionine and whole-grain-rich diets.

The possible action of whole-grain cereal compounds on plasma uric acid level

A recent study on human subjects consuming apples demonstrated that the elevated plasma postprandial antioxidant level (+55 μm trolox equivalents after 1 h and stabilisation at about+20 μm trolox equivalents between 2 and 6 h; ferric-reducing ability of plasma (FRAP) assay) was due to increased uric acid and not to a significant increase in plasma vitamin C or polyphenols(Reference Lotito and Frei213). Fructose was thought to stimulate adenine nucleotide degradation leading to uric acid synthesis(Reference Lotito and Frei214). The authors proposed that the increased plasma antioxidant level following consumption of flavonoid-rich diets is due to an increase in uric acid, while sucrose, sorbitol, lactate and/or methylxanthines are also candidates for endogenous uric acid synthesis(Reference Lotito and Frei214). Uric acid is a powerful antioxidant whose concentration in human plasma can reach 160–450 μm, and can account for as much as 40–90 % of the plasma antioxidant capacity(Reference Lotito and Frei214). A recent study on human subjects has shown that there is little or no correlation between changes in plasma total phenolic acids and antioxidant capacity (FRAP assay) following the consumption of wheat bran, indicating that compounds other than phenolic acids contribute to the postprandial increase in plasma antioxidants to about+50 μm of FRAP between 1 and 3 h(Reference Price, Welch and Lee-Manion146). This increase is in the same range as that found by Lotito & Frei with apples(Reference Lotito and Frei213) and with other values reported by Price et al. with tea, red wine, spinach and strawberries, from+15 to+100 μm increase in plasma FRAP(Reference Price, Welch and Lee-Manion146). This cannot be explained by the low fructose content of wheat bran (about 50 mg/100 g), much lower than that of apples (about 5·7 g/100 g)(Reference Souci, Fachmann and Kraut215). However, whole-grain cereals contain an important package of bioactive compounds other than fructose or polyphenols whose effect upon endogenous antioxidant synthesis has not been explored. It would be therefore relevant to confirm this increase in plasma antioxidant level following wheat bran consumption, and to identify the mechanisms underlying such an increase, which is apparently not due to the increase in circulating plasma polyphenols alone(Reference Price, Welch and Lee-Manion146). Work is also needed to determine whether the consumption of whole-grain cereals and/or bran and germ fractions can significantly increase the plasma uric acid concentration to those produced by coffee (+5 %) or tea (+7 %)(Reference Natella, Nardini and Giannetti216).

Whole-grain cereals as a source of phytic acid and lignins

Phytic acid from whole-grain cereals has long been considered to be nutritionally negative, since it chelates minerals such as Zn, Fe, Ca and/or Mg, thus limiting their intestinal bioavailability(Reference Lopez, Leenhardt and Coudray217). This has been used as an argument for using refined flours instead of wholemeal wheat flours. However, phytic acid is also a strong antioxidant in vitro (Reference Graf and Eaton218), and may reach 6 % in the bran of certain wheat varieties (Table 2). It therefore needs to be determined whether the negative effect of phytic acid on mineral assimilation can be offset by its antioxidant activity and the high content in minerals of whole-grain wheat. Today, the answer to this is undoubtedly ‘yes’. First, the quantity of mineral chelated by phytic acid is apparently not high enough compared with the much greater quantity in whole-grain cereals compared with refined ones. Rats fed whole-wheat flour absorbed more minerals than rats fed white wheat flour(Reference Levrat-Verny, Coudray and Bellanger219). Besides, baking bread according to a sourdough procedure can activate endogenous phytases and lower the pH, thus limiting the chelation of minerals by phytic acid(Reference Leenhardt, Levrat-Verny and Chanliaud220). Second, it is now known that phytic acid can chelate Fe, thus limiting the damage due to the Fenton reaction leading to the production of the very reactive free radical OH. Third, the phytate in whole grain is accompanied by other bioactive compounds that are lost during refining. Phytic acid is therefore a serious candidate as a whole-grain cereal antioxidant acting in vivo. Unfortunately, I know of no studies that have explored the antioxidant effect of this compound from whole-grain cereals in vivo.

The concentration of lignins in whole-grain wheat is 1·9 %: 5·6 % in wheat bran and 1·5 % in germ (Table 1). Lignins are absent from refined flour and are generally considered to be nutritionally inert. However, some studies have demonstrated its potential positive physiological effects. Studies on rats showed that lignin may account for 26–32 % of the enterolactone (a mammalian lignan) formed from cereal bran(Reference Begum, Nicolle and Mila221). Mammalian lignans are antioxidants in vitro at the concentrations (10–100 μm) achievable in vivo (Reference Kitts, Yuan and Wijewickreme222), particularly in the colon(Reference Bach Knudsen, Serena and Kjaer223). A study on rats fed a diet containing 8 % lignin for 21 d showed that lignins can have antioxidant effects on ex vivo fresh lymphocytes by significantly decreasing the peroxide-induced DNA strand breaks and visible light-induced oxidative DNA lesions under the form of oxidised bases via singlet oxygen – 1O2 – production(Reference Labaj, Wsolova and Lazarova224). But I know of no studies on human subjects that have examined the physiological effects of lignins. However, if lignins are partially metabolised to mammalian lignans in humans, as they are in rats, they might add to the protection by lignans observed in human subjects against some cancers(Reference Adlercreutz96). Again, studies are needed to explore the antioxidant effect of whole-grain cereal lignins in vivo.

Whole-grain cereals as a source of bioactive compounds with underestimated physiological effects

Whole-grain cereals as a source of lipotropes and methyl donors: betaine, choline, folates, methionine and myo-inositol

Betaine and choline are now recognised as important in human nutrition: betaine improves the health of the heart, liver and kidneys, while choline is important for lipid metabolism, brain development, the integrity and signalling function of cell membranes, and as a precursor of phosphatidylcholine, acetylcholine and betaine (Table 3)(Reference Craig225, Reference Zeisel and Blusztajn226). The nutritional role of folates (vitamin B9) is also well recognised, particularly in the prevention of neural tube defects and CVD (Table 3). What is more surprising is that their contribution to the health benefits of whole-grain cereals, particularly wheat bran and wheat germ, has not been recognised until very recently (Fig. 2)(Reference Fardet, Rock and Rémésy136, Reference Likes, Madl and Zeisel227). Whole-grain wheat, wheat bran and wheat germ, respectively, contain about 0·28, 1·04 and 1·09 % betaine and choline and about 51, 231 and 420 μg folates/100 g (Tables 1 and 2). However, whole-grain cereals are not very good sources of folates as compared with legumes or vegetables, notably when based on a 100 kcal (420 kJ) content(Reference Cho, Johnson and Song228). The bioavailability of choline and betaine from whole-grain cereal products and fractions is not known. However, its presence as a free soluble osmolyte(Reference Craig225) in cells of the aleurone layer suggests that betaine is readily available, especially compared with fibre-bound antioxidant polyphenols. To my knowledge, only two studies, using the metabonomic approach, have underlined the importance of betaine from whole-grain cereals by showing an increased hepatic, urinary and plasma betaine levels in rats and pigs fed whole-grain wheat flour and high-fibre rye bread(Reference Bertram, Bach Knudsen and Serena229, Reference Fardet, Canlet and Gottardi230). This suggests that betaine from whole-grain cereals is quite available. It has also been recently shown that free betaine can reverse insulin resistance and liver injury in mice fed a high-fat diet, an animal model of non-alcoholic fatty liver disease(Reference Borgschulte, Kathirvel and Herrera231). Thus, the probably high bioavailability of betaine from cereals(Reference Bertram, Bach Knudsen and Serena229, Reference Fardet, Canlet and Gottardi230) combined with its many described health effects(Reference Craig225) suggest that whole-grain cereal betaine may have multivariate health benefits.

Betaine, choline and folates are all methyl donors, able per se to transform homocysteine into methionine, thereby decreasing hyperhomocysteinaemia(Reference Brouwer, van Dusseldorp and Thomas232), a known risk factor for CVD(Reference Graham, Daly and Refsum233), and also for neural tube defects(Reference Mills, McPartlin and Kirke234) and cancers(Reference Wu and Wu235). The dietary intake of whole-grain and bran, but not germ, is significantly and negatively associated with the plasma homocysteine concentration: − 17·4 and − 10·9 % when comparing the highest and lowest quintiles of whole-grain and bran cereal intake, respectively(Reference Jensen, Koh-Banerjee and Franz17). The wide variety of micronutrients may interact in synergy in this effect(Reference Jensen, Koh-Banerjee and Franz17). More precisely, one may hypothesise that folates, betaine and choline would be primarily involved. Besides, since hyperhomocysteinaemia is associated with increased oxidative stress(Reference Loscalzo236, Reference Tyagi, Sedoris and Steed237), betaine and choline may act as indirect antioxidants.

Betaine, choline and folates are also lipotropic compounds, together with methionine and myo-inositol, that are essential for lipid metabolism, DNA methylation and the production of nucleoproteins and membranes(Reference Craig225, Reference Zeisel and Blusztajn226, Reference Christman, Chen and Sheikhnejad238Reference Zeisel, Da Costa and Franklin240). By definition, a lipotrope is a substance that specifically prevents excess fat deposition in the liver by hastening fat removal or by limiting lipid synthesis. However, using this definition sensu strictu, very few studies on human subjects have been published; most have been performed on animals. It is estimated that whole-grain wheat, wheat bran and wheat germ can supply 0·51, 1·31 and 1·59 g lipotropes/100 g, respectively (Table 2). These values could be higher if other compounds with indirect lipotrope-like effects are included (those that indirectly prevent fat accumulation) such as Mg, niacin, pantothenic acid, RS, some flavonoids, PUFA, phytic acid, lignans, some oligosaccharides and fibre. Among lipotropes, as for choline, myo-inositol (a carbocyclic polyol) is derived from several myo-inositol-derived compounds that are essentially free myo-inositol and conjugated myo-inositol, either with glycosylated (for example, galactinol and di-galactosyl myo-inositol) or phosphorylated (for example, phytate or hexakisphosphate) groups. However, the lipotropic effect of phytate has not yet been demonstrated in human subjects and is probably low since human phytases are much less active than those in the rat small intestine(Reference Iqbal, Lewis and Cooper241). In addition, among the nine isomers of inositol, only myo-inositol has been shown to be lipotropic, not chiro-inositol(Reference Okazaki, Setoguchi and Katayama242), which is abundant in the pseudo-cereal buckwheat(Reference Horbowicz and Obendorf243, Reference Steadman, Burgoon and Schuster244) and is mainly known for its action against insulin resistance and its ability to help controlling blood glucose(Reference Kim, Kim and Joo245). Except for myo-inositol phosphate (from hexakisphosphate to monophosphate) contents, there are few data on the free myo-inositol content of whole-grain cereals and their bran and germ fractions before processing. To my knowledge, the only published values are 86·7 mg/100 g for whole-grain amaranth(Reference Becker, Wheeler and Lorenz246), 8·5 mg/100 g for oats(Reference Darbre and Norris247), 30·8–35·4 mg/100 g for whole-grain quinoa(Reference Koziol248) and 52·5 mg/100 g for dry mature wheat embryo(Reference Horbowicz and Obendorf249), which is quite similar to the germ fraction. The same authors also reported that dry mature wheat embryo contained about 56 mg galactinol/100 g(Reference Horbowicz and Obendorf249). Myo-inositol is therefore mainly present in phytate in cereal grains, about 95 % in wheat(Reference Matheson and Strother250). I have used this percentage and the phytic acid content of whole-grain wheat to estimate the free myo-inositol contents of whole-grain wheat, wheat bran and wheat germ (Table 2). The total myo-inositol content of 487 foods was published in 1980, forty-seven of which were processed cereal-based products (twenty-four types of bread, fifteen breakfast cereals and eight kinds of pasta). The total myo-inositol/100 g was 25–1150 mg for wheat breads and 7–35 mg/100 g for wheat-derived breakfast cereals(Reference Clements and Darnell251). Considering all cereal foods, the values given were then within the range 6–1150 mg/100 g for breads and 2–274 mg/100 g for other cereal foods (pasta and breakfast cereals)(Reference Clements and Darnell251). But these values are for total myo-inositol after acid hydrolysis for 40 h at 120°C, which releases myo-inositol from phytate in addition to free myo-inositol(Reference Clements and Darnell251). Nevertheless, hydrolysis of phytic acid within lower inositol phosphate esters (from inositol pentaphosphate to inositol monophosphate and free myo-inositol) by activated endogenous food phytases, through, for example, sourdough baking with natural leaven(Reference Leenhardt, Levrat-Verny and Chanliaud220) and/or simple fermentation with yeast(Reference Reddy, Sathe and Salunkhe252) and/or germination(Reference Darbre and Norris247, Reference Reddy, Sathe and Salunkhe252, Reference Ferrel253), may lead to free myo-inositol formation(Reference Darbre and Norris247, Reference Nakano, Joh and Narita254), as was shown by using different hydrothermal processes with lactic acid and whole barley kernels(Reference Bergman, Fredlund and Reinikainen255). Free myo-inositol may then become available for absorption depending on the quantity not degraded by microflora, either during pre-fermentation or in the colon. Thus, the total free myo-inositol content of wheat products is difficult to ascertain precisely and probably depends on the processing parameters (which would explain the high value ranges found for breads). But it is not insignificant. Once ingested, except for folates whose bioavailability would be low when originating from cereal products, other cereal lipotropic compounds are quite readily available in the digestive tract (Table 2), myo-inositol being likely to be further partly converted into chiro-inositol after absorption, as shown in rats(Reference Pak, Huang and Lilley256).

Wheat bran and germ are rich in choline, which is important in lipid metabolism and DNA methylation. Choline, as choline bitartrate, is often used as a lipotrope in animal diets(Reference Reeves, Nielsen and Fahey257), and rats fed a choline-free diet for 14 months develop severe hepatic lesions, hepatic DNA undermethylation and cellular carcinomas(Reference Locker, Reddy and Lombardi258), DNA undermethylation being related to carcinogenesis development(Reference Gama-Sosa, Slagel and Trewyn259), as demonstrated for benign and malignant human colon neoplasms(Reference Goelz, Vogelstein and Hamilton260). The extent to which lipotropes from whole-grain wheat such as choline help improve lipid status, by preventing fat deposition in the liver, and in balancing DNA methylation in the liver and colon deserve to be explored in prolonged trials with a whole-grain cereal-based diet. In addition to the well-known anti-carcinogenic property of several whole-grain cereal compounds (Table 4), that of choline(Reference Goelz, Vogelstein and Hamilton260) and betaine(Reference Cho, Willett and Colditz121) should be studied more thoroughly, more particularly at the colorectal level.

The specific actions of bound and free ferulic acid

The physiological action of ferulic acid from whole grain has undoubtedly been underestimated because it is poorly absorbed by the small intestine ( < 5 %; Table 2), and because most studies have been conducted with the free compound at high and often unrealistic nutritional levels. These studies have nevertheless underlined the potential role of ferulic acid as an antioxidant, anti-microbial, anti-apoptotic, anti-ageing, anti-inflammatory, neuroprotective, hypotensive, pulmonary-protective and cholesterol-lowering agent in metabolic diseases such as thrombosis, atherosclerosis, cancer and diabetes (Tables 3 and 4)(Reference Barone, Calabrese and Mancuso104, Reference Ou and Kwok261, Reference Srinivasan, Sudheer and Menon262). However, there have been few studies on the capacity of ferulic acid from cereal products to improve some physiological functions in human subjects(Reference Barone, Calabrese and Mancuso104). Ferulic acid may reach up to 0·2 % of whole-grain wheat and over 0·6 % of wheat bran (Table 2), which is quite significant; and 80 % of ferulic acid is in the bran fraction(Reference Rybka, Sitarski and Raczynskabojanowska263). Since no more than 5 % of ferulic acid is absorbed by the intestine(Reference Mateo Anson, van den Berg and Havenaar153), about 95 % reaches the colon bound to fibre where it may act as a natural antioxidant on epithelial cells(Reference Vitaglione, Napolitano and Fogliano150). Thus, both free and metabolised ferulic acid (mainly sulfated and glucuronated) may have a signalling function within cells, and the bound compound might be a strong protective antioxidant and anti-inflammatory agent within the colon. The bacterial esterases in the colon will also partially and relatively slowly solubilise bound ferulic acid, as shown in vitro in a human model colon(Reference Kroon, Faulds and Ryden264). The possible absorption of ferulic acid within the colon and the physiological effects of its metabolites produced by the colon microbiota remain therefore to be quantified and qualified.

The specific actions of lignins

I have discussed the potential role of lignin as an antioxidant. However, lignin is one of the main non-energy-producing compounds in whole grain (about 1·9 % of whole-grain wheat, 5·6 % of wheat bran and 1·5 % of wheat germ) (Table 1). Although generally considered to be nutritionally inert, such a high concentration should have physiological effects, such as protecting the gut epithelium against oxidative damage and protecting other cell wall compounds against fermentation, so increasing faecal bulk and the associated positive health effects (dilution of carcinogens). Some studies support the hypothesis that lignins are not nutritionally inert. For example, bioactive lignophenol derivatives from bamboo lignin are anti-carcinogenic in human neuroblastoma SH-SY5Y cells, where they suppress oxidative stress-induced apoptosis(Reference Akao, Seki and Nakagawa265). It has also been shown that cell walls containing lignins (hydrophobic polymers) favour the adsorption of hydrophobic carcinogens and their release in the faeces(Reference Ferguson and Harris266). Lignins from wheat bran also adsorb bile salts (i.e. bile salt-sequestering agent) such as deoxycholate in vitro, but a link between cholesterol lowering and wheat bran consumption was not demonstrated(Reference Calvert and Yeates267). Lignin may reduce bile salt reabsorption in vivo by adsorbing them(Reference Chang and Johnson268), and may further reduce the formation of carcinogenic metabolites from bile salts by colon bacteria(Reference Drasar and Jenkins269). The lignin nordihydroguairetic acid is also able to prevent changes in renal morphology, by reducing oxidative stress, in rats with diabetic nephropathy for which reactive oxygen species play an important role in its development as a result of chronic hyperglycaemia(Reference Anjaneyulu and Chopra270). Finally, lignins from fractionated hardwood hydrolysate, when consumed during 3 weeks from an 8 % lignin-based diet, are able to decrease H2O2- and visible light-induced DNA damage in ex vivo fresh rat blood lymphocytes(Reference Labaj, Wsolova and Lazarova224) and in testicular cells(Reference Labaj, Slamenova and Lazarova109). This suggests that lignin compounds or some of their metabolites have crossed the epithelial barrier, or at least have been able to induce antioxidant defences in blood by unknown mechanisms. More recently, studies using a liquid chromatography–MS-based metabonomic approach showed that lignins appear not to be metabolised by rats for 2 d, but that they probably had some effects on endogenous metabolism(Reference Fardet, Llorach and Orsoni271). To summarise, lignins might act in many ways: they are metabolised to enterolactone in rats(Reference Begum, Nicolle and Mila221), their antioxidant capacity may protect the gut epithelium, they may act on endogenous metabolism, they may reduce DNA damage in blood or cells via their antioxidant capacity and they may adsorb carcinogens. All these potential physiological effects should be taken into consideration in further in vivo studies, especially towards cancer prevention. Lignins are therefore far from being inert and researchers in nutrition and cereal technology should ask more questions about the nutritional effects of lignins.

The combined effects of B vitamins

Whole-grain wheat, and especially its bran and germ fractions, contains almost all the B-group vitamins, vitamins B1 (thiamin), B2 (riboflavin), B3 (niacin), B5 (panthothenic acid), B6 (pyridoxine), B8 (biotin) and B9 (folates). Whole wheat contains about 9·1 mg B vitamins/100 g, bran about 30·3 mg and germ about 12·3 mg (Table 1). Whole-grain cereals are particularly significant sources of thiamin, niacin, pantothenic acid and biotin compared with other food sources, and wheat germ is rich in nicotinic acid, pantothenic acid and pyridoxine. Cereal products are not a particularly rich source of folates unless fortified with folic acid (the synthetic form of folate), as it is often the case, especially for breakfast cereals. One key issue is the bioavailability of these vitamins in whole-grain cereals, but data are scarce: the few studies on the subject show that the bioavailability of each B vitamin seems to vary greatly, and that it is far from 100 % (Table 2). Thiamin and pyridoxine are the most bioavailable (Table 2). The specific action of each of these vitamins is described in Table 3. Their actions are complex and multi-factorial. The B vitamins are also called the ‘B-complex vitamins’ and they play an important role in maintaining muscle tone in the gastrointestinal tract and promoting the health of the nervous system, skin, hair and liver. Thiamin, nicotinic acid, pyridoxine, pantothenic acid and folates play a positive role in mental health (Tables 3 and 4). For example, folates and pyridoxine are coenzymes in the one-carbon metabolism pathways and are involved in the synthesis of serotonin and other neurotransmitters, deficits of which are implicated in deficient mental health(Reference Hindmarch272). Folates also reduce the risk of neural tube defects in babies when consumed during the periconceptional period(Reference Berry, Li and Erickson273). It was recently suggested that they could be used to treat depression(Reference Coppen and Bolander-Gouaille274, Reference Miller275), as a low folate status is associated with depression(Reference Gilbody, Lightfoot and Sheldon276). Although difficult to demonstrate, it would be particularly interesting to explore the effect of whole-grain cereals on the nervous system and mental health, particularly disorders such as depression, insomnia, cognitive impairment or more generally psychic equilibrium. Other bioactive compounds, such as choline, ferulic acid, Mg, Zn, Cu, inositols, policosanol and melatonin, are also potential candidates for mental health protection and equilibrium (Tables 3 and 4).

The effects of whole-grain cereals on bone, teeth, articulation and tendon health

Whole-grain cereals and their fractions might contribute to the good health of bones, cartilages, teeth, collagen, joints and tendons (Table 3), which are all constituents of the skeleton, by the combined actions of α-linolenic acid, Fe, Zn, Mg, Mn, Cu, P, Ca, K, nicotinic acid, tocotrienols, phylloquinone (vitamin K) and β-cryptoxanthin (Table 4). While P and Ca are components of hydroxyapatite, a major constituent of bones and teeth, the Ca:P ratio in cereals, notably wheat (about 0·08; Table 2), is below the ratio of 0·5–0·8 recommended for a satisfactory Ca use by the body. Ca from whole-grain cereals is therefore unlikely to contribute significantly to the health of bones and teeth. However, the addition of calcium carbonate (CaCO3) to cereal food recipes before processing might be a simple way to achieve the desirable Ca:P ratio without altering product palatability(Reference Lioger, Leenhardt and Demigne277). Whole-grain wheat also contains Ca absorption enhancers such as fructans and/or RS, which increase the apparent absorption of Ca from 20 to 50 % in rats(Reference Lopez, Levrat-Verny and Coudray71Reference Lopez, Coudray and Levrat-Verny73). Similarly, inulin increases Ca absorption by about 12 % in human subjects(Reference Lopez, Levrat-Verny and Coudray71Reference Lopez, Coudray and Levrat-Verny73). However, although whole-grain wheat does not contain inulin, it may contain up to 2·3 g fructans/100 g (Table 2) that might also increase Ca absorption upon fermentation. The effect of indigestible oligosaccharides such as fructans on Ca absorption and metabolism, and bone health (as measured by indices such as bone mineral content and density, and/or bone resorption rate/osteopenia) is more and more recognised today, both in rats and humans(Reference Ohta, Ohtsuki and Hosono278Reference Scholz-Ahrens and Schrezenmeir280).

The results for P are less conclusive; some studies have shown increased P in bone following fructo-oligosaccharide consumption in rats(Reference Ohta, Ohtsuki and Hosono278, Reference Scholz-Ahrens and Schrezenmeir280), while others have found no effect(Reference Scholz-Ahrens and Schrezenmeir280). P is mainly supplied by phytic acid (>85 % of the total P in grain), which has a high affinity for hydroxyapatite(Reference Nordbö and Rolla281). Indeed, the incidence of dental caries has been hypothesised to be concomitant with the change towards dietary habits of Western societies, as was shown with African Bantu acquiring susceptibility to dental decay as they adopted the European diet, through increased consumption of cariogenic refined foods such as refined sugar and white wheat bread in which a dominant caries-preventing factor would be removed during the refining process(Reference McClure282Reference Osborn, Noriskin and Staz284). P, which is abundant in less refined wheat flour, is involved in this effect(Reference Osborn285). Thereafter, several studies on rats using organic and inorganic phosphates and different Ca:P ratios also showed the cariostatic effect of phytic acid(Reference McClure282, Reference McClure286Reference Wynn, Haldi and Bentley289), possibly through its ability to affect organic materials and the adsorption of bacteria to tooth surfaces(Reference Nordbö and Rolla281), and also through its ability to be rapidly adsorbed onto hydroxyapatite, forming a natural barrier resistant to acid attacks(Reference Magrill290) and thus to protect teeth from demineralisation and the formation of cavities by causing the desorption of salivary proteins from hydroxyapatite, the first step in plaque formation(Reference Nordbö and Rolla281, Reference Pruitt, Jamieson and Caldwell291). But, later, Cole & Bowen failed to show a significant effect of feeding monkeys with phytic acid for 2 weeks on the physical properties of plaques (such as dry and wet weights), or their chemical properties (protein, carbohydrate, Ca, Mg and P contents), or the microbial composition(Reference Cole and Bowen292). Further studies in human subjects are therefore needed to ascertain the cariostatic role of phytic acid, and perhaps of other cereal bioactive compounds, in subjects on a regular whole-grain cereal diet.

Whole-grain wheat also contains mammalian lignans (0·2–0·6 mg/100 g; Table 2) that seem to protect against osteoporosis (Table 3), notably in the postmenopausal period. Japanese women consuming high concentrations of phyto-oestrogens were found to have fewer hip fractures than women in the USA or Europe(Reference Adlercreutz and Mazur293). However, the effect of lignans on bone health remains to be confirmed. To my knowledge, no research has answered this particular issue of the role of long-term whole-grain cereal consumption on skeletal health and bone physiology.

Whole-grain cereals as a source of oligosaccharides

It has previously been seen that whole-grain cereals are rich in fibre (including RS) and oligosaccharides that may have both a prebiotic effect by favouring the development of a healthy microbiota(Reference Chanvrier, Appelqvist and Bird294, Reference Swennen, Courtin and Delcour295) and that enhance mineral absorption through hypertrophy of the gut epithelium(Reference Lopez, Coudray and Bellanger70, Reference Lopez, Levrat-Verny and Coudray71). Thus, whole-grain wheat contains 1·9 %, its bran has 3·7 % and the germ fraction 10·1 % of fructans (fructo-oligosaccharide), raffinose and stachyose (Table 1). The average wheat germ raffinose content is about 8 % and may reach 10·9 %, which is quite high (Table 2). Whole-grain wheat contains about 0·4 % of raffinose and wheat bran has 1·2 % (Table 2). The stachyose content is lower: 0·1 % in whole-grain wheat, 0·2 % in wheat bran and no data are available for wheat germ (Table 2). Raffinose is a trisaccharide composed of galactose, glucose and fructose. Stachyose is a tetrasaccharide formed with two galactose molecules, one glucose and one fructose. To my knowledge, there are no published data on the health effects of these whole-grain cereal oligosaccharides, apart from the fact that they are both considered to reinforce the fibre effect of whole-grain cereals, by producing SCFA generally favourable to large-bowel health. They are completely fermented in vitro within 48 h in the presence of a piglet faecal inoculum(Reference Krause, Easter and Mackie296). Rats fed a 3 % raffinose-based diet for 21 d have a significantly reduced weight gain, more lactobacilli and fewer streptococci, greater SCFA production, and, interestingly, a lower plasma TAG concentration with no effect on plasma cholesterol(Reference Tortuero, Fernández and Rupérez297). However, it must be noted that fermented products (notably breads) constitute an important part of the whole-grain cereal food consumption of humans; and fermentation may lead to the partial breakdown of fructans, raffinose and stachyose by bacteria.

The specific action of phytosterol and of little studied bioactive whole-grain cereal compounds: α-linolenic acid, policosanol, melatonin and para-aminobenzoic acid

The concentration of α-linolenic acid, an n-3 fatty acid (18 : 3) with many positive health effects (Table 3), may reach 0·5 % of wheat germ and almost 0·2 % of wheat bran (Table 1). A diet containing about 2·7 g α-linolenic acid-rich wheat germ oil per d has an anti-atherosclerotic effect in mildly hypercholesterolaemic subjects; it acts by inhibiting oxidative stress-mediated synthesis of CD40L (protein involved in the progression of atherosclerosis with inflammatory and prothrombotic properties)(Reference Alessandri, Pignatelli and Loffredo298). Wheat germ contains 0·53 % α-linolenic acid, so one should consume about 500 g/d to reach the 2·7 g tested in the present study, which is not really realistic. However, a regular consumption of wheat germ as a nutritional complement and/or of wheat germ oil is nutritionally relevant.

Phytosterols, policosanol and melatonin, although present at lower concentrations, also possess numerous positive health effects (Table 3). Phytosterols, known for their cholesterol-lowering effect in humans(Reference Farquhar, Smith and Dempsey299, Reference Jones, Ntanios and Raeini-Sarjaz300), are particularly high in wheat germ (430 mg/100 g) (Table 2) but their health effects are not known when they come from whole-grain cereals. Policosanol is a natural mixture of high-molecular-weight aliphatic primary alcohols (C24 to C34) in which octacosanol is the main compound(Reference Kato, Karino and Hasegawa301, Reference Taylor, Rapport and Lockwood302). Although less nutritionally studied, policosanol is also a lipid-lowering agent (for example, total and LDL-cholesterol) in both human subjects and animals at levels of about 10–20 mg daily, and it can also increase HDL-cholesterol up to+30 %(Reference Gouni-Berthold and Berthold303, Reference Varady, Wang and Jones304), making it a promising agent in CVD prevention and treatment(Reference Varady, Wang and Jones304). Whole-grain wheat contains about 3 mg policosanol/100 g (Table 2). One recent study has shown that eating chocolate pellets supplemented with wheat germ policosanol (20 mg/d) for 4 weeks does not reduce blood cholesterol or modify the blood lipid profile of healthy human subjects(Reference Lin, Rudrum and van der Wielen305). A diet containing about 100 mg policosanol/d eaten for 30 d reduced the increase in plasma LDL-cholesterol in hypercholesterolaemic rabbits by reducing cholesterol synthesis in the liver through increased LDL catabolism(Reference Menendez, Arruzazabala and Mas306). Feeding policosanol to rats for up to 4 weeks (250 and 500 mg/kg per d) significantly renders the lipoprotein fractions (VLVL+LDL) resistant to ex vivo Cu-mediated oxidation(Reference Menendez, Fraga and Amor307). In view of these results, the policosanol content of whole-grain wheat seems too low (about 3 mg/100 g) to significantly improve the blood lipid profile in humans. Rather, it is probably the combined action of the different cholesterol-lowering compounds of wheat (for example, SCFA produced by undigestible carbohydrates, soluble fibre, tocotrienols, phytosterols and policosanol) that contributes to improve the blood lipid profile to its optimum.

The concentration of the mammalian pineal hormone melatonin, which can be extracted from numerous plants, is about 0·3 μg/100 g in whole-grain wheat (Table 2)(Reference Hosseinian, Li and Beta308). This compound has a positive effect on human mood, cognitive functions, prolonged sleep period and brain neuromodulation(Reference Asayama, Yamadera and Ito309, Reference Maurizi310), but it may also be an antioxidant(Reference Maurizi310) and anti-carcinogen(Reference Garcia-Navarro, Gonzalez-Puga and Escames311, Reference Shiu312) (Table 3). The health effects of melatonin in humans when originating from whole-grain cereals are not known: as for policosanol and other cholesterol-lowering compounds, due to the low melatonin content of whole-grain wheat (Table 4), this is probably the combined action of melatonin and of other compounds acting positively on mental and brain health that has to be considered first.

Para-aminobenzoic acid has also been detected in cereals. Values are scarce and not recent: reported values are 0·34–0·55, 1·34 and 0·852 mg/100 g for whole-grain wheat, bran and germ fractions, respectively(Reference Calhoun, Bechtel and Bradley313, Reference Calhoun, Hepburn and Bradley314). Para-aminobenzoic acid is best known as a sunscreen agent that protects the skin from UV radiation(Reference Wang, Huang and Tai315), but it also stimulates bacterial growth in the intestine and is an intermediate in the bacterial synthesis of folates. Besides its role in folate formation, para-aminobenzoic acid has long been used to treat rickettsial infections and may lead to a 11·5 % decrease in serum cholesterol in man, when consumed at 8 mg/d in the form of its Na salt(Reference Barbieri, Papadogiannakis and Eneroth316, Reference Failey and Childress317). Para-aminobenzoic acid down-regulates N-acetyltransferase in human cell cultures (peripheral blood mononuclear cells)(Reference Butcher, Ilett and Minchin318) – acetylation plays an important role in the activation of several potential human carcinogens(Reference Hein, Doll and Gray319, Reference Minchin, Reeves and Teitel320), and inhibits the production of thromboxane which participates in blood coagulation (anti-aggregatory effect) and in increased arterial pressure through vasoconstriction(Reference Barbieri, Papadogiannakis and Eneroth321). However, these studies used para-aminobenzoic acid concentrations of 30–100 μm, about 4–137 mg/l, which is far higher than the quantity that can be obtained from eating whole-grain cereal products, as whole-grain wheat containing only 0·34–0·55 mg para-aminobenzoic acid/100 g (Table 2). Thus, like the other bioactive compounds present at low concentrations in whole-grain wheat (for example, policosanol and melatonin), the health benefit of cereal para-aminobenzoic acid has to be considered complementary to that of other cholesterol-lowering, anti-carcinogenic and anti-aggregatory compounds.

The nutrigenomic approach

Nutrigenomics in nutrition is devoted to the study of the influence of dietary interventions on gene transcription (transcriptome), protein synthesis (proteome) and metabolites (metabolome, the whole set of metabolites) in cells, body fluids and tissues(Reference Elliott, Pico and Dommels322Reference Zeisel326). One of the most important objectives of nutrigenomics is to detect and identify early metabolic disturbances and their regulation (for example, in relation to oxidative stress or inflammation) that can lead to more serious chronic diseases. The possibility of detecting some diseases early could change clinical nutrition and public health practices(Reference Zeisel326). This implies studying the effects of bioactive compounds in whole-grain cereals on gene expression, protein synthesis and the metabolome. In the field of nutritional studies, besides the measurement of usual biomarkers such as plasma glucose (for example, GI) or urinary lipid peroxides (oxidative stress index), it seems particularly important to focus on the metabolome, which reflects both the endproducts of metabolism and the changes over time of metabolism following food consumption. While many metabolomic studies have been done with isolated compounds, notably in pharmacology for drug toxicity(Reference Keun327), very few have been done with complex food products. In metabolomics and nutrition, only a few studies have been performed(Reference Rezzi, Ramadan and Fay328): to characterise the metabolic effect of energy restriction(Reference Selman, Kerrison and Cooray329), vitamin deficiency(Reference Griffin, Muller and Woograsingh330) or of intake of PUFA-rich oils(Reference Mutch, Grigorov and Berger331), antioxidant-rich foods such as soya(Reference Solanky, Bailey and Beckwith-Hall332), chamomile(Reference Wang, Tang and Nicholson333) and tea(Reference Van Dorsten, Daykin and Mulder334), or of pure dietary antioxidants such as epicatechin(Reference Solanky, Bailey and Holmes335), catechin(Reference Fardet, Llorach and Martin336) or ferulic and sinapic acids and lignins(Reference Fardet, Llorach and Orsoni271). Studies on rats have been carried out using the metabolomic approach to explore the metabolic fate and the effect on endogenous metabolism of whole-grain and refined wheat flours(Reference Fardet, Canlet and Gottardi230) and of lignin-enriched wheat bran lignins(Reference Fardet, Llorach and Orsoni271). It has thus been shown that whole-grain wheat flour consumption leads to significant increases in liver betaine and GSH and decreases in some liver lipids, but has no effect on conventional lipid and oxidative stress biomarkers. It also causes a greater urinary excretion of tricarboxylic acid cycle intermediates, aromatic amino acids and hippurate (from phenolic acid degradation in the colon). When the diet was changed to refined wheat flour, a new metabolic balance was reached within 48 h, and conversely from refined to whole-grain flour (Fig. 3)(Reference Fardet, Canlet and Gottardi230). The metabolomic approach also showed that rats did not appear to metabolise lignins from wheat bran within 2 d of the regimen, but they are likely to affect endogenous metabolism through mechanisms which need to be elucidated(Reference Fardet, Llorach and Orsoni271). Results are convincing in that new metabolic effects have been unravelled using this new open approach, for example, the role of symbiotic microbiota in triggering diet-induced mechanisms of steatosis(Reference Dumas, Barton and Toye337) or some specific metabolic pathway disturbances in diabetic rats(Reference Zhang, Nagana Gowda and Asiago338), thus improving our understanding of diseases and the mechanisms responsible for them. However, more significant conclusions could be drawn once the databases for compound identification are completed and distributed. To my knowledge, few if any studies have investigated the effect of consuming complex whole-grain cereals and their fractions on gene expression. The tools are now available to study this, which would provide important information about which gene-regulated metabolic pathways are stimulated by the synergetic action of the bioactive compounds in whole-grain cereals, not the restricted action of isolated compounds. Thus, nutrigenomics should enable us to better characterise the metabolic pathways affected in vivo by the antioxidants in whole-grain cereals.

Fig. 3 Linear discriminant (LD) analysis score plot of the 1H NMR urinary spectra highlighting the separation before, between and after the diet change (days 14–15) and between the urine sampling times (postprandial (PP) and post-absorptive (PA)). (- - - -), Refined flour followed by whole-grain flour consumption (RF-WGF) group; (—), whole-grain flour followed by refined flour consumption (WGF-RF) group. Each polygon represents the limits of the metabolic profile obtained for the ten rats of a given group at a given day and urine sampling time. Urine samples were collected from days 13 to 28 (for details, see Fardet et al. (Reference Fardet, Canlet and Gottardi230)).

Conclusion

The metabolic fate and health effects of major compounds such as lignin (up to 9 % in wheat bran), ferulic acid (up to 0·6 % in wheat bran), phytic acid (up to 6 % in wheat bran) and betaine (up to 1·5 % in wheat bran) (Table 2) have been little studied when originating from whole-grain cereals. Yet, these three compounds may account for about 11 % of wheat bran (Table 1), and therefore deserve to be studied more. Wheat germ also merits greater attention since it contains quite significant levels of bioactive compounds such as α-linolenic acid (about 530 mg/100 g), GSH (about 133 mg/100 g), GSSG (about 69 mg/100 g), thiamin (about 1·75 mg/100 g), vitamin E (about 27·1 mg total tocols/100 g), flavonoids (about 300 mg/100 g), betaine (about 851 mg/100 g), choline (about 223 mg/100 g), myo-inositol (>11 mg/100 g) and phytosterols (about 430 mg/100 g) (Table 2). It thus contains 2·5 % of vitamins and minerals, at least 1·6 % of lipotropic compounds and 1·2 % of sulfur compounds. All these compounds are involved in the new hypotheses proposed here and their corresponding physiological mechanisms. Based on past and new hypotheses, a synthetic view of the mechanisms underlying the health benefits of whole-grain cereals and their fractions can be proposed (Fig. 4). The diagram purposefully illustrates the complexity of the mechanisms involved and their obvious synergy and interconnection in vivo. Due to this complexity, whole-grain cereal bioactive compounds are listed in Table 4, ranking according to the five major health outcomes generally considered in the literature: body-weight regulation, CVD, diabetes, cancers, and gut health; mental, brain and skeleton health being new proposed ways to explore. One important question remains: do bioactive compounds exert the same effects when they are free compounds and when they are in whole-grain cereals? This is notable, because their bioavailability in whole-grain cereals is probably lower than the free compounds (Table 2) and because the quantities in whole-grain cereal products do not match the daily human needs. Again, it is probably the summed and combined action of all the bioactive compounds on a particular physiological function (as illustrated in Fig. 4 and Table 4) which leads to improved specific physiological functions such as antioxidant status and glucose homeostasis, especially when whole-grain products are consumed daily, generating long-term health benefits. This is why it is urgent to carry out further in vivo studies both in rats and human subjects, to unravel the complex mechanisms activated by the consumption of highly complex foods such as whole-grain cereal products. Intervention studies on human subjects consuming whole-grain cereals are so rare that they should be carried out first. The non-invasive characteristic and high potential of the metabolomic approach for unravelling new metabolites and metabolic pathways affected by a given diet and its ability to explore the complexity inherent in metabolism means that it should accompany the measurement of the usual biomarkers in order to describe the metabolic actions of whole-grain cereals in all their complexity. The mechanisms described in Fig. 4 are complex, but are above all interconnected as in the whole organism. Metabolomics therefore seems to be the most appropriate tool for studying such an interconnectedness, and so provide a more realistic view of how whole-grain cereal bioactive compounds act in synergy. For example, inflammation, oxidative stress and immune system-related metabolic pathways are generally all involved in cancers, as is the case for other metabolic diseases in which there is a progressive metabolic imbalance following an unhealthy diet. Finally, genomic studies are needed on the action of whole-grain cereals on gene regulation, as bioactive compounds really exert their physiological effects within the cell. While isolated free bioactive compounds may be used for in vitro studies on cell cultures, studies in animals and human subjects should use an integrated ‘complex food approach’.

Fig. 4 Current and new proposed physiological mechanisms involved in protection by whole-grain cereals (adapted from Table 3). The dotted thin arrows () indicate the link between whole-grain bioactive compounds and protective physiological mechanisms, while the plain arrows () indicate the relationship between physiological mechanisms and health outcomes.

Cereals other than wheat

The present review discusses whole-grain wheat, since it is one of the most widely consumed cereals, especially in Western Europe. However, most of the bioactive compounds in wheat are also present in other major cereals such as rice, maize, oats, barley, sorghum and millet. The main differences lie in the relative contents of each of these compounds, their distribution in bran, germ and endosperm and the proportions of the bran and germ fractions. Nevertheless, compounds such as γ-oryzanol, avenanthramides and saponins are specific to cereals other than wheat.

The bran fraction

The proportion of the bran fraction varies with the cereal type: for wheat, rice and maize, it is 10–16 % of the whole grain. The bran fraction in rice contains about 15–20 % oil(Reference Souci, Fachmann and Kraut215, Reference Britz, Prasad and Moreau339). This oil is rich in bioactive compounds and contains more than 100 different antioxidants, such as lipoic acid, a powerful antioxidant(Reference Packer, Witt and Tritschler340, Reference Roy, Sen and Tritschler341) that helps prevent cognitive deficits, is beneficial in the treatment of Alzheimer's disease(Reference Maczurek, Hager and Kenklies342), and may protect against risk factors of CVD(Reference Wollin and Jones343). Rice bran contains tocotrienols (10·6 mg/100 g)(Reference Yu, Nehus and Badger344), γ-oryzanol (281 mg/100 g)(Reference Yu, Nehus and Badger344) and up to 1·2 % phytosterols(Reference Emmons, Peterson and Paul345) such as β-sitosterol, all of which may help improve the blood lipid profile and reduce the risk of CVD(Reference Heinemann, Kullak-Ublick and Pietruck346Reference Wilson, Nicolosi and Woolfrey348). Rice bran also contains up to 21 % dietary fibres(Reference Emmons, Peterson and Paul345). Maize bran has more dietary fibre than wheat and rice bran, about 74–79 %(Reference Souci, Fachmann and Kraut215, Reference Kahlon and Chow349, Reference Saulnier, Vigouroux and Thibault350). It contains about 4 % phenolic acids, about 50 % heteroxylans and about 20 % cellulose, and is almost devoid of lignins(Reference Saulnier, Vigouroux and Thibault350). It is particularly rich in ferulic acid (up to 3 %), mainly in a very resistant (to enzymes) bound form(Reference Saulnier, Marot and Elgorriaga351). And, contrary to wheat for which phytate is essentially in the bran fraction, 90 % of maize phytate is in the germ fraction(Reference O'Dell, De Boland and Koityonann352).

Some specific compounds

Some bioactive compounds are quite specific to certain cereals: γ-oryzanol in rice, avenanthramides and saponins in oats, and, although present in other cereals such as wheat, β(1 → 3)(1 → 4)-glucans in oats and barley, and alkylresorcinols in rye. Their mechanisms of action and health effects are shown in Table 3.

γ-Oryzanol in rice

γ-Oryzanol is derived from rice bran oil and is a mixture of substances including sterols and ferulic acid, and at least ten phytosteryl ferulates (for example, methylsterols esterified to ferulic acid). Its content in whole-grain rice is 18–63 mg/100 g (DW)(Reference Britz, Prasad and Moreau339, Reference Miller and Engel353) and in rice bran 185–421 mg/100 g, depending on the rice variety, milling time, stabilisation process and extraction methods(Reference Yu, Nehus and Badger344, Reference Chen and Bergman354Reference Shin, Godber and Martin356). Its antioxidant activity has been demonstrated in vitro (Reference Juliano, Cossu and Alamanni357). Its health effects are diversified, with positive actions against CVD and hyperlipidaemia, as shown in animal models through cholesterol-lowering, lipid peroxidation reduction and anti-atherogenic effects(Reference Wilson, Nicolosi and Woolfrey348, Reference Rong, Ausman and Nicolosi358Reference Suh, Yoo and Chang360) and in human subjects(Reference Cicero and Gaddi361).

Avenanthramides and saponins in oats

Aventhramides are specific polyphenols from oats. They are substituted cinnamic acid amides of anthranilic acids and there are at least twenty-five distinct entities(Reference Collins362). Total avenanthramide content in five oat cultivars (husked and naked) ranges from 4·2 to 9·1 mg/100 g(Reference Shewry, Piironen and Lampi363), while the oat grain contains 4–13 mg avenanthramide 1/100 g (the major avenanthramide), again depending on the oat cultivar(Reference Dimberg, Theander and Lingnert364). The avenanthramide content in oat bran is 1·3–12·5 mg/100 g according to the type of avenanthramide considered(Reference Dimberg, Theander and Lingnert364, Reference Mattila, Pihlava and Hellstrom365). As polyphenols, they are strong antioxidants both in vitro (Reference Fagerlund, Sunnerheim and Dimberg366, Reference Peterson, Hahn and Emmons367) and in vivo (Reference Chen, Milbury and Collins140). They play a particular role in the prevention of CVD due to their anti-inflammatory and anti-atherogenic effects(Reference Liu, Zubik and Collins368), and by protecting LDL from oxidation, in synergy with vitamin C, as shown on human LDL(Reference Chen, Milbury and Kwak369).

Saponins are glycosides with a steroid or triterpenoid aglycone(Reference Güçlü-Üstündag and Mazza370). They are especially found in oats, which synthesise two families of saponins, the steroidal avenacosides and the triterpenoid avenacins(Reference Osbourn371). The saponin content, depending on the oat cultivar, seems to be situated mainly within the endosperm and has been shown to vary from 0·02 to 0·13 % (DW)(Reference Önning, Asp and Sivik372, Reference Price, Johnson and Fenwick373). Saponins have a wide range of biological activities (about fifty are listed by Güçlü-Üstündag & Mazza(Reference Güçlü-Üstündag and Mazza370)), such as anti-carcinogenic and hypocholesterolaemic(Reference Matsuura374), stimulation of the immune system(Reference Barr, Sjölander and Cox375, Reference Sjölander, Cox and Barr376) and cholesterol-lowering(Reference Oakenfull, Fenwick and Hood377). However, it is not known whether all these properties could be ascribed to cereal saponins. Saponins are also poorly absorbed by the gut(Reference Calvert and Yeates267).

β(1 → 3)(1 → 4)-Glucan in barley and oats

The β(1 →  3)(1 →  4)-glucan content of oats and barley is especially high. Total, insoluble and soluble barley β-glucan contents vary widely with the variety, the presence of hull (i.e. hulled v. hull-less) and the amylose content(Reference Baik and Ullrich378). Thus, the water-soluble β-glucan content of barley is 0·5–8·3 % (w/w, DW)(Reference Baik and Ullrich378Reference Izydorczyk, Storsley and Labossiere385), the insoluble fraction is 1·2–21·7 % (w/w, DW)(Reference Åman and Graham379Reference Gajdosová, Petruláková and Havrlentová381) and the total β-glucan content is 3·0–27·17 % (w/w, DW)(Reference Åman and Graham379Reference Gajdosová, Petruláková and Havrlentová381, Reference Izydorczyk and Dexter383). Total β-glucans contents vary widely and might be attributable, in addition to variety variability, to the method of extraction and possible confusion in some studies where the soluble β-glucan fraction seems to be confounded with the total β-glucans.

The soluble β-glucans content of naked oat grains is 3·9–7·5 %, and in hulled oat grains it is 2·0–7·5 % (w/w, DW); the insoluble content of naked oat grains is 5·2–10·8 % and that of hulled oat grains is 13·8–33·7 % (w/w, DW)(Reference Gajdosová, Petruláková and Havrlentová381, Reference Prentice, Babler and Faber386). Much work has already been done on the health effects of β-glucans, particularly their glycaemia- and cholesterol-lowering properties, having implications for type 2 diabetes(Reference Kim, Stote and Behall387) and CVD(Reference Wood56, Reference Butt, Tahir-Nadeem and Khan388, Reference Kalra and Joad389). As soluble viscous fibre(Reference Izydorczyk and Dexter383), they slow the rate of gastric emptying, and the diffusion of glucose and NEFA into epithelial cells for absorption in both animals and humans(Reference Wood56, Reference Kalra and Joad389). However, a recent study conducted on healthy subjects demonstrated that muesli enriched with oat β-glucans had no more effect on gastric emptying rate than did cornflake-based muesli, despite its plasma glucose-lowering effect(Reference Hlebowicz, Darwiche and Bjorgell390). β-Glucans are also positively involved in the protection against cancers, especially through reactions with mutagenic agents to prevent them interacting with DNA as shown in rodent and human cell lines(Reference Mantovani, Bellini and Angeli391).

Alkylresorcinols in rye

Alkylresorcinols are plant-derived phenolic lipids, especially found in whole-grain cereals. Rye contains the highest concentration of alkylresorcinols, which can be twice that of wheat (up to 320 mg/100 g DW)(Reference Ross, Shepherd and Schupphaus392). They are 1,3-dihydroxybenzene derivatives with an alkyl chain at position 5 of the benzene ring, which gives them an amphiphilic feature. They are apparently relatively well absorbed within the small intestine (about 58 %; Table 2) of ileostomates following the consumption of soft bread enriched with rye bran and whole-grain rye crispbread(Reference Ross, Kamal-Eldin and Lundin393), making them (either intact in plasma or as metabolites in urine) potential biomarkers of whole-grain rye and wheat intake(Reference Landberg, Aman and Friberg394Reference Ross, Kamal-Eldin and Aman396), especially for epidemiological research and observational studies(Reference Ross, Kamal-Eldin and Aman396, Reference Guyman, Adlercreutz and Koskela397). Their biological activity is multifactorial(Reference Ross, Kamal-Eldin and Aman396), from interacting with metabolic enzymes (for example, inhibiting 3-phosphoglycerate dehydrogenase, the key enzyme in TAG synthesis in adipocytes)(Reference Tsuge, Mizokami and Imai398) to decreasing cholesterol in the rat liver(Reference Ross, Chen and Frank399), to anticancer/cytotoxic effects but almost exclusively in vitro (Reference Kozubek and Tyman400, Reference Ross and Kasum401).

New bases for improving the nutritional properties of cereal products

The elucidation of the mechanisms by which whole-grain cereals protect our bodies, together with a better understanding of how bioactive compounds are released from the cereal food matrix and delivered to the bloodstream, will provide important information for the industrial development of cereal products with improved nutritional qualities. Surprisingly, the present supply of cereal products of a good nutritional quality is still limited. I believe that the best way to improve the nutritional quality of cereal products is to combine the preservation of a relatively intact botanical food structure (as far as the recipe allows it), a low-GI feature and a high nutritional density of fibre and bioactive compounds, by using less refined flour with a higher extraction rate. These factors are important but probably not sufficient to ensure that the right macro- or micronutrient reaches the right site of absorption for an optimal physiological effect. This is why more and more private and public research is aimed at modelling the fate of nutrients from complex foods within the intestine so as to predict their bioaccessibility and thus control their delivery for a specific physiological effect(Reference Norton, Moore and Fryer402Reference Tedeschi, Clement and Rouvet404).

Optimising and controlling the delivery of bioactive compounds for improving health

There are great differences between the food content in a defined nutrient and the percentage really metabolised, or even absorbed. This is especially true for cereal products where numerous factors linked to the food matrix may limit the release of macro- and micronutrients. There is increasing evidence that the physical structure of natural cereal food matrices (for example, intact cereal kernels) or the artificial microstructure of processed cereal products may either favour or limit the bioavailability of nutrients, and thus their nutritional effects. However, differences in bioaccessibility–bioavailability of nutrients, particularly micronutrients, at present cannot be correlated with differences in long-term health effects, except for the positive health effects of starch and its so-called slowly digestible fraction(Reference Englyst, Kingman and Cummings405, Reference Lehmann and Robin406). The question is therefore: is there a positive correlation between increased or decreased bioaccessibility of a given nutrient and its health effect? This probably depends on the nutrient considered and on the health status of the subject. For example, the rapid release of glucose from starch digestion into the bloodstream is advantageous in some situations (for example, the urgent need for glucose for brain or muscles to function, as for immediate intellectual and physical efforts), and harmful in other situations (for example, type 2 diabetes). The same approach is now being developed for proteins (slow v. rapid proteins) and lipids for which their physical state and/or their physico-chemical properties may influence the release of amino acids and fatty acids, respectively, into the bloodstream. The resulting significant metabolic impact could be used in some situations such as diabetic subjects(Reference Marangoni, Idziak and Rush407), the elderly(Reference Remond, Machebeuf and Yven408) and for patients on enteral nutrition suffering from pancreatic insufficiency to adequately hydrolyse lipids(Reference Armand, Pasquier and Andre409).

In vitro bioaccessibility and in vivo bioavailability studies with vegetables and whole-grain cereals and/or their fractions have clearly shown that food structure affects the bioavailability of polyphenols, carotenoids, minerals, trace elements and vitamins (Table 2)(Reference Parada and Aguilera403). Table 2 shows the results of bioavailability studies on whole-grain wheat products and wheat bran. Much data are still lacking: studies exploring the bioavailability of compounds in whole-grain cereals are scarce and the products are often consumed as part of a complex diet that also supplies the same bioactive compounds from other foods. For example, studies on mineral or trace element bioavailability in rats often included mineral mixtures that made it difficult to determine the exact apparent absorption of the mineral supplied by the cereals. Thus, radiolabelled cereal products should be used more frequently to answer such questions. The few data obtained show that bioactive compounds are far from being 100 % bioavailable within the small intestine. No more than 5 % of the ferulic acid in wheat bran is released into the small intestine, so that most reaches the colon where it can exert an antioxidant protective action on the gut epithelium. On the other hand, there is convincing evidence that the small proportion absorbed in the small intestine can affect cell signalling and the activation or repression of some genes. Thus, in a way similarly to starch, it seems that two fractions of ferulic acid can be defined: the rapidly available ferulic acid released and absorbed in the small intestine (i.e. free and soluble-conjugated), and slowly available ferulic acid gradually released mainly in the colon (i.e. ester-linked)(Reference Kroon, Faulds and Ryden264), each fraction having its own health benefits.

Betaine (about 0·9 % of wheat bran; Table 1), unlike ferulic acid, is probably much more bioavailable since it is not bound to other constituents: is there a need to slow down its release and to favour a fraction reaching the colon, for example, for improving its anticancer effect(Reference Giovannucci, Rimm and Ascherio410)? The same issue, that is the optimal bioavailability to reach, might be questioned for polyphenols such as lignans and alkylresorcinols, vitamins and minerals, and phytosterols. The problem for phytic acid is slightly different; we need to know the extent to which it is reasonable to pre-hydrolyse it in order to combine a maximum mineral bioavailability with its antioxidant effect in the gut against free radicals produced by microbiota, and from its potential hypoglycaemic effect as well.

Otherwise, the case of fibre is not yet resolved for whole-grain wheat which contains more insoluble fibre than soluble fibre (soluble:total fibre ratio is about 0·16; calculated from Table 2): what would be the optimum ratio of soluble:total fibre to reach? It is not known to what extent it would be beneficial to increase the soluble fibre content, for example, by pre-hydrolysing insoluble arabinoxylans to soluble arabinoxylans (soluble:total arabinoxylans ratio is about 0·18; calculated from Table 2). Soluble fibres may be beneficial to health by reducing the postprandial glucose response through increased viscosity(Reference Lu, Walker and Muir411) (see Tables 3 and 4), but they may also be harmful, by, for example, increasing the risk of colon cancer(Reference Moore, Park and Tsuda412).

Provided it has positive health benefits, the range by which industrial processes can improve the bioaccessibility and bioavailability of cereal bioactive compounds is therefore large. This approach has been applied to starch with success(Reference Björck and Asp413), by controlling its delivery in the gut by rendering it more slowly hydrolysed (i.e. slowly digestible starch) within the small intestine, or by making it inaccessible to α-amylase (i.e. RS), so that a fraction of starch reaches the colon where it is fermented to the anti-carcinogenic molecule butyrate, the preferred fuel for colonocytes (see Whole-grain cereals and butyrate production section). Technologists know how to modulate the proportions of these three fractions in cereal products, i.e. rapidly, slowly and indigestible starch. RS is representative of the different ways it can be used by breeders and technologists to control the delivering of a compound, i.e. starch, within the digestive tract. It has been seen that the RS content of whole-grain products may be very high, up to 12 % in ordinary barley kernels and even 22 % by combining intact botanical structure with a high-amylose barley variety(Reference Nilsson, Ostman and Holst54). The formation of RS can be technologically favoured through starch encapsulation within the cereal food matrix by protein or fibre networks (RS1), restricting starch granule gelatinisation (RS2), the use of high-amylose cereal varieties with a high content of retrograded starch (RS3) and/or chemical modification such as acylation (RS4). RS is now considered to be a prebiotic compound that can positively modify microbiota growth in quality and quantity within the colon(Reference Dongowski, Jacobasch and Schmiedl414, Reference Topping, Fukushima and Bird415). If technologists may be able to modify processing parameters such as temperature, extrusion pressure, retrogradation and/or chemical modification to increase the RS content, breeders can select high-amylose cereal varieties(Reference Chanvrier, Appelqvist and Bird294, Reference Rahman, Bird and Regina416), amylose being more slowly digested than amylopectin(Reference Goddard, Young and Marcus417, Reference Hallfrisch and Behall418).

The traditional use of fermentation and the development of new technologies

Fig. 5 shows the ways in which the nutritional quality of whole-grain cereals can be improved. There are mainly three: the growing conditions, the genetic approach and through technological processes.

Fig. 5 Ways for improving cereal product nutritional quality. RS, resistant starch.

Growing conditions

The growing conditions, for example, the use of adequate fertilisers, can increase the cereal content of Se, Mg, Fe and Zn(Reference Hawkesford and Zhao419Reference Soliman421) with possible modified physiological effects in humans(Reference Fallahi, Mohtadinia and Ali Mahboob422). An increase in environmental stress, for example, water stress, cold or exposure to micro-organisms, may favour the synthesis of antioxidants by the plant to combat this stress. This has been shown with α-tocopherols, carotenoids and betaine in wheat seedlings and sugarbeet roots under temperature- and salt-stressed environments(Reference Hanson and Wyse423, Reference Keles and Öncel424).

Genetic approach

The genetic approach(Reference King425) using conventional tools (indirect action on genes) such as cross-breeding and hybridisation to combine varieties high in some bioactive compounds, for example, Zn, Fe and pro-vitamin A(Reference Cakmak, Ozkan and Braun426, Reference Ortiz-Monasterio, Palacios-Rojas and Meng427), and/or low in others, for example, phytic acid(Reference Mendoza, Viteri and Lonnerdal428, Reference Raboy429), and non-conventional tools (direct action on genes) such as genetic engineering to modify gene expression in relation to the nutrient synthesis and/or metabolism can be used to improve the nutritional quality of whole-grain cereals. By these means, the amylose(Reference Chanvrier, Appelqvist and Bird294, Reference King, Noakes and Bird430, Reference Regina, Bird and Topping431), RS(Reference Rahman, Bird and Regina416), arabinoxylan(Reference Saulnier, Sado and Branlard432) and mineral/vitamin(Reference Hawkesford and Zhao419, Reference Brinch-Pedersen, Borg and Tauris433) contents can be modified (i.e. increased in most cases).

Development of new technologies

Besides growing conditions and genetics, the third way of improving the nutritional quality of cereal products is through technological processes. The literature about them is plethoric, but it is not an objective of the present paper to review them. However, some key issues may be emphasised since they allow optimising the health benefits of cereal by preserving their nutritional density and food structure.

Increasing nutritional density in bioactive compounds through germination, soaking and pre-fermentation of whole-grain cereals and/or their fractions

Cereals are usually processed in two main ways. The first is dry fractionation followed by cooking under different conditions of water content, temperature and pressure, as for pasta, biscuits, breakfast cereals and other cereal products widely consumed in Western countries. The second is fermentation. This is generally used for whole-grain cereals in more traditional procedures used for the many whole-grain foods consumed in developed countries and several alcoholic beverages (for example, beer, sake, whisky, etc) consumed around the world(Reference Hammes, Brandt and Francis434, Reference Nout435). A fermentative step stimulates enzyme activities, which generally increases the content of free bioactive compounds. Bread products combine both approaches by using dry milling, fermentation and cooking.

Due to the plasma cholesterol- and glucose-lowering properties of soluble fibre and to its low content in wheat, due to the numerous health effects of free ferulic acid(Reference Ou and Kwok261, Reference Srinivasan, Sudheer and Menon262), and due to the relative negative effect of phytic acid upon mineral bioavailability(Reference Lopez, Leenhardt and Coudray217), different ways to pre-hydrolyse insoluble fibre (for example, insoluble β-glucans or arabinoxylans) into soluble fibre with endohydrolases(Reference Vitaglione, Napolitano and Fogliano150, Reference Napolitano, Lanzuise and Ruocco436), ester-bound ferulic acid into free ferulic acid with feruloyl-esterases(Reference Faulds and Williamson437, Reference Wang, Geng and Egashira438), and phytic acid with exogenous or endogenous phytases (i.e. through adding degrading fungal and microbial enzymes, genetic engineering to over-express phytase activity and food processes to activate endogenous phytases(Reference Lopez, Leenhardt and Coudray217)) have been considered with the objective of increasing the bioactive potential of whole-grain cereal foods, and in the end their nutritional value.

Practically, this could be also partly achieved by using traditional and natural processes such as germination, soaking and/or fermentation in a highly hydrated medium. The fermentation of whole-grain cereals such as wheat, maize, rice, sorghum and millet, either germinated or not, often in combination with leguminous seeds (for example, soyabean and chickpea), is widespread in developed countries and the Orient for whole-grain cereal-based beverages, gruels and porridges (for example, koko, doro, ogi, akasa, tuo zaafi and togwa in Africa; idli in India; shoyu in the Orient; chicha in South America; or kishk in Arabian countries). It increases the nutritional density of the products, protects against diarrhoea, is easy to apply, allows a good preservation of the products (useful, for example, for long displacements), may improve sensory quality and is inexpensive(Reference Chavan and Kadam439Reference Lioger, Leenhardt and Rémésy441). Before fermentation, whole-grain cereals are generally soaked, germinated, dried and coarsely ground with a grinding stone(Reference Gadaga, Mutukumira and Narvhus440). Fermentation, by activating enzymes, can release bound bioactive compounds, synthetise new bioactive compounds, degrade anti-nutrients and increase protein and starch digestibility(Reference Chavan and Kadam439). This is accompanied by numerous potential positive health effects as recently reviewed, for example, improved gut health or reduction of the rate of starch degradation(Reference Poutanen, Flander and Katina442). Thus, germination and fermentation have been used for whole-grain wheat, rye, maize, sorghum and millet in order to decrease the tannin and phytic acid contents, as both compounds impair mineral bioavailability – leading to Fe-deficiency anaemia in developing countries – and also in order to increase the protein/gluten and starch digestibility and the concentration of free amino acids by enhanced proteolytic and α-amylolytic activities(Reference Hassan and El Tinay177, Reference Matuschek, Towo and Svanberg178, Reference Towo, Matuschek and Svanberg180, Reference Abd Elmoneim, Schiffler and Bernhard443Reference Wedad, El-Tinay and Mustafa449). Sourdough pre-fermentation (incubation for 24 h at 30°C with lactic acid bacteria) for whole-wheat flour degrades about 60–70 % of the phytic acid in bread dough (compared with the initial flour content) in 4 h, so increasing Mg bioaccessibility in vitro (Reference Leenhardt, Levrat-Verny and Chanliaud220, Reference Lopez, Krespine and Guy450) and in vivo in rats(Reference Lopez, Duclos and Coudray451). In another study, the type of starter for sourdough fermentation and the type of raw material (native v. malted or germinated rye) was shown to influence the content in bioactive compounds of the resulting wholemeal rye flour. The combination of germination and fermentation increased the levels of folates (7-fold), free phenolic acids (10-fold), total phenolic compounds (4-fold), lignans (3-fold) and alkylresorcinols, but, to a lesser extent ( < 1·5-fold) the metabolic activities of microbes together with the breakdown and hydrolysis of some cereal cell walls were involved in this effect(Reference Katina, Liukkonen and Kaukovirta-Norja452). Conversely, a 4 h sourdough fermentation of whole-wheat flour leads to losses of alkylresorcinol(Reference Winata and Lorenz453). The fermentation of rye bran also enhances the free ferulic acid and the solubilisation of pentosans through xylanase activation(Reference Katina, Laitila and Juvonen454). Recently, an increased level of free ferulic acid (about a 2-fold increase) has been reported within whole-wheat dough pizza upon 18 and 48 h of fermentation(Reference Moore, Luther and Cheng455), as well as an increase in pentosan solubilisation and prolamin hydrolysis in germinated rye sourdough(Reference Loponen, Kanerva and Zhang446). This could have practical nutritional implications as discussed earlier with free ferulic acid, and also since the soluble fraction of arabinoxylans has been shown to reduce the glycaemic response in either healthy subjects(Reference Lu, Walker and Muir411) or in those with impaired glucose tolerance(Reference Garcia, Otto and Reich456). On the other hand, prolamin proteins are known to trigger coeliac disease (autoimmune disorder due to gluten intolerance) and their intensive pre-hydrolysis during germination and fermentation might render cereal products from these technologies coeliac-safe(Reference Loponen, Kanerva and Zhang446). Lastly, fermentation of whole-grain cereals has been reported in several studies to increase the content of available methionine and B vitamins, such as thiamin, riboflavin, niacin, folates and pantothenic acid, through the action of micro-organisms(Reference Chavan and Kadam439). Despite all these convincing results, the health benefits of hydrolysis and/or the release of free bioactive compounds from whole-grain cereal products through germination and/or fermentation have not been sufficiently explored in human subjects. The addition of a pre-fermentation step before processing other cereal products, such as those usually widely consumed in our Western societies (for example, breakfast cereals or crackers), should also be studied more. A recent study showed that adding a pre-fermentation step while omitting steam cooking before wheat flake processing preserved a satisfactorily nutritional quality by improving the management of the feeling of hunger in the morning and by moderately improving insulin economy, which could be of interest for type 2 diabetic subjects(Reference Lioger, Fardet and Foassert457).

Whole-grain and wholemeal breads are generally made of flours with an extraction rate of 85–90 % (type 80 flours). Baking these flours does not sufficiently degrade phytic acid or hydrate the fibre fraction. These flours also do not generally contain the germ fraction, leading to a loss of B vitamins. One alternative would be to add 20 to 30 % whole-grain flour (with an extraction rate of 100 %) to white wheat flour(Reference Lioger, Leenhardt and Rémésy441). The whole-grain flour could be pre-fermented in a strongly hydrated medium with leaven, and then reincorporated into white flour for baking to avoid hydration competing with gluten and fibre. This adds the germ fraction together with a significant increase in bioactive compounds while partially degrading phytic acid(Reference Lioger, Leenhardt and Rémésy441). Sourdough whole-grain barley and wheat breads also reduce the glycaemic response in healthy subjects through delayed gastric emptying and possibly through a higher content of RS, thus prolonging satiety with potential benefits in weight control(Reference Liljeberg and Bjorck458, Reference Liljeberg, Lonner and Bjorck459).

Reinforcing the food structure cohesiveness in processed cereal products

As preserving intact the botanical structure in whole-grain cereal products and favouring compactness of processed cereal products such as pasta reduces the glycaemic and insulinaemic responses and increases satiety, both of which are useful in the management of type 2 diabetes and weight regulation, processed cereal products with greater cohesiveness need to be identified. This can be achieved artificially by creating protein and/or fibrous networks in the food matrix to hinder enzyme accessibility to its substrate within the small intestine(Reference Brennan, Blake and Ellis460), by using intact cereal kernels with a natural fibrous network(Reference Liljeberg, Granfeldt and Bjorck51, Reference Nilsson, Ostman and Holst54), and/or by altering kneading intensities and proving time during baking to obtain breads with a more dense crumb texture(Reference Burton and Lightowler461). Some have also tried, with relative success, to increase the thickness of breakfast cereal flakes to reduce their glycaemic and insulinaemic indices in healthy subjects(Reference Granfeldt, Eliasson and Bjorck462). The more frequent use of more or less intact whole-grain cereal kernels in food recipes seems the most promising, easiest and cheapest way to explore by technologists.

Isolating the aleurone layer from the wheat bran fraction

Since most of the bioactive compounds are in the aleurone layer of the bran(Reference Antoine, Lullien-Pellerin and Abecassis463) and since the pericarp (especially the outer fraction composed of cellulose, penstosans and lignins is poorly digestible) may contain contaminants (pesticides, mycotoxins and heavy metals), antinutrient compounds, irritants for the digestive epithelium (for example, lignins and insoluble fibre) and may limit the bioavailability of bioactive compounds, different processes for isolating the aleurone layer from wheat bran have been investigated(Reference Buri, von Reding and Gavin464Reference Hemery, Rouau and Lullien-Pellerin466), with the objective of reincorporating it in cereal food recipes. This appears to be a new way of enhancing the nutrition value of cereal products(Reference Buri, von Reding and Gavin464, Reference Hemery, Rouau and Lullien-Pellerin466). The aleurone layer represents approximately 6–9 % of the whole-grain wheat (Fig. 1). Some researchers have studied the nutritional quality of aleurone flour, and shown that the aleurone layer is a rich source of bioavailable folate in humans(Reference Fenech, Noakes and Clifton467), that it lowers plasma homocysteine(Reference Fenech, Noakes and Clifton468), increases SCFA production(Reference Cheng, Trimble and Illman469), reduces colon adenoma in azoxymethane-treated rats(Reference McIntosh, Royle and Pointing470), and that it is more digestible (+17 %) and fermentable (+30 %) than wheat bran, so yielding more butyrate(Reference Amrein, Granicher and Arrigoni471). It also has a higher antioxidant activity than wheat bran (1·5-fold) and whole-grain wheat (2-fold) in vitro (Reference Miller, Rigelhof and Marquart132, Reference Buri, von Reding and Gavin464). However, isolating the aleurone layer from the bran fraction means losing the health benefits of lignins (mainly in the outer pericarp and testa layers of the bran fraction), which seem to be significant and remain largely unknown (see above). The long-term benefit of consuming bran and aleurone fractions on several physiological parameters and major health problems is therefore an important issue that should be explored in order to assess the real nutritional value of lignins and decide whether the few negative physiological effects generally associated with lignins are outweighed by their positive effects. The issue is close to that of phytic acid, which also has both negative and positive physiological effects. However, the issue of preserving the lignin would be the most meaningful in the case of organic whole-grain cereals which should not contain pesticides in their outer pericarp.

Conclusions

The nutritional quality of cereal products may therefore be improved by agricultural conditions, genetics and technological processes. Organic agriculture, genetics, the use of a pre-fermentation step and of a more or less intact grain structure are probably the most promising ways to preserve and enhance the nutritional density of whole-grain foods. Sourdough pre-fermentation could also be used for other whole-grain cereal foods such as breakfast cereals. The first parameter described in Fig. 5 is the milling process, and the best way to preserve a high nutritional density in bioactive compounds is to use flours with high extraction rates. It must be remembered that whole-grain wheat, wheat bran and wheat germ contain, respectively, at least 15, 52 and at least 24 % bioactive compounds and dietary fibre (Table 1). Removing the bran fraction during milling and using it to feed animals is therefore an issue to consider more seriously.

General conclusions

The importance of preserving bran and germ fractions

The bioactive compounds in whole-grain cereals are unevenly distributed (Fig. 1). Some (mainly soluble fibre, Se, some B vitamins, carotenoids and flavonoids) are present in significant quantities in the endosperm, but most are in the bran (especially the aleurone layer) and germ fractions. This fact alone shows the importance of preserving these fractions in cereal products, at least in the most currently consumed forms of breads and breakfast cereals, and to a lesser extent pasta, crackers and biscuits. Some products consumed on special occasions (i.e. generally not at breakfast, lunch or dinner), such as cakes, pastries and viennoiseries, use very refined flours (extraction rate of 70–82 %), and it is probably not meaningful to use less refined flours. To preserve the bran and germ fractions means either reincorporating fractions later in the recipe or using the whole-grain cereal so as to maintain its botanical structure relatively intact during processing. However, reincorporation of the bran and germ fractions implies destroying the botanical structure with the loss of its health benefits (for example, increased satiety or RS content), unless technological processes can yield a cereal product with an artificial compact food structure as for pasta(Reference Fardet, Hoebler and Baldwin472) or breads with decreased loaf volume(Reference Burton and Lightowler461).

The concept of the ‘whole-grain package’

The content of individual bioactive compounds in whole grain often seems too low for them to have any significant or lasting physiological effects. It is becoming more and more evident that the synergetic action of several bioactive compounds contributes to health protection and/or the maintenance of one physiological function, not just one compound. Fig. 1 and Table 4 illustrate this concept of the ‘whole-grain package’: thus, obesity/body-weight regulation, CVD, type 2 diabetes, cancers, gut, mental/nervous system and skeleton health may be potentially protected by at least, respectively, ten, thirty-four, seventeen, thirty-two, ten, twenty-six and sixteen different bioactive compounds and/or groups of compounds (i.e. oligosaccharides, tocols, phenolic acids, flavonoids, saponins, inositols, γ-oryzanol, lignans and alkylresorcinols). Because of their many protective bioactive compounds (at least twenty-six), whole-grain cereals are particularly suitable for protecting the body from CVD, cancers and mental/nervous system disorders. The long-term protection against mental or nervous system disorders by consuming whole-grain cereal products therefore deserves to be studied in human subjects, notably because depression ranks among the major causes of mortality and disability with an overall prevalence of 5–8 %(Reference Coppen and Bolander-Gouaille274). It is also remarkable that at least thirty compounds and/or groups of compounds may participate in antioxidant protection through different mechanisms (Tables 3 and 4), which approximately corresponds to a total of at least 3·9, 13·4 and 6·3 % of the whole-grain wheat, wheat bran and germ fractions (Tables 1 and 2). As most age-related and chronic diseases are associated with increased oxidative stress, the regular consumption of whole-grain cereal products should benefit all of us, but particularly the elderly.

The importance of pesticides and mycotoxins

Since whole-grain cereals include by definition the outer parts of the grain, they may contain pesticides and mycotoxins (for example, zearalenone and deoxynivalenol in wheat or fumonisin in maize). Their presence should not decrease the benefits of bioactive compounds also mainly contained in the outer layers. For example, there may be a relationship between the consumption of fumonisin-contaminated maize in some regions of the world (for example, China and South Africa) and the occurrence of oesophageal cancers(Reference Chu and Li473, Reference Rheeder, Marasas and Thiel474). However, more generally, the consequences of long-term consumption of high quantities of mycotoxin-contaminated cereal grains for human health (i.e. toxicological effects) are not well known. The link between some cancers and exposure to pesticides has been well established, particularly among farmers(Reference Lebailly, Niez and Baldi475). It is therefore particularly relevant that recommendations for the consumption of more whole-grain cereal products should be accompanied by the production of less contaminated cereals, such as those from organic agriculture devoid of pesticides.

Perspectives

It is surprising to note that, although numerous epidemiological surveys have shown a significant and positive association between whole-grain cereal consumption and the prevention of several chronic diseases, fewer studies have been performed on the mechanisms involved. For example, to my knowledge, no more than eleven studies have examined the antioxidant hypothesis by postprandial or intervention studies in human subjects to investigate the antioxidant effect of whole-grain cereals, bran or germ(Reference Fardet, Rock and Rémésy136), with only a recent postprandial study on human subjects consuming wheat bran(Reference Price, Welch and Lee-Manion146). Therefore, there is a real gap between observational studies and the elucidation of the mechanisms involved. The mechanisms are certainly complex, as has been seen. But more data are needed on the mechanisms involved so as to prepare strong, convincing arguments for an increased consumption of whole-grain cereal products by the public, to better inform health professionals about their health benefits, to favour their marketing by the food industry and to develop new health claims in the near future.

Acknowledgements

I thank Dr Christian Rémésy for his constructive criticism of the manuscript and Professor Inger Björck (Department of Applied Nutrition and Food Chemistry, Chemical Centre, Lund University, Sweden) for allowing me to use her original diagram (from the HealthGrain Project, European Community's Sixth Framework Programme, FOOD-CT-2005-514008, 2005–2010) that I have adapted for Fig. 2 of the paper (see original diagram in the brochure ‘Progress in HEALTHGRAIN 2008’ at http://www.healthgrain.org/pub/). The English text of the manuscript has been checked by Dr Owen Parkes.

There are no conflicts of interest and the present review received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

Colour versions of Figs. 1, 2 and 4 can be seen in the online version of the paper.

Appendix 1

References cited for evaluating the range (minimum and maximum values) of bioactive compound contents in whole-grain wheat, and wheat bran and germ fractions (data for Tables 1 and 2)*

* All wheat varieties are included, i.e. durum, soft, hard, spring, winter and pigmented wheats; all data are expressed for 100 g of food. When data are expressed on a DM basis within a reference with no indication of the water content, results are converted on a fresh matter basis considering a mean water content of 13 % for whole-grain wheat, 10 % for wheat bran and 11·4 % for wheat germ (means calculated from US Department of Agriculture database for cereal grains and pasta(479)).

Whole-grain wheat

Reduced glutathione: 1·04–5·74 mg/100 g(Reference Sarwin, Walther and Laskawy210, Reference Archer480)

Oxidised glutathione: 0·86–2·88 mg/100 g(Reference Archer480)

Sulfur amino acids:

Methionine: 0·17–0·24 g/100 g(479, Reference Shewry481Reference Waggle, Lambert and Miller483)

Cystine: 0·19–0·40 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Sugars:

Monosaccharides: 0·26–1·30 g/100 g(Reference Colonna, Buléon, Leloup, Jarrige, Ruckebusch and Demarquilly484, Reference Knudsen485)

Sucrose: 0·60–1·39 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Colonna, Buléon, Leloup, Jarrige, Ruckebusch and Demarquilly484, Reference Knudsen485)

Total fibre (lignin, oligosaccharides, resistant starch and phytic acid included): 9·0–17·3 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Gebruers, Dornez and Boros486Reference Ward, Poutanen and Gebruers492)

Insoluble fibre (lignin included): 9·5–11·4 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Hernot, Boileau and Bauer488, Reference Picolli da Silva and de Lourdes Santorio Ciocca490, Reference Abdel-Aal and Hucl493)

Soluble fibre: 1·1–3·2 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Hernot, Boileau and Bauer488, Reference Picolli da Silva and de Lourdes Santorio Ciocca490, Reference Ragaee, Campbell and Scoles491, Reference Abdel-Aal and Hucl493)

Cellulose: 2·1–2·8 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Anderson and Bridges494)

Hemicellulose: 8·6 g/100 g(Reference Knudsen485)

Lignins: 0·9–2·8 g/100 g(Reference Knudsen485Reference Haskå, Nyman and Andersson487)

Fructans: 0·6–2·3 g/100 g(Reference Knudsen485, Reference Haskå, Nyman and Andersson487, Reference Fretzdorff and Welge495Reference Huynh, Wallwork and Stangoulis497)

Raffinose: 0·13–0·59 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Colonna, Buléon, Leloup, Jarrige, Ruckebusch and Demarquilly484, Reference Knudsen485, Reference Fretzdorff and Welge495, Reference Huynh, Palmer and Mather496)

Stachyose: 0·05–0·17 g/100 g(Reference Colonna, Buléon, Leloup, Jarrige, Ruckebusch and Demarquilly484, Reference Knudsen485)

Total arabinoxylans: 1·2–6·8 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Gebruers, Dornez and Boros486, Reference Haskå, Nyman and Andersson487, Reference Ragaee, Campbell and Scoles491, Reference Henry498, Reference Lempereur, Rouau and Abecassis499)

Water-extractable arabinoxylans: 0·2–1·2 g/100 g(Reference Gebruers, Dornez and Boros486, Reference Ragaee, Campbell and Scoles491)

β-Glucans: 0·2–4·7 g/100 g(Reference Knudsen485, Reference Gebruers, Dornez and Boros486, Reference Ragaee, Campbell and Scoles491, Reference Ward, Poutanen and Gebruers492, Reference Henry498, Reference Genç, Özdemir and Demirbas500)

Phytic acid: 0·28–1·50 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Hemery, Lullien-Pellerin and Rouau501Reference Tariq, Talat and Asia506)

Fe: 1·0–14·2 mg/100 g(Reference Cakmak, Ozkan and Braun426, Reference Ortiz-Monasterio, Palacios-Rojas and Meng427, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference House and Welch502, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507Reference Tang, Zou and He511)

Mg: 17–191 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Tang, Zou and He511, Reference Zook, Greene and Morris512)

Zn: 0·8–8·9 mg/100 g(Reference Cakmak, Ozkan and Braun426, Reference Ortiz-Monasterio, Palacios-Rojas and Meng427, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Lorenz and Loewe509, Reference Tang, Zou and He511Reference Welch and Graham513)

Mn: 0·9–7·8 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Lorenz and Loewe509, Reference Tang, Zou and He511, Reference Zook, Greene and Morris512)

Cu: 0·09–1·21 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Lorenz and Loewe509, Reference Tang, Zou and He511Reference Welch and Graham513)

Se: 0·0003–3·0000 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Davis, Peters and Cain507, Reference Zook, Greene and Morris512, Reference Fan, Zhao and Poulton514, Reference Zhao, McGrath and Gray515)

P: 218–792 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Davis, Peters and Cain507, Reference Tang, Zou and He511)

Ca: 7–70 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Tang, Zou and He511)

Na: 2–16 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

K: 209–635 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference O'Dell, Burpo and Savage504, Reference Davis, Peters and Cain507, Reference Tang, Zou and He511)

Thiamin (vitamin B1): 0·13–0·99 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Batifoulier, Verny and Chanliaud516Reference Ranhotra, Gelroth and Novak519)

Riboflavin (vitamin B2): 0·04–0·31 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Batifoulier, Verny and Chanliaud516Reference Davis, Peters and Letourneau518)

Niacin (vitamin B3): 1·9–11·1 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Davis, Cain and Peters517, Reference Davis, Peters and Letourneau518)

Pantothenic acid (vitamin B5): 0·72–1·99 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Davis, Peters and Letourneau518)

Pyridoxine (vitamin B6): 0·09–0·66 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Batifoulier, Verny and Chanliaud516Reference Davis, Peters and Letourneau518)

Biotin (vitamin B8): 0·002–0·011 mg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482)

Folates (vitamin B9): 0·014–0·087 mg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482, Reference Davis, Peters and Letourneau518, Reference Gujska and Kuncewicz520Reference Piironen, Edelmann and Kariluoto522)

Tocols (vitamin E) = tocopherols+tocotrienols: 2·3–7·1 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Ward, Poutanen and Gebruers492, Reference Lampi, Nurmi and Ollilainen523Reference Panfili, Fratianni and Irano526)

Total tocopherols: 1·06–2·89 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Lampi, Nurmi and Ollilainen523Reference Panfili, Fratianni and Irano526)

α-Tocopherol: 0·34–3·49 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Davis, Peters and Letourneau518, Reference Lampi, Nurmi and Ollilainen523Reference Moore, Hao and Zhou527)

Total tocotrienols: 1·09–4·49 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Lampi, Nurmi and Ollilainen523Reference Panfili, Fratianni and Irano526)

Phylloquinone (vitamin K): 0·002–0·020 mg/100 g(479, Reference Souci, Fachmann and Kraut482)

Total carotenoids: 0·044–0·626 mg/100 g(479, Reference Abdel-Aal and Hucl493, Reference Konopka, Kozirok and Rotkiewicz528Reference Panfili, Fratianni and Irano530)

β-Carotene: 0·005–0·025 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Moore, Hao and Zhou527, Reference Konopka, Kozirok and Rotkiewicz528)

Lutein: 0·026–0·383 mg/100 g(Reference Adom, Sorrells and Liu14, Reference Moore, Hao and Zhou527Reference Roose, Kahl and Ploeger531)

Zeaxanthin: 0·009–0·039 mg/100 g(Reference Adom, Sorrells and Liu14, Reference Moore, Hao and Zhou527, Reference Leenhardt, Lyan and Rock529Reference Roose, Kahl and Ploeger531)

β-Cryptoxanthin: 1·12–13·28 μg/100 g(Reference Adom, Sorrells and Liu14)

Total phenolic acids: 16–102 mg/100 g(Reference Li, Shewry and Ward197, Reference Ward, Poutanen and Gebruers492)

Extractable (free and conjugated) phenolic acids: 5–39 mg/100 g(Reference Li, Shewry and Ward197, Reference Ward, Poutanen and Gebruers492)

Bound phenolic acids: 14–78 mg/100 g(Reference Li, Shewry and Ward197, Reference Ward, Poutanen and Gebruers492)

Total ferulic acid: 16–213 mg/100 g(Reference Li, Shewry and Ward197, Reference Lempereur, Rouau and Abecassis499, Reference Moore, Hao and Zhou527, Reference Adom and Liu532Reference Mpofu, Sapirstein and Beta535)

Free/soluble-conjugated ferulic acid: 0·7–4·9 mg/100 g(Reference Moore, Hao and Zhou527, Reference Adom and Liu532)

Bound ferulic acid: 14–64 mg/100 g(Reference Li, Shewry and Ward197, Reference Moore, Hao and Zhou527, Reference Adom and Liu532)

Total dehydrodiferulic acid: 1·5–76·0 mg/100 g(Reference Li, Shewry and Ward197, Reference Barron, Surget and Rouau533, Reference Lempereur, Surget and Rouau534)

Total dehydrotrimer ferulic acid: 2·6–3·5 mg/100 g(Reference Hemery, Lullien-Pellerin and Rouau501, Reference Barron, Surget and Rouau533)

Total flavonoids: 30–43 mg catechin equivalents/100 g(Reference Adom, Sorrells and Liu14, Reference Adom and Liu532)

Free flavonoids: 2·15–4·86 mg catechin equivalents/100 g(Reference Adom, Sorrells and Liu14, Reference Adom and Liu532)

Bound flavonoids: 28–40 mg catechin equivalents/100 g(Reference Adom, Sorrells and Liu14, Reference Adom and Liu532)

Anthocyanins: 0·45–52·60 mg/100 g(Reference Hosseinian, Li and Beta308, Reference Abdel-Aal and Hucl493, Reference Abdel-Aal and Hucl536, Reference Abdel-Aal, Abou-Arab and Gamel537)

Isoflavonoids:

Daidzein: 2·1 μg/100 g(Reference Liggins, Mulligan and Runswick538)

Genistein: 12·7 μg/100 g(Reference Liggins, Mulligan and Runswick538)

Lignans: 0·199–0·619 mg/100 g(Reference Adlercreutz and Mazur293, Reference Dinelli, Marotti and Bosi539, Reference Milder, Feskens and Arts540)

Alkylresorcinols: 11·6–128·8 mg/100 g(Reference Ross, Shepherd and Schupphaus392, Reference Ross, Kamal-Eldin and Lundin393, Reference Ross, Kamal-Eldin and Aman396, Reference Ross, Chen and Frank399, Reference Ward, Poutanen and Gebruers492, Reference Hemery, Lullien-Pellerin and Rouau501, Reference Andersson, Kamal-Eldin and Fraś541)

Betaine: 22–291 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Waggle, Lambert and Miller483, Reference Patterson and Bhagwat542)

Total choline: 27–195 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Calhoun, Bechtel and Bradley313, Reference Calhoun, Hepburn and Bradley314, Reference Waggle, Lambert and Miller483, Reference Patterson and Bhagwat542)

Phytosterols: 57–98 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Nyström, Paasonen and Lampi489, Reference Ward, Poutanen and Gebruers492, Reference Hakala, Lampi and Ollilainen543Reference Piironen, Toivo and Lampi546)

Total d-chiro-inositol: 17 mg/100 g(Reference Kim, Kim and Joo245)

Policosanol: 0·30–5·62 mg/100 g(Reference Irmak and Dunford547)

Melatonin: 0·2–0·4 μg/100 g(Reference Hosseinian, Li and Beta308)

p-Aminobenzoic acid (PABA): 0·34–0·55 mg/100 g(Reference Calhoun, Bechtel and Bradley313, Reference Calhoun, Hepburn and Bradley314)

Wheat bran

α-Linolenic acid (18 : 3n-3): 0·16 g/100 g(Reference Trautwein548)

Reduced glutathione: about 1·7–19·4 mg/100 g(Reference Every, Morrison and Simmons549)

Oxidised glutathione: about 6·1–21·4 mg/100 g(Reference Every, Morrison and Simmons549)

Sulfur amino acids:

Methionine: 0·20–0·29 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Cystine: 0·32–0·45 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Sugars:

Monosaccharides: 0·14–0·63 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485)

Sucrose: 1·8–3·4 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Fraser and Holmes550, Reference Saunders and Walker551)

Total fibre (lignin, oligosaccharides, resistant starch and phytic acid included): 35·7–52·8 g/100 g(Reference Begum, Nicolle and Mila221, Reference Amrein, Granicher and Arrigoni471, Reference Souci, Fachmann and Kraut482, Reference Haskå, Nyman and Andersson487Reference Nyström, Paasonen and Lampi489, Reference Chen, Haack and Janecky552Reference Maes, Vangeneugden and Delcour554)

Insoluble fibre (lignin included): 32·4–41·6 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Hernot, Boileau and Bauer488, Reference Abdel-Aal and Hucl493, Reference Chen, Haack and Janecky552, Reference Esposito, Arlotti and Bonifati555Reference Morris and Ellis557)

Soluble fibre: 1·3–5·8 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Hernot, Boileau and Bauer488, Reference Abdel-Aal and Hucl493, Reference Chen, Haack and Janecky552, Reference Esposito, Arlotti and Bonifati555)

Cellulose: 6·5–9·9 g/100 g(Reference Amrein, Granicher and Arrigoni471, 479, Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Gordon and Chao556, Reference Anderson and Clydesdale558Reference Heller, Hackler and Rivers561)

Hemicellulose: 20·8–33·0 g/100 g(Reference Knudsen485, Reference Fraser and Holmes550, Reference Anderson and Clydesdale558Reference Heller, Hackler and Rivers561)

Lignins: 2·2–9 g/100 g(Reference Begum, Nicolle and Mila221, Reference Knudsen485, Reference Haskå, Nyman and Andersson487, Reference Chen, Haack and Janecky552, Reference Gordon and Chao556, Reference Anderson and Clydesdale558Reference Maes and Delcour562)

Fructans: 0·6–4·0 g/100 g(Reference Knudsen485, Reference Haskå, Nyman and Andersson487, Reference Saunders and Walker551)

Raffinose: 1·08–1·32 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Fraser and Holmes550, Reference Saunders and Walker551)

Stachyose: 0·04–0·36 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Knudsen485, Reference Saunders and Walker551)

Total arabinoxylans: 5·0–26·9 g/100 g(Reference Amrein, Granicher and Arrigoni471, Reference Gebruers, Dornez and Boros486, Reference Haskå, Nyman and Andersson487, Reference Ward, Poutanen and Gebruers492, Reference Maes, Vangeneugden and Delcour554, Reference Maes and Delcour562, Reference Dornez, Gebruers and Wiame563)

Water-extractable arabinoxylans: 0·1–1·4 g/100 g(Reference Gebruers, Dornez and Boros486, Reference Ward, Poutanen and Gebruers492, Reference Maes and Delcour562, Reference Dornez, Gebruers and Wiame563)

β-Glucans: 1·1–2·6 g/100 g(Reference Amrein, Granicher and Arrigoni471, Reference Knudsen485, Reference Maes and Delcour562)

Phytic acid: 2·3–6·0 g/100 g(Reference Amrein, Granicher and Arrigoni471, Reference Souci, Fachmann and Kraut482, Reference Tabekhia and Donnelly505, Reference Lehrfeld and Wu553, Reference Gordon and Chao556, Reference Morris and Ellis557, Reference Bagheri and Gueguen559, Reference Camire and Clydesdale564Reference Jenab and Thompson566)

Fe: 2·5–19·0 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Monasterio and Graham510, Reference Tang, Zou and He511, Reference Gordon and Chao556, Reference Morris and Ellis557, Reference Liu, Wang and Wang567)

Mg: 390–640 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511, Reference Bagheri and Gueguen559, Reference Bagheri and Guéguen568)

Zn: 2·5–14·1 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Monasterio and Graham510, Reference Tang, Zou and He511, Reference Morris and Ellis557, Reference Bagheri and Gueguen559, Reference Liu, Wang and Wang567, Reference Bagheri and Guéguen568)

Mn: 4–14 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511)

Cu: 0·84–2·20 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511)

Se: 2–78 μg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

P: 900–1500 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511, Reference Bagheri and Gueguen559, Reference Bagheri and Guéguen568)

Ca: 24–150 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511, Reference Bagheri and Gueguen559, Reference Bagheri and Guéguen568)

Na: 2–41 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

K: 1182–1900 mg/100 g(Reference Lopez, Coudray and Bellanger70, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Tang, Zou and He511)

Thiamin (vitamin B1): 0·506–0·800 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Riboflavin (vitamin B2): 0·210–0·800 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Niacin (vitamin B3): 13·6–35·9 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Pantothenic acid (vitamin B5): 2·2–4·1 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Pyridoxine (vitamin B6): 0·704–1·303 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Shils, Olson and Shike569)

Biotin (vitamin B8): 0·044 mg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482)

Folates (vitamin B9): 0·088–0·373 mg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482, Reference Perloff and Butrum521, Reference Mullin and Jui570)

Tocols (vitamin E) = tocopherols+tocotrienols: 9·5 mg/100 g(Reference Souci, Fachmann and Kraut482)

Total tocopherols: 2·4 mg/100 g(Reference Souci, Fachmann and Kraut482)

α-Tocopherol: 0·13–2·84 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Zhou, Su and Yu571, Reference Zhou, Yin and Yu572)

Total tocotrienols: 7·1 mg/100 g(Reference Souci, Fachmann and Kraut482)

Phylloquinone (vitamin K): 0·002–0·083 mg/100 g(479, Reference Souci, Fachmann and Kraut482)

Total carotenoids: 0·25–1·18 mg/100 g(479, Reference Abdel-Aal and Hucl493)

β-Carotene: 0·003–0·010 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Zhou, Yin and Yu572)

Lutein: 0·050–0·180 mg/100 g(Reference Zhou, Su and Yu571, Reference Zhou, Yin and Yu572)

Zeaxanthin: 0·025–0·219 mg/100 g(Reference Zhou, Su and Yu571, Reference Zhou, Yin and Yu572)

β-Cryptoxanthin: 0·018–0·064 mg/100 g(Reference Zhou, Su and Yu571, Reference Zhou, Yin and Yu572)

Total phenolic acids: 761–1384 mg/100 g(Reference Barron, Surget and Rouau533, Reference Robertson, Faulds and Smith573)

Extractable (free and conjugated) phenolic acids: 46–63 mg gallic acid equivalents/100 g(Reference Irmak, Jonnala and MacRitchie574, Reference Kim, Tsao and Yang575)

Bound phenolic acids: 148–340 mg gallic acid equivalents/100 g(Reference Irmak, Jonnala and MacRitchie574, Reference Kim, Tsao and Yang575)

Total ferulic acid: 138–631 mg/100 g(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154, Reference Gallardo, Jiménez and García-Conesa194, Reference Rybka, Sitarski and Raczynskabojanowska263, Reference Kroon, Faulds and Ryden264, Reference Mattila, Pihlava and Hellstrom365, Reference Lempereur, Rouau and Abecassis499, Reference Barron, Surget and Rouau533, Reference Robertson, Faulds and Smith573, Reference Kim, Tsao and Yang575, Reference Siebenhandl, Grausgruber and Pellegrini576)

Free/soluble-conjugated ferulic acid: 1·34–23·05 mg/100 g(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154, Reference Gallardo, Jiménez and García-Conesa194, Reference Zhou, Su and Yu571, Reference Zhou, Yin and Yu572, Reference Irmak, Jonnala and MacRitchie574, Reference Kim, Tsao and Yang575, Reference Apak, Güçlü and Ozyürek577Reference Zhou and Yu579)

Bound ferulic acid: 122–286 mg/100 g(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154, Reference Irmak, Jonnala and MacRitchie574, Reference Kim, Tsao and Yang575)

Total dehydrodiferulic acid: 13–230 mg/100 g(Reference Gallardo, Jiménez and García-Conesa194, Reference Barron, Surget and Rouau533, Reference Lempereur, Surget and Rouau534, Reference Robertson, Faulds and Smith573)

Total dehydrotrimer ferulic acid: 15–25 mg/100 g(Reference Barron, Surget and Rouau533)

Total flavonoids: 14·9–40·6 mg/100 g(Reference Feng and McDonald193)

Anthocyanins: 0·9–48·0 mg/100 g(Reference Abdel-Aal and Hucl493, Reference Abdel-Aal and Hucl536, Reference Abdel-Aal, Abou-Arab and Gamel537, Reference Iqbal, Bhanger and Anwar580)

Isoflavonoids:

Daidzein: 3·5 μg/100 g(Reference Adlercreutz and Mazur293)

Genistein: 3·8–6·9 μg/100 g(Reference Adlercreutz and Mazur293, Reference Liggins, Mulligan and Runswick538)

Lignans: 2·8–6·7 mg/100 g(Reference Begum, Nicolle and Mila221, Reference Smeds, Eklund and Sjoholm581)

Alkylresorcinols: 215–323 mg/100 g(Reference Mattila, Pihlava and Hellstrom365, Reference Ross, Shepherd and Schupphaus392, Reference Kulawinek, Jaromin and Kozubek582)

Betaine: 230–1506 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Zeisel, Mar and Howe477, Reference Waggle, Lambert and Miller483, Reference Graham, Hollis and Migaud583, Reference Slow, Donaggio and Cressey584)

Total choline: 74–270 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Calhoun, Hepburn and Bradley314, Reference Zeisel, Mar and Howe477, Reference Waggle, Lambert and Miller483, Reference Graham, Hollis and Migaud583)

Phytosterols: 121–195 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Hakala, Lampi and Ollilainen543, Reference Piironen, Toivo and Lampi546, Reference Nyström, Lampi and Rita585)

Total d-chiro-inositol: not detected(Reference Kim, Kim and Joo245)

Policosanol: 0·11–3·00 mg/100 g(Reference Irmak, Jonnala and MacRitchie574, Reference Irmak, Dunford and Milligan586)

PABA: 1·34 mg/100 g(Reference Calhoun, Hepburn and Bradley314)

Wheat germ

α-Linolenic acid (18 : 3n-3): 0·47–0·59 mg/100 g(Reference Srivastava, Sudha and Baskaran25, Reference Trautwein548, Reference Moruzzi, Viviani and Sechi587)

Reduced glutathione: about 19·4–245·7 mg/100 g(Reference Every, Morrison and Simmons549)

Oxidised glutathione: about 15·3–122·4 mg/100 g(Reference Every, Morrison and Simmons549)

Sulfur amino acids:

Methionine: 0·39–0·58 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Cystine: 0·35–0·61 g/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Sugars:

Glucose: < 390–700 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Dubois, Geddes and Smith588Reference Linko, Cheng and Milner590)

Fructose: < 200–801 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Dubois, Geddes and Smith588Reference Linko, Cheng and Milner590)

Sucrose: 7·7–16·0 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Fraser and Holmes550, Reference Dubois, Geddes and Smith588Reference Shurpalekar and Rao592)

Total fibre (lignins, oligosaccharides, resistant starch and phytic acid included): 10·6–24·7 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Haskå, Nyman and Andersson487Reference Nyström, Paasonen and Lampi489)

Insoluble fibre: 8·5–18·6 g/100 g(Reference Srivastava, Sudha and Baskaran25, Reference Souci, Fachmann and Kraut482, Reference Hernot, Boileau and Bauer488)

Soluble fibre: 2·1–6·1 g/100 g(Reference Srivastava, Sudha and Baskaran25, Reference Souci, Fachmann and Kraut482, Reference Hernot, Boileau and Bauer488)

Cellulose: 7·5 g/100 g(Reference Fraser and Holmes550)

Hemicellulose: 6·8 g/100 g(Reference Fraser and Holmes550)

Lignins: 1·3–1·6 g/100 g(Reference Haskå, Nyman and Andersson487)

Fructans: 1·7–2·5 g/100 g(Reference Haskå, Nyman and Andersson487)

Raffinose: 5·0–10·9 g/100 g(Reference Fraser and Holmes550, Reference Dubois, Geddes and Smith588Reference Shurpalekar and Rao592)

Total arabinoxylans: 5·6–9·1 g/100 g(Reference Haskå, Nyman and Andersson487, Reference Dornez, Gebruers and Wiame563)

Water-extractable arabinoxylans: 0·37 g/100 g(Reference Dornez, Gebruers and Wiame563)

Phytic acid: 1·3–2·2 g/100 g(Reference Souci, Fachmann and Kraut482, Reference Fretzdorff565, Reference Bilgicli and Ibanoglu593)

Fe: 3·9–10·3 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Mg: 200–340 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Zn: 10–18 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Mn: 9–18 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Inglett and Blessin594)

Cu: 0·70–1·42 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Se: 1–79 μg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

P: 770–1337 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Ca: 36–84 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Na: 2–37 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

K: 842–1300 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Garcia, Gardner and Cavins589, Reference Garcia, Inglett and Blessin594)

Thiamin (vitamin B1): 0·8–2·7 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Riboflavin (vitamin B2): 0·49–0·80 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Niacin (vitamin B3): 4·0–8·5 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Pantothenic acid (vitamin B5): 1–2·7 mg/100 g(Reference Calhoun, Hepburn and Bradley314, 479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Pyridoxine (vitamin B6): 0·49–1·98 mg/100 g(479, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483)

Biotin (vitamin B8): 17·0–17·4 μg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482)

Folates (vitamin B9): 0·14–0·70 mg/100 g(Reference Calhoun, Hepburn and Bradley314, Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Perloff and Butrum521)

Tocols (vitamin E) = tocopherols+tocotrienols: 23·1–31 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Nielsen and Hansen524)

Total tocopherols: 21·5–30·6 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Nielsen and Hansen524)

α-Tocopherol: 3·1–22 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Waggle, Lambert and Miller483, Reference Nielsen and Hansen524, Reference Nielsen and Hansen525, Reference Leenhardt, Fardet and Lyan591)

Total tocotrienols: 1·3–1·6 mg/100 g(Reference Souci, Fachmann and Kraut482, Reference Nielsen and Hansen524)

Phylloquinone (vitamin K): 0·003–0·350 mg/100 g(Reference Souci, Fachmann and Kraut482)

β-Carotene: 0·062 mg/100 g(Reference Souci, Fachmann and Kraut482)

Extractable (free and conjugated) phenolic acids: about 51 mg/100 g(Reference Gallardo, Jiménez and García-Conesa194)

Total ferulic acid: 7–124 mg/100 g(Reference Gallardo, Jiménez and García-Conesa194, Reference Lempereur, Rouau and Abecassis499)

Free/conjugated soluble ferulic acid: about 18 mg/100 g(Reference Gallardo, Jiménez and García-Conesa194)

Total dehydrodiferulic acid: about 9 mg/100 g(Reference Gallardo, Jiménez and García-Conesa194)

Total flavonoids: 300 mg rutin equivalents/100 g(Reference Zhu, Zhou and Qian595)

Lignans: 0·490 mg/100 g(Reference Dodin, Lemay and Jacques596)

Betaine: 306–1395 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Zeisel, Mar and Howe477, Reference Waggle, Lambert and Miller483)

1395 mg/100 g(Reference Zeisel, Mar and Howe477): toasted

Total choline: 152–330 mg/100 g(Reference Likes, Madl and Zeisel227, Reference Calhoun, Hepburn and Bradley314, Reference Waggle, Lambert and Miller483)

152 mg/100 g(Reference Zeisel, Mar and Howe477): toasted

Phytosterols: 410–450 mg/100 g(Reference Nyström, Paasonen and Lampi489, Reference Piironen, Toivo and Lampi546, Reference Ostlund, Racette and Stenson597)

Policosanol: 1·0 mg/100 g(Reference Irmak, Dunford and Milligan586)

PABA: 0·852 mg/100 g(Reference Calhoun, Hepburn and Bradley314)

Appendix 2

References for evaluating the range of compound bioavailability and degree of fibre-type compounds fermentation from whole-grain wheat, wheat bran and/or derived products (data for Table 2).

Whole-grain wheat and derived products

Reduced glutathione: negligible in humans as free compound(Reference Witschi, Reddy and Stofer209)

Stachyose and raffinose:

Completely fermented in vitro within 48 h as free compound(Reference Krause, Easter and Mackie296)

97–99 % in dogs(Reference Mühlum, Ingwersen and Schünemann598)

Total fibre: 34 % in human subjects fed wholemeal bread(Reference Van Dokkum, Pikaar and Thissen599)

Cellulose: 20 % in human subjects fed wholemeal bread(Reference Van Dokkum, Pikaar and Thissen599)

Hemicellulose: 46 % in human subjects fed wholemeal bread(Reference Van Dokkum, Pikaar and Thissen599)

Lignins: 4 % in human subjects fed wholemeal bread(Reference Van Dokkum, Pikaar and Thissen599)

Phytic acid: 54–79 % apparently degraded (faeces recovery) in human subjects fed Hovis bread (whole bread)(Reference McCance and Widdowson600)

Rapidly and almost fully absorbed (about 79 %) in upper part of the gastrointestinal tract of rats fed free compound(Reference Sakamoto, Vucenik and Shamsuddin601)

Small-intestinal phytases have high activity in rats and very much lower activity in human subjects and pigs(Reference Lopez, Leenhardt and Coudray217)

Fe: 1–20 % in human subjects fed usual diets(Reference Martin204)

Mg:

70 % in rats fed whole-wheat flour(Reference Levrat-Verny, Coudray and Bellanger219)

21–28 % in human subjects fed brown bread diet(Reference McCance and Widdowson602)

50 % in human subjects fed a typical diet(603)

57·6 % in human subjects fed a standard diet(Reference Walti, Zimmermann and Walczyk604)

Zn:

16·6 % in human subjects consuming wholemeal bread(Reference Sandstrom, Arvidsson and Cederblad605)

20 % in adult women consuming whole-wheat tortillas(Reference Sundkvist, Dahlin and Nilsson606)

35 % in rats fed whole-wheat flour(Reference Levrat-Verny, Coudray and Bellanger219)

88·9–94·6 % in rats fed whole-wheat flour(Reference Saha, Weaver and Mason607)

18·5 % in rats fed wheatmeal(Reference Fox, Fairweather-Tait and Eagles608)

60–82 % in rats fed whole-grain wheat(Reference Welch, House and Ortiz-Monasterio609)

30–37 % in rats fed whole-wheat flour chapatti(Reference Ahmed, Anjum and Ur Rehman610)

Cu:

62–85 % in human subjects fed whole-wheat bread(Reference Johnson and Lykken611)

16·3–16·5 % in rats fed free compound(Reference Lopez, Levrat-Verny and Coudray71, Reference Lopez, Coudray and Levrat-Verny73)

Se:

81·1–84·5 % in rats fed whole-wheat flour(Reference Saha, Weaver and Mason607)

73–86 % in rats fed whole wheat as compared with sodium selenite(Reference Mutanen, Koivistoinen and Morris612)

100 % in rats fed whole-wheat flour as compared with sodium selenite(Reference Alexander, Whanger and Miller613)

P: 41–55 % in human subjects fed brown bread diet(Reference McCance and Widdowson602)

Ca:

81·7 % in human subjects fed whole-wheat bread(Reference Weaver, Heaney and Martin614)

43–44 % in rats fed whole-wheat flour chapatti(Reference Ahmed, Anjum and Ur Rehman610)

85·7–92·8 % in rats fed whole-wheat flour(Reference Saha, Weaver and Mason607)

Thiamin (vitamin B1): 91 % in rats fed whole-wheat bread compared with free thiamine mononitrate (100 %)(Reference Ranhotra, Gelroth and Novak519)

Riboflavin (vitamin B2): 95 % as oral supplement in human subjects(Reference Zempleni, Galloway and McCormick615)

Niacin (vitamin B3): low(Reference Truswell19)

Pantothenic acid (vitamin B5): about 50 % in human subjects for average American diet(Reference Tarr, Tamura and Stokstad616)

Pyridoxine (vitamin B6): 71–79 % for an average American diet compared with free compound(Reference Tarr, Tamura and Stokstad616)

α-Tocopherol: 70 % in human subjects fed free compound(Reference Kayden and Traber617)

Total ferulic acid: 3·2–3·6 % urinary excretion in rats(Reference Adam, Crespy and Levrat-Verny152)

Free/soluble-conjugated ferulic acid: at least that of wheat bran in rat small intestine(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154)

Bound ferulic acid: a small fraction released within small intestine by intestinal esterases(Reference Andreasen, Kroon and Williamson618)

Alkylresorcinols: 60–79 % from ileal samples in pigs fed whole-grain rye bread(Reference Ross, Shepherd and Knudsen619)

Phytosterols: weakly absorbed from the gut(Reference Nissinen, Gylling and Vuoristo620)

Total free inositols (myo- and chiro-inositol):

Apparently high in rats fed free compounds for myo-inositol(Reference Pak, Huang and Lilley256)

Apparently high in women fed free compounds for chiro-inositol(Reference Nestler, Jakubowicz and Reamer621)

Apparently high in old human subjects fed free compounds for pinitol(Reference Campbell, Haub and Fluckey622)

Wheat bran

Total fibre:

55·6 % neutral sugars in human subjects fed wheat bran(Reference Chen, Haack and Janecky552)

34 % neutral sugars in human subjects fed wheat bran(Reference Nyman, Asp and Cummings623)

35–42 % neutral-detergent fibre in human subjects fed coarse and fine bran(Reference Heller, Hackler and Rivers561)

36·9 and 41·1 % in rats fed coarse and fine brans(Reference Kahlon, Chow and Hoefer624)

39 % in rats fed wheat bran(Reference Nyman, Asp and Cummings623)

49·1 % NSP in rats fed wheat bran(Reference Hansen, Knudsen and Eggum625)

58·8–65·0 % in pigs fed coarse and fine bran cell walls(Reference Ehle, Jeraci and Robertson626)

41·5 % in pigs fed wheat bran-based diet(Reference Robertson, Murison and Chesson627)

Insoluble fibre:

42·3 % in rats fed wheat bran(Reference Hansen, Knudsen and Eggum625)

Cellulose:

6–23 % in human subjects fed coarse and fine bran(Reference Heller, Hackler and Rivers561)

7 % in human subjects fed wheat bran(Reference Nyman, Asp and Cummings623)

13·8–21·9 % in rats fed coarse and fine brans(Reference Kahlon, Chow and Hoefer624)

24·1 % in pigs fed wheat bran-based diet(Reference Robertson, Murison and Chesson627)

18·2–23·7 % in pigs fed coarse and fine brans(Reference Ehle, Jeraci and Robertson626)

Hemicellulose:

50–54 % in human subjects fed coarse and fine brans(Reference Heller, Hackler and Rivers561)

69·4–74·4 % in pigs fed coarse and fine brans(Reference Ehle, Jeraci and Robertson626)

46·5 % non-cellulosic neutral sugar residues in pigs fed wheat bran-based diet(Reference Robertson, Murison and Chesson627)

Lignins:

Undigested in humans(Reference Heller, Hackler and Rivers561)

0 % in rats fed wheat bran(Reference Nyman, Asp and Cummings623)

0–4 % in rats fed processed wheat bran(Reference Nyman and Asp628)

Soluble fibre: 72·9 % in rats fed wheat bran fibre(Reference Hansen, Knudsen and Eggum625)

Total arabinoxylans: 49·2 % arabinose and 71·1 % xylose in human subjects fed wheat bran(Reference Chen, Haack and Janecky552)

Phytic acid:

Phytate from wheat bran without phytase is almost not absorbed at the intestinal level in humans(Reference Sandberg and Andersson629)

58–60 % degraded into lower myo-inositol phosphates in ileostomates fed raw wheat bran(Reference Sandberg and Andersson629, Reference Sandberg, Andersson and Carlsson630) and only 5 % with phytase-deactivated wheat bran(Reference Sandberg and Andersson629, Reference Sandberg, Andersson and Carlsson630)

58 % degraded in ileostomates and 25 % hydrolysed for extruded wheat bran (loss of phytase activity)(Reference Sandberg, Andersson and Carlsson630)

Fe:

3·8 % in human subjects fed rolls made of wheat bran and white wheat flour(Reference Brune, Rossander-Hulten and Hallberg631)

Negative effect of bran on Fe absorption is not observed in rats(Reference Reddy and Cook632)

Se:

About 60 % in rats fed wheat bran compared with sodium selenite and selenomethionine biological value(Reference Reeves, Gregoire and Garvin633)

80 % in rats fed wheat bran as compared with sodium selenite(Reference Alexander, Whanger and Miller613)

P: 41–56 % in human subjects fed sodium phytate+white bread(Reference McCance and Widdowson602)

Ca: 22·3 % in human subjects fed extruded wheat bran cereals(Reference Weaver, Heaney and Martin614)

Niacin (vitamin B3):

27–38 % in human subjects fed a concentrate of bound niacin from wheat bran(Reference Carter and Carpenter634)

17 % in rats fed a concentrate of bound niacin from wheat bran (cited in Carter & Carpenter(Reference Carter and Carpenter634))

Pyridoxine (vitamin B6): unavailable in human subjects fed wheat bran(Reference Kies, Kan and Fox635)

Folates (vitamin B9): low in human subjects fed wheat bran(Reference Fenech, Noakes and Clifton467)

Tocopherols/tocotrienols (vitamin E): not available in rats fed wheat bran(Reference Kahlon, Chow and Hoefer636)

Bound phenolic acids:

32·7 % in pigs fed a wheat bran diet(Reference Robertson, Murison and Chesson627)

Partially and slowly solubilised from wheat bran within a human model colon(Reference Kroon, Faulds and Ryden264)

Total ferulic acid:

 < 5 % in small intestine of rats fed wheat bran-based diet(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154)

3·9 % urinary excretion in rats fed wheat bran(Reference Adam, Crespy and Levrat-Verny152)

1·99–5·65 % urinary excretion in human subjects fed high-bran cereal(Reference Kern, Bennett and Mellon196)

Free/soluble-conjugated ferulic acid:

High in rat small intestine fed wheat bran(Reference Rondini, Peyrat-Maillard and Marsset-Baglieri154)

27·77–78·92 % urinary excretion in human subjects fed high-bran cereal(Reference Kern, Bennett and Mellon196)

Bound ferulic acid: a small fraction (%?) released within rat small intestine by intestinal esterases following wheat bran consumption(Reference Andreasen, Kroon and Williamson618)

Dehydrodiferulic acid:

Undetectable in plasma of human subjects fed high-bran cereal(Reference Kern, Bennett and Mellon196)

Free diferulic acid can be absorbed from the gut in rats fed wheat bran(Reference Andreasen, Kroon and Williamson637)

Alkylresorcinols: 45–71 % from ileostomy effluents in human subjects fed rye bran soft/crisp bread(Reference Ross, Kamal-Eldin and Lundin393)

Phytosterols: weakly absorbed from the gut in human subjects(Reference Nissinen, Gylling and Vuoristo620)

Appendix 3

References for the physiological mechanisms and health effects of bioactive compounds from whole-grain wheat, and wheat bran and germ fractions (data for Tables 3 and 4)*

* Keywords relative to the physiological mechanisms involved, health outcomes associated with bioactive compounds and the corresponding reference(s) are given; the models used, i.e. human, animals or in vitro cultured cells, may be found in references cited.

α-Linolenic acid (18 : 3n-3):

Health and diseases(Reference Connor638); CVD(Reference Trautwein548, Reference Connor638Reference Kang and Leaf641); anti-atherosclerotic(Reference Alessandri, Pignatelli and Loffredo298); depression and anxiety(Reference Edwards, Peet and Shay642, Reference Yehuda, Rabinovitz and Mostofsky643); plasma TAG(Reference Djousse, Folsom and Province644); blood clotting, thrombosis, plasma lipid profile, blood pressure and inflammation(Reference Connor638); colon(Reference Narisawa, Fukaura and Yazawa645) and breast(Reference Klein, Chajes and Germain646) cancers; synthesis of cytokines and mitogens(Reference Connor638); arachidonic acid (20 : 4n-6) and eicosanoids in tissues (such as lung) and plasma phospholipids, and synthesis of pro-thrombotic cyclo-oxygenase-derived products (thromboxane A2 and B2, PGE2)(Reference Hwang, Boudreau and Chanmugam647); immune system, cell signalling and gene expression(Reference Chapkin, McMurray and Davidson648, Reference Enke, Seyfarth and Schleussner649)

Glutathione (reduced, GSH):

Health and diseases(Reference Townsend, Tew and Tapiero650); source of cysteine(Reference Higashi, Tateishi and Naruse651); oral cancer, anti-carcinogen, antioxidant effect, binding with cellular mutagens and GSH transferase activity(Reference Wattenberg110); detoxification of toxic electrolytic metabolites, xenobiotics and reactive oxygen intermediates(Reference Bilzer and Lauterburg652); cellular immune function(Reference Gmünder, Roth and Eck208)

Sulfur amino acids:

Methionine:

Precursor of glutathione(Reference Morand, Rios and Moundras200); precursor of S-adenosyl methionine(Reference Troen, Chao and Crivello653); neural tube defects(Reference Essien and Wannberg654); colon cancer(Reference Giovannucci, Rimm and Ascherio410); cognitive impairment in situation of folate deficiency(Reference Troen, Chao and Crivello653); antioxidant activity(Reference Caylak, Aytekin and Halifeoglu655); lipotrope(Reference Newberne and Rogers239)

Cystine:

Hair and nail development(Reference Khumalo, Dawber and Ferguson656, Reference Sass, Skladal and Zelger657); muscle wasting(Reference Droge and Holm658); antioxidant and cell signalling through reactive cysteine residues in proteins(Reference Netto, de Oliveira and Monteiro659)

Total fibre(Reference Liu18, Reference Slavin, Martini and Jacobs46, Reference Slavin58, Reference Ferguson and Harris266, Reference Marlett, McBurney and Slavin660, Reference Tucker and Thomas661):

Type 2 diabetes risk(Reference Salmeron, Ascherio and Rimm662); risk of weight and fat gains; large bowel cancer(Reference Boffa, Lupton and Mariani66, Reference McIntyre, Gibson and Young75, Reference Alabaster, Tang and Shivapurkar106, Reference Ferguson and Harris266); satiating effect; cholesterol, bile acids, hormonal activity; immune system, toxicant transit; production of SCFA in the colon(Reference Slavin, Jacobs and Marquart663); SCFA, growth of tumour cells, glutathione-S-transferase and genotoxic activity of 4-hydroxynonenal(Reference Glei, Hofmann and Kuster664); dilution of gut substances; energy content and glycaemic index of foods; insulin response; free radicals(Reference Kohlmeier, Simonsen and Mottus93)

Insoluble fibre(Reference Slavin63): antioxidant-bound phenolics and colon(Reference Vitaglione, Napolitano and Fogliano150); faecal wet and dry weight and faecal bulking effect(Reference Marlett, McBurney and Slavin660); intestinal transit(Reference Marlett, McBurney and Slavin660)

Soluble fibre(Reference Slavin63): cholesterol; glucose and insulin responses(Reference Moore, Park and Tsuda412); bowel health(Reference Moore, Park and Tsuda412)

Lignins:

Antioxidant(Reference Stavric112, Reference Dizhbite, Telysheva and Jurkjane149, Reference Labaj, Wsolova and Lazarova224); dietary carcinogens adsorption(Reference Ferguson and Harris69, Reference Ferguson and Harris266); bile acid reabsorption(Reference Chang and Johnson268); bile-salt sequestrating agent(Reference Eastwood and Girdwood107, Reference Eastwood and Hamilton108); fat absorption(Reference Eastwood and Mowbray665); bile salt pool size(Reference Pomare and Heaton666); cholesterol turnover(Reference Eastwood667); formation of carcinogenic metabolites from bile salts(Reference Drasar and Jenkins269); precursor of lignans(Reference Begum, Nicolle and Mila221); anti-carcinogenic(Reference Akao, Seki and Nakagawa265)

Oligosaccharides (raffinose, stachyose and fructans)(Reference Swennen, Courtin and Delcour295):

Serum cholesterol(Reference Slavin, Martini and Jacobs46, Reference Hara, Haga and Aoyama80); gut modifier, enzyme modulator and binding scavenger(Reference Slavin, Martini and Jacobs46)

Fructans(Reference Kaur and Gupta668, Reference Roberfroid and Delzenne669):

Lifespan and weight gain reduction(Reference Rozan, Nejdi and Hidalgo670); prebiotic(Reference Liu18); microbiota(Reference Gibson, Beatty and Wang671); growth of harmful bacteria, immune system, absorption of minerals and synthesis of B vitamins(Reference Liu18); absorption of Ca, Mg and Fe(Reference Liu18, Reference Coudray, Bellanger and Castiglia-Delavaud72, Reference Lopez, Coudray and Levrat-Verny73); butyrate with cancer-preventing properties in the colon(Reference Femia, Luceri and Dolara672); growth of cancer cells(Reference Femia, Luceri and Dolara672Reference Avivi-Green, Polak-Charcon and Madar674); glycaemia and insulinaemia(Reference Kaur and Gupta668); plasma TAG and total/LDL-cholesterol(Reference Brighenti, Casiraghi and Canzi675, Reference Williams676); lipid metabolism(Reference Beylot677); hepatic gluconeogenesis and glycolysis(Reference Roberfroid and Delzenne669)

Raffinose: weight gain(Reference Tortuero, Fernández and Rupérez297)

Arabinoxylans(Reference Glei, Hofmann and Kuster664):

Colon cancer growth and progression(Reference Pai, Tarnawski and Tran678); glucose response(Reference Lu, Walker and Muir411); chemoprotection and fermentation products(Reference Glei, Hofmann and Kuster664); bile acids(Reference Glei, Hofmann and Kuster664); anti-proliferative properties of butyrate(Reference McMillan, Butcher and Wallis679)

β-Glucans(Reference Wood56):

Satiety(Reference Nilsson, Ostman and Holst54); blood sugar and gastric emptying rate(Reference Liu18); blood cholesterol(Reference Liu18); hypoglycaemic and hypoinsulinaemic(Reference Braaten, Wood and Scott680Reference Tappy, Gugolz and Wursch682); hypocholesterolaemic(Reference Wood56, Reference Maki, Shinnick and Seeley683); propionate, hepatocyte lipid synthesis and cholesterolaemia(Reference Wright, Anderson and Bridges684); anti-carcinogenic(Reference Mantovani, Bellini and Angeli391); immune system(Reference Mantovani, Bellini and Angeli391); peripheral blood monocytes and breast cancer(Reference Demir, Klein and Mandel-Molinas685); anti-bacterial, anti-parasitic, anti-fungal and anti-viral(Reference Mantovani, Bellini and Angeli391)

Phytic acid:

Risk of colon(Reference Ullah and Shamsuddin100) and breast(Reference Vucenik, Yang and Shamsuddin101) cancers; anti-cancer agent(Reference Shamsuddin95, Reference Reddy99, Reference Alabaster, Tang and Shivapurkar106, Reference Vucenik and Shamsuddin686); antioxidant activity(Reference Graf, Empson and Eaton148); chelation with various metals and Fenton reaction(Reference Shamsuddin95); oxidative damage to the intestinal epithelium and neighbouring cells (cited in Slavin(Reference Slavin63)); lipid peroxidation (cited in Ferguson & Harris(Reference Ferguson and Harris69)); formation of ADP-iron-oxygen complexes that initiate lipid peroxidation(Reference Muraoka and Miura687); cellular and nuclear signalling pathways(Reference Shamsuddin95); plasma glucose (cited in Yoon et al. (Reference Yoon, Thompson and Jenkins182)); insulin and/or plasma cholesterol and TAG(Reference Lee, Park and Chun688Reference Onomi, Okazaki and Katayama690); lipid levels in liver and serum(Reference Lee, Park and Cho691); detoxification capacity of liver and levels of GSH transferase and cytochrome P-450(Reference Singh, Prakash Singh and Bamezai692); immune response(Reference Reddy99); renal stones(Reference Grases, Simonet and March693); calcification of cardiovascular system(Reference Grases, Sanchis and Perello694); dental caries and platelet aggregation, treatment of hypercalciura and kidney stones, and Pb poisoning(Reference Graf and Eaton218); gene expression(Reference Shen, Xiao and Ranallo695, Reference Steger, Haswell and Miller696)

Resistant starch(Reference Sajilata, Singhal and Kulkarni697):

Physically inaccessible within small intestine(Reference Liu18); prebiotic(Reference Topping, Fukushima and Bird415); glycaemic response(Reference Nilsson, Ostman and Granfeldt52); glucose metabolism and plasma NEFA(Reference Nilsson, Ostman and Holst54); energy intake; SCFA, butyrate and colon health, and SCFA and serum cholesterol(Reference Brouns, Kettlitz and Arrigoni65, Reference Hara, Haga and Aoyama80); lipid oxidation and metabolism(Reference Higgins, Higbee and Donahoo67); gallstones(Reference Malhotra698)

Fe:

Neural functioning(Reference Beard and Connor699); catalase cofactor(700); lipid peroxidation(Reference Uehara, Chiba and Mogi701); cofactor, enzymes and energy metabolism(Reference Rosenzweig and Volpe702); cellular energy metabolism(Reference Oexle, Gnaiger and Weiss703); infection and mental function(Reference Ramdath and Golden704); cognitive development and intellectual performance(Reference Lozoff, Jimenez and Hagen705, Reference Oski, Honig and Helu706); collagen synthesis(Reference Prockop707); bone health(Reference Katsumata, Katsumata-Tsuboi and Uehara708); aerobic endurance exercise(Reference Willis, Dallman and Brooks709); immunity and infection(Reference Cook and Lynch710); vitamin metabolism(Reference Rosales, Jang and Pinero711); serum and liver TAG, phospholipid, and cholesterol(Reference Uehara, Chiba and Mogi701); obesity(Reference McClung and Karl712)

Mg(Reference Martin204, 603):

Metalloenzymes(Reference Shils, Olson and Shike569); alkaline phosphatase (bone health)(Reference Clancaglini, Plzauro and Curti713); antioxidant(Reference Bussiere, Gueux and Rock714); lipid peroxidation(Reference Olatunji and Soladoye715); hypertriacylglycerolaemia(Reference Kisters, Spieker and Tepel716) and insulin resistance(Reference McCarty156, Reference Paolisso, Sgambato and Pizza159, Reference Olatunji and Soladoye715, Reference Barbagallo and Dominguez717, Reference Colditz, Manson and Stampfer718); diabetes(Reference Durlach and Collery157, Reference Nadler, Balon and Rude719Reference van Dam, Hu and Rosenberg722); glucose uptake(Reference Paolisso, Sgambato and Gambardella158), glucose metabolic clearance rate and insulin response(Reference Paolisso, Sgambato and Gambardella158, Reference Paolisso, Sgambato and Pizza159), and oxidative glucose metabolism(Reference Paolisso, Dimaro and Cozzolino723); platelet aggregability(Reference Shechter, Merz and Paul-Labrador170); blood pressure regulation(Reference Kawano, Matsuoka and Takishita171); coronary atherosclerosis and acute thrombosis(Reference Liao, Folsom and Brancati169); vascular function(Reference Nadler, Buchanan and Natarajan724); blood pressure(Reference Ascherio, Rimm and Giovannucci725); cardiovascular death rate(Reference Rubenowitz, Axelsson and Rylander726); osteoporosis(Reference Cohen727); angiogenesis and inflammation(Reference Bernardini, Nasulewicz and Mazur728); stone formation(Reference Reungjui, Prasongwatana and Premgamone729)

Zn(Reference Martin204, 700):

Alkaline phosphatase cofactor; antioxidant and superoxide dismutase (SOD) cofactor(Reference Bray and Bettger730, Reference Zago and Oteiza731); skeletal growth and maturation, and bone metabolism(Reference Beattie and Avenell732); chemical inactivator(Reference Slavin, Martini and Jacobs46); formation of active carcinogenic compounds(Reference Kohlmeier, Simonsen and Mottus93); Zn-binding compounds and cancer cell death(Reference Ding, Yu and Lind733); oesophagus cancer(Reference Guo, Zhao and Jiang734); Zn sensing receptor and cell signalling(Reference Hershfinkel, Silverman and Sekler735); immune functions(Reference Bogden, Oleske and Munves736); inflammatory diseases and cell signalling mechanisms(Reference Shen, Oesterling and Stromberg737); type 2 diabetes(Reference Mocchegiani, Giacconi and Malavolta738); food intake(Reference Ohinata, Takemoto and Kawanago739)

Mn(Reference Shils, Olson and Shike569, 700):

Antioxidant(Reference Robinson740); metalloenzyme constituent and enzyme activation(Reference Shils, Olson and Shike569); bone health(Reference Beattie and Avenell732, Reference Freeland-Graves and Turnlund741); manganese-SOD, NF-κB activation and carcinogenic process(Reference Cho, Park and Kang742); manganese-SOD and tumour growth(Reference Kattan, Minig and Dauça743)

Cu(Reference Martin204, 700):

Antioxidant(Reference Johnson, Fischer and Kays744); Cu-containing/binding proteins(Reference Shils, Olson and Shike569); bone health(Reference Beattie and Avenell732, Reference Baker, Harvey and Majask-Newman745); central nervous system dysfunction(700); immune and cardiac dysfunctions(700, Reference Lukaski, Klevay and Milne746, Reference Milne747); heart health(Reference Klevay748, Reference Zhou, Jiang and Kang749); anti-cancer effect and DNA binding(Reference Hammud, Nemer and Sawma750); risk of CHD(Reference Klevay751, Reference Klevay752)

Se(Reference Martin204):

Glutathione peroxidase and thioredoxin reductase cofactor; antioxidant(Reference Slavin, Martini and Jacobs46, Reference Kohlmeier, Simonsen and Mottus93, Reference Tapiero, Townsend and Tew753); constituent of selenoproteins(Reference Levander754); tumour growth(Reference Slavin, Martini and Jacobs46, Reference Wattenberg110, Reference Levander754, Reference Burk755); prostate and colon cancer (cited in Reeves et al. (Reference Reeves, Gregoire and Garvin633)); susceptibility to carcinogens(Reference Jacobs756, Reference Jacobs, Forst and Beams757); apoptotic effects(Reference Jariwalla, Gangapurkar and Nakamura758); anti-carcinogenic(Reference Gromadzinska, Reszka and Bruzelius759); cell membranes and oxidation damage(Reference Levander and Morris760); anti-infective(Reference Arvilommi, Poikonen and Jokinen761, Reference Boyne and Arthur762); plasma, liver and erythrocyte GSH peroxidase activity(Reference Ciappellano, Testolin and Porrini763); insulin resistance and vascular endothelium(Reference Douillet, Bost and Accominotti764, Reference Stapleton765); platelet aggregation(Reference Tapiero, Townsend and Tew753)

P(Reference Martin204, 603, Reference Uribarri766):

Kidney health(Reference Uribarri766, Reference Loghman-Adham767); colorectal adenoma(Reference Kesse, Boutron-Ruault and Norat768); tooth development(Reference Arnold and Gaengler769)

Ca(Reference Martin204, 603):

Colorectal cancer(Reference Bostick, Potter and Fosdick770, Reference Ishihara, Inoue and Iwasaki771); signal transduction element(Reference Mariot, Vanoverberghe and Lalevee772); cell signalling(Reference Taylor, Zeng and Pottle773); mitotic events and cell cycle(Reference Ciapa, Pesando and Wilding774); hypertension(603, Reference Bucher, Cook and Guyatt775, Reference Gillman, Hood and Moore776); stroke risk(Reference Umesawa, Iso and Ishihara777); diabetes risk(Reference Colditz, Manson and Stampfer718); tooth development(Reference Arnold and Gaengler769); energy balance and obesity(Reference Astrup778, Reference Major, Chaput and Ledoux779)

Na(Reference Martin204):

Fluid balance(Reference Sharp780); blood pressure(Reference Hollenberg781); CVD(Reference Alderman782); osteoporosis and bone health(Reference Heaney783)

K(Reference Martin204, Reference Demigne, Sabboh and Remesy784, Reference He and MacGregor785):

Diabetes risk(Reference Colditz, Manson and Stampfer718); insulin secretion(Reference Durlach and Collery157, Reference Sjogren, Floren and Nilsson786); blood pressure(Reference Appel, Moore and Obarzanek787); CVD(Reference Chang, Hu and Yue788Reference Young and Ma790); cardiac arrhythmias(Reference Nolan, Batin and Andrews791); kidney health(Reference Tobian, Macneill and Johnson792) and stones(Reference Curhan, Willett and Rimm793); bone health(Reference Marangella, Di Stefano and Casalis794); hypercalciura(Reference Lemann, Pleuss and Gray795)

Thiamin (vitamin B1)(Reference Martin204, 796, Reference Singleton and Martin797):

Antioxidant(Reference Lukienko, Mel'nichenko and Zverinskii798); glucose metabolism and Krebs cycle(Reference Andreasen, Christensen and Meyer799); mental and neuronal health(Reference Ambrose, Bowden and Whelan800)

Riboflavin (vitamin B2)(Reference Martin204, 796):

Haematopoiesis(Reference Fairweather-Tait, Powers and Minski801, Reference Sirivech, Driskell and Frieden802); gastrointestinal development(Reference Yates, Evans and Powers803); mental health(Reference Sterner and Price804); vision(Reference Miyamoto and Sancar805); cardiovascular protection(Reference Mack, Hultquist and Shlafer806, Reference Powers807); cancer(Reference Siassi and Ghadirian808, Reference Webster, Gawde and Bhattacharya809)

Niacin (vitamin B3)(Reference Martin204, 796):

Hypolipidaemic and cardiovascular protection(Reference Figge, Figge and Souney810, Reference Hodis811); cancers(Reference Jacobson, Dame and Pyrek812); AIDS(Reference Pontes Monteiro, Ferreira da Cunha and Correia Filho813); arthritis(Reference Jonas, Rapoza and Blair814); catecholamine stimulation of lipolysis(Reference Carlson815, Reference Davies and Souness816) (cited in Marcus et al. (Reference Marcus, Sonnenfeld and Karpe817) and Figge et al. (Reference Figge, Figge and Souney810))

Pantothenic acid (vitamin B5)(Reference Martin204, 796)

Pyridoxine (vitamin B6)(Reference Martin204, 796):

Colorectal cancer(Reference Anguita, Gasa and Martin-Orue818); asthma and CVD(Reference Ubbink, Becker and Vermaak819); impaired homocysteine metabolism and occlusive arterial disease(Reference Brattström, Israelsson and Norrving820)

Biotin (vitamin B8)(Reference Martin204, 796, Reference McMahon821Reference Sweetman and Nyhan823):

Regulation of gene expression(Reference Rodriguez-Melendez and Zempleni824); cell proliferation(Reference Manthey, Griffin and Zempleni825); dermatological abnormalities; immune response(Reference Baez-Saldana, Diaz and Espinoza826, Reference Rabin827)

Folates (vitamin B9)(Reference Martin204, 796, Reference Kamen828):

Plasma homocysteinaemia(Reference Moat, Hill and McDowell829, Reference Ward, McNulty and McPartlin830); neural tube defects(Reference Berry, Li and Erickson273, Reference Shaw, Schaffer and Velie831); biochemistry of nucleic acid(Reference Kamen828); colon cancer risk(Reference Giovannucci, Rimm and Ascherio410, Reference Giovannucci832); anti-carcinogenic(Reference Jennings833, Reference Macgregor, Schlegel and Wehr834); megaloblastic anaemia(Reference Akilzhanova, Takamura and Aoyagi835); depression(Reference Coppen and Bolander-Gouaille274Reference Gilbody, Lightfoot and Sheldon276); fertility(Reference Ebisch, Thomas and Peters836); lipotrope(Reference Newberne and Rogers239); methylation and related epigenetic effects on gene expression(Reference Zeisel837)

Tocopherols and tocotrienols (vitamin E)(Reference Martin204):

Cardiovascular risk(Reference Gey838, Reference Leger839); antioxidant(Reference Bowry and Ingold840Reference Poulin, Cover and Gustafson842); Se and reduced state (cited in Slavin(Reference Slavin63)); formation of nitrosamines (cited in Slavin(Reference Slavin63)); formation of carcinogens (cited in Slavin et al. (Reference Slavin, Jacobs and Marquart663)); apoptosis(Reference Yu, Simmons-Menchaca and Gapor843)

Tocopherols:

Non-antioxidant effects(Reference Azzi and Stocker844); chemical inactivator (cited in Kohlmeier et al. (Reference Kohlmeier, Simonsen and Mottus93)); protein kinase C regulation(Reference Azzi and Stocker844, Reference Tasinato, Boscoboinik and Bartoli845); monocyte superoxide anion and IL-1(Reference Devaraj and Jialal846); gene expression and cell signalling(Reference Azzi and Stocker844, Reference Ricciarelli, Zingg and Azzi847, Reference Teupser, Thiery and Seidel848); peroxynitrite-derived nitrating species(Reference Christen, Woodall and Shigenaga849, Reference Wolf850); cell proliferation(Reference Chatelain, Boscoboinik and Bartoli851); pancreatic carcinogenesis(Reference Stolzenberg-Solomon, Sheffler-Collins and Weinstein852); type 2 diabetes-induced oxidative stress(Reference Laight, Desai and Gopaul853)

Tocotrienols(Reference Sen, Khanna and Roy347):

Neurodegeneration and immune responses(Reference Sen, Khanna and Roy347); cancer(Reference Nesaretnam, Yew and Wahid94, Reference Sen, Khanna and Roy347, Reference Chatelain, Boscoboinik and Bartoli851); cholesterol(Reference Sen, Khanna and Roy347); risk of heart disease; obesity and osteoporosis/bone calcification(Reference Ima-Nirwana and Suhaniza854, Reference Norazlina, Ima-Nirwana and Gapor855)

Phylloquinone (vitamin K)(Reference Martin204, 700, Reference Suttie856):

Coenzyme and formation of γ-carboxyglutamate residues(Reference Suttie857); osteoporosis(Reference Hodges, Akesson and Vergnaud858); atherosclerosis(Reference Luo, Ducy and McKee859)

β-Carotene:

Cancer(Reference Mayne860); colon cancer(Reference Alabaster, Tang and Shivapurkar106, Reference Baron, Cole and Mott861); lung cancer(Reference Holick, Michaud and Stolzenberg-Solomon862Reference Touvier, Kesse and Clavel-Chapelon864); tumour growth suppressor(Reference Rodriguez-Melendez and Zempleni824, Reference Rettura, Duttagupta and Listowsky865); apoptosis(Reference Cui, Lu and Bai866); immune function(Reference Prabhala, Braune and Garewal867); antioxidant(Reference Packer, Mahood and Mora-Arellano868); coronary artery disease risk(Reference Osganian, Stampfer and Rimm869)

Lutein (xanthophyll family)(Reference Granado, Olmedilla and Blanco870, Reference Stringheta, Nachtigall and Oliveira871):

Ocular function(Reference Richer, Stiles and Statkute872); age-related macular degeneration(Reference Seddon, Ajani and Sperduto873); cataract(Reference Olmedilla, Granado and Blanco874); macular pigment density(Reference Curran-Celentano, Hammond and Ciulla875); antioxidant(Reference Stringheta, Nachtigall and Oliveira871, 876, Reference Schäffer, Roy and Mukherjee877); CVD, stroke and lung cancer(Reference Holick, Michaud and Stolzenberg-Solomon862, Reference Michaud, Feskanich and Rimm863); skin protection(Reference Stahl and Sies878); colon cancer(Reference Slattery, Benson and Curtin879); atherosclerosis(Reference Dwyer, Navab and Dwyer880)

Zeaxanthin (xanthophyll family):

Age-related macular degeneration(Reference Seddon, Ajani and Sperduto873); cataract(Reference Yeum, Shang and Schalch881); macular pigment density(Reference Curran-Celentano, Hammond and Ciulla875); antioxidant(Reference Stringheta, Nachtigall and Oliveira871, 876, Reference Schäffer, Roy and Mukherjee877); CVD and stroke (cited in Anonymous(876)); skin protection(Reference Stahl and Sies878); lung cancer(Reference Holick, Michaud and Stolzenberg-Solomon862)

β-Cryptoxanthin:

Anabolic effects on bone components and bone loss/resorption(Reference Uchiyama, Sumida and Yamaguchi882, Reference Yamaguchi and Uchiyama883); anti-proliferative/chemopreventive agent and lung cancer(Reference Michaud, Feskanich and Rimm863, Reference Kohno, Taima and Sumida884Reference Yuan, Ross and Chu886); carcinogenesis(Reference Tanaka, Kohno and Murakami887); control of differentiation and apoptosis(Reference Nogushi, Sumida and Ogawa888); antioxidant (cited in Castelao & Olmedilla(Reference Castelao and Olmedilla889))

Phenolic acids:

Antioxidant(Reference Rice-Evans, Miller and Paganga890); insulin secretion(Reference Adisakwattana, Moonsan and Yibchok-anun891); plasma glucose, insulin, cholesterol and TAG (cited in Slavin et al. (Reference Slavin, Martini and Jacobs46)); cancer and action as blocking compounds(Reference Tanaka, Kojima and Kawamori892); carcinogens binding to targets and release of phenolic-bound antioxidant(Reference Vitaglione, Napolitano and Fogliano150, Reference Ferguson, Zhu and Harris893); tumour growth suppressor (cited in Slavin et al. (Reference Slavin, Martini and Jacobs46) and Thompson(Reference Thompson173)); enzyme modulators (cited in Slavin et al. (Reference Slavin, Martini and Jacobs46)); dyslipidaemia, hepatosteatosis and oxidative stress(Reference Hsu, Wu and Huang894); cell signalling(Reference Maggi-Capeyron, Ceballos and Cristol186, Reference Rahman, Biswas and Kirkham189)

Ferulic acid(Reference Barone, Calabrese and Mancuso104, Reference Ou and Kwok261, Reference Srinivasan, Sudheer and Menon262):

Antioxidant(Reference Sri Balasubashini, Rukkumani and Viswanathan895); HDL-cholesterol(Reference Kamal-Eldin, Frank and Razdan896); hyperlipidaemia(Reference Sri Balasubashini, Rukkumani and Menon897); anti-carcinogenic(Reference Ferguson and Harris69), for example, tongue cancer(Reference Tanaka, Kojima and Kawamori892); hypotensive and vascular relaxation(Reference Suzuki, Kagawa and Fujii898); hypoglycaemia(Reference Jung, Kim and Hwang899); neurodegenerative disorders (cited in Barone et al. (Reference Barone, Calabrese and Mancuso104))

Flavonoids:

Antioxidant(Reference Ferguson and Harris69, Reference Rice-Evans, Miller and Paganga890); enzyme modulator, antioxidant and tumour growth suppressor (cited in Kohlmeier et al. (Reference Kohlmeier, Simonsen and Mottus93)); anti-carcinogenic (cited in Ferguson & Harris(Reference Ferguson and Harris69) and Thompson(Reference Thompson173)); CVD(Reference Hollman and Katan900); signalling molecules(Reference Moskaug, Carlsen and Myhrstad188, Reference Rahman, Biswas and Kirkham189, Reference Williams, Spencer and Rice-Evans191); cell signalling, gene regulation, angiogenesis and other biological processes(Reference Lotito and Frei214); inflammation(Reference Rahman, Biswas and Kirkham189); platelet aggregation(Reference Van Wauwe and Goossens901); anti-microbial(Reference Cushnie and Lamb902); production of urate(Reference Lotito and Frei214); bone resorption(Reference Zhang, Qin and Hung903); dyslipidaemia, hepatosteatosis and oxidative stress(Reference Hsu, Wu and Huang894)

Anthocyanins:

Antioxidants(Reference Chiang, Wu and Yeh904Reference Nam, Choi and Kang906); anti-inflammatory(Reference Tsuda, Horio and Osawa907, Reference Xia, Ling and Ma908); anti-carcinogenic(Reference Hyun and Chung909, Reference Zhao, Giusti and Malik910); hypoglycaemic(Reference Tsuda, Horio and Uchida911)

Isoflavonoids:

Hormone-like diphenolic phyto-oestrogens(Reference Adlercreutz and Mazur293); cancer and atherosclerosis(Reference Adlercreutz and Mazur293); osteoporosis(Reference Adlercreutz and Mazur293); trabecular connectivity and thickness(Reference Kaludjerovic and Ward912)

Lignans:

Hormone-like diphenolic phyto-oestrogens(Reference Adlercreutz and Mazur293); antioxidant(Reference Liu18, Reference Slavin45, Reference Ferguson and Harris69, Reference Adlercreutz96); hormonally mediated diseases(Reference Adlercreutz and Mazur293); cell proliferation(Reference Adlercreutz, Mousavi and Clark97); tumour growth suppressor(Reference Jenab and Thompson913); precursors of enterolactone and enterodiol(Reference Adlercreutz96, Reference Oikarinen, Pajari and Mutanen914, Reference Prasad915); cancers(Reference Adlercreutz96); osteoporosis(Reference Adlercreutz and Mazur293); rheumatoid arthritis, gastric and duodenal ulcers, skin health, diuretic, antagonistic action of platelet-activating factor receptor and action on superoxide production (cited in Thompson(Reference Thompson173))

Alkylresorcinols(Reference Ross, Kamal-Eldin and Aman396):

Antioxidant(Reference Kamal-Eldin, Pouru and Eliasson916, Reference Kozubek and Nienartowicz917); anti-carcinogenic, anti-microbial, anti-parasitic and cytotoxic, structure and metabolism of nucleic acids, phospholipid bilayer properties(Reference Kozubek and Tyman400); anti-mutagenic(Reference Gasiorowski, Szyba and Brokos918); 3-phosphoglycerate dehydrogenase (key enzyme of TAG synthesis in adipocytes)(Reference Tsuge, Mizokami and Imai398); liver cholesterol(Reference Ross, Chen and Frank399)

Betaine:

Fatty deposits in the liver and hyperhomocysteinaemia(Reference Olthof, van Vliet and Boelsma919); osmoprotectant, performance (for example, athletic)(Reference Craig225); organic osmolyte(Reference Handler and Kwon920); CVD(Reference Konstantinova, Tell and Vollset921); homocysteine and inflammatory markers related to atherosclerosis (C-reactive protein and TNF-α)(Reference Detopoulou, Panagiotakos and Antonopoulou922, Reference Lv, Fan and Du923); sulfur amino acid homeostasis(Reference Delgado-Reyes and Garrow924); colorectal adenoma(Reference Cho, Willett and Colditz121); antioxidant and non-alcoholic fatty liver diseases(Reference Kwon, Jung and Kim925)

Choline(Reference Zeisel and Blusztajn226, 796):

Brain development and normal memory function(Reference Albright, Tsai and Friedrich926Reference Sanders and Zeisel928); plasma homocysteine level(Reference Cho, Zeisel and Jacques929); antioxidant(Reference Sachan, Hongu and Johnsen930); carnitine conservation(Reference Dodson and Sachan931); body fat and fatty acid oxidation(Reference Daily, Hongu and Mynatt932, Reference Sachan and Hongu933); precursor for the cell membrane phospholipids phosphatidylcholine(Reference Exton934), sphingomyelin(Reference Zeisel and Blusztajn226, Reference Hannun935), brain acetylcholine(Reference Cohen and Wurtman936) and for platelet-activating-factor formation(Reference Frenkel, Muguruma and Johnston937); synthesis and release of acetylcholine(Reference Cohen and Wurtman936, Reference Haubrich, Wedeking and Wang938); lipid metabolism, hepatic secretion of VLDL, nerve function and integrity of cell membranes(Reference Zeisel and Blusztajn226); neural tube development(Reference Zeisel939); lipotrope and methyl donor(Reference Zeisel, Da Costa and Franklin240); DNA hypomethylation and tumour development in the liver(Reference Zeisel and Blusztajn226, Reference Newberne and Rogers239, Reference Locker, Reddy and Lombardi258, Reference Henning and Swendseid940); epigenetic regulator of gene expression(Reference Niculescu, Craciunescu and Zeisel941)

Phytosterols(Reference Liu18, Reference Brufau, Canela and Rafecas942, Reference de Jong, Plat and Mensink943):

Total and LDL serum cholesterol(Reference Brufau, Canela and Rafecas942, Reference Batta, Xu and Bollineni944Reference Yankah and Jones947); micelle formation, dietary and biliary cholesterol absorption and LDL-cholesterol(Reference Demonty, Ras and van der Knaap948); vascular smooth muscle cell hyperproliferation(Reference Awad, Smith and Fink949); immunosuppression associated with excessive physical stress(Reference Bouic, Clark and Lamprecht950); anti-inflammatory, anti-pyretic, immunomodulator and anti-diabetic (cited in Brufau et al. (Reference Brufau, Canela and Rafecas942)); anti-diabetic and hypoglycaemic(Reference Tanaka, Misawa and Ito951)

β-Sitosterol:

Growth of colon cancer line(Reference Awad, Chen and Fink952, Reference Raicht, Cohen and Fazzini953); prostate cancer(Reference Berges, Windeler and Trampisch954); carcinogen-induced neoplasia (cited in Wattenberg(Reference Wattenberg110)); apoptosis(Reference Rubis, Paszel and Kaczmarek955) through caspase activation(Reference Awad, Roy and Fink956)

Inositols:

Chiro-inositol:

Insulin, signal transduction and mimetic of insulin action(Reference Larner957); type 2 diabetes(Reference Kim, Kim and Joo245, Reference Asplin, Galasko and Larner958Reference Sun, Heimark and Nguygen961); ovulatory functions and serum androgen concentrations, blood pressure and plasma TAG(Reference Nestler, Jakubowicz and Reamer621); folate-resistant neural tube defects(Reference Cogram, Tesh and Tesh962); pinitol and glucose metabolism(Reference Campbell, Haub and Fluckey622)

Myo-inositol:

Metabolism(Reference Holub963); TAG and total lipid liver, hepatic activities of glucose-6-phosphate dehydrogenase, malic enzyme, fatty acid synthetase and citrate cleavage enzyme(Reference Okazaki, Setoguchi and Katayama242, Reference Katayama964); conversion into chiro-inositol and precursor for several phospholipids (cited in Larner(Reference Larner957), Novak et al. (Reference Novak, Scott Turner and Agranoff965) and Pak et al. (Reference Pak, Huang and Lilley256)); mental health(Reference Fux, Levine and Aviv966, Reference Palatnik, Frolov and Fux967); osmotic demyelination syndrome(Reference Silver, Schroeder and Sterns968); volume regulation during persistent osmotic stress(Reference Nakanishi, Turner and Burg969); cancer(Reference Vucenik and Shamsuddin686); diabetic polyneuropathy and nerve conduction(Reference Greene and Lattimer970); intestinal lipodystrophy(Reference Holub963)

Policosanol:

Octacosanol in human health(Reference Taylor, Rapport and Lockwood302); CVD(Reference Varady, Wang and Jones304); lipid, cholesterol and LDL(Reference Gouni-Berthold and Berthold303, Reference Menendez, Arruzazabala and Mas306, Reference Castano, Mas and Fernandez971Reference Menendez, Fernandez and Del Rio973); cholesterol biosynthesis and LDL catabolism(Reference Menendez, Fernandez and Del Rio973); hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase(Reference McCarty974); LDL peroxidation(Reference Menendez, Fraga and Amor307); membrane lipid peroxidation(Reference Fraga, Menendez and Amor975); lipid metabolism(Reference Kato, Karino and Hasegawa301); platelet aggregation and thromboxane generation, endothelial damage and foam cell formation(Reference Arruzazabala, Molina and Mas976, Reference Carbajal, Arruzazabala and Valdés977); cytoprotection in gastric ulcer(Reference Carbajal, Molina and Valdes978); athletic performances(Reference Saint-John and McNaughton979); cardiac events, and cholesterol and anti-aggregatory effects (cited in Taylor et al. (Reference Taylor, Rapport and Lockwood302)); smooth muscle cell proliferation(Reference Noa, Más and Mesa980); anti-fatigue drug(Reference Taylor, Rapport and Lockwood302)

Melatonin(Reference Ferrari981):

Mood, happiness, sleep and brain neuromodulation in Alzheimer's disease(Reference Asayama, Yamadera and Ito309, Reference Maurizi310); antioxidant(Reference Reiter982, Reference Reiter, Tang and Garcia983); corticoid receptor(Reference Sainz, Mayo and Reiter984); scavenger of hydroxyl radicals(Reference Hardeland, Reiter and Poeggeler985); brain GSH peroxidase activity(Reference Barlow-Walden, Reiter and Abe986); gene expression for antioxidant enzyme(Reference Kotler, Rodríguez and Sáinz987); sleep–wake regulation(Reference Asayama, Yamadera and Ito309, Reference Zhdanova, an and Morabito988); DNA damage(Reference Reiter, Melchiorri and Sewerynek989); lifespan(Reference Anisimov, Zavarzina and Zabezhinski990); oncostatic role and anti-proliferative effect(Reference Garcia-Navarro, Gonzalez-Puga and Escames311, Reference Shiu312); cancers(Reference Srinivasan, Spence and Pandi-Perumal991)

para-Aminobenzoic acid (PABA):

Acetylation in blood and other tissues(Reference Wang, Huang and Tai315, Reference Barbieri, Papadogiannakis and Eneroth321, Reference Hearse and Weber992, Reference Lindsay, McLaren and Baty993); peroxisomal β-oxidation and PABA acetylation(Reference Barbieri, Papadogiannakis and Eneroth316); N-acetyltransferase regulation(Reference Butcher, Ilett and Minchin994); acetylation(Reference Hein, Doll and Gray319, Reference Minchin, Reeves and Teitel320); rickettsial infections and collagen diseases(Reference Failey and Childress995); serum cholesterol(Reference Failey and Childress995); folate formation(Reference Barbieri, Papadogiannakis and Eneroth316); treatment of vitiligo, leukaemia, rheumatic fever and in rickettsial diseases(cited in Barbieri et al. (Reference Barbieri, Papadogiannakis and Eneroth316)); production of thromboxane(Reference Barbieri, Papadogiannakis and Eneroth321); anti-aggregatory(Reference Barbieri, Stain-Malmgren and Papadogiannakis996); UV protection of the skin (cited in Barbieri et al. (Reference Barbieri, Stain-Malmgren and Papadogiannakis996) and Wang et al. (Reference Wang, Huang and Tai315)); liver folic acid metabolism (cited in Russell et al. (Reference Russell, Craig and Rawlings997))

γ-Oryzanol(Reference Cicero and Gaddi361):

Cholesterol and rice bran oil(Reference Berger, Rein and Schafer998); cholesterol absorption and aortic fatty streaks(Reference Wilson, Nicolosi and Woolfrey348, Reference Rong, Ausman and Nicolosi358); lipid metabolism(Reference Ishihara, Ito and Nakakita999); autonomic nervous unbalance and menopausal troubles (climacteric disturbance)(Reference Ishihara, Ito and Nakakita999, Reference Ishihara1000); anti-ulcerogenic(Reference Itaya, Kiyonaga and Ishikawa1001); antioxidant(Reference Juliano, Cossu and Alamanni357, Reference Suh, Yoo and Chang360, Reference Parrado, Miramontes and Jover1002); gene expression and oxidative stress(Reference Lee, Pugh and Klopp1003); glycaemia control(Reference Eason, Archer and Akhtar1004, Reference Khanna, Roy and Packer1005); platelet aggregation(Reference Seetharamaiah, Krishnakantha and Chandrasekhara1006); anxiety and stress ulcer(Reference Itaya, Kiyonaga and Ishikawa1001, Reference Cai, Bi and Zhao1007Reference Jabeen, Badaruddin and Ali1009)

Avenanthramides:

Anti-inflammatory and anti-atherogenic(Reference Liu, Zubik and Collins368); smooth muscle cell proliferation and NO production(Reference Nie, Wise and Peterson1010, Reference Nie, Wise and Peterson1011); antioxidant(Reference Chen, Milbury and Collins140, Reference Peterson, Hahn and Emmons367, Reference Chen, Milbury and Kwak369)

Saponins(Reference Güçlü-Üstündag and Mazza370, Reference Francis, Kerem and Makkar1012, Reference Mimaki, Yokosuka and Kuroda1013):

Hypercholesterolaemia(Reference Thompson173, Reference Matsuura374, Reference Oakenfull, Fenwick and Hood377, Reference Potter, Jimenez-Flores and Pollack1014); lipase activity and fat absorption(Reference Han, Xu and Kimura1015); transcriptional activity of Cu,Zn-SOD gene(Reference Kim, Park and Rho1016); scavenger and superoxides(Reference Yoshiki and Okubo1017); hypoglycaemia(Reference Arai, Osawa and Ohigashi1018, Reference Jie1019); gastric emptying rate and glucose transport across the brush border of the small intestine(Reference Arai, Osawa and Ohigashi1018, Reference Matsuda, Li and Murakami1020); anti-fungal(Reference Matsuura374); anti-viral(Reference Sindambiwe, Calomme and Geerts1021); diabetes(Reference Xi, Hai and Tang1022); anti-inflammatory(Reference Jie1019); anti-carcinogenic(Reference Matsuura374); tumour growth and cytostatic effect(Reference Mimaki, Yokosuka and Kuroda1013, Reference Cai, Liu and Wang1023Reference Podolak, Elas and Cieszka1027); bile acid binding (cited in Mimaki et al. (Reference Mimaki, Yokosuka and Kuroda1013)); cell-mediated immune system and antibody production(Reference Barr, Sjölander and Cox375); nervous system functioning(Reference Francis, Kerem and Makkar1012, Reference Zhang, Dou and Zhang1028); blood coagulation(Reference Peng, Chen and Qiao1029)

References

1Koh-Banerjee, P & Rimm, EB (2003) Whole grain consumption and weight gain: a review of the epidemiological evidence, potential mechanisms and opportunities for future research. Proc Nutr Soc 62, 2529.Google ScholarPubMed
2van de Vijver, LPL, van den Bosch, LMC, van den Brandt, PA, et al. . (2009) Whole-grain consumption, dietary fibre intake and body mass index in the Netherlands cohort study. Eur J Clin Nutr 63, 3138.CrossRefGoogle ScholarPubMed
3Esmaillzadeh, A, Mirmiran, P & Azizi, F (2005) Whole-grain consumption and the metabolic syndrome: a favorable association in Tehranian adults. Eur J Clin Nutr 59, 353362.CrossRefGoogle ScholarPubMed
4Sahyoun, NR, Jacques, PF, Zhang, XL, et al. . (2006) Whole-grain intake is inversely associated with the metabolic syndrome and mortality in older adults. Am J Clin Nutr 83, 124131.CrossRefGoogle ScholarPubMed
5de Munter, JS, Hu, FB, Spiegelman, D, et al. . (2007) Whole grain, bran, and germ intake and risk of type 2 diabetes: a prospective cohort study and systematic review. PLoS Med 4, e261.CrossRefGoogle ScholarPubMed
6Murtaugh, MA, Jacobs, DR, Jacob, B, et al. . (2007) Epidemiological support for the protection of whole grains against diabetes. Proc Nutr Soc 62, 143149.CrossRefGoogle Scholar
7Mellen, PB, Walsh, TF & Herrington, DM (2008) Whole grain intake and cardiovascular disease: a meta-analysis. Nutr Metab Cardiovasc Dis 18, 283290.CrossRefGoogle ScholarPubMed
8Chan, JM, Wang, F & Holly, EA (2007) Whole grains and risk of pancreatic cancer in a large population-based case–control study in the San Francisco Bay area, California. Am J Epidemiol 166, 11741185.CrossRefGoogle Scholar
9Chatenoud, L, Tavani, A, La Vecchia, C, et al. . (1998) Whole grain food intake and cancer risk. Int J Cancer 77, 2428.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
10Jacobs, DR Jr, Marquart, L, Slavin, J, et al. . (1998) Whole-grain intake and cancer: an expanded review and meta-analysis. Nutr Cancer 30, 8596.CrossRefGoogle ScholarPubMed
11Larsson, SC, Giovannucci, E, Bergkvist, L, et al. . (2005) Whole grain consumption and risk of colorectal cancer: a population-based cohort of 60 000 women. Br J Cancer 92, 18031807.CrossRefGoogle Scholar
12Schatzkin, A, Park, Y, Leitzmann, MF, et al. . (2008) Prospective study of dietary fiber, whole grain foods, and small intestinal cancer. Gastroenterology 135, 11631167.CrossRefGoogle ScholarPubMed
13Jacobs, DR Jr, Andersen, LF & Blomhoff, R (2007) Whole-grain consumption is associated with a reduced risk of noncardiovascular, noncancer death attributed to inflammatory diseases in the Iowa Women's Health Study. Am J Clin Nutr 85, 16061614.CrossRefGoogle ScholarPubMed
14Adom, KK, Sorrells, ME & Liu, RH (2003) Phytochemical profiles and antioxidant activity of wheat varieties. J Agric Food Chem 51, 78257834.CrossRefGoogle ScholarPubMed
15Jacobs, DR, Meyer, HE & Solvoll, K (2001) Reduced mortality among whole grain bread eaters in men and women in the Norwegian County Study. Eur J Clin Nutr 55, 137143.CrossRefGoogle ScholarPubMed
16Chatenoud, L, La Vecchia, C, Franceschi, S, et al. . (1999) Refined-cereal intake and risk of selected cancers in Italy. Am J Clin Nutr 70, 11071110.CrossRefGoogle ScholarPubMed
17Jensen, MK, Koh-Banerjee, P, Franz, M, et al. . (2006) Whole grains, bran, and germ in relation to homocysteine and markers of glycemic control, lipids, and inflammation. Am J Clin Nutr 83, 275283.CrossRefGoogle ScholarPubMed
18Liu, RH (2007) Whole grain phytochemicals and health. J Cereal Sci 46, 207219.CrossRefGoogle Scholar
19Truswell, AS (2002) Cereal grains and coronary heart disease. Eur J Clin Nutr 56, 114.CrossRefGoogle ScholarPubMed
20AACC International (1999) Definition of whole grain. http://www.aaccnet.org/definitions/wholegrain.asp (accessed January 2010).Google Scholar
21European Food Information Council (EUFIC) (2009) Whole grain fact sheet. http://www.eufic.org/article/en/page/BARCHIVE/expid/Whole-grain-Fact-Sheet/ (accessed January 2010).Google Scholar
22Food and Drug Adminstration (2006) Draft Guidance for Industry and FDA Staff: Whole Grain Label Statements. http://www.fda.gov/Food/GuidanceComplianceRegulatoryInformation/GuidanceDocuments/FoodLabelingNutrition/UCM059088 (accessed January 2010).Google Scholar
23AACC International (2006) AACC International comments on Part III of the Draft Guidance on Whole Grain Label Statements. http://www.aaccnet.org/definitions/pdfs/AACCIntlWholeGrainComments.pdf (accessed January 2010).Google Scholar
24Jones, JM (2008) Whole grains – issues and deliberations from the Whole Grain Task Force. Cereal Foods World 53, 260264.Google Scholar
25Srivastava, AK, Sudha, ML, Baskaran, V, et al. . (2007) Studies on heat stabilized wheat germ and its influence on rheological characteristics of dough. Eur Food Res Technol 224, 365372.CrossRefGoogle Scholar
26Food and Drug Administration (1999) Health Claim Notification for Whole Grain Foods. http://www.fda.gov/Food/LabelingNutrition/LabelClaims/FDAModernizationActFDAMAClaims/UCM073639.htm (accessed January 2010).Google Scholar
27Jacobs, DR Jr, Meyer, KA, Kushi, LH, et al. . (1998) Whole-grain intake may reduce the risk of ischemic heart disease death in postmenopausal women: the Iowa Women's Health Study. Am J Clin Nutr 68, 248257.CrossRefGoogle ScholarPubMed
28Liu, SM, Manson, JE, Stampfer, MJ, et al. . (2000) Whole grain consumption and risk of ischemic stroke in women – a prospective study. JAMA 284, 15341540.CrossRefGoogle ScholarPubMed
29Thane, CW, Jones, AR, Stephen, AM, et al. . (2005) Whole-grain intake of British young people aged 4–18 years. Br J Nutr 94, 825831.CrossRefGoogle ScholarPubMed
30Thane, CW, Stephen, AM & Jebb, SA (2009) Whole grains and adiposity: little association among British adults. Eur J Clin Nutr 63, 229237.CrossRefGoogle ScholarPubMed
31Cleveland, LE, Moshfegh, AJ, Albertson, AM, et al. . (2000) Dietary intake of whole grains. J Am Coll Nutr 19, 331S338S.CrossRefGoogle ScholarPubMed
32Lang, R & Jebb, SA (2003) Who consumes whole grains, and how much? Proc Nutr Soc 62, 123127.CrossRefGoogle ScholarPubMed
33Welsh, S, Shaw, A & Davis, C (1994) Achieving dietary recommendations – whole-grain foods in the food guide pyramid. Crit Rev Food Sci Nutr 34, 441451.CrossRefGoogle ScholarPubMed
34Albertson, AM & Tobelmann, RC (1995) Consumption of grain and whole-grain foods by an American population during the years 1990 to 1992. J Am Diet Assoc 95, 703704.CrossRefGoogle ScholarPubMed
35National Council on Nutrition and Physical Activity and the Institute for Nutrition Research (1998) Development of the Norwegian Diet. Oslo: University of Oslo.Google Scholar
36Prättälä, R, Helasoja, V & Mykkänen, H (2007) The consumption of rye bread and white bread as dimensions of health lifestyles in Finland. Public Health Nutr 4, 813819.CrossRefGoogle Scholar
37Adams, JF & Engstrom, A (2000) Helping consumers achieve recommended intakes of whole grain foods. J Am Coll Nutr 19, 339S344S.CrossRefGoogle ScholarPubMed
38Jenkins, DJ, Wesson, V, Wolever, TM, et al. . (1988) Wholemeal versus wholegrain breads: proportion of whole or cracked grain and the glycaemic response. BMJ 297, 958960.CrossRefGoogle ScholarPubMed
39Jacobs, DR Jr, Pereira, MA, Stumpf, K, et al. . (2002) Whole grain food intake elevates serum enterolactone. Br J Nutr 88, 111116.CrossRefGoogle ScholarPubMed
40Lutsey, PL, Jacobs, DR, Kori, S, et al. . (2007) Whole grain intake and its cross-sectional association with obesity, insulin resistance, inflammation, diabetes and subclinical CVD: The MESA Study. Br J Nutr 98, 397405.CrossRefGoogle ScholarPubMed
41McKeown, NM, Meigs, JB, Liu, S, et al. . (2002) Whole-grain intake is favorably associated with metabolic risk factors for type 2 diabetes and cardiovascular disease in the Framingham Offspring Study. Am J Clin Nutr 76, 390398.CrossRefGoogle ScholarPubMed
42Newby, P, Maras, J, Bakun, P, et al. . (2007) Intake of whole grains, refined grains, and cereal fiber measured with 7-d diet records and associations with risk factors for chronic disease. Am J Clin Nutr 86, 17451753.CrossRefGoogle ScholarPubMed
43Vanharanta, M, Voutilainen, S, Lakka, TA, et al. . (1999) Risk of acute coronary events according to serum concentrations of enterolactone: a prospective population-based case–control study. Lancet 354, 21122115.CrossRefGoogle ScholarPubMed
44Levi, F, Pasche, C, Lucchini, F, et al. . (2000) Refined and whole grain cereals and the risk of oral, oesophageal and laryngeal cancer. Eur J Clin Nutr 54, 487489.CrossRefGoogle ScholarPubMed
45Slavin, JL (2000) Mechanisms for the impact of whole grain foods on cancer risk. J Am Coll Nutr 19, 300S307S.CrossRefGoogle ScholarPubMed
46Slavin, JL, Martini, MC, Jacobs, DR Jr, et al. . (1999) Plausible mechanisms for the protectiveness of whole grains. Am J Clin Nutr 70, 459S463S.CrossRefGoogle ScholarPubMed
47Haber, GB, Heaton, KW, Murphy, D, et al. . (1977) Depletion and disruption of dietary fibre. Effects on satiety, plasma-glucose, and serum-insulin. Lancet ii, 679682.CrossRefGoogle Scholar
48Read, NW, Welch, IM, Austen, CJ, et al. . (1986) Swallowing food without chewing; a simple way to reduce postprandial glycaemia. Br J Nutr 55, 4347.CrossRefGoogle ScholarPubMed
49Fardet, A, Leenhardt, F, Lioger, D, et al. . (2006) Parameters controlling the glycaemic response to breads. Nutr Res Rev 19, 1825.CrossRefGoogle ScholarPubMed
50Granfeldt, Y, Hagander, B & Bjorck, I (1995) Metabolic responses to starch in oat and wheat products. On the importance of food structure, incomplete gelatinization or presence of viscous dietary fibre. Eur J Clin Nutr 49, 189199.Google ScholarPubMed
51Liljeberg, H, Granfeldt, Y & Bjorck, I (1992) Metabolic responses to starch in bread containing intact kernels versus milled flour. Eur J Clin Nutr 46, 561575.Google ScholarPubMed
52Nilsson, AC, Ostman, EM, Granfeldt, Y, et al. . (2008) Effect of cereal test breakfasts differing in glycemic index and content of indigestible carbohydrates on daylong glucose tolerance in healthy subjects. Am J Clin Nutr 87, 645654.CrossRefGoogle ScholarPubMed
53American Association of Cereal Chemists (2001) The definition of dietary fiber. Cereal Foods World 46, 112129.Google Scholar
54Nilsson, AC, Ostman, EM, Holst, JJ, et al. . (2008) Including indigestible carbohydrates in the evening meal of healthy subjects improves glucose tolerance, lowers inflammatory markers, and increases satiety after a subsequent standardized breakfast. J Nutr 138, 732739.CrossRefGoogle ScholarPubMed
55Topping, D (2007) Cereal complex carbohydrates and their contribution to human health. J Cereal Sci 46, 220229.CrossRefGoogle Scholar
56Wood, PJ (2007) Cereal β-glucans in diet and health. J Cereal Sci 46, 230238.CrossRefGoogle Scholar
57Koh-Banerjee, P, Franz, M, Sampson, L, et al. . (2004) Changes in whole-grain, bran, and cereal fiber consumption in relation to 8-y weight gain among men. Am J Clin Nutr 80, 12371245.CrossRefGoogle ScholarPubMed
58Slavin, JL (2005) Dietary fiber and body weight. Nutrition 21, 411418.CrossRefGoogle ScholarPubMed
59Jenkins, AL, Jenkins, DJ, Zdravkovic, U, et al. . (2002) Depression of the glycemic index by high levels of β-glucan fiber in two functional foods tested in type 2 diabetes. Eur J Clin Nutr 56, 622628.CrossRefGoogle ScholarPubMed
60Zhang, JX, Hallmans, G, Andersson, H, et al. . (1992) Effect of oat bran on plasma cholesterol and bile acid excretion in nine subjects with ileostomies. Am J Clin Nutr 56, 99105.CrossRefGoogle ScholarPubMed
61Lia, A, Hallmans, G, Sandberg, AS, et al. . (1995) Oat β-glucan increases bile acid excretion and a fiber-rich barley fraction increases cholesterol excretion in ileostomy subjects. Am J Clin Nutr 62, 12451251.CrossRefGoogle Scholar
62Scheppach, W, Bartram, HP & Richter, F (1995) Role of short-chain fatty acids in the prevention of colorectal cancer. Eur J Cancer 31, 10771080.CrossRefGoogle Scholar
63Slavin, J (2003) Why whole grains are protective: biological mechanisms. Proc Nutr Soc 62, 129134.CrossRefGoogle ScholarPubMed
64Costabile, A, Klinder, A, Fava, F, et al. . (2008) Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: a double-blind, placebo-controlled, crossover study. Br J Nutr 99, 110120.CrossRefGoogle Scholar
65Brouns, F, Kettlitz, B & Arrigoni, E (2002) Resistant starch and ‘the butyrate revolution’. Trends Food Sci Technol 13, 251261.CrossRefGoogle Scholar
66Boffa, LC, Lupton, JR, Mariani, MR, et al. . (1992) Modulation of colonic epithelial cell proliferation, histone acetylation, and luminal short chain fatty acids by variation of dietary fiber (wheat bran) in rats. Cancer Res 52, 59065912.Google ScholarPubMed
67Higgins, J, Higbee, D, Donahoo, W, et al. . (2004) Resistant starch consumption promotes lipid oxidation. Nutr Metab 1, 8.CrossRefGoogle ScholarPubMed
68McIntosh, GH, Noakes, M, Royle, PJ, et al. . (2003) Whole-grain rye and wheat foods and markers of bowel health in overweight middle-aged men. Am J Clin Nutr 77, 967974.CrossRefGoogle ScholarPubMed
69Ferguson, LR & Harris, PJ (1999) Protection against cancer by wheat bran: role of dietary fibre and phytochemicals. Eur J Cancer Prev 8, 1725.CrossRefGoogle ScholarPubMed
70Lopez, HW, Coudray, C, Bellanger, J, et al. . (2000) Resistant starch improves mineral assimilation in rats adapted to a wheat bran diet. Nutr Res 20, 141155.CrossRefGoogle Scholar
71Lopez, HW, Levrat-Verny, MA, Coudray, C, et al. . (2001) Class 2 resistant starches lower plasma and liver lipids and improve mineral retention in rats. J Nutr 131, 12831289.CrossRefGoogle ScholarPubMed
72Coudray, C, Bellanger, J, Castiglia-Delavaud, C, et al. . (1997) Effect of soluble or partly soluble dietary fibres supplementation on absorption and balance of calcium, magnesium, iron and zinc in healthy young men. Eur J Clin Nutr 51, 375380.CrossRefGoogle ScholarPubMed
73Lopez, HW, Coudray, C, Levrat-Verny, MA, et al. . (2000) Fructooligosaccharides enhance mineral apparent absorption and counteract the deleterious effects of phytic acid on mineral homeostasis in rats. J Nutr Biochem 11, 500508.CrossRefGoogle ScholarPubMed
74Reddy, B, Hamid, R & Rao, C (1997) Effect of dietary oligofructose and inulin on colonic preneoplastic aberrant crypt foci inhibition. Carcinogenesis 18, 13711374.CrossRefGoogle ScholarPubMed
75McIntyre, A, Gibson, PR & Young, GP (1993) Butyrate production from dietary fiber and protection against large bowel cancer in a rat model. Gut 34, 386391.CrossRefGoogle ScholarPubMed
76Heerdt, BG, Houston, MA, Anthony, GM, et al. . (1999) Initiation of growth arrest and apoptosis of MCF-7 mammary carcinoma cells by tributyrin, a triglyceride analogue of the short-chain fatty acid butyrate, is associated with mitochondrial activity. Cancer Res 59, 15841591.Google ScholarPubMed
77Ellerhorst, J, Nguyen, T, Cooper, DNW, et al. . (1999) Induction of differentiation and apoptosis in the prostate cancer cell line LNCaP by sodium butyrate and galectin-1. Int J Oncol 14, 225232.Google ScholarPubMed
78Bird, AR, Vuaran, MS, King, RA, et al. . (2008) Wholegrain foods made from a novel high-amylose barley variety (Himalaya 292) improve indices of bowel health in human subjects. Br J Nutr 99, 10321040.CrossRefGoogle ScholarPubMed
79Liljeberg, H & Bjorck, I (1994) Bioavailability of starch in bread products. Postprandial glucose and insulin responses in healthy subjects and in vitro resistant starch content. Eur J Clin Nutr 48, 151163.Google ScholarPubMed
80Hara, H, Haga, S, Aoyama, Y, et al. . (1999) Short-chain fatty acids suppress cholesterol synthesis in rat liver and intestine. J Nutr 129, 942948.CrossRefGoogle ScholarPubMed
81Jenkins, DJA, Wolever, TMS, Nineham, R, et al. . (1980) Improved glucose tolerance four hours after taking guar with glucose. Diabetologia 19, 2124.CrossRefGoogle ScholarPubMed
82Jenkins, D, Wolever, T, Taylor, R, et al. . (1982) Slow release dietary carbohydrate improves second meal tolerance. Am J Clin Nutr 35, 13391346.CrossRefGoogle ScholarPubMed
83Nilsson, A, Granfeldt, Y, Ostman, E, et al. . (2006) Effects of GI and content of indigestible carbohydrates of cereal-based evening meals on glucose tolerance at a subsequent standardised breakfast. Eur J Clin Nutr 60, 10921099.CrossRefGoogle Scholar
84Cherbut, C (2003) Motor effects of short-chain fatty acids and lactate in the gastrointestinal tract. Proc Nutr Soc 62, 9599.CrossRefGoogle ScholarPubMed
85Wolever, T, Spadafora, P & Eshuis, H (1991) Interaction between colonic acetate and propionate in humans. Am J Clin Nutr 53, 681687.CrossRefGoogle ScholarPubMed
86Homko, CJ, Cheung, P & Boden, G (2003) Effects of free fatty acids on glucose uptake and utilization in healthy women. Diabetes 52, 487491.CrossRefGoogle ScholarPubMed
87Anderson, JW & Bridges, SR (1984) Short-chain fatty acid fermentation products of plant fiber affect glucose metabolism of isolated rat hepatocytes. Proc Soc Exp Biol Med 177, 372376.CrossRefGoogle ScholarPubMed
88Liu, RH (2004) Potential synergy of phytochemicals in cancer prevention: mechanism of action. J Nutr 134, 3479S3485S.CrossRefGoogle ScholarPubMed
89Reddy, BS, Hirose, Y, Cohen, LA, et al. . (2000) Preventive potential of wheat bran fractions against experimental colon carcinogenesis: implications for human colon cancer prevention. Cancer Res 60, 47924797.Google ScholarPubMed
90Sang, SM, Ju, JY, Lambert, JD, et al. . (2006) Wheat bran oil and its fractions inhibit human colon cancer cell growth and intestinal tumorigenesis in Apc(min/+) mice. J Agric Food Chem 54, 97929797.CrossRefGoogle ScholarPubMed
91Bartsch, H & Nair, J (2006) Chronic inflammation and oxidative stress in the genesis and perpetuation of cancer: role of lipid peroxidation, DNA damage, and repair. Langenbecks Arch Surg 391, 499510.CrossRefGoogle ScholarPubMed
92Graf, E & Eaton, JW (1985) Dietary suppression of colonic cancer; fiber or phytate? Cancer 56, 717718.3.0.CO;2-4>CrossRefGoogle ScholarPubMed
93Kohlmeier, L, Simonsen, N & Mottus, K (1995) Dietary modifiers of carcinogenesis. Environ Health Perspect 103, Suppl. 8, 177184.Google ScholarPubMed
94Nesaretnam, K, Yew, WW & Wahid, MB (2007) Tocotrienols and cancer: beyond antioxidant activity. Eur J Lipid Sci Technol 109, 445452.CrossRefGoogle Scholar
95Shamsuddin, AM (2002) Anti-cancer function of phytic acid. Int J Food Sci Technol 37, 769782.CrossRefGoogle Scholar
96Adlercreutz, H (2002) Phyto-oestrogens and cancer. Lancet Oncol 3, 364373.CrossRefGoogle ScholarPubMed
97Adlercreutz, H, Mousavi, Y, Clark, J, et al. . (1992) Dietary phytoestrogens and cancer: in vitro and in vivo studies. J Steroid Biochem Molec Biol 41, 331337.CrossRefGoogle ScholarPubMed
98Markaverich, BM, Webb, B, Densmore, CL, et al. . (1995) Effects of coumestrol on estrogen receptor function and uterine growth in ovariectomized rats. Environ Health Perspect 103, 574581.CrossRefGoogle ScholarPubMed
99Reddy, BS (1999) Prevention of colon carcinogenesis by components of dietary fiber. Anticancer Res 19, 36813683.Google ScholarPubMed
100Ullah, A & Shamsuddin, AM (1990) Dose-dependent inhibition of large intestinal cancer by inositol hexaphosphate in F344 rats. Carcinogenesis 11, 22192222.CrossRefGoogle ScholarPubMed
101Vucenik, I, Yang, G & Shamsuddin, AM (1997) Comparison of pure inositol hexaphosphate and high-bran diet in the prevention of DMBA-induced rat mammary carcinogenesis. Nutr Cancer 28, 713.CrossRefGoogle ScholarPubMed
102Hollman, P & Katan, M (1997) Absorption, metabolism and health effects of dietary flavonoids in man. Biomed Pharmacother 51, 305310.CrossRefGoogle ScholarPubMed
103Edenharder, R, Rauscher, R & Platt, KL (1997) The inhibition by flavonoids of 2-amino-3-methylimidazo[4,5-f]quinoline metabolic activation to a mutagen: a structure–activity relationship study. Mutat Res 379, 2132.CrossRefGoogle Scholar
104Barone, E, Calabrese, V & Mancuso, C (2009) Ferulic acid and its therapeutic potential as a hormetin for age-related diseases. Biogerontology 10, 97108.CrossRefGoogle ScholarPubMed
105Kawabata, K, Yamamoto, T, Hara, A, et al. . (2000) Modifying effects of ferulic acid on azoxymethane-induced colon carcinogenesis in F344 rats. Cancer Lett 157, 1521.CrossRefGoogle ScholarPubMed
106Alabaster, O, Tang, ZC & Shivapurkar, N (1996) Dietary fiber and the chemopreventive modelation of colon carcinogenesis. Mutat Res 350, 185197.CrossRefGoogle ScholarPubMed
107Eastwood, MA & Girdwood, RH (1968) Lignin: a bile-salt sequestrating agent. Lancet ii, 11701172.CrossRefGoogle Scholar
108Eastwood, MA & Hamilton, D (1968) Studies on the adsorption of bile salts to non-absorbed components of diet. Biochim Biophys Acta 152, 165173.CrossRefGoogle ScholarPubMed
109Labaj, J, Slamenova, D, Lazarova, M, et al. . (2004) Lignin-stimulated reduction of oxidative DNA lesions in testicular cells and lymphocytes of Sprague–Dawley rats in vitro and ex vivo. Nutr Cancer 50, 198205.CrossRefGoogle ScholarPubMed
110Wattenberg, LW (1985) Chemoprevention of cancer. Cancer Res 45, 18.Google ScholarPubMed
111Jablonska, E, Gromadzinska, J, Sobala, W, et al. . (2008) Joint effect of GPx1 polymorphism and selenium status on lung cancer risk. Eur J Cancer Suppl 6, 203.CrossRefGoogle Scholar
112Stavric, B (1994) Antimutagens and anticarcinogens in foods. Food Chem Toxicol 32, 7990.CrossRefGoogle ScholarPubMed
113Harris, PJ & Ferguson, LR (1993) Dietary fiber – its composition and role in protection against colorectal cancer. Mutat Res 290, 97110.CrossRefGoogle ScholarPubMed
114Harris, PJ, Roberton, AM, Watson, ME, et al. . (1993) The effects of soluble-fiber polysaccharides on the adsorption of a hydrophobic carcinogen to an insoluble dietary fiber. Nutr Cancer 19, 4354.CrossRefGoogle Scholar
115Morita, T, Tanabe, H, Sugiyama, K, et al. . (2004) Dietary resistant starch alters the characteristics of colonic mucosa and exerts a protective effect on trinitrobenzene sulfonic acid-induced colitis in rats. Biosci Biotechnol Biochem 68, 21552164.CrossRefGoogle ScholarPubMed
116Toden, S, Bird, AR, Topping, DL, et al. . (2007) Dose-dependent reduction of dietary protein-induced colonocyte DNA damage by resistant starch in rats correlates more highly with caecal butyrate than with other short chain fatty acids. Cancer Biol Ther 6, 253258.CrossRefGoogle ScholarPubMed
117Story, JA & Kritchevsky, D (1994) Denis Parsons Burkitt (1911–1993). J Nutr 124, 15511554.CrossRefGoogle ScholarPubMed
118Bauer-Marinovic, M, Florian, S, Muller-Schmehl, K, et al. . (2006) Dietary resistant starch type 3 prevents tumor induction by 1,2-dimethylhydrazine and alters proliferation, apoptosis and dedifferentiation in rat colon. Carcinogenesis 27, 18491859.CrossRefGoogle ScholarPubMed
119Bingham, SA (2007) Mechanisms and experimental and epidemiological evidence relating dietary fibre (non-starch polysaccharides) and starch to protection against large bowel cancer. Proc Nutr Soc 49, 153171.CrossRefGoogle Scholar
120O'Keefe, SJD, Kidd, M, Espitalier-Noel, G, et al. . (1999) Rarity of colon cancer in Africans is associated with low animal product consumption, not fiber. Am J Gastroenterol 94, 13731380.CrossRefGoogle Scholar
121Cho, E, Willett, WC, Colditz, GA, et al. . (2007) Dietary choline and betaine and the risk of distal colorectal adenoma in women. J Natl Cancer Inst 99, 12241231.CrossRefGoogle ScholarPubMed
122Klaunig, JE, Xu, Y, Isenberg, JS, et al. . (1998) The role of oxidative stress in chemical carcinogenesis. Environ Health Perspect 106, 289295.Google ScholarPubMed
123Harris, PJ & Ferguson, LR (1999) Dietary fibres may protect or enhance carcinogenesis. Mutat Res 443, 95110.CrossRefGoogle ScholarPubMed
124Ford, ES, Mokdad, AH, Giles, WH, et al. . (2003) The metabolic syndrome and antioxidant concentrations: findings from the Third National Health and Nutrition Examination Survey. Diabetes 52, 23462352.CrossRefGoogle ScholarPubMed
125Higdon, JV & Frei, B (2003) Obesity and oxidative stress – a direct link to CVD? Arterioscler Thromb Vasc Biol 23, 365367.CrossRefGoogle ScholarPubMed
126Keaney, JF, Larson, MG, Vasan, RS, et al. . (2002) Obesity as a source of systemic oxidative stress: clinical correlates of oxidative stress in the Framingham Study. Circulation 106, 467.Google Scholar
127Evans, JL, Goldfine, ID, Maddux, BA, et al. . (2002) Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocr Rev 23, 599622.CrossRefGoogle ScholarPubMed
128Maiese, K, Morhan, SD & Chong, ZZ (2007) Oxidative stress biology and cell injury during type 1 and type 2 diabetes mellitus. Curr Neurovasc Res 4, 6371.CrossRefGoogle ScholarPubMed
129Cai, H & Harrison, DG (2000) Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res 87, 840844.CrossRefGoogle ScholarPubMed
130Castelao, JE & Gago-Dominguez, M (2008) Risk factors for cardiovascular disease in women: relationship to lipid peroxidation and oxidative stress. Med Hypotheses 71, 3944.CrossRefGoogle ScholarPubMed
131Martinez-Tome, M, Murcia, MA, Frega, N, et al. . (2004) Evaluation of antioxidant capacity of cereal brans. J Agric Food Chem 52, 46904699.CrossRefGoogle ScholarPubMed
132Miller, HE, Rigelhof, F, Marquart, L, et al. . (2000) Antioxidant content of whole grain breakfast cereals, fruits and vegetables. J Am Coll Nutr 19, 312S319S.CrossRefGoogle ScholarPubMed
133Perez-Jimenez, J & Saura-Calixto, F (2005) Literature data may underestimate the actual antioxidant capacity of cereals. J Agric Food Chem 53, 50365040.CrossRefGoogle ScholarPubMed
134Serpen, A, Gökmen, V, Pellegrini, N, et al. . (2008) Direct measurement of the total antioxidant capacity of cereal products. J Cereal Sci 48, 816820.CrossRefGoogle Scholar
135Zielinski, H & Kozlowska, H (2000) Antioxidant activity and total phenolics in selected cereal grains and their different morphological fractions. J Agric Food Chem 48, 20082016.CrossRefGoogle ScholarPubMed
136Fardet, A, Rock, E & Rémésy, C (2008) Is the in vitro antioxidant potential of whole-grain cereals and cereal products well reflected in vivo? J Cereal Sci 48, 258276.CrossRefGoogle Scholar
137Andersson, A, Tengblad, S, Karlstrom, B, et al. . (2007) Whole-grain foods do not affect insulin sensitivity or markers of lipid peroxidation and inflammation in healthy, moderately overweight subjects. J Nutr 137, 14011407.CrossRefGoogle ScholarPubMed
138Beattie, RK, Lee, AM, Strain, JJ, et al. . (2003) Evaluation of the in vivo antioxidant activity of wheat bran in human subjects. Proc Nutr Soc 62, 17A.Google Scholar
139Bruce, B, Spiller, GA, Klevay, LM, et al. . (2000) A diet high in whole and unrefined foods favorably alters lipids, antioxidant defenses, and colon function. J Am Coll Nutr 19, 6167.CrossRefGoogle ScholarPubMed
140Chen, CYO, Milbury, PE, Collins, FW, et al. . (2007) Avenanthramides are bioavailable and have antioxidant activity in humans after acute consumption of an enriched mixture from oats. J Nutr 137, 13751382.CrossRefGoogle ScholarPubMed
141Hamill, LL, Keaveney, EM, Price, RK, et al. . (2007) Assessment of antioxidant biomarkers in human plasma and urine after consumption of wheat bran and aleurone fractions. Proc Nutr Soc 66, 88A.Google Scholar
142Jang, Y, Lee, JH, Kim, OY, et al. . (2001) Consumption of whole grain and legume powder reduces insulin demand, lipid peroxidation, and plasma homocysteine concentrations in patients with coronary artery disease: randomized controlled clinical trial. Arterioscler Thromb Vasc Biol 21, 20652071.CrossRefGoogle ScholarPubMed
143Kim, JY, Kim, JH, Lee, DH, et al. . (2008) Meal replacement with mixed rice is more effective than white rice in weight control, while improving antioxidant enzyme activity in obese women. Nutr Res 28, 6671.CrossRefGoogle Scholar
144Lewis, S, Bolton, C & Heaton, K (1996) Lack of influence of intestinal transit on oxidative status in premenopausal women. Eur J Clin Nutr 50, 565568.Google ScholarPubMed
145Maki, KC, Galant, R, Samuel, P, et al. . (2007) Effects of consuming foods containing oat β-glucan on blood pressure, carbohydrate metabolism and biomarkers of oxidative stress in men and women with elevated blood pressure. Eur J Clin Nutr 61, 786795.CrossRefGoogle ScholarPubMed
146Price, RK, Welch, RW, Lee-Manion, AM, et al. . (2008) Total phenolics and antioxidant potential in plasma and urine of humans after consumption of wheat bran. Cereal Chem 85, 152157.CrossRefGoogle Scholar
147Wang, Q, Han, PH, Zhang, MW, et al. . (2007) Supplementation of black rice pigment fraction improves antioxidant and anti-inflammatory status in patients with coronary heart disease. Asia Pac J Clin Nutr 16, 295301.Google ScholarPubMed
148Graf, E, Empson, KL & Eaton, JW (1987) Phytic acid. A natural antioxidant. J Biol Chem 262, 1164711650.CrossRefGoogle ScholarPubMed
149Dizhbite, T, Telysheva, G, Jurkjane, V, et al. . (2004) Characterization of the radical scavenging activity of lignins – natural antioxidants. Bioresour Technol 95, 309317.CrossRefGoogle ScholarPubMed
150Vitaglione, P, Napolitano, A & Fogliano, V (2008) Cereal dietary fibre: a natural functional ingredient to deliver phenolic compounds into the gut. Trends Food Sci Technol 19, 451463.CrossRefGoogle Scholar
151Babbs, CF (1990) Free radicals and the etiology of colon cancer. Free Radicic Biol Med 8, 191200.CrossRefGoogle ScholarPubMed
152Adam, A, Crespy, V, Levrat-Verny, MA, et al. . (2002) The bioavailability of ferulic acid is governed primarily by the food matrix rather than its metabolism in intestine and liver in rats. J Nutr 132, 19621968.CrossRefGoogle ScholarPubMed
153Mateo Anson, N, van den Berg, R, Havenaar, R, et al. . (2009) Bioavailability of ferulic acid is determined by its bioaccessibility. J Cereal Sci 49, 296300.CrossRefGoogle Scholar
154Rondini, L, Peyrat-Maillard, MN, Marsset-Baglieri, A, et al. . (2004) Bound ferulic acid from bran is more bioavailable than the free compound in rat. J Agric Food Chem 52, 43384343.CrossRefGoogle Scholar
155Pellegrini, N, Serafini, M, Salvatore, S, et al. . (2006) Total antioxidant capacity of spices, dried fruits, nuts, pulses, cereals and sweets consumed in Italy assessed by three different in vitro assays. Mol Nutr Food Res 50, 10301038.CrossRefGoogle ScholarPubMed
156McCarty, MF (2005) Magnesium may mediate the favorable impact of whole grains on insulin sensitivity by acting as a mild calcium antagonist. Med Hypotheses 64, 619627.CrossRefGoogle ScholarPubMed
157Durlach, J & Collery, P (1984) Magnesium and potassium in diabetes and carbohydrate metabolism. Review of the present status and recent results. Magnesium 3, 315323.Google ScholarPubMed
158Paolisso, G, Sgambato, S, Gambardella, A, et al. . (1992) Daily magnesium supplements improve glucose handling in elderly subjects. Am J Clin Nutr 55, 11611167.CrossRefGoogle ScholarPubMed
159Paolisso, G, Sgambato, S, Pizza, G, et al. . (1989) Improved insulin response and action by chronic magnesium administration in aged NIDDM subjects. Diabetes Care 12, 265269.CrossRefGoogle ScholarPubMed
160Pereira, MA, Jacobs, DR Jr, Pins, JJ, et al. . (2002) Effect of whole grains on insulin sensitivity in overweight hyperinsulinemic adults. Am J Clin Nutr 75, 848855.CrossRefGoogle ScholarPubMed
161Balon, T, Jasman, A, Scott, S, et al. . (1994) Dietary magnesium prevents fructose-induced insulin insensitivity in rats. Hypertension 23, 10361039.CrossRefGoogle ScholarPubMed
162Balon, TW, Gu, JL, Tokuyama, Y, et al. . (1995) Magnesium supplementation reduces development of diabetes in a rat model of spontaneous NIDDM. Am J Physiol Endocrinol Metab 269, E745E752.CrossRefGoogle Scholar
163Gould, MK & Chaudry, IH (1970) The action of insulin on glucose uptake by isolated rat soleus muscle. 1. Effects of cations. Biochim Biophys Acta 215, 249257.CrossRefGoogle Scholar
164Weglicki, WB, Mak, IT, Kramer, JH, et al. . (1996) Role of free radicals and substance P in magnesium deficiency. Cardiovasc Res 31, 677682.CrossRefGoogle ScholarPubMed
165Ceriello, A, Bortolotti, N, Crescentini, A, et al. . (1998) Antioxidant defences are reduced during the oral glucose tolerance test in normal and non-insulin-dependent diabetic subjects. Eur J Clin Invest 28, 329333.CrossRefGoogle ScholarPubMed
166Pereira, EC, Ferderbar, S, Bertolami, MC, et al. . (2008) Biomarkers of oxidative stress and endothelial dysfunction in glucose intolerance and diabetes mellitus. Clin Biochem 41, 14541460.CrossRefGoogle ScholarPubMed
167Iseri, LT & French, JH (1984) Magnesium – Nature's physiologic calcium blocker. Am Heart J 108, 188193.CrossRefGoogle ScholarPubMed
168Resnick, LM (1992) Cellular calcium and magnesium metabolism in the pathophysiology and treatment of hypertension and related metabolic disorders. Am J Med 93, S11S20.CrossRefGoogle ScholarPubMed
169Liao, F, Folsom, AR & Brancati, FL (1998) Is low magnesium concentration a risk factor for coronary heart disease? The Atherosclerosis Risk in Communities (ARIC) Study. Am Heart J 136, 480490.CrossRefGoogle ScholarPubMed
170Shechter, M, Merz, CNB, Paul-Labrador, M, et al. . (1999) Oral magnesium supplementation inhibits platelet-dependent thrombosis in patients with coronary artery disease. Am J Cardiol 84, 152156.CrossRefGoogle ScholarPubMed
171Kawano, Y, Matsuoka, H, Takishita, S, et al. . (1998) Effects of magnesium supplementation in hypertensive patients: assessment by office, home, and ambulatory blood pressures. Hypertension 32, 260265.CrossRefGoogle ScholarPubMed
172Al-Mamary, M, Al-Habori, M, Al-Aghbari, A, et al. . (2001) In vivo effects of dietary sorghum tannins on rabbit digestive enzymes and mineral absorption. Nutr Res 21, 13931401.CrossRefGoogle Scholar
173Thompson, LU (1993) Potential health benefits and problems associated with antinutrients in foods. Food Res Int 26, 131149.CrossRefGoogle Scholar
174Gillooly, M, Bothwell, TH, Charlton, RW, et al. . (1984) Factors affecting the absorption of iron from cereals. Br J Nutr 51, 3746.CrossRefGoogle ScholarPubMed
175Tatala, S, Svanberg, U & Mduma, B (1998) Low dietary iron availability is a major cause of anemia: a nutrition survey in the Lindi District of Tanzania. Am J Clin Nutr 68, 171178.CrossRefGoogle Scholar
176Lestienne, I, Besancon, P, Caporiccio, B, et al. . (2005) Iron and zinc in vitro availability in pearl millet flours (Pennisetum glaucum) with varying phytate, tannin, and fiber contents. J Agric Food Chem 53, 32403247.CrossRefGoogle ScholarPubMed
177Hassan, IAG & El Tinay, AH (1995) Effect of fermentation on tannin content and in-vitro protein and starch digestibilities of two sorghum cultivars. Food Chem 53, 149151.CrossRefGoogle Scholar
178Matuschek, E, Towo, E & Svanberg, U (2001) Oxidation of polyphenols in phytate-reduced high-tannin cereals: effect on different phenolic groups and on in vitro accessible iron. J Agric Food Chem 49, 56305638.CrossRefGoogle ScholarPubMed
179Mbithi-Mwikya, S, Van Camp, J, Yiru, Y, et al. . (2000) Nutrient and antinutrient changes in finger millet (Eleusine coracan) during sprouting. Lebensm-Wiss Technol Food Sci Technol 33, 914.CrossRefGoogle Scholar
180Towo, E, Matuschek, E & Svanberg, U (2006) Fermentation and enzyme treatment of tannin sorghum gruels: effects on phenolic compounds, phytate and in vitro accessible iron. Food Chem 94, 369376.CrossRefGoogle Scholar
181Thompson, LU (1988) Antinutrients and blood glucose. Food Technol 42, 123132.Google Scholar
182Yoon, J, Thompson, L & Jenkins, D (1983) The effect of phytic acid on in vitro rate of starch digestibility and blood glucose response. Am J Clin Nutr 38, 835842.CrossRefGoogle ScholarPubMed
183Manach, C, Williamson, G, Morand, C, et al. . (2005) Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr 81, 230S242S.CrossRefGoogle ScholarPubMed
184Choi, J-S, Choi, Y-J, Shin, S-Y, et al. . (2008) Dietary flavonoids differentially reduce oxidized LDL-induced apoptosis in human endothelial cells: role of MAPK- and JAK/STAT-signaling. J Nutr 138, 983990.CrossRefGoogle ScholarPubMed
185Crespo, I, García-Mediavilla, MV, Gutiérrez, B, et al. . (2008) A comparison of the effects of kaempferol and quercetin on cytokine-induced pro-inflammatory status of cultured human endothelial cells. Br J Nutr 100, 968976.CrossRefGoogle ScholarPubMed
186Maggi-Capeyron, MF, Ceballos, P, Cristol, JP, et al. . (2001) Wine phenolic antioxidants inhibit AP-1 transcriptional activity. J Agric Food Chem 49, 56465652.CrossRefGoogle ScholarPubMed
187Yun, K-J, Koh, D-J, Kim, S-H, et al. . (2008) Anti-inflammatory effects of sinapic acid through the suppression of inducible nitric oxide synthase, cyclooxygase-2, and proinflammatory cytokines expressions via nuclear factor-κB inactivation. J Agric Food Chem 56, 1026510272.CrossRefGoogle ScholarPubMed
188Moskaug, JO, Carlsen, H, Myhrstad, MC, et al. . (2005) Polyphenols and glutathione synthesis regulation. Am J Clin Nutr 81, 277S283S.CrossRefGoogle ScholarPubMed
189Rahman, I, Biswas, SK & Kirkham, PA (2006) Regulation of inflammation and redox signaling by dietary polyphenols. Biochem Pharmacol 72, 14391452.CrossRefGoogle Scholar
190Ramos, S (2008) Cancer chemoprevention and chemotherapy: dietary polyphenols and signalling pathways. Mol Nutr Food Res 52, 507526.CrossRefGoogle ScholarPubMed
191Williams, RJ, Spencer, JPE & Rice-Evans, C (2004) Flavonoids: antioxidants or signalling molecules? Free Radic Biol Med 36, 838849.CrossRefGoogle ScholarPubMed
192McCallum, JA & Walker, JRL (1990) Proanthocyanidins in wheat bran. Cereal Chem 67, 282285.Google Scholar
193Feng, Y & McDonald, CE (1989) Comparison of flavonoids in bran of four classes of wheat. Cereal Chem 66, 516518.Google Scholar
194Gallardo, C, Jiménez, L & García-Conesa, M-T (2006) Hydroxycinnamic acid composition and in vitro antioxidant activity of selected grain fractions. Food Chem 99, 455463.CrossRefGoogle Scholar
195Myhrstad, MCW, Carlsen, H, Nordstrom, O, et al. . (2002) Flavonoids increase the intracellular glutathione level by transactivation of the γ-glutamylcysteine synthetase catalytical subunit promoter. Free Radic Biol Med 32, 386393.CrossRefGoogle ScholarPubMed
196Kern, SM, Bennett, RN, Mellon, FA, et al. . (2003) Absorption of hydroxycinnamates in humans after high-bran cereal consumption. J Agric Food Chem 51, 60506055.CrossRefGoogle ScholarPubMed
197Li, L, Shewry, PR & Ward, JL (2008) Phenolic acids in wheat varieties in the HEALTHGRAIN diversity screen. J Agric Food Chem 56, 97329739.CrossRefGoogle ScholarPubMed
198Métayer, S, Seiliez, I, Collin, A, et al. . (2008) Mechanisms through which sulfur amino acids control protein metabolism and oxidative status. J Nutr Biochem 19, 207215.CrossRefGoogle ScholarPubMed
199Tesseraud, S, Métayer Coustard, S, Collin, A, et al. . (2009) Role of sulfur amino acids in controlling nutrient metabolism and cell functions: implications for nutrition. Br J Nutr 101, 11321139.CrossRefGoogle ScholarPubMed
200Morand, C, Rios, L, Moundras, C, et al. . (1997) Influence of methionine availability on glutathione synthesis and delivery by the liver. J Nutr Biochem 8, 246255.CrossRefGoogle Scholar
201Nkabyo, YS, Gu, LH, Jones, DP, et al. . (2006) Thiol/disulfide redox status is oxidized in plasma and small intestinal and colonic mucosa of rats with inadequate sulfur amino acid intake. J Nutr 136, 12421248.CrossRefGoogle ScholarPubMed
202Tateishi, N, Hirasawa, M, Higashi, T, et al. . (1982) The l-methionine-sparing effect of dietary glutathione in rats. J Nutr 112, 22172226.CrossRefGoogle ScholarPubMed
203Flagg, EW, Coates, RJ, Eley, JW, et al. . (1994) Dietary glutathione intake in humans and the relationship between intake and plasma total glutathione level. Nutr Cancer 21, 3346.CrossRefGoogle ScholarPubMed
204Martin, A (2001) Apports nutritionnels conseillés pour la population française (Recommended Dietary Allowances for the French Population), 3rd ed.Paris: Editions TEC & DOC.Google Scholar
205US Department of Agriculture ARS & Nutrient Data Laboratory (2005) USDA National Nutrient Database for Standard Reference, release 18, Baked products. http://www.nal.usda.gov/fnic/foodcomp/Data/SR18/reports/sr18page.htm (accessed January 2010).Google Scholar
206Smith, AT, Kuznesof, S, Richardson, DP, et al. . (2003) Behavioural, attitudinal and dietary responses to the consumption of wholegrain foods. Proc Nutr Soc 62, 455467.CrossRefGoogle Scholar
207Hagen, TM, Wierzbicka, GT, Bowman, BB, et al. . (1990) Fate of dietary glutathione: disposition in the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 259, G530G535.CrossRefGoogle ScholarPubMed
208Gmünder, H, Roth, S, Eck, H-P, et al. . (1990) Interleukin-2 mRNA expression, lymphokine production and DNA synthesis in glutathione-depleted T cells. Cell Immunol 130, 520528.CrossRefGoogle ScholarPubMed
209Witschi, A, Reddy, S, Stofer, B, et al. . (1992) The systemic availability of oral glutathione. Eur J Clin Pharmacol 43, 667669.CrossRefGoogle ScholarPubMed
210Sarwin, R, Walther, C, Laskawy, G, et al. . (1992) Determination of free reduced and total glutathione in wheat flours by an isotope-dilution assay. Z Lebens Unters Forsch 195, 2732.CrossRefGoogle Scholar
211Li, W, Bollecker, SS & Schofield, JD (1995) Glutathione and related thiol compounds. I. Glutathione and related thiol compounds in flour. J Cereal Sci 39, 205212.CrossRefGoogle Scholar
212Weber, F & Grosch, W (1978) Determination of reduced and oxidized glutathione in wheat flours and doughs. Z Lebens Unters Forsch 167, 8792.CrossRefGoogle ScholarPubMed
213Lotito, SB & Frei, B (2004) The increase in human plasma antioxidant capacity after apple consumption is due to the metabolic effect of fructose on urate, not apple-derived antioxidant flavonoids. Free Radic Biol Med 37, 251258.CrossRefGoogle Scholar
214Lotito, SB & Frei, B (2006) Consumption of flavonoid-rich foods and increased plasma antioxidant capacity in humans: cause, consequence, or epiphenomenon? Free Radic Biol Med 41, 17271746.CrossRefGoogle ScholarPubMed
215Souci, SW, Fachmann, W & Kraut, H (2000) Food Composition and Nutritional Tables. Stuttgart: Medpharm Scientific Publishers.Google Scholar
216Natella, F, Nardini, M, Giannetti, I, et al. . (2002) Coffee drinking influences plasma antioxidant capacity in humans. J Agric Food Chem 50, 62116216.CrossRefGoogle ScholarPubMed
217Lopez, HW, Leenhardt, F, Coudray, C, et al. . (2002) Minerals and phytic acid interactions: is it a real problem for human nutrition? Int J Food Sci Technol 37, 727739.CrossRefGoogle Scholar
218Graf, E & Eaton, GW (1990) Antioxidant functions of phytic acid. Free Radic Biol Med 8, 6169.CrossRefGoogle ScholarPubMed
219Levrat-Verny, MA, Coudray, C, Bellanger, J, et al. . (1999) Wholewheat flour ensures higher mineral absorption and bioavailability than white wheat flour in rats. Br J Nutr 82, 1721.CrossRefGoogle ScholarPubMed
220Leenhardt, F, Levrat-Verny, MA, Chanliaud, E, et al. . (2005) Moderate decrease of pH by sourdough fermentation is sufficient to reduce phytate content of whole wheat flour through endogenous phytase activity. J Agric Food Chem 53, 98102.CrossRefGoogle ScholarPubMed
221Begum, AN, Nicolle, C, Mila, I, et al. . (2004) Dietary lignins are precursors of mammalian lignans in rats. J Nutr 134, 120127.CrossRefGoogle ScholarPubMed
222Kitts, DD, Yuan, YV, Wijewickreme, AN, et al. . (1999) Antioxidant activity of the flaxseed lignan secoisolariciresinol diglycoside and its mammalian lignan metabolites enterodiol and enterolactone. Mol Cell Biochem 202, 91100.CrossRefGoogle ScholarPubMed
223Bach Knudsen, KE, Serena, A, Kjaer, AKB, et al. . (2003) Rye bread in the diet of pigs enhances the formation of enterolactone and increases its levels in plasma, urine and feces. J Nutr 133, 13681375.CrossRefGoogle ScholarPubMed
224Labaj, J, Wsolova, L, Lazarova, M, et al. . (2004) Repair of oxidative DNA lesions in blood lymphocytes isolated from Sprague–Dawley rats; the influence of dietary intake of lignin. Neoplasma 51, 450455.Google ScholarPubMed
225Craig, SAS (2004) Betaine in human nutrition. Am J Clin Nutr 80, 539549.CrossRefGoogle ScholarPubMed
226Zeisel, SH & Blusztajn, JK (1994) Choline and human nutrition. Annu Rev Nutr 14, 269296.CrossRefGoogle ScholarPubMed
227Likes, R, Madl, RL, Zeisel, SH, et al. . (2007) The betaine and choline content of a whole wheat flour compared to other mill streams. J Cereal Sci 46, 9395.CrossRefGoogle ScholarPubMed
228Cho, S, Johnson, G & Song, WO (2002) Folate content of foods: comparison between databases compiled before and after new FDA fortification requirements. J Food Comp Anal 15, 293307.CrossRefGoogle Scholar
229Bertram, HC, Bach Knudsen, KE, Serena, A, et al. . (2006) NMR-based metabonomic studies reveal changes in the biochemical profile of plasma and urine from pigs fed high-fibre rye bread. Br J Nutr 95, 955962.CrossRefGoogle ScholarPubMed
230Fardet, A, Canlet, C, Gottardi, G, et al. . (2007) Whole grain and refined wheat flours show distinct metabolic profiles in rats as assessed by a 1H NMR-based metabonomic approach. J Nutr 4, 923929.CrossRefGoogle Scholar
231Borgschulte, G, Kathirvel, E, Herrera, M, et al. . (2008) Betaine treatment reverses insulin resistance and fatty liver disease without reducing oxidative stress or endoplasmic reticulum stress in an animal model of NAFLD. Gastroenterology 134, A414A415.CrossRefGoogle Scholar
232Brouwer, IA, van Dusseldorp, M, Thomas, CM, et al. . (1999) Low-dose folic acid supplementation decreases plasma homocysteine concentrations: a randomized trial. Am J Clin Nutr 69, 99104.CrossRefGoogle ScholarPubMed
233Graham, IM, Daly, LE, Refsum, HM, et al. . (1997) Plasma homocysteine as a risk factor for vascular disease. The European Concerted Action Project. JAMA 277, 17751781.CrossRefGoogle ScholarPubMed
234Mills, JL, McPartlin, JM, Kirke, PN, et al. . (1995) Homocysteine metabolism in pregnancies complicated by neural-tube defects. Lancet 345, 149151.CrossRefGoogle ScholarPubMed
235Wu, LL & Wu, JT (2002) Hyperhomocysteinemia is a risk factor for cancer and a new potential tumor marker. Clin Chim Acta 322, 2128.CrossRefGoogle Scholar
236Loscalzo, J (1996) The oxidant stress of hyperhomocyst(e)inemia. J Clin Invest 98, 57.CrossRefGoogle ScholarPubMed
237Tyagi, N, Sedoris, KC, Steed, M, et al. . (2005) Mechanisms of homocysteine-induced oxidative stress. Am J Physiol Heart Circ Physiol 289, H2649H2656.CrossRefGoogle ScholarPubMed
238Christman, JK, Chen, M-L, Sheikhnejad, G, et al. . (1993) Methyl deficiency, DNA methylation, and cancer: studies on the reversibility of the effects of a lipotrope-deficient diet. J Nutr Biochem 4, 672680.CrossRefGoogle Scholar
239Newberne, PM & Rogers, AE (1986) Labile methyl groups and the promotion of cancer. Annu Rev Nutr 6, 407432.CrossRefGoogle ScholarPubMed
240Zeisel, S, Da Costa, K, Franklin, P, et al. . (1991) Choline, an essential nutrient for humans. FASEB J 5, 20932098.CrossRefGoogle ScholarPubMed
241Iqbal, TH, Lewis, KO & Cooper, BT (1994) Phytase activity in the human and rat small intestine. Gut 35, 12331236.CrossRefGoogle ScholarPubMed
242Okazaki, Y, Setoguchi, T & Katayama, T (2006) Effects of dietary myo-inositol, d-chiro-inositol and l-chiro-inositol on hepatic lipids with reference to the hepatic myo-inositol status in rats fed on 1,1,1-trichloro-2,2-bis (p-chlorophenyl) ethane. Biosci Biotechnol Biochem 70, 27662770.CrossRefGoogle Scholar
243Horbowicz, M & Obendorf, RL (2005) Fagopyritol accumulation and germination of buckwheat seeds matured at 15, 22, and 30°C. Crop Sci 45, 12641270.CrossRefGoogle Scholar
244Steadman, KJ, Burgoon, MS, Schuster, RL, et al. . (2000) Fagopyritols, d-chiro-inositol, and other soluble carbohydrates in buckwheat seed milling fractions. J Agric Food Chem 48, 28432847.CrossRefGoogle ScholarPubMed
245Kim, JI, Kim, JC, Joo, HJ, et al. . (2005) Determination of total chiro-inositol content in selected natural materials and evaluation of the antihyperglycemic effect of pinitol isolated from soybean and carob. Food Sci Biotechnol 14, 441445.Google Scholar
246Becker, R, Wheeler, EL, Lorenz, K, et al. . (1981) A compositional study of amaranth grain. J Food Sci 46, 11751180.CrossRefGoogle Scholar
247Darbre, A & Norris, FW (1956) Vitamins in germination – determination of free and combined inositol in germinating oats. Biochem J 64, 441446.CrossRefGoogle ScholarPubMed
248Koziol, MJ (1992) Chemical composition and nutritional evaluation of quinoa (Chenopodium quinoa Willd.). J Food Comp Anal 5, 3568.CrossRefGoogle Scholar
249Horbowicz, M & Obendorf, RL (1994) Seed desiccation tolerance and storability: dependence on flatulence-producing oligosaccharides and cyclitols? – review and survey. Seed Sci Res 4, 385405.CrossRefGoogle Scholar
250Matheson, NK & Strother, S (1969) The utilization of phytate by germinating wheat. Phytochemistry 8, 13491356.CrossRefGoogle Scholar
251Clements, R Jr & Darnell, B (1980) Myo-inositol content of common foods: development of a high-myo-inositol diet. Am J Clin Nutr 33, 19541967.CrossRefGoogle ScholarPubMed
252Reddy, NR, Sathe, SK & Salunkhe, DK (1982) Phytates in legumes and cereals. Adv Food Res 28, 192.CrossRefGoogle ScholarPubMed
253Ferrel, RE (1978) Distribution of bean and wheat inositol phosphate esters during autolysis and germination. J Food Sci 43, 563565.CrossRefGoogle Scholar
254Nakano, T, Joh, T, Narita, K, et al. . (2000) The pathway of dephosphorylation of myo-inositol hexakisphosphate by phytases from wheat bran of Triticum aestivum L. cv. Nourin #61. Biosci Biotechnol Biochem 64, 9951003.CrossRefGoogle Scholar
255Bergman, E-L, Fredlund, K, Reinikainen, P, et al. . (1999) Hydrothermal processing of barley (cv. Blenheim): optimisation of phytate degradation and increase of free myo-inositol. J Cereal Sci 29, 261272.CrossRefGoogle Scholar
256Pak, Y, Huang, L, Lilley, K, et al. . (1992) In vivo conversion of [3H]myoinositol to [3H]chiroinositol in rat tissues. J Biol Chem 267, 1690416910.CrossRefGoogle ScholarPubMed
257Reeves, PG, Nielsen, FH & Fahey, GC Jr (1993) AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr 123, 19391951.CrossRefGoogle Scholar
258Locker, J, Reddy, TV & Lombardi, B (1986) DNA methylation and hepatocarcinogenesis in rats fed a choline-devoid diet. Carcinogenesis 7, 13091312.CrossRefGoogle ScholarPubMed
259Gama-Sosa, MA, Slagel, VA, Trewyn, RW, et al. . (1983) The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res 11, 68836894.CrossRefGoogle ScholarPubMed
260Goelz, S, Vogelstein, B, Hamilton, SR, et al. . (1985) Hypomethylation of DNA from benign and malignant human colon neoplasms. Science 228, 187190.CrossRefGoogle ScholarPubMed
261Ou, SY & Kwok, KC (2004) Ferulic acid: pharmaceutical functions, preparation and applications in foods. J Sci Food Agric 84, 12611269.CrossRefGoogle Scholar
262Srinivasan, M, Sudheer, AR & Menon, VP (2007) Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 40, 92100.CrossRefGoogle ScholarPubMed
263Rybka, K, Sitarski, J & Raczynskabojanowska, K (1993) Ferulic acid in rye and wheat-grain and grain dietary fiber. Cereal Chem 70, 5559.Google Scholar
264Kroon, PA, Faulds, CB, Ryden, P, et al. . (1997) Release of covalently bound ferulic acid from fiber in the human colon. J Agric Food Chem 45, 661667.CrossRefGoogle Scholar
265Akao, Y, Seki, N, Nakagawa, Y, et al. . (2004) A highly bioactive lignophenol derivative from bamboo lignin exhibits a potent activity to suppress apoptosis induced by oxidative stress in human neuroblastoma SH-SY5Y cells. Bioorg Med Chem 12, 47914801.CrossRefGoogle ScholarPubMed
266Ferguson, LR & Harris, PJ (1996) Studies on the role of specific dietary fibres in protection against colorectal cancer. Mutat Res 350, 173184.CrossRefGoogle ScholarPubMed
267Calvert, GD & Yeates, RA (1982) Adsorption of bile salts by soya-bean flour, wheat bran, lucerne (Medicago sativa), sawdust and lignin: the effect of saponins and other plant constituents. Br J Nutr 47, 4552.CrossRefGoogle ScholarPubMed
268Chang, MLW & Johnson, MA (1980) Effect of lignin versus cellulose on the absorption of taurocholate and lipid metabolism in rats fed cholesterol diet. Nutr Rep Int 21, 513518.Google Scholar
269Drasar, B & Jenkins, D (1976) Bacteria, diet, and large bowel cancer. Am J Clin Nutr 29, 14101416.CrossRefGoogle ScholarPubMed
270Anjaneyulu, M & Chopra, K (2004) Nordihydroguairetic acid, a lignin, prevents oxidative stress and the development of diabetic nephropathy in rats. Pharmacology 72, 4250.CrossRefGoogle ScholarPubMed
271Fardet, A, Llorach, R, Orsoni, A, et al. . (2008) Metabolomics provide new insight on the metabolism of dietary phytochemicals in rats. J Nutr 138, 12821287.CrossRefGoogle Scholar
272Hindmarch, I (2002) Beyond the monoamine hypothesis: mechanisms, molecules and methods. Eur Psychiatry 17, 294299.CrossRefGoogle ScholarPubMed
273Berry, RJ, Li, Z, Erickson, JD, et al. . (1999) Prevention of neural-tube defects with folic acid in China. N Engl J Med 341, 14851490.CrossRefGoogle ScholarPubMed
274Coppen, A & Bolander-Gouaille, C (2005) Treatment of depression: time to consider folic acid and vitamin B12. J Psychopharmacol 19, 5965.CrossRefGoogle ScholarPubMed
275Miller, AL (2008) The methylation, neurotransmitter, and antioxidant connections between folate and depression. Altern Med Rev 13, 216226.Google ScholarPubMed
276Gilbody, S, Lightfoot, T & Sheldon, T (2007) Is low folate a risk factor for depression? A meta-analysis and exploration of heterogeneity. J Epidemiol Community Health 61, 631637.CrossRefGoogle ScholarPubMed
277Lioger, D, Leenhardt, F, Demigne, C, et al. . (2007) Sourdough fermentation of wheat fractions rich in fibres before their use in processed food. J Sci Food Agric 87, 13681373.CrossRefGoogle Scholar
278Ohta, A, Ohtsuki, M, Hosono, A, et al. . (1998) Dietary fructooligosaccharides prevent osteopenia after gastrectomy in rats. J Nutr 128, 106110.CrossRefGoogle ScholarPubMed
279Scholz-Ahrens, KE, Ade, P, Marten, B, et al. . (2007) Prebiotics, probiotics, and synbiotics affect mineral absorption, bone mineral content, and bone structure. J Nutr 137, 838S846S.CrossRefGoogle ScholarPubMed
280Scholz-Ahrens, KE & Schrezenmeir, J (2007) Inulin and oligofructose and mineral metabolism: the evidence from animal trials. J Nutr 137, 2513S2523S.CrossRefGoogle Scholar
281Nordbö, H & Rolla, G (1972) Desorption of salivary proteins from hydroxyapatite by phytic acid and glycerophosphate and the plaque-inhibiting effect of the two compounds in vivo. J Dent Res 51, 800802.CrossRefGoogle ScholarPubMed
282McClure, FJ (1964) Cariostatic effect of phosphates. Science 144, 13371338.CrossRefGoogle ScholarPubMed
283Osborn, TWB & Noriskin, JN (1937) The relation between diet and caries in the South African Bantu. J Dent Res 16, 431441.CrossRefGoogle Scholar
284Osborn, TWB, Noriskin, JN & Staz, J (1937) A comparison of crude and refined sugar and cereals in their ability to produce in vitro decalcification of teeth. J Dent Res 16, 165171.CrossRefGoogle Scholar
285Osborn, TWB (1941) Further studies on the in vitro decalcification of teeth. J Dent Res 20, 5969.CrossRefGoogle Scholar
286McClure, FJ (1960) The cariostatic effect in white rats of phosphorus and calcium supplements added to the flour of bread formulas and to bread diets. J Nutr 72, 131136.CrossRefGoogle Scholar
287McClure, FJ (1963) Further studies on the cariostatic effect of organic and inorganic phosphates. J Dent Res 42, 693699.CrossRefGoogle Scholar
288McClure, FJ & Muller, A Jr (1959) Further observations on the cariostatic effect of phosphates. J Dent Res 38, 776781.CrossRefGoogle Scholar
289Wynn, W, Haldi, J, Bentley, KD, et al. . (1956) Dental caries in the albino rat in relation to the chemical composition of the teeth and of the diet: II. Variations in the Ca/P ratio of the diet induced by changing the phosphorus content. J Nutr 58, 325333.CrossRefGoogle Scholar
290Magrill, DS (1973) The reduction of the solubility of hydroxyapatite in acid by adsorption of phytate from solution. Arch Oral Biol 18, 591600.CrossRefGoogle ScholarPubMed
291Pruitt, KM, Jamieson, AD & Caldwell, RC (1970) Possible basis for the cariostatic effect of inorganic phosphates. Nature 225, 1249.CrossRefGoogle ScholarPubMed
292Cole, MF & Bowen, WH (1975) Effect of sodium phytate on the chemical and microbial composition of dental plaque in the monkey (Macaca fascicularis). J Dent Res 54, 449457.CrossRefGoogle ScholarPubMed
293Adlercreutz, H & Mazur, W (1997) Phyto-oestrogens and Western diseases. Ann Med 29, 95120.CrossRefGoogle ScholarPubMed
294Chanvrier, H, Appelqvist, IA, Bird, AR, et al. . (2007) Processing of novel elevated amylose wheats: functional properties and starch digestibility of extruded products. J Agric Food Chem 55, 1024810257.CrossRefGoogle ScholarPubMed
295Swennen, K, Courtin, CM & Delcour, JA (2006) Non-digestible oligosaccharides with prebiotic properties. Crit Rev Food Sci Nutr 46, 459471.CrossRefGoogle ScholarPubMed
296Krause, DO, Easter, RA & Mackie, RI (1994) Fermentation of stachyose and raffinose by hind-gut bacteria of the weanling pig. Lett Appl Microbiol 18, 349352.CrossRefGoogle Scholar
297Tortuero, F, Fernández, E, Rupérez, P, et al. . (1997) Raffinose and lactic bacteria influence caecal fermentation and serum cholesterol in rats. Nutr Res 17, 4149.CrossRefGoogle Scholar
298Alessandri, C, Pignatelli, P, Loffredo, L, et al. . (2006) α-Linolenic acid-rich wheat germ oil decreases oxidative stress and CD40 ligand in patients with mild hypercholesterolemia. Arterioscler Thromb Vasc Biol 26, 25772578.CrossRefGoogle ScholarPubMed
299Farquhar, JW, Smith, RE & Dempsey, ME (1956) The effect of beta sitosterol on the serum lipids of young men with arteriosclerotic heart disease. Circulation 14, 7782.CrossRefGoogle Scholar
300Jones, PJ, Ntanios, FY, Raeini-Sarjaz, M, et al. . (1999) Cholesterol-lowering efficacy of a sitostanol-containing phytosterol mixture with a prudent diet in hyperlipidemic men. Am J Clin Nutr 69, 11441150.CrossRefGoogle ScholarPubMed
301Kato, S, Karino, K-I, Hasegawa, S, et al. . (1995) Octacosanol affects lipid metabolism in rats fed on a high-fat diet. Br J Nutr 73, 433441.CrossRefGoogle ScholarPubMed
302Taylor, JC, Rapport, L & Lockwood, GB (2003) Octacosanol in human health. Nutrition 19, 192195.CrossRefGoogle ScholarPubMed
303Gouni-Berthold, I & Berthold, HK (2002) Policosanol: clinical pharmacology and therapeutic significance of a new lipid-lowering agent. Am Heart J 143, 356365.CrossRefGoogle ScholarPubMed
304Varady, KA, Wang, Y & Jones, PJH (2003) Role of policosanols in the prevention and treatment of cardiovascular disease. Nutr Rev 61, 376383.CrossRefGoogle ScholarPubMed
305Lin, YG, Rudrum, M, van der Wielen, RPJ, et al. . (2004) Wheat germ policosanol failed to lower plasma cholesterol in subjects with normal to mildly elevated cholesterol concentrations. Metabolism 53, 13091314.CrossRefGoogle ScholarPubMed
306Menendez, R, Arruzazabala, L, Mas, R, et al. . (1997) Cholesterol-lowering effect of policosanol on rabbits with hypercholesterolaemia induced by a wheat starch–casein diet. Br J Nutr 77, 923932.CrossRefGoogle ScholarPubMed
307Menendez, R, Fraga, V, Amor, AM, et al. . (1999) Oral administration of policosanol inhibits in vitro copper ion-induced rat lipoprotein peroxidation. Physiol Behav 67, 17.CrossRefGoogle ScholarPubMed
308Hosseinian, FS, Li, W & Beta, T (2008) Measurement of anthocyanins and other phytochemicals in purple wheat. Food Chem 109, 916924.CrossRefGoogle ScholarPubMed
309Asayama, K, Yamadera, H, Ito, T, et al. . (2003) Double blind study of melatonin effects on the sleep–wake rhythm, cognitive and non-cognitive functions in Alzheimer type dementia. J Nippon Med Sch 70, 334341.CrossRefGoogle ScholarPubMed
310Maurizi, CP (2001) Alzheimer's disease: roles for mitochondrial damage, the hydroxyl radical, and cerebrospinal fluid deficiency of melatonin. Med Hypotheses 57, 156160.CrossRefGoogle ScholarPubMed
311Garcia-Navarro, A, Gonzalez-Puga, C, Escames, G, et al. . (2007) Cellular mechanisms involved in the melatonin inhibition of HT-29 human colon cancer cell proliferation in culture. J Pineal Res 43, 195205.CrossRefGoogle ScholarPubMed
312Shiu, SYW (2007) Towards rational and evidence-based use of melatonin in prostate cancer prevention and treatment. J Pineal Res 43, 19.CrossRefGoogle ScholarPubMed
313Calhoun, WK, Bechtel, WG & Bradley, WB (1958) The vitamin content of wheat, flour, and bread. Cereal Chem 35, 350359.Google Scholar
314Calhoun, WK, Hepburn, FN & Bradley, WB (1960) The distribution of the vitamins of wheat in commercial mill products. Cereal Chem 37, 755761.Google Scholar
315Wang, LH, Huang, WS & Tai, HM (2007) Simultaneous determination of p-aminobenzoic acid and its metabolites in the urine of volunteers, treated with p-aminobenzoic acid sunscreen formulation. J Pharm Biomed Anal 43, 14301436.CrossRefGoogle ScholarPubMed
316Barbieri, B, Papadogiannakis, N, Eneroth, P, et al. . (1995) Arachidonic acid is a preferred acetyl donor among fatty acids in the acetylation of p-aminobenzoic acid by human lymphoid cells. Biochim Biophys Acta 1257, 157166.CrossRefGoogle ScholarPubMed
317Failey, RB & Childress, RH (1962) The effect of para-aminobenzoic acid on the serum cholesterol level in man. Am J Clin Nutr 10, 158162.CrossRefGoogle ScholarPubMed
318Butcher, NJ, Ilett, KF & Minchin, RF (2000) Inactivation of human arylamine N-acetyltransferase 1 by the hydroxylamine of p-aminobenzoic acid. Biochem Pharmacol 60, 18291836.CrossRefGoogle ScholarPubMed
319Hein, DW, Doll, MA, Gray, K, et al. . (1993) Metabolic activation of N-hydroxy-2-aminofluorene and N-hydroxy-2-acetylaminofluorene by monomorphic N-acetyltransferase (NAT1) and polymorphic N-acetyltransferase (NAT2) in colon cytosols of syrian hamsters congenic at the NAT2 locus. Cancer Res 53, 509514.Google ScholarPubMed
320Minchin, RF, Reeves, PT, Teitel, CH, et al. . (1992) N- and O-acetylation of aromatic and heterocyclic amine carcinogens by human monomorphic and polymorphic acetyltransferases expressed in COS-1 cells. Biochem Biophys Res Commun 185, 839844.CrossRefGoogle ScholarPubMed
321Barbieri, B, Papadogiannakis, N, Eneroth, P, et al. . (1997) p-Aminobenzoic acid, but not its metabolite p-acetamidobenzoic acid, inhibits thrombin induced thromboxane formation in human platelets in a non NSAID like manner. Thromb Res 86, 127140.CrossRefGoogle ScholarPubMed
322Elliott, R, Pico, C, Dommels, Y, et al. . (2007) Nutrigenomic approaches for benefit–risk analysis of foods and food components: defining markers of health. Br J Nutr 98, 10951100.CrossRefGoogle ScholarPubMed
323Steiner, C, Arnould, S, Scalbert, A, et al. . (2008) Isoflavones and the prevention of breast and prostate cancer: new perspectives opened by nutrigenomics. Br J Nutr 99, E Suppl. 1, ES78ES108.CrossRefGoogle ScholarPubMed
324Trujillo, E, Davis, C & Milner, J (2006) Nutrigenomics, proteomics, metabolomics, and the practice of dietetics. J Am Diet Assoc 106, 403413.CrossRefGoogle ScholarPubMed
325van Ommen, B (2004) Nutrigenomics: exploiting systems biology in the nutrition and health arenas. Nutrition 20, 48.CrossRefGoogle ScholarPubMed
326Zeisel, SH (2007) Nutrigenomics and metabolomics will change clinical nutrition and public health practice: insights from studies on dietary requirements for choline. Am J Clin Nutr 86, 542548.CrossRefGoogle Scholar
327Keun, HC (2006) Metabonomic modeling of drug toxicity. Pharmacol Ther 109, 92106.CrossRefGoogle ScholarPubMed
328Rezzi, S, Ramadan, Z, Fay, LB, et al. . (2007) Nutritional metabonomics: applications and perspectives. J Proteome Res 6, 513525.CrossRefGoogle Scholar
329Selman, C, Kerrison, ND, Cooray, A, et al. . (2006) Coordinated multitissue transcriptional and plasma metabonomic profiles following acute caloric restriction in mice. Physiol Genomics 27, 187200.CrossRefGoogle ScholarPubMed
330Griffin, JL, Muller, D, Woograsingh, R, et al. . (2002) Vitamin E deficiency and metabolic deficits in neuronal ceroid lipofuscinosis described by bioinformatics. Physiol Genomics 11, 195203.CrossRefGoogle ScholarPubMed
331Mutch, DM, Grigorov, M, Berger, A, et al. . (2005) An integrative metabolism approach identifies stearoyl-CoA desaturase as a target for an arachidonate-enriched diet. FASEB J 19, 599619.CrossRefGoogle ScholarPubMed
332Solanky, KS, Bailey, NJC, Beckwith-Hall, BM, et al. . (2003) Application of biofluid H-1 nuclear magnetic resonance-based metabonomic techniques for the analysis of the biochemical effects of dietary isoflavones on human plasma profile. Anal Biochem 323, 197204.CrossRefGoogle Scholar
333Wang, Y, Tang, H, Nicholson, JK, et al. . (2005) A metabonomic strategy for the detection of the metabolic effects of chamomile (Matricaria recutita L.) ingestion. J Agric Food Chem 53, 191196.CrossRefGoogle ScholarPubMed
334Van Dorsten, FA, Daykin, CA, Mulder, TPJ, et al. . (2006) Metabonomics approach to determine metabolic differences between green tea and black tea consumption. J Agric Food Chem 54, 69296938.CrossRefGoogle ScholarPubMed
335Solanky, KS, Bailey, NJC, Holmes, E, et al. . (2003) NMR-based metabonomic studies on the biochemical effects of epicatechin in the rat. J Agric Food Chem 51, 41394145.CrossRefGoogle ScholarPubMed
336Fardet, A, Llorach, R, Martin, JF, et al. . (2008) A liquid chromatography–quadrupole time-of-flight (LC-QTOF)-based metabolomic approach reveals new metabolic effects of catechin in rats fed high-fat diets. J Proteome Res 7, 23882398.CrossRefGoogle ScholarPubMed
337Dumas, ME, Barton, RH, Toye, A, et al. . (2006) Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A 103, 1251112516.CrossRefGoogle ScholarPubMed
338Zhang, S, Nagana Gowda, GA, Asiago, V, et al. . (2008) Correlative and quantitative 1H NMR-based metabolomics reveals specific metabolic pathway disturbances in diabetic rats. Anal Biochem 383, 7684.CrossRefGoogle ScholarPubMed
339Britz, SJ, Prasad, PVV, Moreau, RA, et al. . (2007) Influence of growth temperature on the amounts of tocopherols, tocotrienols, and γ-oryzanol in brown rice. J Agric Food Chem 55, 75597565.CrossRefGoogle ScholarPubMed
340Packer, L, Witt, EH & Tritschler, HJ (1995) α-Lipoic acid as a biological antioxidant. Free Radic Biol Med 19, 227250.CrossRefGoogle ScholarPubMed
341Roy, S, Sen, CK, Tritschler, HJ, et al. . (1997) Modulation of cellular reducing equivalent homeostasis by α-lipoic acid: mechanisms and implications for diabetes and ischemic injury. Biochem Pharmacol 53, 393399.CrossRefGoogle ScholarPubMed
342Maczurek, A, Hager, K, Kenklies, M, et al. . (2008) Lipoic acid as an anti-inflammatory and neuroprotective treatment for Alzheimer's disease. Adv Drug Deliv Rev 60, 14631470.CrossRefGoogle ScholarPubMed
343Wollin, SD & Jones, PJH (2003) α-Lipoic acid and cardiovascular disease. J Nutr 133, 33273330.CrossRefGoogle ScholarPubMed
344Yu, SG, Nehus, ZT, Badger, TM, et al. . (2007) Quantification of vitamin E and γ-oryzanol components in rice germ and bran. J Agric Food Chem 55, 73087313.CrossRefGoogle ScholarPubMed
345Emmons, CL, Peterson, DM & Paul, GL (1999) Antioxidant capacity of oat (Avena sativa L.) extracts. 2. In vitro antioxidant activity and contents of phenolic and tocol antioxidants. J Agric Food Chem 47, 48944898.CrossRefGoogle ScholarPubMed
346Heinemann, T, Kullak-Ublick, G-A, Pietruck, B, et al. . (1991) Mechanisms of action of plant sterols on inhibition of cholesterol absorption. Eur J Clin Pharmacol 40, S59S63.CrossRefGoogle ScholarPubMed
347Sen, CK, Khanna, S & Roy, S (2006) Tocotrienols: vitamin E beyond tocopherols. Life Sci 78, 20882098.CrossRefGoogle ScholarPubMed
348Wilson, TA, Nicolosi, RJ, Woolfrey, B, et al. . (2007) Rice bran oil and oryzanol reduce plasma lipid and lipoprotein cholesterol concentrations and aortic cholesterol ester accumulation to a greater extent than ferulic acid in hypercholesterolemic hamsters. J Nutr Biochem 18, 105112.CrossRefGoogle ScholarPubMed
349Kahlon, TS & Chow, FI (2000) In vitro binding of bile acids by rice bran, oat bran, wheat bran, and corn bran. Cereal Chem 77, 518521.CrossRefGoogle Scholar
350Saulnier, L, Vigouroux, J & Thibault, J-F (1995) Isolation and partial characterization of feruloylated oligosaccharides from maize bran. Carbohydr Res 272, 241253.CrossRefGoogle ScholarPubMed
351Saulnier, L, Marot, C, Elgorriaga, M, et al. . (2001) Thermal and enzymatic treatments for the release of free ferulic acid from maize bran. Carbohydr Polym 45, 269275.CrossRefGoogle Scholar
352O'Dell, BL, De Boland, AR & Koityonann, SR (1972) Distribution of phytate and nutritionally important elements among the morphological components of cereals grains. J Agric Food Chem 20, 718721.CrossRefGoogle Scholar
353Miller, A & Engel, KH (2006) Content of γ-oryzanol and composition of steryl ferulates in brown rice (Oryza sativa L.) of European origin. J Agric Food Chem 54, 81278133.CrossRefGoogle Scholar
354Chen, MH & Bergman, CJ (2005) A rapid procedure for analysing rice bran tocopherol, tocotrienol and γ-oryzanol contents. J Food Comp Anal 18, 139151.CrossRefGoogle Scholar
355Schramm, R, Abadie, A, Hua, N, et al. . (2007) Fractionation of the rice bran layer and quantification of vitamin E, oryzanol, protein, and rice bran saccharide. J Biol Eng 1, 9.CrossRefGoogle ScholarPubMed
356Shin, T-S, Godber, JS, Martin, DE, et al. . (1997) Hydrolytic stability and changes in E vitamers and oryzanol of extruded rice bran during storage. J Food Sci 62, 704728.CrossRefGoogle Scholar
357Juliano, C, Cossu, M, Alamanni, MC, et al. . (2005) Antioxidant activity of γ-oryzanol: mechanism of action and its effect on oxidative stability of pharmaceutical oils. Int J Pharm 299, 146154.CrossRefGoogle ScholarPubMed
358Rong, N, Ausman, L & Nicolosi, R (1997) Oryzanol decreases cholesterol absorption and aortic fatty streaks in hamsters. Lipids 32, 303309.CrossRefGoogle ScholarPubMed
359Seetharamaiah, GS & Chandrasekhara, N (1993) Comparative hypocholesterolemic activities of oryzanol, curcumin and ferulic acid in rats. J Food Sci Technol Mysore 30, 249252.Google Scholar
360Suh, MH, Yoo, SH, Chang, PS, et al. . (2005) Antioxidative activity of microencapsulated γ-oryzanol on high cholesterol-fed rats. J Agric Food Chem 53, 97479750.CrossRefGoogle ScholarPubMed
361Cicero, AFG & Gaddi, A (2001) Rice bran oil and γ-oryzanol in the treatment of hyperlipoproteinaemias and other conditions. Phytother Res 15, 277289.CrossRefGoogle ScholarPubMed
362Collins, FW (1989) Oat phenolics: avenanthramides, novel substituted N-cinnamoylanthranilate alkaloids from oat groats and hulls. J Agric Food Chem 37, 6066.CrossRefGoogle Scholar
363Shewry, PR, Piironen, V, Lampi, A-M, et al. . (2008) Phytochemical and fiber components in oat varieties in the HEALTHGRAIN diversity screen. J Agric Food Chem 56, 97779784.CrossRefGoogle ScholarPubMed
364Dimberg, LH, Theander, O & Lingnert, H (1993) Avenanthramides – a group of phenolic antioxidants in oats. Cereal Chem 70, 637641.Google Scholar
365Mattila, P, Pihlava, J-M & Hellstrom, J (2005) Contents of phenolic acids, alkyl- and alkenylresorcinols, and avenanthramides in commercial grain products. J Agric Food Chem 53, 82908295.CrossRefGoogle ScholarPubMed
366Fagerlund, A, Sunnerheim, K & Dimberg, LH (2009) Radical-scavenging and antioxidant activity of avenanthramides. Food Chem 113, 550556.CrossRefGoogle Scholar
367Peterson, DM, Hahn, MJ & Emmons, CL (2002) Oat avenanthramides exhibit antioxidant activities in vitro. Food Chem 79, 473478.CrossRefGoogle Scholar
368Liu, L, Zubik, L, Collins, FW, et al. . (2004) The antiatherogenic potential of oat phenolic compounds. Atherosclerosis 175, 3949.CrossRefGoogle ScholarPubMed
369Chen, CY, Milbury, PE, Kwak, HK, et al. . (2004) Avenanthramides and phenolic acids from oats are bioavailable and act synergistically with vitamin C to enhance hamster and human LDL resistance to oxidation. J Nutr 134, 14591466.CrossRefGoogle ScholarPubMed
370Güçlü-Üstündag, Ö & Mazza, G (2007) Saponins: properties, applications and processing. Crit Rev Food Sci Nutr 47, 231258.CrossRefGoogle ScholarPubMed
371Osbourn, AE (2003) Saponins in cereals. Phytochemistry 62, 14.CrossRefGoogle ScholarPubMed
372Önning, G, Asp, N-G & Sivik, B (1993) Saponin content in different oat varieties and in different fractions of oat grain. Food Chem 48, 251254.CrossRefGoogle Scholar
373Price, KR, Johnson, IT & Fenwick, GR (1987) The chemistry and biological significance of saponins in foods and feedingstuffs. Crit Rev Food Sci Nutr 26, 27135.CrossRefGoogle ScholarPubMed
374Matsuura, H (2001) Saponins in garlic as modifiers of the risk of cardiovascular disease. J Nutr 131, 1000S1005S.CrossRefGoogle ScholarPubMed
375Barr, IG, Sjölander, A & Cox, JC (1998) ISCOMs and other saponin based adjuvants. Adv Drug Deliv Rev 32, 247271.CrossRefGoogle Scholar
376Sjölander, A, Cox, J & Barr, I (1998) ISCOMs: an adjuvant with multiple functions. J Leukoc Biol 64, 713723.CrossRefGoogle ScholarPubMed
377Oakenfull, DG, Fenwick, DE, Hood, RL, et al. . (1979) Effects of saponins on bile acids and plasma lipids in the rat. Br J Nutr 42, 209216.CrossRefGoogle ScholarPubMed
378Baik, B-K & Ullrich, SE (2008) Barley for food: characteristics, improvement, and renewed interest. J Cereal Sci 48, 233242.CrossRefGoogle Scholar
379Åman, P & Graham, H (1987) Analysis of total and insoluble mixed-linked (1 → 3),(1 → 4)-β-d-glucans in barley and oats. J Agric Food Chem 35, 704709.CrossRefGoogle Scholar
380Anker-Nilssen, K, Sahlstrøm, S, Knutsen, SH, et al. . (2008) Influence of growth temperature on content, viscosity and relative molecular weight of water-soluble β-glucans in barley (Hordeum vulgare L.). J Cereal Sci 48, 670677.CrossRefGoogle Scholar
381Gajdosová, A, Petruláková, Z, Havrlentová, M, et al. . (2007) The content of water-soluble and water-insoluble β-d-glucans in selected oats and barley varieties. Carbohydr Polym 70, 4652.CrossRefGoogle Scholar
382Holtekjølen, AK, Uhlen, AK, Bråthen, E, et al. . (2006) Contents of starch and non-starch polysaccharides in barley varieties of different origin. Food Chem 94, 348358.CrossRefGoogle Scholar
383Izydorczyk, MS & Dexter, JE (2008) Barley β-glucans and arabinoxylans: molecular structure, physicochemical properties, and uses in food products – a review. Food Res Int 41, 850868.CrossRefGoogle Scholar
384Izydorczyk, MS, Macri, LJ & MacGregor, AW (1998) Structure and physicochemical properties of barley non-starch polysaccharides – II. Alkaliextractable β-glucans and arabinoxylans. Carbohydr Polym 35, 259269.CrossRefGoogle Scholar
385Izydorczyk, MS, Storsley, J, Labossiere, D, et al. . (2000) Variation in total and soluble β-glucan content in hulless barley: effects of thermal, physical, and enzymic treatments. J Agric Food Chem 48, 982989.CrossRefGoogle ScholarPubMed
386Prentice, N, Babler, S & Faber, S (1980) Enzymic analysis of β-d-glucans in cereal grains. Cereal Chem 57, 198202.Google Scholar
387Kim, H, Stote, KS, Behall, KM, et al. . (2009) Glucose and insulin responses to whole grain breakfasts varying in soluble fiber, β-glucan: a doase response study in obese women with increased risk for insulin resistance. Eur J Nutr 48, 170175.CrossRefGoogle ScholarPubMed
388Butt, MS, Tahir-Nadeem, M, Khan, MKI, et al. . (2008) Oat: unique among the cereals. Eur J Nutr 47, 6879.CrossRefGoogle Scholar
389Kalra, S & Joad, S (2000) Effect of dietary barley β-glucan on cholesterol and lipoprotein fractions in rats. J Cereal Sci 31, 141145.CrossRefGoogle Scholar
390Hlebowicz, J, Darwiche, G, Bjorgell, O, et al. . (2008) Effect of muesli with 4 g oat β-glucan on postprandial blood glucose, gastric emptying and satiety in healthy subjects: a randomized crossover trial. J Am Coll Nutr 27, 470475.CrossRefGoogle ScholarPubMed
391Mantovani, MS, Bellini, MF, Angeli, JPF, et al. . (2008) β-Glucans in promoting health: prevention against mutation and cancer. Mutat Res 658, 154161.CrossRefGoogle ScholarPubMed
392Ross, AB, Shepherd, MJ, Schupphaus, M, et al. . (2003) Alkylresorcinols in cereals and cereal products. J Agric Food Chem 51, 41114118.CrossRefGoogle ScholarPubMed
393Ross, AB, Kamal-Eldin, A, Lundin, EA, et al. . (2003) Cereal alkylresorcinols are absorbed by humans. J Nutr 133, 22222224.CrossRefGoogle ScholarPubMed
394Landberg, R, Aman, P, Friberg, LE, et al. . (2009) Dose response of whole-grain biomarkers: alkylresorcinols in human plasma and their metabolites in urine in relation to intake. Am J Clin Nutr 89, 290296.CrossRefGoogle ScholarPubMed
395Linko-Parvinen, A-M, Landberg, R, Tikkanen, MJ, et al. . (2007) Alkylresorcinols from whole-grain wheat and rye are transported in human plasma lipoproteins. J Nutr 137, 11371142.CrossRefGoogle ScholarPubMed
396Ross, AB, Kamal-Eldin, A & Aman, P (2004) Dietary alkylresorcinols: absorption, bioactivities, and possible use as biomarkers of whole-grain wheat- and rye-rich foods. Nutr Rev 62, 8195.CrossRefGoogle ScholarPubMed
397Guyman, LA, Adlercreutz, H, Koskela, A, et al. . (2008) Urinary 3-(3,5-dihydroxyphenyl)-1-propanoic acid, an alkylresorcinol metabolite, is a potential biomarker of whole-grain intake in a U.S. population. J Nutr 138, 19571962.CrossRefGoogle Scholar
398Tsuge, N, Mizokami, M, Imai, S, et al. . (1992) Adipostatin-A and adipostatin-B, new inhibitors of glycerol-3-phosphate dehydrogenase. J Antibiot 45, 886891.CrossRefGoogle ScholarPubMed
399Ross, AB, Chen, Y, Frank, J, et al. . (2004) Cereal alkylresorcinols elevate γ-tocopherol levels in rats and inhibit γ-tocopherol metabolism in vitro. J Nutr 134, 506510.Google ScholarPubMed
400Kozubek, A & Tyman, JHP (1999) Resorcinolic lipids, the natural non-isoprenoid phenolic amphiphiles and their biological activity. Chem Rev 99, 125.CrossRefGoogle ScholarPubMed
401Ross, JA & Kasum, CM (2002) Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr 22, 1934.CrossRefGoogle ScholarPubMed
402Norton, I, Moore, S & Fryer, P (2007) Understanding food structuring and breakdown: engineering approaches to obesity. Obes Rev 8, Suppl. 1, 8388.CrossRefGoogle ScholarPubMed
403Parada, J & Aguilera, JM (2007) Food microstructure affects the bioavailability of several nutrients. J Food Sci 72, R21R32.CrossRefGoogle ScholarPubMed
404Tedeschi, C, Clement, V, Rouvet, M, et al. . (2009) Dissolution tests as a tool for predicting bioaccessibility of nutrients during digestion. Food Hydrocolloids 23, 12281235.CrossRefGoogle Scholar
405Englyst, H, Kingman, S & Cummings, J (1992) Classification and measurement of nutritionally important starch fractions. Eur J Clin Nutr 46, S33S50.Google ScholarPubMed
406Lehmann, U & Robin, F (2007) Slowly digestible starch – its structure and health implications: a review. Trends Food Sci Technol 18, 346355.CrossRefGoogle Scholar
407Marangoni, A, Idziak, S & Rush, J (2008) Controlled release of food lipids using monoglyceride gel phases regulates lipid and insulin metabolism in humans. Food Biophys 3, 241245.CrossRefGoogle Scholar
408Remond, D, Machebeuf, M, Yven, C, et al. . (2007) Postprandial whole-body protein metabolism after a meat meal is influenced by chewing efficiency in elderly subjects. Am J Clin Nutr 85, 12861292.CrossRefGoogle ScholarPubMed
409Armand, M, Pasquier, B, Andre, M, et al. . (1999) Digestion and absorption of 2 fat emulsions with different droplet sizes in the human digestive tract. Am J Clin Nutr 70, 10961106.CrossRefGoogle ScholarPubMed
410Giovannucci, E, Rimm, EB, Ascherio, A, et al. . (1995) Alcohol, low-methionine–low-folate diets, and risk of colon cancer in men. J Natl Cancer Inst 87, 265273.CrossRefGoogle ScholarPubMed
411Lu, ZX, Walker, KZ, Muir, JG, et al. . (2000) Arabinoxylan fiber, a byproduct of wheat flour processing, reduces the postprandial glucose response in normoglycemic subjects. Am J Clin Nutr 71, 11231128.CrossRefGoogle ScholarPubMed
412Moore, MA, Park, CB & Tsuda, H (1998) Soluble and insoluble fiber influences on cancer development. Crit Rev Oncol Hematol 27, 229242.CrossRefGoogle ScholarPubMed
413Björck, I & Asp, N-G (1994) Controlling the nutritional properties of starch in foods – a challenge to the food industry. Trends Food Sci Technol 5, 213218.CrossRefGoogle Scholar
414Dongowski, G, Jacobasch, G & Schmiedl, D (2005) Structural stability and prebiotic properties of resistant starch type 3 increase bile acid turnover and lower secondary bile acid formation. J Agric Food Chem 53, 92579267.CrossRefGoogle ScholarPubMed
415Topping, DL, Fukushima, M & Bird, AR (2003) Resistant starch as a prebiotic and synbiotic: state of the art. Proc Nutr Soc 62, 171176.CrossRefGoogle ScholarPubMed
416Rahman, S, Bird, A, Regina, A, et al. . (2007) Resistant starch in cereals: exploiting genetic engineering and genetic variation. J Cereal Sci 46, 251260.CrossRefGoogle Scholar
417Goddard, MS, Young, G & Marcus, R (1984) The effect of amylose content on insulin and glucose responses to ingested rice. Am J Clin Nutr 39, 388392.Google ScholarPubMed
418Hallfrisch, J & Behall, KM (2000) Mechanisms of the effects of grains on insulin and glucose responses. J Am Coll Nutr 19, 320S325S.CrossRefGoogle ScholarPubMed
419Hawkesford, MJ & Zhao, F-J (2007) Strategies for increasing the selenium content of wheat. J Cereal Sci 46, 282292.CrossRefGoogle Scholar
420Singh, BR (1991) Selenium content of wheat as affected by selenate and selenite contained in a Cl- or SO4-based NPK fertilizer. Fertilizer Res 30, 17.CrossRefGoogle Scholar
421Soliman, MF (1980) Zinc uptake by wheat plants as influenced by nitrogen fertilizers and calcium carbonate. Agric Res Rev 58, 113121.Google Scholar
422Fallahi, E, Mohtadinia, J & Ali Mahboob, S (2005) Effect of consumption of whole bread baked from cultivated wheat with micronutrient fertilizers on blood indices of iron. J Food Agric Environ 3, 3942.Google Scholar
423Hanson, AD & Wyse, R (1982) Biosynthesis, translocation, and accumulation of betaine in sugar beet and its progenitors in relation to salinity. Plant Physiol 70, 11911198.CrossRefGoogle ScholarPubMed
424Keles, Y & Öncel, I (2002) Response of antioxidative defence system to temperature and water stress combinations in wheat seedlings. Plant Sci 163, 783790.CrossRefGoogle Scholar
425King, JC (2002) Biotechnology: a solution for improving nutrient bioavailability. Int J Vitam Nutr Res 72, 712.CrossRefGoogle ScholarPubMed
426Cakmak, I, Ozkan, H, Braun, HJ, et al. . (2000) Zinc and iron concentrations in seeds of wild, primitive, and modern wheats. Food Nutr Bull 21, 401403.CrossRefGoogle Scholar
427Ortiz-Monasterio, JI, Palacios-Rojas, N, Meng, E, et al. . (2007) Enhancing the mineral and vitamin content of wheat and maize through plant breeding. J Cereal Sci 46, 293307.CrossRefGoogle Scholar
428Mendoza, C, Viteri, F, Lonnerdal, B, et al. . (1998) Effect of genetically modified, low-phytic acid maize on absorption of iron from tortillas. Am J Clin Nutr 68, 11231127.CrossRefGoogle ScholarPubMed
429Raboy, V (2002) Progress in breeding low phytate crops. J Nutr 132, 503S505S.CrossRefGoogle ScholarPubMed
430King, RA, Noakes, M, Bird, AR, et al. . (2008) An extruded breakfast cereal made from a high amylose barley cultivar has a low glycemic index and lower plasma insulin response than one made from a standard barley. J Cereal Sci 48, 526530.CrossRefGoogle Scholar
431Regina, A, Bird, A, Topping, D, et al. . (2006) High-amylose wheat generated by RNA interference improves indices of large-bowel health in rats. Proc Natl Acad Sci U S A 103, 35463551.CrossRefGoogle ScholarPubMed
432Saulnier, L, Sado, P-E, Branlard, G, et al. . (2007) Wheat arabinoxylans: exploiting variation in amount and composition to develop enhanced varieties. J Cereal Sci 46, 261281.CrossRefGoogle Scholar
433Brinch-Pedersen, H, Borg, S, Tauris, B, et al. . (2007) Molecular genetic approaches to increasing mineral availability and vitamin content of cereals. J Cereal Sci 46, 308326.CrossRefGoogle Scholar
434Hammes, WP, Brandt, MJ, Francis, KL, et al. . (2005) Microbial ecology of cereal fermentations. Trends Food Sci Technol 16, 411.CrossRefGoogle Scholar
435Nout, MJR (2009) Rich nutrition from the poorest – cereal fermentations in Africa and Asia. Food Microbiology 26, 685692.CrossRefGoogle ScholarPubMed
436Napolitano, A, Lanzuise, S, Ruocco, M, et al. . (2006) Treatment of cereal products with a tailored preparation of Trichoderma enzymes increases the amount of soluble dietary fiber. J Agric Food Chem 54, 78637869.CrossRefGoogle ScholarPubMed
437Faulds, CB & Williamson, G (1995) Release of ferulic acid from wheat bran by a ferulic acid esterase (FAE-III) from Aspergillus niger. Appl Microbiol Biotechnol 43, 10821087.CrossRefGoogle ScholarPubMed
438Wang, XK, Geng, X, Egashira, Y, et al. . (2005) Release of ferulic acid from wheat bran by an inducible feruloyl esterase from an intestinal bacterium Lactobacillus acidophilus. Food Sci Technol Res 11, 241247.CrossRefGoogle Scholar
439Chavan, JK & Kadam, SS (1989) Nutritional improvement of cereals by fermentation. Crit Rev Food Sci Nutr 28, 349400.CrossRefGoogle ScholarPubMed
440Gadaga, TH, Mutukumira, AN, Narvhus, JA, et al. . (1999) A review of traditional fermented foods and beverages of Zimbabwe. Int J Food Microbiol 53, 111.CrossRefGoogle ScholarPubMed
441Lioger, D, Leenhardt, F & Rémésy, C (2006) Intérêt de la fermentation, en milieu très hydraté, des produits céréaliers riches en fibres pour améliorer leur valeur nutritionnelle (Interest of fibre-rich cereal products fermentation in very hydrated environment to improve their nutritional value). Ind Cér 149, 1422.Google Scholar
442Poutanen, K, Flander, L & Katina, K (2009) Sourdough and cereal fermentation in a nutritional perspective. Food Microbiol 26, 693699.CrossRefGoogle Scholar
443Abd Elmoneim, OE, Schiffler, B & Bernhard, R (2004) Effect of fermentation on the starch digestibility, resistant starch and some physicochemical properties of sorghum flour. Nahrung/Food 48, 9194.Google Scholar
444Eklund-Jonsson, C, Sandberg, A-S & Larsson Alminger, M (2006) Reduction of phytate content while preserving minerals during whole grain cereal tempe fermentation. J Cereal Sci 44, 154160.CrossRefGoogle Scholar
445El Hag, ME, El Tinay, AH & Yousif, NE (2002) Effect of fermentation and dehulling on starch, total polyphenols, phytic acid content and in vitro protein digestibility of pearl millet. Food Chem 77, 193196.CrossRefGoogle Scholar
446Loponen, J, Kanerva, P, Zhang, C, et al. . (2009) Prolamin hydrolysis and pentosan solubilization in germinated-rye sourdoughs determined by chromatographic and immunological methods. J Agric Food Chem 57, 746753.CrossRefGoogle ScholarPubMed
447Mugula, JK, Sorhaug, T & Stepaniak, L (2003) Proteolytic activities in togwa, a Tanzanian fermented food. Int J Food Microbiol 84, 112.CrossRefGoogle ScholarPubMed
448Thiele, C, Grassl, S & Ganzle, M (2004) Gluten hydrolysis and depolymerization during sourdough fermentation. J Agric Food Chem 52, 13071314.CrossRefGoogle ScholarPubMed
449Wedad, HA, El-Tinay, AH, Mustafa, AI, et al. . (2008) Effect of fermentation, malt-pretreatment and cooking on antinutritional factors and protein digestibility of sorghum cultivars. Pak J Nutr 7, 335341.CrossRefGoogle Scholar
450Lopez, HW, Krespine, V, Guy, C, et al. . (2001) Prolonged fermentation of whole wheat sourdough reduces phytate level and increases soluble magnesium. J Agric Food Chem 49, 26572662.CrossRefGoogle ScholarPubMed
451Lopez, HW, Duclos, V, Coudray, C, et al. . (2003) Making bread with sourdough improves mineral bioavailability from reconstituted whole wheat flour in rats. Nutrition 19, 524530.CrossRefGoogle ScholarPubMed
452Katina, K, Liukkonen, K-H, Kaukovirta-Norja, A, et al. . (2007) Fermentation-induced changes in the nutritional value of native or germinated rye. J Cereal Sci 46, 348355.CrossRefGoogle Scholar
453Winata, A & Lorenz, K (1997) Effects of fermentation and baking of whole wheat and whole rye sourdough breads on cereal alkylresorcinols. Cereal Chem 74, 284287.CrossRefGoogle Scholar
454Katina, K, Laitila, A, Juvonen, R, et al. . (2007) Bran fermentation as a means to enhance technological properties and bioactivity of rye. Food Microbiol 24, 175186.CrossRefGoogle ScholarPubMed
455Moore, J, Luther, M, Cheng, Z, et al. . (2009) Effects of baking conditions, dough fermentation, and bran particle size on antioxidant properties of whole-wheat pizza crusts. J Agric Food Chem 57, 832839.CrossRefGoogle ScholarPubMed
456Garcia, AL, Otto, B, Reich, SC, et al. . (2007) Arabinoxylan consumption decreases postprandial serum glucose, serum insulin and plasma total ghrelin response in subjects with impaired glucose tolerance. Eur J Clin Nutr 61, 334341.CrossRefGoogle ScholarPubMed
457Lioger, D, Fardet, A, Foassert, P, et al. . (2009) Influence of sourdough prefermentation, of steam cooking suppression and of decreased sucrose content during wheat flakes processing on the plasma glucose and insulin responses and satiety of healthy subjects. J Am Coll Nutr 28, 3036.CrossRefGoogle ScholarPubMed
458Liljeberg, H & Bjorck, I (1996) Delayed gastric emptying rate as a potential mechanism for lowered glycemia after eating sourdough bread: studies in humans and rats using test products with added organic acids or an organic salt. Am J Clin Nutr 64, 886893.CrossRefGoogle ScholarPubMed
459Liljeberg, HG, Lonner, CH & Bjorck, IM (1995) Sourdough fermentation or addition of organic acids or corresponding salts to bread improves nutritional properties of starch in healthy humans. J Nutr 125, 15031511.Google ScholarPubMed
460Brennan, CS, Blake, DE, Ellis, PR, et al. . (1996) Effects of guar galactomannan on wheat bread microstructure and on the in vitro and in vivo digestibility of starch in bread. J Cereal Sci 24, 151160.CrossRefGoogle Scholar
461Burton, P & Lightowler, HJ (2006) Influence of bread volume on glycaemic response and satiety. Br J Nutr 96, 877882.CrossRefGoogle ScholarPubMed
462Granfeldt, Y, Eliasson, AC & Bjorck, I (2000) An examination of the possibility of lowering the glycemic index of oat and barley flakes by minimal processing. J Nutr 130, 22072214.CrossRefGoogle ScholarPubMed
463Antoine, C, Lullien-Pellerin, V, Abecassis, J, et al. . (2002) Nutritional interest of the wheat seed aleurone layer. Sci Alim 22, 545556.Google Scholar
464Buri, RC, von Reding, W & Gavin, MH (2004) Description and characterization of wheat aleurone. Cereal Foods World 49, 274282.Google Scholar
465Harris, PJ, Chavan, RR & Ferguson, LR (2005) Production and characterisation of two wheat-bran fractions: an aleurone-rich and a pericarp-rich fraction. Mol Nutr Food Res 49, 536545.CrossRefGoogle Scholar
466Hemery, Y, Rouau, X, Lullien-Pellerin, V, et al. . (2007) Dry processes to develop wheat fractions and products with enhanced nutritional quality. J Cereal Sci 46, 327347.CrossRefGoogle Scholar
467Fenech, M, Noakes, M, Clifton, P, et al. . (1999) Aleurone flour is a rich source of bioavailable folate in humans. J Nutr 129, 11141119.CrossRefGoogle ScholarPubMed
468Fenech, M, Noakes, M, Clifton, P, et al. . (2005) Aleurone flour increases red-cell folate and lowers plasma homocyst(e)ine substantially in man. Br J Nutr 93, 353360.CrossRefGoogle ScholarPubMed
469Cheng, BO, Trimble, RP, Illman, RJ, et al. . (1987) Comparative effects of dietary wheat bran and its morphological components (aleurone and pericarp-seed coat) on volatile fatty acid concentrations in the rat. Br J Nutr 57, 6976.CrossRefGoogle ScholarPubMed
470McIntosh, GH, Royle, PJ & Pointing, G (2001) Wheat aleurone flour increases cecal β-glucuronidase activity and butyrate concentration and reduces colon adenoma burden in azoxymethane-treated rats. J Nutr 131, 127131.CrossRefGoogle ScholarPubMed
471Amrein, TM, Granicher, P, Arrigoni, E, et al. . (2003) In vitro digestibility and colonic fermentability of aleurone isolated from wheat bran. Lebensm-Wiss Technol Food Sci Technol 36, 451460.CrossRefGoogle Scholar
472Fardet, A, Hoebler, C, Baldwin, PM, et al. . (1998) Involvement of the protein network in the in vitro degradation of starch from spaghetti and lasagne: a microscopic and enzymic study. J Cereal Sci 27, 133145.CrossRefGoogle Scholar
473Chu, FS & Li, GY (1994) Simultaneous occurrence of fumonisin B1 and other mycotoxins in moldy corn collected from the People's Republic of China in regions with high incidences of esophageal cancer. Appl Environ Microbiol 60, 847852.CrossRefGoogle ScholarPubMed
474Rheeder, JP, Marasas, WFO, Thiel, PG, et al. . (1992) Fusarium moniliforme and fumonisins in corn in relation to human esophageal cancer in Transkei. Phytopathology 82, 353357.CrossRefGoogle Scholar
475Lebailly, P, Niez, E & Baldi, I (2007) Données épidémiologiques sur le lien entre cancers et pesticides (Epidemiological data on the link between cancer and pesticides). Oncologie 9, 361369.CrossRefGoogle Scholar
476Surget, A & Barron, C (2005) Histologie du grain de blé (Histology of the wheat grain). Ind Cér 145, 37.Google Scholar
477Zeisel, SH, Mar, MH, Howe, JC, et al. . (2003) Concentrations of choline-containing compounds and betaine in common foods. J Nutr 133, 13021307.CrossRefGoogle ScholarPubMed
478Poutanen, K, Shepherd, R, Shewry, PR, et al. . (2008) Beyond whole grain: The European HEALTHGRAIN project aims at healthier cereal foods. Cereal Foods World 53, 3235.Google Scholar
479US Department of Agriculture (2005) USDA National Nutrient Database for Standard Reference, Release 18, Cereal grains and pasta. http://www.nal.usda.gov/fnic/foodcomp/Data/SR18/reports/sr18page.htm.Google Scholar
480Archer, MJ (1972) Relationship between free glutathione content and quality assessment parameters of wheat cultivars (Triticum aestivum L.). J Sci Food Agric 23, 485491.CrossRefGoogle Scholar
481Shewry, PR (2007) Improving the protein content and composition of cereal grain. J Cereal Sci 46, 239250.CrossRefGoogle Scholar
482Souci, SW, Fachmann, W & Kraut, H (2008) Food Composition and Nutritional Tables, 7th ed.. Stuttgart, Germany: MedPharm Scientific Publishers.Google Scholar
483Waggle, DH, Lambert, MA, Miller, GD, et al. . (1967) Extensive analyses of flours and millfeeds made from nine different wheat mixes. II. amino acids, minerals, vitamins, and gross energy. Cereal Chem 44, 4860.Google Scholar
484Colonna, P, Buléon, A, Leloup, V, et al. (1995) Constituants des céréales, des graines, des fruits et de leurs sous-produits (Constituents of grains, seeds, fruits and their by-products). In Nutrition des ruminants domestiques. Ingestion et digestion (Nutrition of domestic ruminants. Ingestion and digestion), chapter 3 [Jarrige, R, Ruckebusch, Y and Demarquilly, C, et al. ., editors]. Paris: INRA.Google Scholar
485Knudsen, KEB (1997) Carbohydrate and lignin contents of plant materials used in animal feeding. Anim Feed Sci Tech 67, 319338.CrossRefGoogle Scholar
486Gebruers, K, Dornez, E, Boros, D, et al. . (2008) Variation in the content of dietary fiber and components thereof in wheats in the HEALTHGRAIN Diversity Screen. J Agric Food Chem 56, 97409749.CrossRefGoogle ScholarPubMed
487Haskå, L, Nyman, M & Andersson, R (2008) Distribution and characterisation of fructan in wheat milling fractions. J Cereal Sci 48, 768774.CrossRefGoogle Scholar
488Hernot, DC, Boileau, TW, Bauer, LL, et al. . (2008) In vitro digestion characteristics of unprocessed and processed whole grains and their components. J Agric Food Chem 56, 1072110726.CrossRefGoogle ScholarPubMed
489Nyström, L, Paasonen, A, Lampi, A-M, et al. . (2007) Total plant sterols, steryl ferulates and steryl glycosides in milling fractions of wheat and rye. J Cereal Sci 45, 106115.CrossRefGoogle Scholar
490Picolli da Silva, L & de Lourdes Santorio Ciocca, M (2005) Total, insoluble and soluble dietary fiber values measured by enzymatic-gravimetric method in cereal grains. J Food Comp Anal 18, 113120.CrossRefGoogle Scholar
491Ragaee, SM, Campbell, GL, Scoles, GJ, et al. . (2001) Studies on rye (Secale cereale L.) lines exhibiting a range of extract viscosities. 1. Composition, molecular weight distribution of water extracts, and biochemical characteristics of purified water-extractable arabinoxylan. J Agric Food Chem 49, 24372445.CrossRefGoogle ScholarPubMed
492Ward, JL, Poutanen, K, Gebruers, K, et al. . (2008) The HEALTHGRAIN Cereal Diversity Screen: concept, results, and prospects. J Agric Food Chem 56, 96999709.CrossRefGoogle Scholar
493Abdel-Aal, E-SM & Hucl, P (1999) A rapid method for quantifying total anthocyanins in blue aleurone and purple pericarp wheats. Cereal Chem 76, 350354.CrossRefGoogle Scholar
494Anderson, J & Bridges, S (1988) Dietary fiber content of selected foods. Am J Clin Nutr 47, 440447.CrossRefGoogle ScholarPubMed
495Fretzdorff, B & Welge, N (2003) Fructan and raffinose contents in cereals and pseudo-cereal grains. Getreide, Mehl und Brot 57, 38.Google Scholar
496Huynh, BL, Palmer, L, Mather, DE, et al. . (2008) Genotypic variation in wheat grain fructan content revealed by a simplified HPLC method. J Cereal Sci 48, 369378.CrossRefGoogle Scholar
497Huynh, BL, Wallwork, H, Stangoulis, JCR, et al. . (2008) Quantitative trait loci for grain fructan concentration in wheat (Triticum aestivum L.). Theor Appl Genet 117, 701709.CrossRefGoogle ScholarPubMed
498Henry, RJ (1987) Pentosan and (1 → 3),(1 → 4)-β-glucan concentrations in endosperm and wholegrain of wheat, barley, oats and rye. J Cereal Sci 6, 253258.CrossRefGoogle Scholar
499Lempereur, I, Rouau, X & Abecassis, J (1997) Genetic and agronomic variation in arabinoxylan and ferulic acid contents of durum wheat (Triticum durum L.) grain and its milling fractions. J Cereal Sci 25, 103110.CrossRefGoogle Scholar
500Genç, H, Özdemir, M & Demirbas, A (2001) Analysis of mixed-linked (1 → 3), (1 → 4)-β-d-glucans in cereal grains from Turkey. Food Chem 73, 221224.CrossRefGoogle Scholar
501Hemery, Y, Lullien-Pellerin, V, Rouau, X, et al. . (2009) Biochemical markers: efficient tools for the assessment of wheat grain tissue proportions in milling fractions. J Cereal Sci 49, 5564.CrossRefGoogle Scholar
502House, WA & Welch, RM (1987) Bioavailability to rats of iron in 6 varieties of wheat-grain intrinsically labeled with radioiron. J Nutr 117, 476480.CrossRefGoogle Scholar
503Lopez, HW, Krespine, V, Lemaire, A, et al. . (2003) Wheat variety has a major influence on mineral bioavailability; studies in rats. J Cereal Sci 37, 257266.CrossRefGoogle Scholar
504O'Dell, BL, Burpo, CE & Savage, JE (1972) Evaluation of zinc availability in foodstuffs of plant and animal origin. J Nutr 102, 653660.CrossRefGoogle ScholarPubMed
505Tabekhia, MM & Donnelly, BJ (1982) Phytic acid in durum-wheat and its milled products. Cereal Chem 59, 105107.Google Scholar
506Tariq, M, Talat, M, Asia, L, et al. . (2007) Influence of processing and cooking methodologies for reduction of phytic acid content in wheat (Triticum aestivum) varieties. J Food Process Preserv 31, 583594.Google Scholar
507Davis, KR, Peters, LJ, Cain, RF, et al. . (1984) Evaluation of the nutrient composition of wheat. III. Minerals. Cereal Foods World 29, 246248.Google Scholar
508Frossard, E, Bucher, M, Mächler, F, et al. . (2000) Potential for increasing the content and bioavailability of Fe, Zn and Ca in plants for human nutrition. J Sci Food Agric 80, 861879.3.0.CO;2-P>CrossRefGoogle Scholar
509Lorenz, K & Loewe, R (1977) Mineral composition of U.S. and Canadian wheats and wheat blends. J Agric Food Chem 25, 806809.CrossRefGoogle ScholarPubMed
510Monasterio, I & Graham, RD (2000) Breeding for trace minerals in wheat. Food Nutr Bull 21, 392396.CrossRefGoogle Scholar
511Tang, J, Zou, C, He, Z, et al. . (2008) Mineral element distributions in milling fractions of Chinese wheats. J Cereal Sci 48, 821828.CrossRefGoogle Scholar
512Zook, EG, Greene, FE & Morris, ER (1970) Nutrient composition of selected wheats and wheat products. VI. Distribution of manganese, copper, nickel, zinc, magnesium, lead, tin, cadmium, chromium, and selenium as determined by atomic absorption spectroscopy and colorimetry. Cereal Chem 47, 720731.Google Scholar
513Welch, RM & Graham, RD (2000) A new paradigm for world agriculture: productive, sustainable, nutritious, healthful food systems. Food Nutr Bull 21, 361366.CrossRefGoogle Scholar
514Fan, MS, Zhao, FJ, Poulton, PR, et al. . (2008) Historical changes in the concentrations of selenium in soil and wheat grain from the Broadbalk experiment over the last 160 years. Sci Total Environ 389, 532538.CrossRefGoogle ScholarPubMed
515Zhao, F, McGrath, S, Gray, C, et al. . (2007) Selenium concentrations in UK wheat and biofortification strategies. Comp Biochem Physiol A Mol Integr Physiol 146, Suppl. 1, S246.CrossRefGoogle Scholar
516Batifoulier, F, Verny, M-A, Chanliaud, E, et al. . (2005) Effect of different breadmaking methods on thiamine, riboflavin and pyridoxine contents of wheat bread. J Cereal Sci 42, 101108.CrossRefGoogle Scholar
517Davis, KR, Cain, RF, Peters, LJ, et al. . (1981) Evaluation of the nutrient composition of wheat. 2. Proximate analysis, thiamin, riboflavin, niacin, and pyridoxine. Cereal Chem 58, 116120.Google Scholar
518Davis, KR, Peters, LJ & Letourneau, D (1984) Variability of the vitamin content in wheat. Cereal Foods World 29, 364370.Google Scholar
519Ranhotra, G, Gelroth, J, Novak, F, et al. . (1985) Bioavailability for rats of thiamin in whole wheat and thiamin-restored white bread. J Nutr 115, 601606.CrossRefGoogle ScholarPubMed
520Gujska, E & Kuncewicz, A (2005) Determination of folate in some cereals and commercial cereal-grain products consumed in Poland using trienzyme extraction and high-performance liquid chromatography methods. Eur Food Res Technol 221, 208213.CrossRefGoogle Scholar
521Perloff, BP & Butrum, RR (1977) Folacin in selected foods. J Am Diet Assoc 70, 161172.CrossRefGoogle ScholarPubMed
522Piironen, V, Edelmann, M, Kariluoto, S, et al. . (2008) Folate in wheat genotypes in the HEALTHGRAIN Diversity Screen. J Agric Food Chem 56, 97269731.CrossRefGoogle ScholarPubMed
523Lampi, A-M, Nurmi, T, Ollilainen, V, et al. . (2008) Tocopherols and tocotrienols in wheat genotypes in the HEALTHGRAIN Diversity Screen. J Agric Food Chem 56, 97169721.CrossRefGoogle ScholarPubMed
524Nielsen, MM & Hansen, A (2008) Stability of vitamin E in wheat flour and whole wheat flour during storage. Cereal Chem 85, 716720.CrossRefGoogle Scholar
525Nielsen, MM & Hansen, A (2008) Rapid high-performance liquid chromatography determination of tocopherols and tocotrienols in cereals. Cereal Chem 85, 248251.CrossRefGoogle Scholar
526Panfili, G, Fratianni, A & Irano, M (2003) Normal phase high-performance liquid chromatography method for the determination of tocopherols and tocotrienols in cereals. J Agric Food Chem 51, 39403944.CrossRefGoogle ScholarPubMed
527Moore, J, Hao, Z, Zhou, K, et al. . (2005) Carotenoid, tocopherol, phenolic acid, and antioxidant properties of Maryland-grown soft wheat. J Agric Food Chem 53, 66496657.CrossRefGoogle ScholarPubMed
528Konopka, I, Kozirok, W & Rotkiewicz, D (2004) Lipids and carotenoids of wheat grain and flour and attempt of correlating them with digital image analysis of kernel surface and cross-sections. Food Res Int 37, 429438.CrossRefGoogle Scholar
529Leenhardt, F, Lyan, B, Rock, E, et al. . (2006) Genetic variability of carotenoid concentration, and lipoxygenase and peroxidase activities among cultivated wheat species and bread wheat varieties. Eur J Agron 25, 170176.CrossRefGoogle Scholar
530Panfili, G, Fratianni, A & Irano, M (2004) Improved normal-phase high-performance liquid chromatography procedure for the determination of carotenoids in cereals. J Agric Food Chem 52, 63736377.CrossRefGoogle ScholarPubMed
531Roose, M, Kahl, J & Ploeger, A (2009) Influence of the farming system on the xanthophyll content of soft and hard wheat. J Agric Food Chem 57, 182188.CrossRefGoogle ScholarPubMed
532Adom, KK & Liu, RH (2002) Antioxidant activity of grains. J Agric Food Chem 50, 61826187.CrossRefGoogle ScholarPubMed
533Barron, C, Surget, A & Rouau, X (2007) Relative amounts of tissues in mature wheat (Triticum aestivum L.) grain and their carbohydrate and phenolic acid composition. J Cereal Sci 45, 8896.CrossRefGoogle Scholar
534Lempereur, I, Surget, A & Rouau, X (1998) Variability in dehydrodiferulic acid composition of durum wheat (Triticum durum Desf.) and distribution in milling fractions. J Cereal Sci 28, 251258.CrossRefGoogle Scholar
535Mpofu, A, Sapirstein, HD & Beta, T (2006) Genotype and environmental variation in phenolic content, phenolic acid composition, and antioxidant activity of hard spring wheat. J Agric Food Chem 54, 12651270.CrossRefGoogle ScholarPubMed
536Abdel-Aal, ESM & Hucl, P (2003) Composition and stability of anthocyanins in blue-grained wheat. J Agric Food Chem 51, 21742180.CrossRefGoogle ScholarPubMed
537Abdel-Aal, E-SM, Abou-Arab, AA, Gamel, TH, et al. . (2008) Fractionation of blue wheat anthocyanin compounds and their contribution to antioxidant properties. J Agric Food Chem 56, 1117111177.CrossRefGoogle ScholarPubMed
538Liggins, J, Mulligan, A, Runswick, S, et al. . (2002) Daidzein and genistein content of cereals. Eur J Clin Nutr 56, 961966.CrossRefGoogle ScholarPubMed
539Dinelli, G, Marotti, I, Bosi, S, et al. . (2007) Lignan profile in seeds of modern and old Italian soft wheat (Triticum aestivum L.) cultivars as revealed by CE-MS analyses. Electrophoresis 28, 42124219.CrossRefGoogle ScholarPubMed
540Milder, IEJ, Feskens, EJM, Arts, ICW, et al. . (2005) Intake of the plant lignans secoisolariciresinol, matairesinol, lariciresinol, and pinoresinol in Dutch men and women. J Nutr 135, 12021207.CrossRefGoogle ScholarPubMed
541Andersson, AAM, Kamal-Eldin, A, Fraś, A, et al. . (2008) Alkylresorcinols in wheat varieties in the HEALTHGRAIN Diversity Screen. J Agric Food Chem 56, 97229725.CrossRefGoogle ScholarPubMed
542US Department of Agriculture ARS, Nutrient Data Laboratory, Patterson, KY & Bhagwat, SA et al. . (2008) USDA database for the choline content of common foods, release 2. http://www.nal.usda.gov/fnic/foodcomp/Data/Choline/Choln02.pdf (accessed 2008).Google Scholar
543Hakala, P, Lampi, A-M, Ollilainen, V, et al. . (2002) Steryl phenolic acid esters in cereals and their milling fractions. J Agric Food Chem 50, 53005307.CrossRefGoogle ScholarPubMed
544Iafelice, G, Verardo, V, Marconi, E, et al. . (2009) Characterization of total, free and esterified phytosterols in tetraploid and hexaploid wheats. J Agric Food Chem 57, 22672273.CrossRefGoogle ScholarPubMed
545Nurmi, T, Nyström, L, Edelmann, M, et al. . (2008) Phytosterols in wheat genotypes in the HEALTHGRAIN Diversity Screen. J Agric Food Chem 56, 97109715.CrossRefGoogle ScholarPubMed
546Piironen, V, Toivo, J & Lampi, AM (2002) Plant sterols in cereals and cereal products. Cereal Chem 79, 148154.CrossRefGoogle Scholar
547Irmak, S & Dunford, NT (2005) Policosanol contents and compositions of wheat varieties. J Agric Food Chem 53, 55835586.CrossRefGoogle ScholarPubMed
548Trautwein, EA (2001) n-3 Fatty acids – physiological and technical aspects for their use in food. Eur J Lipid Sci Technol 103, 4555.3.0.CO;2-9>CrossRefGoogle Scholar
549Every, D, Morrison, SC, Simmons, LD, et al. . (2006) Distribution of glutathione in millstreams and relationships to chemical and baking properties of flour. Cereal Chem 83, 5761.CrossRefGoogle Scholar
550Fraser, JR & Holmes, DC (1959) Proximate analysis of wheat flour carbohydrates. IV. – analysis of wholemeal flour and some of its fractions. J Sci Food Agric 10, 506512.CrossRefGoogle Scholar
551Saunders, RM & Walker, HG (1969) Sugars of wheat bran. Cereal Chem 46, 85.Google Scholar
552Chen, H, Haack, V, Janecky, C, et al. . (1998) Mechanisms by which wheat bran and oat bran increase stool weight in humans. Am J Clin Nutr 68, 711719.CrossRefGoogle ScholarPubMed
553Lehrfeld, J & Wu, YV (1991) Distribution of phytic acid in milled fractions of Scout-66 hard red winter-wheat. J Agric Food Chem 39, 18201824.CrossRefGoogle Scholar
554Maes, C, Vangeneugden, B & Delcour, JA (2004) Relative activity of two endoxylanases towards water-unextractable arabinoxylans in wheat bran. J Cereal Sci 39, 181186.CrossRefGoogle Scholar
555Esposito, F, Arlotti, G, Bonifati, AM, et al. . (2005) Antioxidant activity and dietary fibre in durum wheat bran by-products. Food Res Int 38, 11671173.CrossRefGoogle Scholar
556Gordon, DT & Chao, LS (1984) Relationship of components in wheat bran and spinach to iron bioavailability in the anemic rat. J Nutr 114, 526535.CrossRefGoogle ScholarPubMed
557Morris, ER & Ellis, R (1980) Bioavailability to rats of iron and zinc in wheat bran – response to low-phytate bran and effect of the phytate-zinc molar ratio. J Nutr 110, 20002010.CrossRefGoogle ScholarPubMed
558Anderson, NE & Clydesdale, FM (1980) An analysis of the dietary fiber content of a standard wheat bran. J Food Sci 45, 336340.CrossRefGoogle Scholar
559Bagheri, SM & Gueguen, L (1982) Bioavailability to rats of calcium, magnesium, phosphorus and zinc in wheat bran diets containing equal amounts of these minerals. Nutr Rep Int 25, 583589.Google Scholar
560Falcao-e-Cunha, L, Peres, H, Freire, JPB, et al. . (2004) Effects of alfalfa, wheat bran or beet pulp, with or without sunflower oil, on caecal fermentation and on digestibility in the rabbit. Anim Feed Sci Tech 117, 131149.CrossRefGoogle Scholar
561Heller, S, Hackler, L, Rivers, J, et al. . (1980) Dietary fiber: the effect of particle size of wheat bran on colonic function in young adult men. Am J Clin Nutr 33, 17341744.CrossRefGoogle ScholarPubMed
562Maes, C & Delcour, JA (2002) Structural characterisation of water-extractable and water-unextractable arabinoxylans in wheat bran. J Cereal Sci 35, 315326.CrossRefGoogle Scholar
563Dornez, E, Gebruers, K, Wiame, S, et al. . (2006) Insight into the distribution of arabinoxylans, endoxylanases, and endoxylanase inhibitors in industrial wheat roller mill streams. J Agric Food Chem 54, 85218529.CrossRefGoogle ScholarPubMed
564Camire, AL & Clydesdale, FM (1982) Analysis of phytic acid in foods by HPLC. J Food Sci 47, 575578.CrossRefGoogle Scholar
565Fretzdorff, B (1989) Phytic acid in wheat bran and germ products – how to remove phytic acid from these products. Z Lebens Unters Forsch 189, 110112.CrossRefGoogle Scholar
566Jenab, M & Thompson, LU (2000) Phytic acid in wheat bran affects colon morphology, cell differentiation and apoptosis. Carcinogenesis 21, 15471552.CrossRefGoogle ScholarPubMed
567Liu, ZH, Wang, HY, Wang, XE, et al. . (2008) Effect of wheat pearling on flour phytase activity, phytic acid, iron, and zinc content. LWT Food Sci Technol 41, 521527.CrossRefGoogle Scholar
568Bagheri, S & Guéguen, L (1985) Effect of wheat bran and pectin on the absorption and retention of phosphorus, calcium, magnesium and zinc by the growing-pig. Reprod Nutr Dev 25, 705716.CrossRefGoogle ScholarPubMed
569Shils, ME, Olson, JA & Shike, M (editors) (1994) Modern Nutrition in Health and Disease, 8th ed.Philadelphia, PA: Lea and Febiger.Google Scholar
570Mullin, WJ & Jui, PY (1986) Folate content of bran from different wheat classes. Cereal Chem 63, 516518.Google Scholar
571Zhou, K, Su, L & Yu, LL (2004) Phytochemicals and antioxidant properties in wheat bran. J Agric Food Chem 52, 61086114.CrossRefGoogle ScholarPubMed
572Zhou, KQ, Yin, JJ & Yu, LL (2005) Phenolic acid, tocopherol and carotenoid compositions, and antioxidant functions of hard red winter wheat bran. J Agric Food Chem 53, 39163922.CrossRefGoogle ScholarPubMed
573Robertson, JA, Faulds, CB, Smith, AC, et al. . (2008) Peroxidase-mediated oxidative cross-linking and its potential to modify mechanical properties in water-soluble polysaccharide extracts and cereal grain residues. J Agric Food Chem 56, 17201726.CrossRefGoogle ScholarPubMed
574Irmak, S, Jonnala, RS & MacRitchie, F (2008) Effect of genetic variation on phenolic acid and policosanol contents of Pegaso wheat lines. J Cereal Sci 48, 2026.CrossRefGoogle Scholar
575Kim, KH, Tsao, R, Yang, R, et al. . (2006) Phenolic acid profiles and antioxidant activities of wheat bran extracts and the effect of hydrolysis conditions. Food Chem 95, 466473.CrossRefGoogle Scholar
576Siebenhandl, S, Grausgruber, H, Pellegrini, N, et al. . (2007) Phytochemical profile of main antioxidants in different fractions of purple and blue wheat, and black barley. J Agric Food Chem 55, 85418547.CrossRefGoogle ScholarPubMed
577Apak, R, Güçlü, K, Ozyürek, M, et al. . (2005) Total antioxidant capacity assay of human serum using copper(II)-neocuproine as chromogenic oxidant: the CUPRAC method. Free Radic Res 39, 949961.CrossRefGoogle ScholarPubMed
578Moore, J, Liu, JG, Zhou, KQ, et al. . (2006) Effects of genotype and environment on the antioxidant properties of hard winter wheat bran. J Agric Food Chem 54, 53135322.CrossRefGoogle ScholarPubMed
579Zhou, K & Yu, L (2004) Antioxidant properties of bran extracts from Trego wheat grown at different locations. J Agric Food Chem 52, 11121117.CrossRefGoogle ScholarPubMed
580Iqbal, S, Bhanger, MI & Anwar, F (2007) Antioxidant properties and components of bran extracts from selected wheat varieties commercially available in Pakistan. LWT Food Sci Technol 40, 361367.CrossRefGoogle Scholar
581Smeds, AI, Eklund, PC, Sjoholm, RE, et al. . (2007) Quantification of a broad spectrum of lignans in cereals, oilseeds, and nuts. J Agric Food Chem 55, 13371346.CrossRefGoogle ScholarPubMed
582Kulawinek, M, Jaromin, A, Kozubek, A, et al. . (2008) Alkylresorcinols in selected Polish rye and wheat cereals and whole-grain cereal products. J Agric Food Chem 56, 72367242.CrossRefGoogle ScholarPubMed
583Graham, SF, Hollis, JH, Migaud, M, et al. . (2009) Analysis of betaine and choline contents of aleurone, bran, and flour fractions of wheat (Triticum aestivum L.) using 1H nuclear magnetic resonance (NMR) spectroscopy. J Agric Food Chem 57, 19481951.CrossRefGoogle ScholarPubMed
584Slow, S, Donaggio, M, Cressey, PJ, et al. . (2005) The betaine content of New Zealand foods and estimated intake in the New Zealand diet. J Food Comp Anal 18, 473485.CrossRefGoogle Scholar
585Nyström, L, Lampi, AM, Rita, H, et al. . (2007) Effects of processing on availability of total plant sterols, steryl ferulates and steryl glycosides from wheat and rye bran. J Agric Food Chem 55, 90599065.CrossRefGoogle ScholarPubMed
586Irmak, S, Dunford, NT & Milligan, J (2006) Policosanol contents of beeswax, sugar cane and wheat extracts. Food Chem 95, 312318.CrossRefGoogle Scholar
587Moruzzi, G, Viviani, R, Sechi, AM, et al. . (1969) Studies on compounds and individual lipids of wheat germ. J Food Sci 34, 581584.CrossRefGoogle Scholar
588Dubois, M, Geddes, WF & Smith, F (1960) The carbohydrates of the Gramineae. X. A quantitative study of the carbohydrates of wheat germ. Cereal Chem 37, 557567.Google Scholar
589Garcia, WJ, Gardner, HW, Cavins, JF, et al. . (1972) Composition of air-classified defatted corn and wheat germ flours. Cereal Chem 49, 499507.Google Scholar
590Linko, P, Cheng, Y-Y & Milner, M (1960) Changes in the soluble carbohydrates during browning of wheat embryos. Cereal Chem 37, 548556.Google Scholar
591Leenhardt, F, Fardet, A, Lyan, B, et al. . (2008) Wheat germ supplementation of a low vitamin E diet in rats affords effective antioxidant protection in tissues. J Am Coll Nutr 27, 222228.CrossRefGoogle ScholarPubMed
592Shurpalekar, SR & Rao, PH (1977) Wheat germ. Adv Food Res 23, 187304.CrossRefGoogle ScholarPubMed
593Bilgicli, N & Ibanoglu, S (2007) Effect of wheat germ and wheat bran on the fermentation activity, phytic acid content and colour of tarhana, a wheat flour–yoghurt mixture. J Food Eng 78, 681686.CrossRefGoogle Scholar
594Garcia, WJ, Inglett, GE & Blessin, CW (1972) Mineral constituents in corn and wheat-germ by atomic-absorption spectroscopy. Cereal Chem 49, 158167.Google Scholar
595Zhu, KX, Zhou, HM & Qian, HF (2006) Comparative study of chemical composition and physicochemical properties of defatted wheat germ flour and its protein isolate. J Food Biochem 30, 329341.CrossRefGoogle Scholar
596Dodin, S, Lemay, A, Jacques, H, et al. . (2005) The effects of flaxseed dietary supplement on lipid profile, bone mineral density, and symptoms in menopausal women: a randomized, double-blind, wheat germ placebo-controlled clinical trial. J Clin Endocrinol Metab 90, 13901397.CrossRefGoogle ScholarPubMed
597Ostlund, RE Jr, Racette, SB & Stenson, WF (2003) Inhibition of cholesterol absorption by phytosterol-replete wheat germ compared with phytosterol-depleted wheat germ. Am J Clin Nutr 77, 13851389.CrossRefGoogle ScholarPubMed
598Mühlum, A, Ingwersen, M, Schünemann, C, et al. . (1989) Precaecal and postileal digestion of sucrose, lactose, stachyose and raffinose. Adv Anim Physiol Anim Nutr 19, 3143.Google Scholar
599Van Dokkum, W, Pikaar, NA & Thissen, JT (1983) Physiological effects of fibre-rich types of bread. 2. Dietary fibre from bread: digestibility by the intestinal microflora and water-holding capacity in the colon of human subjects. Br J Nutr 50, 6174.Google ScholarPubMed
600McCance, RA & Widdowson, EM (1935) Phytin in human nutrition. Biochem J 29, 26942699.CrossRefGoogle ScholarPubMed
601Sakamoto, K, Vucenik, I & Shamsuddin, AM (1993) [3H]Phytic acid (inositol hexaphosphate) is absorbed and distributed to various tissues in rats. J Nutr 123, 713720.CrossRefGoogle ScholarPubMed
602McCance, RA & Widdowson, EM (1942) Mineral metabolism of healthy adults on white and brown bread dietaries. J Physiol 101, 4485.CrossRefGoogle Scholar
603Institute of Medicine (1997) Dietary Reference Intakes for Calcium, Phosphorus, Magnesium, Vitamin D, and Fluoride. Washington, DC: National Academy Press.Google Scholar
604Walti, MK, Zimmermann, MB, Walczyk, T, et al. . (2003) Measurement of magnesium absorption and retention in type 2 diabetic patients with the use of stable isotopes. Am J Clin Nutr 78, 448453.CrossRefGoogle ScholarPubMed
605Sandstrom, B, Arvidsson, B, Cederblad, A, et al. . (1980) Zinc absorption from composite meals. 1. The significance of wheat extraction rate, zinc, calcium, and protein-content in meals based on bread. Am J Clin Nutr 33, 739745.CrossRefGoogle Scholar
606Sundkvist, G, Dahlin, LB, Nilsson, H, et al. . (2000) Sorbitol and myo-inositol levels and morphology of sural nerve in relation to peripheral nerve function and clinical neuropathy in men with diabetic, impaired, and normal glucose tolerance. Diabet Med 17, 259268.CrossRefGoogle ScholarPubMed
607Saha, PR, Weaver, CM & Mason, AC (1994) Mineral bioavailability in rats from intrinsically labeled whole wheat-flour of various phytate levels. J Agric Food Chem 42, 25312535.CrossRefGoogle Scholar
608Fox, TE, Fairweather-Tait, SJ, Eagles, J, et al. . (1994) Assessment of zinc bioavailability – studies in rats on zinc absorption from wheat using radio-isotopes and stable-isotopes. Br J Nutr 71, 95101.CrossRefGoogle Scholar
609Welch, RM, House, WA, Ortiz-Monasterio, I, et al. . (2005) Potential for improving bioavailable zinc in wheat grain (Triticum species) through plant breeding. J Agric Food Chem 53, 21762180.CrossRefGoogle ScholarPubMed
610Ahmed, A, Anjum, F, Ur Rehman, S, et al. . (2008) Bioavailability of calcium, iron and zinc fortified whole wheat flour chapatti. Plant Foods Hum Nutr 63, 713.CrossRefGoogle ScholarPubMed
611Johnson, PE & Lykken, GI (1988) Copper-65 absorption by men fed intrinsically and extrinsically labeled whole wheat bread. J Agric Food Chem 36, 537540.CrossRefGoogle Scholar
612Mutanen, M, Koivistoinen, P, Morris, VC, et al. . (2007) Relative nutritional availability to rats of selenium in Finnish spring wheat (Triticum aestivum L.) fertilized or sprayed with sodium selenate and in an American winter bread wheat naturally high in Se. Br J Nutr 57, 319329.CrossRefGoogle Scholar
613Alexander, AR, Whanger, PD & Miller, LT (1983) Bioavailability to rats of selenium in various tuna and wheat products. J Nutr 113, 196204.CrossRefGoogle ScholarPubMed
614Weaver, CM, Heaney, RP, Martin, BR, et al. . (1991) Human calcium absorption from whole-wheat products. J Nutr 121, 17691775.CrossRefGoogle ScholarPubMed
615Zempleni, J, Galloway, J & McCormick, D (1996) Pharmacokinetics of orally and intravenously administered riboflavin in healthy humans. Am J Clin Nutr 63, 5466.CrossRefGoogle ScholarPubMed
616Tarr, J, Tamura, T & Stokstad, E (1981) Availability of vitamin B6 and pantothenate in an average American diet in man. Am J Clin Nutr 34, 13281337.CrossRefGoogle Scholar
617Kayden, H & Traber, M (1993) Absorption, lipoprotein transport, and regulation of plasma concentrations of vitamin E in humans. J Lipid Res 34, 343358.CrossRefGoogle ScholarPubMed
618Andreasen, MF, Kroon, PA, Williamson, G, et al. . (2001) Esterase activity able to hydrolyze dietary antioxidant hydroxycinnamates is distributed along the intestine of mammals. J Agric Food Chem 49, 56795684.CrossRefGoogle ScholarPubMed
619Ross, AB, Shepherd, MJ, Knudsen, KEB, et al. . (2003) Absorption of dietary alkylresorcinols in ileal-cannulated pigs and rats. Br J Nutr 90, 787794.CrossRefGoogle Scholar
620Nissinen, M, Gylling, H, Vuoristo, M, et al. . (2002) Micellar distribution of cholesterol and phytosterols after duodenal plant stanol ester infusion. Am J Physiol Gastrointest Liver Physiol 282, G1009G1015.CrossRefGoogle ScholarPubMed
621Nestler, JE, Jakubowicz, DJ, Reamer, P, et al. . (1999) Ovulatory and metabolic effects of d-chiro-inositol in the polycystic ovary syndrome. N Engl J Med 340, 13141320.CrossRefGoogle ScholarPubMed
622Campbell, WW, Haub, MD, Fluckey, JD, et al. . (2004) Pinitol supplementation does not affect insulin-mediated glucose metabolism and muscle insulin receptor content and phosphorylation in older humans. J Nutr 134, 29983003.Google Scholar
623Nyman, M, Asp, N-G, Cummings, J, et al. . (1986) Fermentation of dietary fibre in the intestinal tract: comparison between man and rat. Br J Nutr 55, 487496.CrossRefGoogle Scholar
624Kahlon, TS, Chow, FI, Hoefer, JL, et al. . (2001) Effect of wheat bran fiber and bran particle size on fat and fiber digestibility and gastrointestinal tract measurements in the rat. Cereal Chem 78, 481484.CrossRefGoogle Scholar
625Hansen, I, Knudsen, KEB & Eggum, BO (2007) Gastrointestinal implications in the rat of wheat bran, oat bran and pea fibre. Br J Nutr 68, 451462.CrossRefGoogle Scholar
626Ehle, FR, Jeraci, JL, Robertson, JB, et al. . (1982) The influence of dietary fiber on digestibility, rate of passage and gastrointestinal fermentation in pigs. J Anim Sci 55, 10711081.CrossRefGoogle Scholar
627Robertson, JA, Murison, SD & Chesson, A (1992) Particle-size distribution and solubility of dietary fiber in swede-bran (Brassica napus) based and wheat-bran-based diets during gastrointestinal transit in the pig. J Sci Food Agric 58, 197205.CrossRefGoogle Scholar
628Nyman, M & Asp, N-G (1985) Dietary fibre fermentation in the rat intestinal tract: effect of adaptation period, protein and fibre levels, and particle size. Br J Nutr 54, 635643.CrossRefGoogle ScholarPubMed
629Sandberg, AS & Andersson, H (1988) Effect of dietary phytase on the digestion of phytate in the stomach and small intestine of humans. J Nutr 118, 469473.CrossRefGoogle ScholarPubMed
630Sandberg, A-S, Andersson, H, Carlsson, N-G, et al. . (1987) Degradation products of bran phytate formed during digestion in the human small intestine: effect of extrusion cooking on digestibility. J Nutr 117, 20612065.CrossRefGoogle ScholarPubMed
631Brune, M, Rossander-Hulten, L, Hallberg, L, et al. . (1992) Iron absorption from bread in humans: inhibiting effects of cereal fiber, phytate and inositol phosphates with different numbers of phosphate groups. J Nutr 122, 442449.CrossRefGoogle ScholarPubMed
632Reddy, M & Cook, J (1991) Assessment of dietary determinants of nonheme-iron absorption in humans and rats. Am J Clin Nutr 54, 723728.CrossRefGoogle ScholarPubMed
633Reeves, PG, Gregoire, BR, Garvin, DF, et al. . (2007) Determination of selenium bioavailability from wheat mill fractions in rats by using the slope-ratio assay and a modified Torula yeast-based diet. J Agric Food Chem 55, 516522.CrossRefGoogle Scholar
634Carter, EGA & Carpenter, KJ (1982) The bioavailability for humans of bound niacin from wheat bran. Am J Clin Nutr 36, 855861.CrossRefGoogle ScholarPubMed
635Kies, C, Kan, S & Fox, HM (1984) Vitamin B6 availability from wheat, rice and corn brans for humans. Nutr Rep Int 30, 483491.Google Scholar
636Kahlon, TS, Chow, FI, Hoefer, JL, et al. . (1986) Bioavailability of vitamin A and vitamin E as influenced by wheat bran and bran particle size. Cereal Chem 63, 490493.Google Scholar
637Andreasen, MF, Kroon, PA, Williamson, G, et al. . (2001) Intestinal release and uptake of phenolic antioxidant diferulic acids. Free Radic Biol Med 31, 304314.CrossRefGoogle ScholarPubMed
638Connor, WE (2000) Importance of n-3 fatty acids in health and disease. Am J Clin Nutr 71, 171S175S.CrossRefGoogle ScholarPubMed
639Brouwer, IA, Katan, MB & Zock, PL (2004) Dietary α-linolenic acid is associated with reduced risk of fatal coronary heart disease, but increased prostate cancer risk: a meta-analysis. J Nutr 134, 919922.CrossRefGoogle ScholarPubMed
640Hu, FB, Stampfer, MJ, Manson, JE, et al. . (1999) Dietary intake of α-linolenic acid and risk of fatal ischemic heart disease among women. Am J Clin Nutr 69, 890897.CrossRefGoogle ScholarPubMed
641Kang, JX & Leaf, A (1996) Antiarrhythmic effects of polyunsaturated fatty acids: recent studies. Circulation 94, 17741780.CrossRefGoogle ScholarPubMed
642Edwards, R, Peet, M, Shay, J, et al. . (1998) Omega-3 polyunsaturated fatty acid levels in the diet and in red blood cell membranes of depressed patients. J Affect Disord 48, 149155.CrossRefGoogle ScholarPubMed
643Yehuda, S, Rabinovitz, S & Mostofsky, DI (2005) Mixture of essential fatty acids lowers test anxiety. Nutr Neurosci 8, 265267.CrossRefGoogle ScholarPubMed
644Djousse, L, Folsom, AR, Province, MA, et al. . (2003) Dietary linolenic acid and carotid atherosclerosis: the National Heart, Lung, and Blood Institute Family Heart Study. Am J Clin Nutr 77, 819825.CrossRefGoogle ScholarPubMed
645Narisawa, T, Fukaura, Y, Yazawa, K, et al. . (1994) Colon cancer prevention with a small amount of dietary perilla oil high in α-linolenic acid in an animal model. Cancer 73, 20692075.3.0.CO;2-1>CrossRefGoogle ScholarPubMed
646Klein, V, Chajes, V, Germain, E, et al. . (2000) Low α-linolenic acid content of adipose breast tissue is associated with an increased risk of breast cancer. Eur J Cancer 36, 335340.CrossRefGoogle ScholarPubMed
647Hwang, DH, Boudreau, M & Chanmugam, P (1988) Dietary linolenic acid and longer-chain n-3 fatty acids: comparison of effects on arachidonic acid metabolism in rats. J Nutr 118, 427437.CrossRefGoogle ScholarPubMed
648Chapkin, RS, McMurray, DN, Davidson, LA, et al. . (2008) Bioactive dietary long-chain fatty acids: emerging mechanisms of action. Br J Nutr 100, 11521157.CrossRefGoogle ScholarPubMed
649Enke, U, Seyfarth, L, Schleussner, E, et al. . (2008) Impact of PUFA on early immune and fetal development. Br J Nutr 100, 11581168.CrossRefGoogle ScholarPubMed
650Townsend, DM, Tew, KD & Tapiero, H (2003) The importance of glutathione in human disease. Biomed Pharmacother 57, 145155.CrossRefGoogle ScholarPubMed
651Higashi, T, Tateishi, N, Naruse, A, et al. . (1977) A novel physiological role of liver glutathione as a reservoir of l-cysteine. J Biochem 82, 117124.CrossRefGoogle ScholarPubMed
652Bilzer, M & Lauterburg, BH (1991) Effects of hypochlorous acid and chloramines on vascular resistance, cell integrity, and biliary glutathione disulfide in the perfused rat-liver – modulation by glutathione. J Hepatol 13, 8489.CrossRefGoogle ScholarPubMed
653Troen, AM, Chao, W-H, Crivello, NA, et al. . (2008) Cognitive impairment in folate-deficient rats corresponds to depleted brain phosphatidylcholine and is prevented by dietary methionine without lowering plasma homocysteine. J Nutr 138, 25022509.CrossRefGoogle ScholarPubMed
654Essien, FB & Wannberg, SL (1993) Methionine but not folinic acid or vitamin B-12 alters the frequency of neural tube defects in axd mutant mice. J Nutr 123, 2734.CrossRefGoogle ScholarPubMed
655Caylak, E, Aytekin, M & Halifeoglu, I (2008) Antioxidant effects of methionine, α-lipoic acid, N-acetylcysteine and homocysteine on lead-induced oxidative stress to erythrocytes in rats. Exp Toxicol Pathol 60, 289294.CrossRefGoogle ScholarPubMed
656Khumalo, N, Dawber, R & Ferguson, D (2005) Apparent fragility of African hair is unrelated to the cystine-rich protein distribution: a cytochemical electron microscopic study. Exp Dermatol 14, 311314.CrossRefGoogle Scholar
657Sass, J, Skladal, D, Zelger, B, et al. . (2004) Trichothiodystrophy: quantification of cysteine in human hair and nails by application of sodium azide-dependent oxidation to cysteic acid. Arch Dermatol Res 296, 188191.Google ScholarPubMed
658Droge, W & Holm, E (1997) Role of cysteine and glutathione in HIV infection and other diseases associated with muscle wasting and immunological dysfunction. FASEB J 11, 10771089.CrossRefGoogle ScholarPubMed
659Netto, LES, de Oliveira, MA, Monteiro, G, et al. . (2007) Reactive cysteine in proteins: protein folding, antioxidant defense, redox signaling and more. Comp Biochem Physiol C Toxicol Pharmacol 146, 180193.CrossRefGoogle ScholarPubMed
660Marlett, JA, McBurney, MI & Slavin, JL (2002) Position of the American Dietetic Association: health implications of dietary fiber. J Am Diet Assoc 102, 9931000.CrossRefGoogle ScholarPubMed
661Tucker, LA & Thomas, KS (2009) Increasing total fiber intake reduces risk of weight and fat gains in women. J Nutr 139, 576581.CrossRefGoogle ScholarPubMed
662Salmeron, J, Ascherio, A, Rimm, EB, et al. . (1997) Dietary fiber, glycemic load, and risk of non-insulin-dependent diabetes mellitus in women. Diabetes Care 277, 472477.Google ScholarPubMed
663Slavin, JL, Jacobs, D, Marquart, L, et al. . (2001) The role of whole grains in disease prevention. J Am Diet Assoc 101, 780785.CrossRefGoogle ScholarPubMed
664Glei, M, Hofmann, T, Kuster, K, et al. . (2006) Both wheat (Triticum aestivum) bran arabinoxylans and gut flora-mediated fermentation products protect human colon cells from genotoxic activities of 4-hydroxynonenal and hydrogen peroxide. J Agric Food Chem 54, 20882095.CrossRefGoogle ScholarPubMed
665Eastwood, M & Mowbray, L (1976) The binding of the components of mixed micelle to dietary fiber. Am J Clin Nutr 29, 14611467.CrossRefGoogle ScholarPubMed
666Pomare, EW & Heaton, KW (1973) Alteration of bile salt metabolism by dietary fibre (bran). Gut 14, 826.Google ScholarPubMed
667Eastwood, MA (1975) Vegetable dietary fiber – potent pith. J R Soc Health 95, 188190.CrossRefGoogle Scholar
668Kaur, N & Gupta, AK (2002) Applications of inulin and oligofructose in health and nutrition. J Biosci 27, 703714.CrossRefGoogle ScholarPubMed
669Roberfroid, MB & Delzenne, NM (1998) Dietary fructans. Annu Rev Nutr 18, 117143.CrossRefGoogle ScholarPubMed
670Rozan, P, Nejdi, A, Hidalgo, S, et al. . (2008) Effects of lifelong intervention with an oligofructose-enriched inulin in rats on general health and lifespan. Br J Nutr 100, 11921199.CrossRefGoogle ScholarPubMed
671Gibson, GR, Beatty, ER, Wang, X, et al. . (1995) Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology 108, 975982.CrossRefGoogle ScholarPubMed
672Femia, AP, Luceri, C, Dolara, P, et al. . (2002) Antitumorigenic activity of the prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis on azoxymethane-induced colon carcinogenesis in rats. Carcinogenesis 23, 19531960.CrossRefGoogle ScholarPubMed
673Archer, SY, Meng, S, Shei, A, et al. . (1998) p21WAF1 is required for butyrate-mediated growth inhibition of human colon cancer cells. Proc Natl Acad Sci U S A 95, 67916796.CrossRefGoogle ScholarPubMed
674Avivi-Green, C, Polak-Charcon, S, Madar, Z, et al. . (2002) Different molecular events account for butyrate-induced apoptosis in two human colon cancer cell lines. J Nutr 132, 18121818.CrossRefGoogle ScholarPubMed
675Brighenti, F, Casiraghi, MC, Canzi, E, et al. . (1999) Effect of consumption of a ready-to-eat breakfast cereal containing inulin on the intestinal milieu and blood lipids in healthy male volunteers. Eur J Clin Nutr 53, 726733.CrossRefGoogle ScholarPubMed
676Williams, CM (1999) Effects of inulin on lipid parameters in humans. J Nutr 129, 1471S1473S.CrossRefGoogle ScholarPubMed
677Beylot, M (2005) Effects of inulin-type fructans on lipid metabolism in man and in animal models. Br J Nutr 93, S163S168.CrossRefGoogle ScholarPubMed
678Pai, R, Tarnawski, AS & Tran, T (2004) Deoxycholic acid activates β-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Mol Biol Cell 15, 21562163.CrossRefGoogle ScholarPubMed
679McMillan, L, Butcher, S, Wallis, Y, et al. . (2000) Bile acids reduce the apoptosis-inducing effects of sodium butyrate on human colon adenoma (AA/C1) cells: implications for colon carcinogenesis. Biochem Biophys Res Commun 273, 4549.CrossRefGoogle ScholarPubMed
680Braaten, JT, Wood, PJ, Scott, FW, et al. . (1991) Oat gum lowers glucose and insulin after an oral glucose load. Am J Clin Nutr 53, 14251430.CrossRefGoogle ScholarPubMed
681Ostman, E, Rossi, E, Larsson, H, et al. . (2006) Glucose and insulin responses in healthy men to barley bread with different levels of (1 →  3;1 →  4)-β-glucans; predictions using fluidity measurements of in vitro enzyme digests. J Cereal Sci 43, 230235.CrossRefGoogle Scholar
682Tappy, L, Gugolz, E & Wursch, P (1996) Effects of breakfast cereals containing various amounts of β-glucan fibers on plasma glucose and insulin responses in NIDDM subjects. Diabetes Care 19, 831834.CrossRefGoogle ScholarPubMed
683Maki, KC, Shinnick, F, Seeley, MA, et al. . (2003) Food products containing free tall oil-based phytosterols and oat β-glucan lower serum total and LDL cholesterol in hypercholesterolemic adults. J Nutr 133, 808813.CrossRefGoogle ScholarPubMed
684Wright, RS, Anderson, JW & Bridges, SR (1990) Propionate inhibits hepatocyte lipid synthesis. Proc Soc Exp Biol Med 195, 2629.CrossRefGoogle ScholarPubMed
685Demir, G, Klein, HO, Mandel-Molinas, N, et al. . (2007) β Glucan induces proliferation and activation of monocytes in peripheral blood of patients with advanced breast cancer. Int Immunopharmacol 7, 113116.CrossRefGoogle ScholarPubMed
686Vucenik, I & Shamsuddin, AM (2003) Cancer inhibition by inositol hexaphosphate (IP6) and inositol: from laboratory to clinic. J Nutr 133, 3778S3784S.CrossRefGoogle ScholarPubMed
687Muraoka, S & Miura, T (2004) Inhibition of xanthine oxidase by phytic acid and its antioxidative action. Life Sci 74, 16911700.CrossRefGoogle ScholarPubMed
688Lee, SH, Park, HJ, Chun, HK, et al. . (2006) Dietary phytic acid lowers the blood glucose level in diabetic KK mice. Nutr Res 26, 474479.CrossRefGoogle Scholar
689Lee, S-H, Park, H-J, Chun, H-K, et al. . (2007) Dietary phytic acid improves serum and hepatic lipid levels in aged ICR mice fed a high-cholesterol diet. Nutr Res 27, 505510.CrossRefGoogle Scholar
690Onomi, S, Okazaki, Y & Katayama, T (2004) Effect of dietary level of phytic acid on hepatic and serum lipid status in rats fed a high-sucrose diet. Biosci Biotechnol Biochem 68, 13791381.CrossRefGoogle ScholarPubMed
691Lee, SH, Park, HJ, Cho, SY, et al. . (2005) Effects of dietary phytic acid on serum and hepatic lipid levels in diabetic KK mice. Nutr Res 25, 869876.CrossRefGoogle Scholar
692Singh, A, Prakash Singh, S & Bamezai, R (1997) Modulatory influence of arecoline on the phytic acid-altered hepatic biotransformation system enzymes, sulfhydryl content and lipid peroxidation in a murine system. Cancer Lett 117, 16.CrossRefGoogle Scholar
693Grases, F, Simonet, BM, March, JG, et al. . (2000) Inositol hexakisphosphate in urine: the relationship between oral intake and urinary excretion. BJU Int 85, 138142.CrossRefGoogle ScholarPubMed
694Grases, F, Sanchis, P, Perello, J, et al. . (2008) Phytate reduces age-related cardiovascular calcification. Front Biosci 13, 71157122.CrossRefGoogle ScholarPubMed
695Shen, XT, Xiao, H, Ranallo, R, et al. . (2003) Modulation of ATP-dependent chromatin remodeling complexes by inositol polyphosphates. Science 299, 112114.CrossRefGoogle ScholarPubMed
696Steger, DJ, Haswell, ES, Miller, AL, et al. . (2003) Regulation of chromatin remodeling by inositol polyphosphates. Science 299, 114116.CrossRefGoogle ScholarPubMed
697Sajilata, MG, Singhal, RS & Kulkarni, PR (2006) Resistant starch: a review. Compr Rev Food Sci Food Saf 5, 117.CrossRefGoogle ScholarPubMed
698Malhotra, SL (1968) Epidemiological study of cholelithiasis among railroad workers in India with special reference to causation. Gut 9, 290295.CrossRefGoogle ScholarPubMed
699Beard, JL & Connor, JR (2003) Iron status and neural functioning. Annu Rev Nutr 23, 4158.CrossRefGoogle ScholarPubMed
700Institute of Medicine (2001) Dietary Reference Intake for Vitamin A, Vitamin K, Arsenic, Baron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc. Washington, DC: National Academy Press.Google Scholar
701Uehara, M, Chiba, H, Mogi, H, et al. . (1997) Induction of increased phosphatidylcholine hydroperoxide by an iron-deficient diet in rats. J Nutr Biochem 8, 385391.CrossRefGoogle Scholar
702Rosenzweig, P & Volpe, S (1999) Iron, thermoregulation, and metabolic rate. Crit Rev Food Sci Nutr 39, 131148.CrossRefGoogle ScholarPubMed
703Oexle, H, Gnaiger, E & Weiss, G (1999) Iron-dependent changes in cellular energy metabolism: influence on citric acid cycle and oxidative phosphorylation. Biochim Biophys Acta 1413, 99107.CrossRefGoogle ScholarPubMed
704Ramdath, DD & Golden, MHN (1989) Non-haematological aspects of iron nutrition. Nutr Res Rev 2, 2949.CrossRefGoogle ScholarPubMed
705Lozoff, B, Jimenez, E, Hagen, J, et al. . (2000) Poorer behavioral and developmental outcome more than 10 years after treatment for iron deficiency in infancy. Pediatrics 105, E51.CrossRefGoogle ScholarPubMed
706Oski, FA, Honig, AS, Helu, B, et al. . (1983) Effect of iron therapy on behavior performance in nonanemic, iron-deficient infants. Pediatrics 71, 877880.CrossRefGoogle Scholar
707Prockop, DJ (1971) Role of iron in synthesis of collagen in connective tissue. Fed Proc 30, 984990.Google ScholarPubMed
708Katsumata, S, Katsumata-Tsuboi, R, Uehara, M, et al. . (2009) Severe iron deficiency decreases both bone formation and bone resorption in rats. J Nutr 139, 238243.CrossRefGoogle ScholarPubMed
709Willis, WT, Dallman, PR & Brooks, GA (1988) Physiological and biochemical correlates of increased work in trained iron-deficient rats. J Appl Physiol 65, 256263.CrossRefGoogle ScholarPubMed
710Cook, J & Lynch, S (1986) The liabilities of iron deficiency. Blood 68, 803809.CrossRefGoogle ScholarPubMed
711Rosales, FJ, Jang, J-T, Pinero, DJ, et al. . (1999) Iron deficiency in young rats alters the distribution of vitamin A between plasma and liver and between hepatic retinol and retinyl esters. J Nutr 129, 12231228.CrossRefGoogle ScholarPubMed
712McClung, JP & Karl, JP (2009) Iron deficiency and obesity: the contribution of inflammation and diminished iron absorption. Nutr Rev 67, 100104.CrossRefGoogle Scholar
713Clancaglini, P, Plzauro, JM, Curti, C, et al. . (1990) Effect of membrane moiety and magnesium ions on the inhibition of matrix-induced alkaline phosphatase by zinc ions. Int J Biochem 22, 747751.CrossRefGoogle Scholar
714Bussiere, FI, Gueux, E, Rock, E, et al. . (2002) Increased phagocytosis and production of reactive oxygen species by neutrophils during magnesium deficiency in rats and inhibition by high magnesium concentration. Br J Nutr 87, 107113.CrossRefGoogle ScholarPubMed
715Olatunji, LA & Soladoye, AO (2007) Increased magnesium intake prevents hyperlipidemia and insulin resistance and reduces lipid peroxidation in fructose-fed rats. Pathophysiology 14, 1115.CrossRefGoogle ScholarPubMed
716Kisters, K, Spieker, C, Tepel, M, et al. . (1993) New data about the effects of oral physiological magnesium supplementation on several cardiovascular risk factors (lipids and blood pressure). Magnes Res 6, 355360.Google ScholarPubMed
717Barbagallo, M & Dominguez, LJ (2007) Magnesium metabolism in type 2 diabetes mellitus, metabolic syndrome and insulin resistance. Arch Biochem Biophys 458, 4047.CrossRefGoogle ScholarPubMed
718Colditz, G, Manson, J, Stampfer, M, et al. . (1992) Diet and risk of clinical diabetes in women. Am J Clin Nutr 55, 10181023.CrossRefGoogle ScholarPubMed
719Nadler, JL, Balon, TW & Rude, R (1997) Fiber intake and risk of developing non-insulin-dependent diabetes mellitus. JAMA 277, 17611762.CrossRefGoogle ScholarPubMed
720Paolisso, G, Scheen, A, D'Onofrio, F, et al. . (1990) Magnesium and glucose homeostasis. Diabetologia 33, 511514.CrossRefGoogle ScholarPubMed
721Schulze, MB, Schulz, M, Heidemann, C, et al. . (2007) Fiber and magnesium intake and incidence of type 2 diabetes: a prospective study and meta-analysis. Arch Intern Med 167, 956965.CrossRefGoogle ScholarPubMed
722van Dam, RM, Hu, FB, Rosenberg, L, et al. . (2006) Dietary calcium and magnesium, major food sources, and risk of type 2 diabetes in U.S. black women. Diabetes Care 29, 22382243.CrossRefGoogle ScholarPubMed
723Paolisso, G, Dimaro, G, Cozzolino, D, et al. . (1992) Chronic magnesium administration enhances oxidative glucose metabolism in thiazide treated hypertensive patients. Am J Hypertens 5, 681686.CrossRefGoogle ScholarPubMed
724Nadler, J, Buchanan, T, Natarajan, R, et al. . (1993) Magnesium deficiency produces insulin resistance and increased thromboxane synthesis. Hypertension 21, 10241029.CrossRefGoogle ScholarPubMed
725Ascherio, A, Rimm, E, Giovannucci, E, et al. . (1992) A prospective study of nutritional factors and hypertension among US men. Circulation 86, 14751484.CrossRefGoogle ScholarPubMed
726Rubenowitz, E, Axelsson, G & Rylander, R (1996) Magnesium in drinking water and death from acute myocardial infarction. Am J Epidemiol 143, 456462.CrossRefGoogle ScholarPubMed
727Cohen, L (1988) Recent data on magnesium and osteoporosis. Magnes Res 1, 8587.Google ScholarPubMed
728Bernardini, D, Nasulewicz, A, Mazur, A, et al. . (2005) Magnesium and microvascular endothelial cells: a role in inflammation and angiogenesis. Front Biosci 10, 11771182.CrossRefGoogle ScholarPubMed
729Reungjui, S, Prasongwatana, V, Premgamone, A, et al. . (2002) Magnesium status of patients with renal stones and its effect on urinary citrate excretion. BJU Int 90, 635639.CrossRefGoogle ScholarPubMed
730Bray, TM & Bettger, WJ (1990) The physiological role of zinc as an antioxidant. Free Radic Biol Med 8, 281291.CrossRefGoogle ScholarPubMed
731Zago, MP & Oteiza, PI (2001) The antioxidant properties of zinc: interactions with iron and antioxidants. Free Radic Biol Med 31, 266274.CrossRefGoogle ScholarPubMed
732Beattie, JH & Avenell, A (1992) Trace element nutrition and bone metabolism. Nutr Res Rev 5, 167188.CrossRefGoogle ScholarPubMed
733Ding, W-Q, Yu, H-J & Lind, SE (2008) Zinc-binding compounds induce cancer cell death via distinct modes of action. Cancer Lett 271, 251259.CrossRefGoogle ScholarPubMed
734Guo, W, Zhao, Y-P, Jiang, Y-G, et al. . (2008) Restoring the metabolic disturbance of zinc: may not only contribute to the prevention of esophageal squamous cell cancer. Med Hypotheses 71, 957959.CrossRefGoogle ScholarPubMed
735Hershfinkel, M, Silverman, WF & Sekler, I (2007) The zinc sensing receptor, a link between zinc and cell signaling. Mol Med 13, 331336.CrossRefGoogle ScholarPubMed
736Bogden, J, Oleske, J, Munves, E, et al. . (1987) Zinc and immunocompetence in the elderly: baseline data on zinc nutriture and immunity in unsupplemented subjects. Am J Clin Nutr 46, 101109.CrossRefGoogle ScholarPubMed
737Shen, H, Oesterling, E, Stromberg, A, et al. . (2008) Zinc deficiency induces vascular pro-inflammatory parameters associated with NF-κB and PPAR signaling. J Am Coll Nutr 27, 577587.CrossRefGoogle ScholarPubMed
738Mocchegiani, E, Giacconi, R & Malavolta, M (2008) Zinc signalling and subcellular distribution: emerging targets in type 2 diabetes. Trends Mol Med 14, 419428.CrossRefGoogle ScholarPubMed
739Ohinata, K, Takemoto, M, Kawanago, M, et al. . (2009) Orally administered zinc increases food intake via vagal stimulation in rats. J Nutr 139, 611616.CrossRefGoogle ScholarPubMed
740Robinson, BH (1998) The role of manganese superoxide dismutase in health and disease. J Inher Metab Dis 21, 598603.CrossRefGoogle ScholarPubMed
741Freeland-Graves, JH & Turnlund, JR (1996) Deliberations and evaluations of the approaches, endpoints and paradigms for manganese and molybdenum dietary recommendations. J Nutr 126, 2435S2440S.CrossRefGoogle ScholarPubMed
742Cho, SJ, Park, JW, Kang, JS, et al. . (2008) Nuclear factor-κB dependency of doxorubicin sensitivity in gastric cancer cells is determined by manganese superoxide dismutase expression. Cancer Sci 99, 11171124.CrossRefGoogle ScholarPubMed
743Kattan, Z, Minig, V, Dauça, M, et al. . (2007) Role of manganese superoxide dismutase on growth and invasive properties of human estrogen-independent breast cancer cells. Eur J Cancer Suppl 5, 7677.CrossRefGoogle Scholar
744Johnson, MA, Fischer, JG & Kays, SE (1992) Is copper an antioxidant nutrient? Crit Rev Food Sci Nutr 32, 131.CrossRefGoogle ScholarPubMed
745Baker, A, Harvey, L, Majask-Newman, G, et al. . (1999) Effect of dietary copper intakes on biochemical markers of bone metabolism in healthy adult males. Eur J Clin Nutr 53, 408412.CrossRefGoogle ScholarPubMed
746Lukaski, HC, Klevay, LM & Milne, DB (1988) Effects of dietary copper on human autonomic cardiovascular function. EurJ Appl Physiol 58, 7480.CrossRefGoogle ScholarPubMed
747Milne, D (1998) Copper intake and assessment of copper status. Am J Clin Nutr 67, 1041S1045S.CrossRefGoogle ScholarPubMed
748Klevay, LM (2006) Heart failure improvement from a supplement containing copper. Eur Heart J 27, 117118.CrossRefGoogle ScholarPubMed
749Zhou, Y, Jiang, Y & Kang, YJ (2008) Copper reverses cardiomyocyte hypertrophy through vascular endothelial growth factor-mediated reduction in the cell size. J Mol Cell Cardiol 45, 106117.CrossRefGoogle ScholarPubMed
750Hammud, HH, Nemer, G, Sawma, W, et al. . (2008) Copper–adenine complex, a compound, with multi-biochemical targets and potential anti-cancer effect. Chem Biol Interact 173, 8496.CrossRefGoogle ScholarPubMed
751Klevay, L (1975) Coronary heart disease: the zinc/copper hypothesis. Am J Clin Nutr 28, 764774.CrossRefGoogle ScholarPubMed
752Klevay, LM (1977) Hypo-cholesterolemia due to sodium phytate. Nutr Rep Int 15, 587595.Google Scholar
753Tapiero, H, Townsend, DM & Tew, KD (2003) The antioxidant role of selenium and seleno-compounds. Biomed Pharmacother 57, 134144.CrossRefGoogle ScholarPubMed
754Levander, OA (1992) Selenium and sulfur in antioxidant protective systems – relationships with vitamin E and malaria. Proc Soc Exp Biol Med 200, 255259.CrossRefGoogle ScholarPubMed
755Burk, RF (1990) Protection against free-radical injury by selenoenzymes. Pharmacol Ther 45, 383385.CrossRefGoogle ScholarPubMed
756Jacobs, MM (1977) Inhibitory effects of selenium on 1,2-dimethylhydrazine and methylazoxymethanol colon carcinogenesis. Correlative studies on selenium effects on the mutagenicity and sister chromatid exchange rates of selected carcinogens. Cancer 40, 25572564.3.0.CO;2-T>CrossRefGoogle Scholar
757Jacobs, MM, Forst, CF & Beams, FA (1981) Biochemical and clinical effects of selenium on dimethylhydrazine-induced colon cancer in rats. Cancer Res 41, 44584465.Google ScholarPubMed
758Jariwalla, RJ, Gangapurkar, B & Nakamura, D (2009) Differential sensitivity of various human tumour-derived cell types to apoptosis by organic derivatives of selenium. Br J Nutr 101, 182189.CrossRefGoogle ScholarPubMed
759Gromadzinska, J, Reszka, E, Bruzelius, K, et al. . (2008) Selenium and cancer: biomarkers of selenium status and molecular action of selenium supplements. Eur J Nutr 47, 2950.CrossRefGoogle ScholarPubMed
760Levander, O & Morris, V (1984) Dietary selenium levels needed to maintain balance in North American adults consuming self-selected diets. Am J Clin Nutr 39, 809815.CrossRefGoogle ScholarPubMed
761Arvilommi, H, Poikonen, K, Jokinen, I, et al. . (1983) Selenium and immune functions in humans. Infect Immun 41, 185189.CrossRefGoogle ScholarPubMed
762Boyne, R & Arthur, JR (1986) The response of selenium-deficient mice to Candida albicans infection. J Nutr 116, 816822.CrossRefGoogle ScholarPubMed
763Ciappellano, S, Testolin, G & Porrini, M (1989) Effects of durum wheat dietary selenium on glutathione peroxidase activity and Se content in long-term-fed rats. Ann Nutr Metab 33, 2230.CrossRefGoogle ScholarPubMed
764Douillet, C, Bost, M, Accominotti, M, et al. . (1998) Effect of selenium and vitamin E supplementation on lipid abnormalities in plasma, aorta, and adipose tissue of Zucker rats. Biol Trace Elem Res 65, 221236.CrossRefGoogle ScholarPubMed
765Stapleton, SR (2000) Selenium: an insulin mimetic. Cell Mol Life Sci 57, 18741879.CrossRefGoogle ScholarPubMed
766Uribarri, J (2007) Phosphorus homeostasis in normal health and in chronic kidney disease patients with special emphasis on dietary phosphorus intake. Semin Dial 20, 295301.CrossRefGoogle ScholarPubMed
767Loghman-Adham, M (1997) Adaptation to changes in dietary phosphorus intake in health and in renal failure. J Lab Clin Med 129, 176188.CrossRefGoogle ScholarPubMed
768Kesse, E, Boutron-Ruault, M-C, Norat, T, et al. . (2005) Dietary calcium, phosphorus, vitamin D, dairy products and the risk of colorectal adenoma and cancer among French women of the E3N-EPIC prospective study. Int J Cancer 117, 137144.CrossRefGoogle ScholarPubMed
769Arnold, WH & Gaengler, P (2007) Quantitative analysis of the calcium and phosphorus content of developing and permanent human teeth. Ann Anat 189, 183190.CrossRefGoogle ScholarPubMed
770Bostick, RM, Potter, JD, Fosdick, L, et al. . (1993) Calcium and colorectal epithelial cell proliferation: a preliminary randomized, double-blinded, placebo-controlled clinical trial. J Natl Cancer Inst 85, 132141.CrossRefGoogle ScholarPubMed
771Ishihara, J, Inoue, M, Iwasaki, M, et al. . (2008) Dietary calcium, vitamin D, and the risk of colorectal cancer. Am J Clin Nutr 88, 15761583.CrossRefGoogle ScholarPubMed
772Mariot, P, Vanoverberghe, K, Lalevee, N, et al. . (2002) Overexpression of an α 1H (Cav3.2) T-type calcium channel during neuroendocrine differentiation of human prostate cancer cells. J Biol Chem 277, 1082410833.CrossRefGoogle ScholarPubMed
773Taylor, JT, Zeng, XB, Pottle, JE, et al. . (2008) Calcium signaling and T-type calcium channels in cancer cell cycling. World J Gastroenterol 14, 49844991.CrossRefGoogle ScholarPubMed
774Ciapa, B, Pesando, D, Wilding, M, et al. . (1994) Cell-cycle calcium transients driven by cyclic changes in inositol trisphosphate levels. Nature 368, 875878.CrossRefGoogle ScholarPubMed
775Bucher, HC, Cook, RJ, Guyatt, GH, et al. . (1996) Effects of dietary calcium supplementation on blood pressure. A meta-analysis of randomized controlled trials. JAMA 275, 10161022.CrossRefGoogle ScholarPubMed
776Gillman, MW, Hood, MY, Moore, LL, et al. . (1995) Effect of calcium supplementation on blood pressure in children. J Pediatr 127, 186192.CrossRefGoogle ScholarPubMed
777Umesawa, M, Iso, H, Ishihara, J, et al. . (2008) Dietary calcium intake and risks of stroke, its subtypes, and coronary heart disease in Japanese: The JPHC Study Cohort. Stroke 39, 24492456.CrossRefGoogle ScholarPubMed
778Astrup, A (2008) The role of calcium in energy balance and obesity: the search for mechanisms. Am J Clin Nutr 88, 873874.CrossRefGoogle ScholarPubMed
779Major, GC, Chaput, JP, Ledoux, M, et al. . (2008) Recent developments in calcium-related obesity research. Obes Rev 9, 428445.CrossRefGoogle ScholarPubMed
780Sharp, RL (2006) Role of sodium in fluid homeostasis with exercise. J Am Coll Nutr 25, 231S239S.CrossRefGoogle ScholarPubMed
781Hollenberg, NK (2006) The influence of dietary sodium on blood pressure. J Am Coll Nutr 25, 240S246S.CrossRefGoogle ScholarPubMed
782Alderman, MH (2006) Evidence relating dietary sodium to cardiovascular disease. J Am Coll Nutr 25, 256S261S.CrossRefGoogle ScholarPubMed
783Heaney, RP (2006) Role of dietary sodium in osteoporosis. J Am Coll Nutr 25, 271S276S.CrossRefGoogle ScholarPubMed
784Demigne, C, Sabboh, H, Remesy, C, et al. . (2004) Protective effects of high dietary potassium: nutritional and metabolic aspects. J Nutr 134, 29032906.CrossRefGoogle ScholarPubMed
785He, FJ & MacGregor, GA (2008) Beneficial effects of potassium on human health. Physiologia Plantarum 133, 725735.CrossRefGoogle ScholarPubMed
786Sjogren, A, Floren, CH & Nilsson, A (1988) Oral administration of magnesium hydroxide to subjects with insulin-dependent diabetes mellitus – effects on magnesium and potassium levels and on insulin requirements. Magnesium 7, 117122.Google ScholarPubMed
787Appel, LJ, Moore, TJ, Obarzanek, E, et al. . (1997) A clinical trial of the effects of dietary patterns on blood pressure. N Engl J Med 336, 11171124.CrossRefGoogle ScholarPubMed
788Chang, H-Y, Hu, Y-W, Yue, C-SJ, et al. . (2006) Effect of potassium-enriched salt on cardiovascular mortality and medical expenses of elderly men. Am J Clin Nutr 83, 12891296.CrossRefGoogle ScholarPubMed
789Ishimitsu, T, Tobian, L, Sugimoto, KI, et al. . (1995) High potassium diets reduce macrophage adherence to the vascular wall in stroke-prone spontaneously hypertensive rats. J Vasc Res 32, 406412.CrossRefGoogle Scholar
790Young, DB & Ma, G (1999) Vascular protective effects of potassium. Semin Nephrol 19, 477486.Google ScholarPubMed
791Nolan, J, Batin, PD, Andrews, R, et al. . (1998) Prospective study of heart rate variability and mortality in chronic heart failure: results of the United Kingdom Heart Failure Evaluation and Assessment of Risk Trial (UK-Heart). Circulation 98, 15101516.CrossRefGoogle ScholarPubMed
792Tobian, L, Macneill, D, Johnson, MA, et al. . (1984) Potassium protection against lesions of the renal tubules, arteries, and glomeruli and nephron loss in salt-loaded hypertensive Dahl S-rats. Hypertension 6, I170I176.CrossRefGoogle ScholarPubMed
793Curhan, GC, Willett, WC, Rimm, EB, et al. . (1993) A prospective study of dietary calcium and other nutrients and the risk of symptomatic kidney stones. N Engl J Med 328, 833838.CrossRefGoogle ScholarPubMed
794Marangella, M, Di Stefano, M, Casalis, S, et al. . (2004) Effects of potassium citrate supplementation on bone metabolism. Calcif Tissue Int 74, 330335.CrossRefGoogle ScholarPubMed
795Lemann, J Jr, Pleuss, JA, Gray, RW, et al. . (1991) Potassium administration increases and potassium deprivation reduces urinary calcium excretion in healthy adults. Kidney Int 39, 973983.CrossRefGoogle ScholarPubMed
796Institute of Medicine (1998) Dietary Reference Intakes for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and Choline. Washington, DC: National Academy Press.Google Scholar
797Singleton, CK & Martin, PR (2001) Molecular mechanisms of thiamine utilization. Curr Mol Med 1, 197207.CrossRefGoogle ScholarPubMed
798Lukienko, P, Mel'nichenko, N, Zverinskii, I, et al. . (2000) Antioxidant properties of thiamine. Bull Exp Biol Med 130, 874876.CrossRefGoogle ScholarPubMed
799Andreasen, MF, Christensen, LP, Meyer, AS, et al. . (2000) Content of phenolic acids and ferulic acid dehydrodimers in 17 rye (Secale cereale L.) varieties. J Agric Food Chem 48, 28372842.CrossRefGoogle ScholarPubMed
800Ambrose, ML, Bowden, SC & Whelan, G (2001) Thiamin treatment and working memory function of alcohol-dependent people: preliminary findings. Alcohol Clin Exp Res 25, 112116.Google ScholarPubMed
801Fairweather-Tait, SJ, Powers, HJ, Minski, MJ, et al. . (1992) Riboflavin deficiency and iron absorption in adult Gambian men. Ann Nutr Metab 36, 3440.CrossRefGoogle ScholarPubMed
802Sirivech, S, Driskell, J & Frieden, E (1977) NADH-FMN oxidoreductase activity and iron content of organs from riboflavin- and iron-deficient rats. J Nutr 107, 739745.CrossRefGoogle ScholarPubMed
803Yates, CA, Evans, GS & Powers, HJ (2001) Riboflavin deficiency: early effects on post-weaning development of the duodenum in rats. Br J Nutr 86, 593599.CrossRefGoogle ScholarPubMed
804Sterner, RT & Price, WR (1973) Restricted riboflavin: within-subject behavioral effects in humans. Am J Clin Nutr 26, 150160.CrossRefGoogle ScholarPubMed
805Miyamoto, Y & Sancar, A (1998) Vitamin B2-based blue-light photoreceptors in the retinohypothalamic tract as the photoactive pigments for setting the circadian clock in mammals. Proc Natl Acad Sci U S A 95, 60976102.CrossRefGoogle ScholarPubMed
806Mack, CP, Hultquist, DE & Shlafer, M (1995) Myocardial flavin reductase and riboflavin: a potential role in decreasing reoxygenation injury. Biochem Biophys Res Commun 212, 3540.CrossRefGoogle ScholarPubMed
807Powers, HJ (2003) Riboflavin (vitamin B-2) and health. Am J Clin Nutr 77, 13521360.CrossRefGoogle ScholarPubMed
808Siassi, F & Ghadirian, P (2005) Riboflavin deficiency and esophageal cancer: a case control-household study in the Caspian Littoral of Iran. Cancer Detect Prev 29, 464469.CrossRefGoogle ScholarPubMed
809Webster, RP, Gawde, MD & Bhattacharya, RK (1996) Modulation of carcinogen-induced DNA damage and repair enzyme activity by dietary riboflavin. Cancer Lett 98, 129135.CrossRefGoogle ScholarPubMed
810Figge, HL, Figge, J, Souney, PF, et al. . (1988) Nicotinic acid – a review of its clinical use in the treatment of lipid disorders. Pharmacotherapy 8, 287294.CrossRefGoogle ScholarPubMed
811Hodis, HN (1995) Reversibility of atherosclerosis – evolving perspectives from two arterial imaging clinical trials: The Cholesterol Lowering Atherosclerosis Regression study and the Monitored Atherosclerosis Regression study. J Cardiovasc Pharmacol 25, S25S31.CrossRefGoogle ScholarPubMed
812Jacobson, EL, Dame, AJ, Pyrek, JS, et al. . (1995) Evaluating the role of niacin in human carcinogenesis. Biochimie 77, 394398.CrossRefGoogle ScholarPubMed
813Pontes Monteiro, J, Ferreira da Cunha, D, Correia Filho, D, et al. . (2004) Niacin metabolite excretion in alcoholic pellagra and AIDS patients with and without diarrhea. Nutrition 20, 778782.CrossRefGoogle Scholar
814Jonas, W, Rapoza, C & Blair, W (1996) The effect of niacinamide on osteoarthritis: a pilot study. Inflamm Res 45, 330334.CrossRefGoogle ScholarPubMed
815Carlson, LA (1963) Studies on effect of nicotinic acid on catecholamine stimulated lipolysis in adipose tissue in vitro. Acta Med Scand 173, 719722.CrossRefGoogle ScholarPubMed
816Davies, JI & Souness, JE (1981) The mechanisms of hormone and drug actions on fatty acid release from adipose tissue. Rev Pure Appl Pharmacol Sci 2, 1112.Google ScholarPubMed
817Marcus, C, Sonnenfeld, T, Karpe, B, et al. . (1989) Inhibition of lipolysis by agents acting via adenylate cyclase in fat cells from infants and adults. Pediatr Res 26, 255259.CrossRefGoogle Scholar
818Anguita, M, Gasa, J, Martin-Orue, SM, et al. . (2006) Study of the effect of technological processes on starch hydrolysis, non-starch polysaccharides solubilization and physicochemical properties of different ingredients using a two-step in vitro system. Anim Feed Sci Tech 129, 99115.CrossRefGoogle Scholar
819Ubbink, JB, Becker, PJ & Vermaak, WJH (1996) Will an increased dietary folate intake reduce the incidence of cardiovascular disease? Nutr Rev 54, 213216.CrossRefGoogle ScholarPubMed
820Brattström, L, Israelsson, B, Norrving, B, et al. . (1990) Impaired homocysteine metabolism in early-onset cerebral and peripheral occlusive arterial disease. Effects of pyridoxine and folic acid treatment. Atherosclerosis 81, 5160.CrossRefGoogle ScholarPubMed
821McMahon, RJ (2002) Biotin in metabolism and molecular biology. Annu Rev Nutr 22, 221239.CrossRefGoogle ScholarPubMed
822Said, HM (2009) Cell and molecular aspects of human intestinal biotin absorption. J Nutr 139, 158162.CrossRefGoogle ScholarPubMed
823Sweetman, L & Nyhan, WL (1986) Inheritable biotin-treatable disorders and associated phenomena. Annu Rev Nutr 6, 317343.CrossRefGoogle ScholarPubMed
824Rodriguez-Melendez, R & Zempleni, J (2003) Regulation of gene expression by biotin (review). J Nutr Biochem 14, 680690.CrossRefGoogle ScholarPubMed
825Manthey, KC, Griffin, JB & Zempleni, J (2002) Biotin supply affects expression of biotin transporters, biotinylation of carboxylases and metabolism of interleukin-2 in Jurkat cells. J Nutr 132, 887892.CrossRefGoogle ScholarPubMed
826Baez-Saldana, A, Diaz, G, Espinoza, B, et al. . (1998) Biotin deficiency induces changes in subpopulations of spleen lymphocytes in mice. Am J Clin Nutr 67, 431437.CrossRefGoogle ScholarPubMed
827Rabin, BS (1983) Inhibition of experimentally induced autoimmunity in rats by biotin deficiency. J Nutr 113, 23162322.CrossRefGoogle ScholarPubMed
828Kamen, B (1997) Folate and antifolate pharmacology. Semin Oncol 24, 3039.Google ScholarPubMed
829Moat, SJ, Hill, MH, McDowell, IFW, et al. . (2003) Reduction in plasma total homocysteine through increasing folate intake in healthy individuals is not associated with changes in measures of antioxidant activity or oxidant damage. Eur J Clin Nutr 57, 483489.CrossRefGoogle ScholarPubMed
830Ward, M, McNulty, H, McPartlin, J, et al. . (1997) Plasma homocysteine, a risk factor for cardiovascular disease, is lowered by physiological doses of folic acid. QJM 90, 519524.CrossRefGoogle ScholarPubMed
831Shaw, GM, Schaffer, D, Velie, EM, et al. . (1995) Periconceptional vitamin use, dietary folate, and the occurrence of neural tube defects. Epidemiology 6, 219226.CrossRefGoogle ScholarPubMed
832Giovannucci, E (2002) Epidemiologic studies of folate and colorectal neoplasia: a review. J Nutr 132, 2350S2355S.CrossRefGoogle ScholarPubMed
833Jennings, E (1995) Folic acid as a cancer-preventing agent. Med Hypotheses 45, 297303.CrossRefGoogle ScholarPubMed
834Macgregor, JT, Schlegel, R, Wehr, CM, et al. . (1990) Cytogenetic damage induced by folate deficiency in mice is enhanced by caffeine. Proc Natl Acad Sci U S A 87, 99629965.CrossRefGoogle ScholarPubMed
835Akilzhanova, A, Takamura, N, Aoyagi, K, et al. . (2006) Folic acid deficiency: main etiological factor of megaloblastic anemia in Kazakhstan? Am J Hematol 81, 471.CrossRefGoogle ScholarPubMed
836Ebisch, IMW, Thomas, CMG, Peters, WHM, et al. . (2007) The importance of folate, zinc and antioxidants in the pathogenesis and prevention of subfertility. Hum Reprod Update 13, 163174.CrossRefGoogle ScholarPubMed
837Zeisel, SH (2009) Importance of methyl donors during reproduction. Am J Clin Nutr 89, 673S677S.CrossRefGoogle ScholarPubMed
838Gey, KF (1998) Vitamins E plus C and interacting conutrients required for optimal health: a critical and constructive review of epidemiology and supplementation data regarding cardiovascular disease and cancer. Biofactors 7, 113174.Google Scholar
839Leger, CL (2000) Prevention of cardiovascular risk by vitamin E. Ann Biol Clin 58, 527540.Google ScholarPubMed
840Bowry, VW & Ingold, KU (1999) The unexpected role of vitamin E (α-tocopherol) in the peroxidation of human low-density lipoprotein. Acc Chem Res 32, 2734.CrossRefGoogle Scholar
841Duthie, SJ, Ma, A, Ross, MA, et al. . (1996) Antioxidant supplementation decreases oxidative DNA damage in human lymphocytes. Cancer Res 56, 12911295.Google ScholarPubMed
842Poulin, JE, Cover, C, Gustafson, MR, et al. . (1996) Vitamin E prevents oxidative modification of brain and lymphocyte band 3 proteins during aging. Proc Natl Acad Sci U S A 93, 56005603.CrossRefGoogle ScholarPubMed
843Yu, WP, Simmons-Menchaca, M, Gapor, A, et al. . (1999) Induction of apoptosis in human breast cancer cells by tocopherols and tocotrienols. Nutr Cancer 33, 2632.CrossRefGoogle ScholarPubMed
844Azzi, A & Stocker, A (2000) Vitamin E: non-antioxidant roles. Prog Lipid Res 39, 231255.CrossRefGoogle ScholarPubMed
845Tasinato, A, Boscoboinik, D, Bartoli, GM, et al. . (1995) d-α-Tocopherol inhibition of vascular smooth muscle cell proliferation occurs at physiological concentrations, correlates with protein kinase C inhibition, and is independent of its antioxidant properties. Proc Natl Acad Sci U S A 92, 1219012194.CrossRefGoogle ScholarPubMed
846Devaraj, S & Jialal, I (1999) α-Tocopherol decreases interleukin-1β release from activated human monocytes by inhibition of 5-lipoxygenase. Arterioscler Thromb Vasc Biol 19, 11251133.CrossRefGoogle ScholarPubMed
847Ricciarelli, R, Zingg, J-M & Azzi, A (2000) Vitamin E reduces the uptake of oxidized LDL by inhibiting CD36 scavenger receptor expression in cultured aortic smooth muscle cells. Circulation 102, 8287.CrossRefGoogle ScholarPubMed
848Teupser, D, Thiery, J & Seidel, D (1999) α-Tocopherol down-regulates scavenger receptor activity in macrophages. Atherosclerosis 144, 109115.CrossRefGoogle ScholarPubMed
849Christen, S, Woodall, AA, Shigenaga, MK, et al. . (1997) γ-Tocopherol traps mutagenic electrophiles such as NOx and complements α-tocopherol: physiological implications. Proc Natl Acad Sci U S A 94, 32173222.CrossRefGoogle ScholarPubMed
850Wolf, G (1997) γ-Tocopherol: an efficient protector of lipids against nitric oxide-initiated peroxidative damage. Nutr Rev 55, 376378.CrossRefGoogle ScholarPubMed
851Chatelain, E, Boscoboinik, DO, Bartoli, GM, et al. . (1993) Inhibition of smooth muscle cell proliferation and protein kinase C activity by tocopherols and tocotrienols. Biochim Biophys Acta 1176, 8389.CrossRefGoogle ScholarPubMed
852Stolzenberg-Solomon, RZ, Sheffler-Collins, S, Weinstein, S, et al. . (2009) Vitamin E intake, α-tocopherol status, and pancreatic cancer in a cohort of male smokers. Am J Clin Nutr 89, 584591.CrossRefGoogle Scholar
853Laight, DW, Desai, KM, Gopaul, NK, et al. . (1999) F2-isoprostane evidence of oxidant stress in the insulin resistant, obese Zucker rat: effects of vitamin E. Eur J Pharmacol 377, 8992.CrossRefGoogle ScholarPubMed
854Ima-Nirwana, S & Suhaniza, S (2004) Effects of tocopherols and tocotrienols on body composition and bone calcium content in adrenalectomized rats replaced with dexamethasone. J Med Food 7, 4551.CrossRefGoogle ScholarPubMed
855Norazlina, M, Ima-Nirwana, S, Gapor, MTA, et al. . (2002) Tocotrienols are needed for normal bone calcification in growing female rats. Asia Pac J Clin Nutr 11, 194199.CrossRefGoogle ScholarPubMed
856Suttie, JW (1992) Vitamin K and human nutrition. J Am Diet Assoc 92, 585590.CrossRefGoogle ScholarPubMed
857Suttie, J (1993) Synthesis of vitamin K-dependent proteins. FASEB J 7, 445452.CrossRefGoogle ScholarPubMed
858Hodges, SJ, Akesson, K, Vergnaud, P, et al. . (1993) Circulating levels of vitamin K1 and vitamin K2 decreased in elderly women with hip fracture. J Bone Miner Res 8, 12411245.CrossRefGoogle ScholarPubMed
859Luo, G, Ducy, P, McKee, MD, et al. . (1997) Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 386, 7881.CrossRefGoogle ScholarPubMed
860Mayne, S (1996) β-Carotene, carotenoids, and disease prevention in humans. FASEB J 10, 690701.CrossRefGoogle ScholarPubMed
861Baron, JA, Cole, BF, Mott, L, et al. . (2003) Neoplastic and antineoplastic effects of β-carotene on colorectal adenoma recurrence: results of a randomized trial. J Natl Cancer Inst 95, 717722.CrossRefGoogle ScholarPubMed
862Holick, CN, Michaud, DS, Stolzenberg-Solomon, R, et al. . (2002) Dietary carotenoids, serum β-carotene, and retinol and risk of lung cancer in the Alpha-Tocopherol, Beta-Carotene Cohort Study. Am J Epidemiol 156, 536547.CrossRefGoogle ScholarPubMed
863Michaud, DS, Feskanich, D, Rimm, EB, et al. . (2000) Intake of specific carotenoids and risk of lung cancer in 2 prospective US cohorts. Am J Clin Nutr 72, 990997.CrossRefGoogle ScholarPubMed
864Touvier, M, Kesse, E, Clavel-Chapelon, F, et al. . (2005) Dual association of β-carotene with risk of tobacco-related cancers in a cohort of French women. J Natl Cancer Inst 97, 13381344.CrossRefGoogle Scholar
865Rettura, G, Duttagupta, C, Listowsky, P, et al. . (1983) Dimethylbenz(a)anthracene (DMBA)-induced tumors: prevention by supplemental β-carotene (BC). Fed Proc 42, 786.Google Scholar
866Cui, Y, Lu, Z, Bai, L, et al. . (2007) β-Carotene induces apoptosis and up-regulates peroxisome proliferator-activated receptor γ expression and reactive oxygen species production in MCF-7 cancer cells. Eur J Cancer 43, 25902601.CrossRefGoogle ScholarPubMed
867Prabhala, RH, Braune, LM, Garewal, HS, et al. . (1993) Influence of β-carotene on immune functions. Ann N Y Acad Sci 691, 262263.CrossRefGoogle ScholarPubMed
868Packer, JE, Mahood, JS, Mora-Arellano, VO, et al. . (1981) Free radicals and singlet oxygen scavengers: reaction of a peroxy-radical with β-carotene, diphenyl furan and 1,4-diazobicyclo(2,2,2)-octane. Biochem Biophys Res Commun 98, 901906.CrossRefGoogle Scholar
869Osganian, SK, Stampfer, MJ, Rimm, E, et al. . (2003) Dietary carotenoids and risk of coronary artery disease in women. Am J Clin Nutr 77, 13901399.CrossRefGoogle ScholarPubMed
870Granado, F, Olmedilla, B & Blanco, I (2003) Nutritional and clinical relevance of lutein in human health. Br J Nutr 90, 487502.CrossRefGoogle ScholarPubMed
871Stringheta, PC, Nachtigall, AM, Oliveira, TT, et al. . (2006) Lutein: antioxidant properties and health benefits. Alimentos e Nutricao 17, 229237.Google Scholar
872Richer, S, Stiles, W, Statkute, L, et al. . (2004) Double-masked, placebo-controlled, randomized trial of lutein and antioxidant supplementation in the intervention of atrophic age-related macular degeneration: the Veterans LAST study (Lutein Antioxidant Supplementation Trial). Optometry 75, 216229.CrossRefGoogle ScholarPubMed
873Seddon, JM, Ajani, UA, Sperduto, RD, et al. . (1994) Dietary carotenoids, vitamins A, C, and E, and advanced age-related macular degeneration. Eye Disease Case–Control Study Group. JAMA 272, 14131420.CrossRefGoogle Scholar
874Olmedilla, B, Granado, F, Blanco, I, et al. . (2003) Lutein, but not α-tocopherol, supplementation improves visual function in patients with age-related cataracts: a 2-y double-blind, placebo-controlled pilot study. Nutrition 19, 2124.CrossRefGoogle Scholar
875Curran-Celentano, J, Hammond, BR Jr, Ciulla, TA, et al. . (2001) Relation between dietary intake, serum concentrations, and retinal concentrations of lutein and zeaxanthin in adults in a Midwest population. Am J Clin Nutr 74, 796802.CrossRefGoogle Scholar
876Anonymous (2005) Lutein and zeaxanthin. Monograph. Altern Med Rev 10, 128135.Google Scholar
877Schäffer, MW, Roy, SS, Mukherjee, S, et al. . (2008) Identification of lutein, a dietary antioxidant carotenoid in guinea pig tissues. Biochem Biophys Res Commun 374, 378381.CrossRefGoogle ScholarPubMed
878Stahl, W & Sies, H (2002) Carotenoids and protection against solar UV radiation. Skin Pharmacol Appl Skin Physiol 15, 291296.CrossRefGoogle ScholarPubMed
879Slattery, ML, Benson, J, Curtin, K, et al. . (2000) Carotenoids and colon cancer. Am J Clin Nutr 71, 575582.CrossRefGoogle ScholarPubMed
880Dwyer, JH, Navab, M, Dwyer, KM, et al. . (2001) Oxygenated carotenoid lutein and progression of early atherosclerosis: The Los Angeles Atherosclerosis Study. Circulation 103, 29222927.CrossRefGoogle ScholarPubMed
881Yeum, K-J, Shang, F, Schalch, W, et al. . (1999) Fat-soluble nutrient concentrations in different layers of human cataractous lens. Curr Eye Res 19, 502505.CrossRefGoogle ScholarPubMed
882Uchiyama, S, Sumida, T & Yamaguchi, M (2004) Oral administration of β-cryptoxanthin induces anabolic effects on bone components in the femoral tissues of rats in vivo. Biol Pharm Bull 27, 232235.CrossRefGoogle Scholar
883Yamaguchi, M & Uchiyama, S (2004) β-Cryptoxanthin stimulates bone formation and inhibits bone resorption in tissue culture in vitro. Mol Cell Biochem 258, 137144.CrossRefGoogle ScholarPubMed
884Kohno, H, Taima, M, Sumida, T, et al. . (2001) Inhibitory effect of mandarin juice rich in β-cryptoxanthin and hesperidin on 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced pulmonary tumorigenesis in mice. Cancer Lett 174, 141150.CrossRefGoogle Scholar
885Lian, F, Kang-Quan, Hu K-Q, Russell, RM, et al. . (2006) β-Cryptoxanthin suppresses the growth of immortalized human bronchial epithelial cells and non-small-cell lung cancer cells and up-regulates retinoic acid receptor β expression. Int J Cancer 119, 20842089.CrossRefGoogle ScholarPubMed
886Yuan, J-M, Ross, RK, Chu, X-D, et al. . (2001) Prediagnostic levels of serum β-cryptoxanthin and retinol predict smoking-related lung cancer risk in Shanghai, China. Cancer Epidemiol Biomarkers Prev 10, 767773.Google ScholarPubMed
887Tanaka, T, Kohno, H, Murakami, M, et al. . (2000) Suppression of azoxymethane-induced colon carcinogenesis in male F344 rats by mandarin juices rich in β-cryptoxanthin and hesperidin. Int J Cancer 88, 146150.3.0.CO;2-I>CrossRefGoogle Scholar
888Nogushi, S, Sumida, T, Ogawa, H, et al. . (2003) Effects of oxygenated carotenoid β-cryptoxanthin on morphological differentiation and apoptosis in Neuro2a neuroblastoma cells. Biosci Biotechnol Biochem 67, 24672469.CrossRefGoogle Scholar
889Castelao, JE & Olmedilla, B (2002) Vitamins A, C and E, folate and most carotenoids do not influence bladder cancer risk. Evid Based Oncol 3, 5859.CrossRefGoogle Scholar
890Rice-Evans, CA, Miller, NJ & Paganga, G (1997) Antioxidant properties of phenolic compounds. Trends Plant Sci 2, 152159.CrossRefGoogle Scholar
891Adisakwattana, S, Moonsan, P & Yibchok-anun, S (2008) Insulin-releasing properties of a series of cinnamic acid derivatives in vitro and in vivo. J Agric Food Chem 56, 78387844.CrossRefGoogle ScholarPubMed
892Tanaka, T, Kojima, T, Kawamori, T, et al. . (1993) Inhibition of 4-nitroquinoline-1-oxide-induced rat tongue carcinogenesis by the naturally occurring plant phenolics caffeic, ellagic, chlorogenic and ferulic acids. Carcinogenesis 14, 13211325.CrossRefGoogle ScholarPubMed
893Ferguson, LR, Zhu, ST & Harris, PJ (2005) Antioxidant and antigenotoxic effects of plant cell wall hydroxycinnamic acids in cultured HT-29 cells. Mol Nutr Food Res 49, 585593.CrossRefGoogle ScholarPubMed
894Hsu, C-L, Wu, C-H, Huang, S-L, et al. . (2009) Phenolic compounds rutin and o-coumaric acid ameliorate obesity induced by high-fat diet in rats. J Agric Food Chem 57, 425431.CrossRefGoogle ScholarPubMed
895Sri Balasubashini, M, Rukkumani, R, Viswanathan, P, et al. . (2004) Ferulic acid alleviates lipid peroxidation in diabetic rats. Phytother Res 18, 310314.CrossRefGoogle Scholar
896Kamal-Eldin, A, Frank, J, Razdan, A, et al. . (2000) Effects of dietary phenolic compounds on tocopherol, cholesterol, and fatty acids in rats. Lipids 35, 427435.CrossRefGoogle ScholarPubMed
897Sri Balasubashini, M, Rukkumani, R & Menon, VP (2003) Protective effects of ferulic acid on hyperlipidemic diabetic rats. Acta Diabetol 40, 118122.CrossRefGoogle ScholarPubMed
898Suzuki, A, Kagawa, D, Fujii, A, et al. . (2002) Short- and long-term effects of ferulic acid on blood pressure in spontaneously hypertensive rats. Am J Hypertens 15, 351357.CrossRefGoogle Scholar
899Jung, EH, Kim, SR, Hwang, IK, et al. . (2007) Hypoglycemic effects of a phenolic acid fraction of rice bran and ferulic acid in C57BL/KsJ-db/db mice. J Agric Food Chem 55, 98009804.CrossRefGoogle ScholarPubMed
900Hollman, PCH & Katan, MB (1997) Absorption, metabolism and health effects of dietary flavonoids in man. Biomed Pharmacother 51, 305310.CrossRefGoogle ScholarPubMed
901Van Wauwe, J & Goossens, J (1983) Effects of antioxidants on cyclooxygenase and lipoxygenase activities in intact human platelets: comparison with indomethacin and ETYA. Prostaglandins 26, 725730.CrossRefGoogle ScholarPubMed
902Cushnie, TPT & Lamb, AJ (2005) Antimicrobial activity of flavonoids. Int J Antimicrob Agents 26, 343356.CrossRefGoogle ScholarPubMed
903Zhang, G, Qin, L, Hung, WY, et al. . (2006) Flavonoids derived from herbal Epimedium Brevicornum Maxim prevent OVX-induced osteoporosis in rats independent of its enhancement in intestinal calcium absorption. Bone 38, 818825.CrossRefGoogle ScholarPubMed
904Chiang, AN, Wu, HL, Yeh, HI, et al. . (2006) Antioxidant effects of black rice extract through the induction of superoxide dismutase and catalase activities. Lipids 41, 797803.CrossRefGoogle ScholarPubMed
905Ling, WH, Wang, LL & Ma, J (2002) Supplementation of the black rice outer layer fraction to rabbits decreases atherosclerotic plaque formation and increases antioxidant status. J Nutr 132, 2026.CrossRefGoogle ScholarPubMed
906Nam, SH, Choi, SP, Kang, MY, et al. . (2006) Antioxidative activities of bran extracts from twenty one pigmented rice cultivars. Food Chem 94, 613620.CrossRefGoogle Scholar
907Tsuda, T, Horio, F & Osawa, T (2002) Cyanidin 3-O-β-d-glucoside suppresses nitric oxide production during a zymosan treatment in rats. J Nutr Sci Vitaminol 48, 305310.CrossRefGoogle ScholarPubMed
908Xia, XD, Ling, WH, Ma, J, et al. . (2006) An anthocyanin-rich extract from black rice enhances atherosclerotic plaque stabilization in apolipoprotein E-deficient mice. J Nutr 136, 22202225.CrossRefGoogle ScholarPubMed
909Hyun, JW & Chung, HS (2004) Cyanidin and malvidin from Oryza sativa cv. heugjinjubyeo mediate cytotoxicity against human monocytic leukemia cells by arrest of G2/M phase and induction of apoptosis. J Agric Food Chem 52, 22132217.CrossRefGoogle Scholar
910Zhao, C, Giusti, MM, Malik, M, et al. . (2004) Effects of commercial anthocyanin-rich extracts on colonic cancer and nontumorigenic colonic cell growth. J Agric Food Chem 52, 61226128.CrossRefGoogle ScholarPubMed
911Tsuda, T, Horio, F, Uchida, K, et al. . (2003) Dietary cyanidin 3-O-β-d-glucoside-rich purple corn color prevents obesity and ameliorates hyperglycemia in mice. J Nutr 133, 21252130.CrossRefGoogle ScholarPubMed
912Kaludjerovic, J & Ward, WE (2009) Neonatal exposure to daidzein, genistein, or the combination modulates bone development in female CD-1 mice. J Nutr 139, 467473.CrossRefGoogle ScholarPubMed
913Jenab, M & Thompson, LU (1996) The influence of flaxseed and lignans on colon carcinogenesis and β-glucuronidase activity. Carcinogenesis 17, 13431348.CrossRefGoogle ScholarPubMed
914Oikarinen, SI, Pajari, A-M & Mutanen, M (2000) Chemopreventive activity of crude hydroxsymatairesinol (HMR) extract in ApcMin mice. Cancer Lett 161, 253258.CrossRefGoogle Scholar
915Prasad, K (2005) Hypocholesterolemic and antiatherosclerotic effect of flax lignan complex isolated from flaxseed. Atherosclerosis 179, 269275.CrossRefGoogle ScholarPubMed
916Kamal-Eldin, A, Pouru, A, Eliasson, C, et al. . (2001) Alkylresorcinols as antioxidants: hydrogen donation and peroxyl radical-scavenging effects. J Sci Food Agric 81, 353356.3.0.CO;2-X>CrossRefGoogle Scholar
917Kozubek, A & Nienartowicz, B (1995) Cereal grain resorcinolic lipids inhibit H2O2-induced peroxidation of biological membranes. Acta Biochim Pol 42, 309315.CrossRefGoogle ScholarPubMed
918Gasiorowski, K, Szyba, K, Brokos, B, et al. . (1996) Antimutagenic activity of alkylresorcinols from cereal grains. Cancer Lett 106, 109115.CrossRefGoogle ScholarPubMed
919Olthof, MR, van Vliet, T, Boelsma, E, et al. . (2003) Low dose betaine supplementation leads to immediate and long term lowering of plasma homocysteine in healthy men and women. J Nutr 133, 41354138.CrossRefGoogle ScholarPubMed
920Handler, JS & Kwon, HM (2001) Cell and molecular biology of organic osmolyte accumulation in hypertonic renal cells. Nephron 87, 106110.CrossRefGoogle ScholarPubMed
921Konstantinova, SV, Tell, GS, Vollset, SE, et al. . (2008) Divergent associations of plasma choline and betaine with components of metabolic syndrome in middle age and elderly men and women. J Nutr 138, 914920.CrossRefGoogle ScholarPubMed
922Detopoulou, P, Panagiotakos, DB, Antonopoulou, S, et al. . (2008) Dietary choline and betaine intakes in relation to concentrations of inflammatory markers in healthy adults: the ATTICA study. Am J Clin Nutr 87, 424430.CrossRefGoogle ScholarPubMed
923Lv, S, Fan, R, Du, Y, et al. . (2009) Betaine supplementation attenuates atherosclerotic lesion in apolipoprotein E-deficient mice. Eur J Nutr 48, 205212.CrossRefGoogle ScholarPubMed
924Delgado-Reyes, CV & Garrow, TA (2005) High sodium chloride intake decreases betaine-homocysteine S-methyltransferase expression in guinea pig liver and kidney. Am J Physiol Regul Integr Comp Physiol 288, R182R187.CrossRefGoogle ScholarPubMed
925Kwon, DY, Jung, YS, Kim, SJ, et al. . (2009) Impaired sulfur-amino acid metabolism and oxidative stress in nonalcoholic fatty liver are alleviated by betaine supplementation in rats. J Nutr 139, 6368.CrossRefGoogle ScholarPubMed
926Albright, CD, Tsai, AY, Friedrich, CB, et al. . (1999) Choline availability alters embryonic development of the hippocampus and septum in the rat. Dev Brain Res 113, 1320.CrossRefGoogle ScholarPubMed
927Meck, WH & Williams, CL (2003) Metabolic imprinting of choline by its availability during gestation: implications for memory and attentional processing across the lifespan. Neurosci Biobehav Rev 27, 385399.CrossRefGoogle ScholarPubMed
928Sanders, LM & Zeisel, SH (2007) Choline. Nutr Today 42, 181186.CrossRefGoogle ScholarPubMed
929Cho, E, Zeisel, SH, Jacques, P, et al. . (2006) Dietary choline and betaine assessed by food-frequency questionnaire in relation to plasma total homocysteine concentration in the Framingham Offspring Study. Am J Clin Nutr 83, 905911.CrossRefGoogle ScholarPubMed
930Sachan, DS, Hongu, N & Johnsen, M (2005) Decreasing oxidative stress with choline and carnitine in women. J Am Coll Nutr 24, 172176.CrossRefGoogle ScholarPubMed
931Dodson, W & Sachan, D (1996) Choline supplementation reduces urinary carnitine excretion in humans. Am J Clin Nutr 63, 904910.CrossRefGoogle ScholarPubMed
932Daily, JW, Hongu, N, Mynatt, RL, et al. . (1998) Choline supplementation increases tissue concentrations of carnitine and lowers body fat in guinea pigs. J Nutr Biochem 9, 464470.CrossRefGoogle Scholar
933Sachan, DS & Hongu, N (2000) Increases in VO2max and metabolic markers of fat oxidation by caffeine, carnitine, and choline supplementation in rats. J Nutr Biochem 11, 521526.CrossRefGoogle ScholarPubMed
934Exton, JH (1994) Phosphatidylcholine breakdown and signal transduction. Biochim Biophys Acta 1212, 2642.CrossRefGoogle ScholarPubMed
935Hannun, Y (1994) The sphingomyelin cycle and the second messenger function of ceramide. J Biol Chem 269, 31253128.CrossRefGoogle ScholarPubMed
936Cohen, EL & Wurtman, RJ (1975) Brain acetylcholine: increase after systematic choline administration. Life Sci 16, 10951102.CrossRefGoogle Scholar
937Frenkel, RA, Muguruma, K & Johnston, JM (1996) The biochemical role of platelet-activating factor in reproduction. Prog Lipid Res 35, 155168.CrossRefGoogle ScholarPubMed
938Haubrich, DR, Wedeking, PW & Wang, PFL (1974) Increase in tissue concentration of acetylcholine in guinea pigs in vivo induced by administration of choline. Life Sci 14, 921927.CrossRefGoogle ScholarPubMed
939Zeisel, SH (2006) Choline: critical role during fetal development and dietary requirements in adults. Annu Rev Nutr 26, 229250.CrossRefGoogle ScholarPubMed
940Henning, SM & Swendseid, ME (1996) The role of folate, choline, and methionine in carcinogenesis induced by methyl-deficient diets. Adv Exp Med Biol 399, 143155.CrossRefGoogle ScholarPubMed
941Niculescu, MD, Craciunescu, CN & Zeisel, SH (2006) Dietary choline deficiency alters global and gene-specific DNA methylation in the developing hippocampus of mouse fetal brains. FASEB J 20, 4349.CrossRefGoogle ScholarPubMed
942Brufau, G, Canela, MA & Rafecas, M (2008) Phytosterols: physiologic and metabolic aspects related to cholesterol-lowering properties. Nutr Res 28, 217225.CrossRefGoogle ScholarPubMed
943de Jong, A, Plat, J & Mensink, RP (2003) Metabolic effects of plant sterols and stanols (review). J Nutr Biochem 14, 362369.CrossRefGoogle ScholarPubMed
944Batta, AK, Xu, GR, Bollineni, JS, et al. . (2005) Effect of high plant sterol-enriched diet and cholesterol absorption inhibitor, SCH 58235, on plant sterol absorption and plasma concentrations in hypercholesterolemic wild-type Kyoto rats. Metabolism 54, 3848.CrossRefGoogle ScholarPubMed
945Jones, PJH, MacDougall, DE, Ntanios, F, et al. . (1997) Dietary phytosterols as cholesterol-lowering agents in humans. Can J Physiol Pharmacol 75, 217227.CrossRefGoogle ScholarPubMed
946Lee, Y-M, Haastert, B, Scherbaum, W, et al. . (2003) A phytosterol-enriched spread improves the lipid profile of subjects with type 2 diabetes mellitus. Eur J Nutr 42, 111117.CrossRefGoogle ScholarPubMed
947Yankah, VV & Jones, PJH (2001) Phytosterols and health implications – efficacy and nutritional aspects. Inform 12, 899903.Google Scholar
948Demonty, I, Ras, RT, van der Knaap, HCM, et al. . (2009) Continuous dose–response relationship of the LDL-cholesterol-lowering effect of phytosterol intake. J Nutr 139, 271284.CrossRefGoogle ScholarPubMed
949Awad, AB, Smith, AJ & Fink, CS (2001) Plant sterols regulate rat vascular smooth muscle cell growth and prostacyclin release in culture. Prostaglandins Leukot Essent Fatty Acids 64, 323330.CrossRefGoogle ScholarPubMed
950Bouic, PJD, Clark, A, Lamprecht, J, et al. . (1999) The effects of B-sitosterol (BSS) and B-sitosterol glucoside (BSSG) mixture on selected immune parameters of marathon runners: inhibition of post marathon immune suppression and inflammation. Int J Sports Med 20, 258262.CrossRefGoogle ScholarPubMed
951Tanaka, M, Misawa, E, Ito, Y, et al. . (2006) Identification of five phytosterols from aloe vera gel as anti-diabetic compounds. Biol Pharm Bull 29, 14181422.CrossRefGoogle ScholarPubMed
952Awad, AB, Chen, YC, Fink, CS, et al. . (1996) β-Sitosterol inhibits HT-29 human colon cancer cell growth and alters membrane lipids. Anticancer Res 16, 27972804.Google ScholarPubMed
953Raicht, RF, Cohen, BI, Fazzini, EP, et al. . (1980) Protective effect of plant sterols against chemically induced colon tumors in rats. Cancer Res 40, 403405.Google ScholarPubMed
954Berges, RR, Windeler, J, Trampisch, HJ, et al. . (1995) Randomised, placebo-controlled, double-blind clinical trial of β-sitosterol in patients with benign prostatic hyperplasia. Lancet 345, 15291532.CrossRefGoogle ScholarPubMed
955Rubis, B, Paszel, A, Kaczmarek, M, et al. . (2008) Beneficial or harmful influence of phytosterols on human cells? Br J Nutr 100, 11831191.CrossRefGoogle ScholarPubMed
956Awad, AB, Roy, R & Fink, CS (2003) β-Sitosterol, a plant sterol, induces apoptosis and activates key caspases in MDA-MB-231 human breast cancer cells. Oncol Rep 10, 497500.Google ScholarPubMed
957Larner, J (2002) d-Chiro-inositol – its functional role in insulin action and its deficit in insulin resistance. Int J Exp Diabetes Res 3, 4760.CrossRefGoogle ScholarPubMed
958Asplin, I, Galasko, G & Larner, J (1993) Chiro-inositol deficiency and insulin resistance: a comparison of the chiro-inositol- and the myo-inositol-containing insulin mediators isolated from urine, hemodialysate, and muscle of control and type II diabetic subjects. Proc Natl Acad Sci U S A 90, 59245928.CrossRefGoogle ScholarPubMed
959Brautigan, DL, Brown, M, Grindrod, S, et al. . (2005) Allosteric activation of protein phosphatase 2C by d-chiro-inositol-galactosamine, a putative mediator mimetic of insulin action. Biochemistry 44, 1106711073.CrossRefGoogle ScholarPubMed
960Kawa, JM, Przybylski, R & Taylor, CG (2003) Urinary chiro-inositol and myo-inositol excretion is elevated in the diabetic db/db mouse and streptozotocin diabetic rat. Exp Biol Med 228, 907914.CrossRefGoogle ScholarPubMed
961Sun, T-H, Heimark, DB, Nguygen, T, et al. . (2002) Both myo-inositol to chiro-inositol epimerase activities and chiro-inositol to myo-inositol ratios are decreased in tissues of GK type 2 diabetic rats compared to Wistar controls. Biochem Biophys Res Commun 293, 10921098.CrossRefGoogle ScholarPubMed
962Cogram, P, Tesh, S, Tesh, J, et al. . (2002) d-Chiro-inositol is more effective than myo-inositol in preventing folate-resistant mouse neural tube defects. Hum Reprod 17, 24512458.CrossRefGoogle ScholarPubMed
963Holub, BJ (1986) Metabolism and function of myo-inositol and inositol phospholipids. Annu Rev Nutr 6, 563597.CrossRefGoogle ScholarPubMed
964Katayama, T (1997) Effects of dietary myo-inositol or phytic acid on hepatic concentrations of lipids and hepatic activities of lipogenic enzymes in rats fed on corn starch or sucrose. Nutr Res 17, 721728.CrossRefGoogle Scholar
965Novak, JE, Scott Turner, R, Agranoff, BW, et al. . (1999) Differentiated human NT2-N neurons possess a high intracellular content of myo-inositol. J Neurochem 72, 14311440.CrossRefGoogle ScholarPubMed
966Fux, M, Levine, J, Aviv, A, et al. . (1996) Inositol treatment of obsessive-compulsive disorder. Am J Psychiatry 153, 12191221.Google ScholarPubMed
967Palatnik, A, Frolov, K, Fux, M, et al. . (2001) Double-blind, controlled, crossover trial of inositol versus fluvoxamine for the treatment of panic disorder. J Clin Psychopharmacol 21, 335339.CrossRefGoogle ScholarPubMed
968Silver, SM, Schroeder, BM, Sterns, RH, et al. . (2006) Myoinositol administration improves survival and reduces myelinolysis after rapid correction of chronic hyponatremia in rats. J Neuropathol Exp Neurol 65, 3744.CrossRefGoogle ScholarPubMed
969Nakanishi, T, Turner, RJ & Burg, MB (1989) Osmoregulatory changes in myo-inositol transport by renal cells. Proc Natl Acad Sci U S A 86, 60026006.CrossRefGoogle ScholarPubMed
970Greene, DA & Lattimer, SA (1983) Impaired rat sciatic nerve sodium potassium adenosine triphosphatase in acute streptozocin diabetes and its correction by dietary myoinositol supplementation. J Clin Invest 72, 10581063.CrossRefGoogle Scholar
971Castano, G, Mas, R, Fernandez, JC, et al. . (2001) Effects of policosanol in older patients with type II hypercholesterolemia and high coronary risk. J Gerontol A Biol Sci Med Sci 56, M186M193.CrossRefGoogle ScholarPubMed
972Castaño, G, Tula, L, Canetti, M, et al. . (1996) Effects of policosanol in hypertensive patients with type II hypercholesterolemia. Curr Ther Res 57, 691699.CrossRefGoogle Scholar
973Menendez, R, Fernandez, SI, Del Rio, A, et al. . (1994) Policosanol inhibits cholesterol biosynthesis and enhances low density lipoprotein processing in cultured human fibroblasts. Biol Res 27, 199203.Google ScholarPubMed
974McCarty, MF (2002) Policosanol safely down-regulates HMG-CoA reductase – potential as a component of the Esselstyn regimen. Med Hypotheses 59, 268279.CrossRefGoogle ScholarPubMed
975Fraga, V, Menendez, R, Amor, AM, et al. . (1997) Effect of policosanol on in vitro and in vivo rat liver microsomal lipid peroxidation. Arch Med Res 28, 355360.Google ScholarPubMed
976Arruzazabala, ML, Molina, V, Mas, R, et al. . (2002) Antiplatelet effects of policosanol (20 and 40 mg/day) in healthy volunteers and dyslipidaemic patients. Clin Exp Pharmacol Physiol 29, 891897.CrossRefGoogle ScholarPubMed
977Carbajal, D, Arruzazabala, ML, Valdés, S, et al. . (1998) Effect of policosanol on platelet aggregation and serum levels of arachidonic acid metabolites in healthy volunteers. Prostaglandins Leukot Essent Fatty Acids 58, 6164.CrossRefGoogle ScholarPubMed
978Carbajal, D, Molina, V, Valdes, S, et al. . (1995) Antiulcer activity of higher primary alcohols of beeswax. J Pharm Pharmacol 47, 731733.CrossRefGoogle ScholarPubMed
979Saint-John, M & McNaughton, L (1986) Octacosanol ingestion and its effects on metabolic responses to submaximal cycle ergometry, reaction time and chest and grip strength. Int Clin Nutr Rev 6, 8187.Google Scholar
980Noa, M, Más, R & Mesa, R (2001) A comparative study of policosanol vs lovastatin on intimal thickening in rabbit cuffed carotid artery. Pharmacol Res 43, 3137.CrossRefGoogle ScholarPubMed
981Ferrari, C (2004) Functional foods, herbs and nutraceuticals: towards biochemical mechanisms of healthy aging. Biogerontology 5, 275289.CrossRefGoogle ScholarPubMed
982Reiter, R (1995) Oxidative processes and antioxidative defense mechanisms in the aging brain. FASEB J 9, 526533.CrossRefGoogle ScholarPubMed
983Reiter, R, Tang, L, Garcia, JJ, et al. . (1997) Pharmacological actions of melatonin in oxygen radical pathophysiology. Life Sci 60, 22552271.CrossRefGoogle ScholarPubMed
984Sainz, RM, Mayo, JC, Reiter, RJ, et al. . (1999) Melatonin regulates glucocorticoid receptor: an answer to its antiapoptotic action in thymus. FASEB J 13, 15471556.CrossRefGoogle ScholarPubMed
985Hardeland, R, Reiter, RJ, Poeggeler, B, et al. . (1993) The significance of the metabolism of the neurohormone melatonin: antioxidative protection and formation of bioactive substances. Neurosci Biobehav Rev 17, 347357.CrossRefGoogle Scholar
986Barlow-Walden, LR, Reiter, RJ, Abe, M, et al. . (1995) Melatonin stimulates brain glutathione peroxidase activity. Neurochem Int 26, 497502.CrossRefGoogle ScholarPubMed
987Kotler, M, Rodríguez, C, Sáinz, RM, et al. . (1998) Melatonin increases gene expression for antioxidant enzymes in rat brain cortex. J Pineal Res 24, 8389.CrossRefGoogle ScholarPubMed
988Zhdanova, Wurtm IV, an, RJ, Morabito, C, et al. . (1996) Effects of low oral doses of melatonin, given 2–4 hours before habitual bedtime, on sleep in normal young humans. Sleep 19, 423431.CrossRefGoogle ScholarPubMed
989Reiter, RJ, Melchiorri, D, Sewerynek, E, et al. . (1995) A review of the evidence supporting melatonin's role as an antioxidant. J Pineal Res 18, 111.CrossRefGoogle ScholarPubMed
990Anisimov, VN, Zavarzina, NY, Zabezhinski, MA, et al. . (2001) Melatonin increases both life span and tumor incidence in female CBA mice. J Gerontol A Biol Sci Med Sci 56, B311B323.CrossRefGoogle ScholarPubMed
991Srinivasan, V, Spence, DW, Pandi-Perumal, SR, et al. . (2008) Therapeutic actions of melatonin in cancer: possible mechanisms. Integr Cancer Ther 7, 189203.CrossRefGoogle ScholarPubMed
992Hearse, DJ & Weber, WW (1973) Multiple N-acetyltransferases and drug metabolism. Tissue distribution, characterization and significance of mammalian N-acetyltransferase. Biochem J 132, 519526.CrossRefGoogle ScholarPubMed
993Lindsay, RM, McLaren, AM & Baty, JD (1988) Effect of hemolysis on the acetylation of p-aminobenzoic acid by human whole-blood and washed erythrocytes in vitro. Biochem Soc Trans 16, 1024.CrossRefGoogle Scholar
994Butcher, NJ, Ilett, KF & Minchin, RF (2000) Substrate-dependent regulation of human arylamine N-acetyltransferase-1 in cultured cells. Mol Pharmacol 57, 468473.CrossRefGoogle ScholarPubMed
995Failey, RB Jr & Childress, RH (1962) The effect of para-aminobenzoic acid on the serum cholesterol level in man. Am J Clin Nutr 10, 158162.CrossRefGoogle ScholarPubMed
996Barbieri, B, Stain-Malmgren, R & Papadogiannakis, N (1999) p-Aminobenzoic acid and its metabolite p-acetamidobenzoic acid inhibit agonist-induced aggregation and arachidonic acid-induced Ca2+ (i) transients in human platelets. Thromb Res 95, 235243.CrossRefGoogle ScholarPubMed
997Russell, K, Craig, ID, Rawlings, JM, et al. . (2001) The use of p-aminobenzoic acid and chromic oxide to confirm complete excreta collection in a carnivore, Felis silvestris catus. Comp Biochem Physiol C Toxicol Pharmacol 130, 339345.CrossRefGoogle Scholar
998Berger, A, Rein, D, Schafer, A, et al. . (2005) Similar cholesterol-lowering properties of rice bran oil, with varied γ-oryzanol, in mildly hypercholesterolemic men. Eur J Nutr 44, 163173.CrossRefGoogle ScholarPubMed
999Ishihara, M, Ito, Y, Nakakita, T, et al. . (1982) Clinical effect of γ-oryzanol on climacteric disturbance - on serum lipid peroxides (article in Japanese). Nippon Sanka Fujinka Gakkai Zasshi 34, 243251.Google Scholar
Ishihara, M (1984) Effect of γ-oryzanol on serum lipid peroxide level and clinical symptoms of patients with climacteric disturbances. Asia Oceania J Obstet Gynaecol 10, 317323.CrossRefGoogle ScholarPubMed
Itaya, K, Kiyonaga, J & Ishikawa, M (1976) Studies of γ-oryzanol (1). Effects on stress-induced ulcer. Nippon Yakurigaku Zasshi 72, 10011011.CrossRefGoogle ScholarPubMed
Parrado, J, Miramontes, E, Jover, M, et al. . (2003) Prevention of brain protein and lipid oxidation elicited by a water-soluble oryzanol enzymatic extract derived from rice bran. Eur J Nutr 42, 307314.CrossRefGoogle ScholarPubMed
Lee, CK, Pugh, TD, Klopp, RG, et al. . (2004) The impact of α-lipoic acid, coenzyme Q10, and caloric restriction on life span and gene expression patterns in mice. Free Radic Biol Med 36, 10431057.CrossRefGoogle ScholarPubMed
Eason, RC, Archer, HE, Akhtar, S, et al. . (2002) Lipoic acid increases glucose uptake by skeletal muscles of obese-diabetic ob/ob mice. Diabetes Obes Metab 4, 2935.CrossRefGoogle ScholarPubMed
Khanna, S, Roy, S, Packer, L, et al. . (1999) Cytokine-induced glucose uptake in skeletal muscle: redox regulation and the role of α-lipoic acid. Am J Physiol Regul Integr Comp Physiol 276, R1327R1333.CrossRefGoogle ScholarPubMed
Seetharamaiah, GS, Krishnakantha, TP & Chandrasekhara, N (1990) Influence of oryzanol on platelet aggregation in rats. J Nutr Sci Vitaminol 36, 291297.CrossRefGoogle ScholarPubMed
Cai, X-J, Bi, X-P, Zhao, Z, et al. . (2006) The effects of antidepressant treatment on efficacy of antihypertensive therapy in elderly hypertension. Zhonghua Nei Ke Za Zhi 45, 639641.Google ScholarPubMed
Ichimaru, Y, Moriyama, M, Ichimaru, M, et al. . (1984) Effects of γ-oryzanol on gastric lesions and small intestinal propulsive activity in mice. Nippon Yakurigaku Zasshi 84, 537542.CrossRefGoogle ScholarPubMed
Jabeen, B, Badaruddin, M, Ali, R, et al. . (2007) Attenuation of restraint-induced behavioral deficits and serotonergic responses by stabilized rice bran in rats. Nutr Neurosci 10, 1116.CrossRefGoogle ScholarPubMed
Nie, L, Wise, M, Peterson, D, et al. . (2006) Mechanism by which avenanthramide-c, a polyphenol of oats, blocks cell cycle progression in vascular smooth muscle cells. Free Radic Biol Med 41, 702708.CrossRefGoogle ScholarPubMed
Nie, L, Wise, ML, Peterson, DM, et al. . (2006) Avenanthramide, a polyphenol from oats, inhibits vascular smooth muscle cell proliferation and enhances nitric oxide production. Atherosclerosis 186, 260266.CrossRefGoogle ScholarPubMed
Francis, G, Kerem, Z, Makkar, HPS, et al. . (2007) The biological action of saponins in animal systems: a review. Br J Nutr 88, 587605.CrossRefGoogle Scholar
Mimaki, Y, Yokosuka, A, Kuroda, M, et al. . (2001) Cytotoxic activities and structure–cytotoxic relationships of steroidal saponins. Biol Pharm Bull 24, 12861289.CrossRefGoogle ScholarPubMed
Potter, SM, Jimenez-Flores, R, Pollack, J, et al. . (1993) Protein–saponin interaction and its influence on blood lipids. J Agric Food Chem 41, 12871291.CrossRefGoogle Scholar
Han, L-K, Xu, B-J, Kimura, Y, et al. . (2000) Platycodi radix affects lipid metabolism in mice with high fat diet-induced obesity. J Nutr 130, 27602764.CrossRefGoogle ScholarPubMed
Kim, YH, Park, KH & Rho, HM (1996) Transcriptional activation of the Cu,Zn-superoxide dismutase gene through the AP2 site by ginsenoside Rb2 extracted from a medicinal plant, Panax ginseng. J Biol Chem 271, 2453924543.CrossRefGoogle ScholarPubMed
Yoshiki, Y & Okubo, K (1995) Active oxygen scavenging activity of DDMP (2,3-dihydro-2,5-dihydroxy-6-methyl-4H-pyran-4-one) saponin in soybean seed. Biosci Biotechnol Biochem 59, 15561557.CrossRefGoogle Scholar
Arai, S, Osawa, T, Ohigashi, H, et al. . (2001) A mainstay of functional food science in Japan – history, present status, and future outlook. Biosci Biotechnol Biochem 65, 13.CrossRefGoogle ScholarPubMed
Jie, L (1995) Pharmacology of oleanolic acid and ursolic acid. J Ethnopharmacol 49, 5768.CrossRefGoogle Scholar
Matsuda, H, Li, Y, Murakami, T, et al. . (1999) Structure-related inhibitory activity of oleanolic acid glycosides on gastric emptying in mice. Bioorg Med Chem 7, 323327.CrossRefGoogle ScholarPubMed
Sindambiwe, JB, Calomme, M, Geerts, S, et al. . (1998) Evaluation of biological activities of triterpenoid saponins from Maesa lanceolata. J Nat Prod 61, 585590.CrossRefGoogle ScholarPubMed
Xi, MM, Hai, CX, Tang, HF, et al. . (2008) Antioxidant and antiglycation properties of total saponins extracted from traditional Chinese medicine used to treat diabetes mellitus. Phytother Res 22, 228237.CrossRefGoogle ScholarPubMed
Cai, J, Liu, M, Wang, Z, et al. . (2002) Apoptosis induced by dioscin in HeLa cells. Biol Pharm Bull 25, 193196.CrossRefGoogle ScholarPubMed
Hanausek, M, Ganesh, P, Walaszek, Z, et al. . (2001) Avicins, a family of triterpenoid saponins from Acacia victoriae (Bentham), suppress H-ras mutations and aneuploidy in a murine skin carcinogenesis model. Proc Natl Acad Sci U S A 98, 1155111556.CrossRefGoogle Scholar
Liu, WK, Xu, SX & Che, CT (2000) Anti-proliferative effect of ginseng saponins on human prostate cancer cell line. Life Sci 67, 12971306.CrossRefGoogle ScholarPubMed
Mujoo, K, Haridas, V, Hoffmann, JJ, et al. . (2001) Triterpenoid saponins from Acacia victoriae (Bentham) decrease tumor cell proliferation and induce apoptosis. Cancer Res 61, 54865490.Google ScholarPubMed
Podolak, I, Elas, M & Cieszka, K (1998) In vitro antifungal and cytotoxic activity of triterpene saponosides and quinoid pigments from Lysimachia vulgaris L. Phytother Res 12, S70S73.3.0.CO;2-9>CrossRefGoogle Scholar
Zhang, YW, Dou, DQ, Zhang, L, et al. . (2001) Effects of ginsenosides from Panax ginseng on cell-to-cell communication function mediated by gap junctions. Planta Med 67, 417422.CrossRefGoogle ScholarPubMed
Peng, JP, Chen, H, Qiao, YQ, et al. . (1996) Two new steroidal saponins from Allium sativum and their inhibitory effects on blood coagulability (article in Chinese). Yao Xue Xue Bao 31, 607612.Google Scholar
Figure 0

Fig. 1 The three wheat fraction (bran, germ and endosperm) with their main bioactive compounds as obtained from Tables 1 and 2. Whole-grain wheat has an heterogeneous struture with bioactive compounds unevenly distributed within its different parts (with permission from Surget & Barron for original image(476), and adapted from the brochure ‘Progress in HEALTHGRAIN 2008’, HealthGrain Project, European Community's Sixth Framework Programme, FOOD-CT-2005-514008, 2005–2010). * No published data on the precise locations of policosanol and phytosterols in a specific layer of the wheat bran fraction.

Figure 1

Table 1 Average content of the major bioactive compounds in whole-grain wheat and wheat bran and germ fractions (%)*

Figure 2

Table 2 Content, apparent absorption and fermentability of bioactive compounds and fibre from whole-grain wheat and wheat bran and germ fractions*

Figure 3

Table 3 Main physiological functions, potential protective mechanisms and health benefits of isolated bioactive compounds found in whole-grain wheat, rice and oat*

Figure 4

Table 4 Whole-grain cereal bioactive compounds potentially involved in the prevention of major health outcomes and in antioxidant protection*

Figure 5

Fig. 2 Current accepted mechanisms for how whole grain protects against major chronic diseases (modified with permission from Professor I. Björck (University of Lund, Sweden); see the HealthGrain brochure for original diagram: ‘Progress in HEALTHGRAIN 2008’, a project from the European Community's Sixth Framework Programme, FOOD-CT-2005-514008, 2005–2010; see Poutanen et al.(478) for more details about the Project). GI, glycaemic index; II, insulinaemic index.

Figure 6

Fig. 3 Linear discriminant (LD) analysis score plot of the 1H NMR urinary spectra highlighting the separation before, between and after the diet change (days 14–15) and between the urine sampling times (postprandial (PP) and post-absorptive (PA)). (- - - -), Refined flour followed by whole-grain flour consumption (RF-WGF) group; (—), whole-grain flour followed by refined flour consumption (WGF-RF) group. Each polygon represents the limits of the metabolic profile obtained for the ten rats of a given group at a given day and urine sampling time. Urine samples were collected from days 13 to 28 (for details, see Fardet et al.(230)).

Figure 7

Fig. 4 Current and new proposed physiological mechanisms involved in protection by whole-grain cereals (adapted from Table 3). The dotted thin arrows () indicate the link between whole-grain bioactive compounds and protective physiological mechanisms, while the plain arrows () indicate the relationship between physiological mechanisms and health outcomes.

Figure 8

Fig. 5 Ways for improving cereal product nutritional quality. RS, resistant starch.