Abstract
Various secondary plant metabolites or phytochemicals, including polyphenols and carotenoids, have been associated with a variety of health benefits, such as reduced incidence of type 2 diabetes, cardiovascular diseases, and several types of cancer, most likely due to their involvement in ameliorating inflammation and oxidative stress. However, discrepancies exist between their putative effects when comparing observational and intervention studies, especially when using pure compounds. These discrepancies may in part be explained by differences in intake levels and their bioavailability. Prior to exerting their bioactivity, these compounds must be made bioavailable, and considerable differences may arise due to their matrix release, changes during digestion, uptake, metabolism, and biodistribution, even before considering dose‐ and host‐related factors. Though many insights have been gained on factors affecting secondary plant metabolite bioavailability, many gaps still exist in our knowledge. In this position paper, we highlight several major gaps in our understanding of phytochemical bioavailability, including effects of food processing, changes during digestion, involvement of cellular transporters in influx/efflux through the gastrointestinal epithelium, changes during colonic fermentation, and their phase I and phase II metabolism following absorption.
Keywords: Biotransformation, Food processing, Microbiota, Mixed diet, Transporters
Abbreviations
- CVD
cardiovascular diseases
- MCTs
monocarboxylic acid transporters
- MRP
multidrug resistance protein
- NEPP
nonextractable polyphenols
- NPC1L1
Niemann‐Pick C1‐Like 1
- SGLT‐1
sodium‐glucose linked transporter 1
- T2D
type 2 diabetes
- TOF
time‐of‐flight
1. Introduction
Phytochemicals comprise a diverse group of secondary plant compounds, including polyphenols, carotenoids, triterpenes, phytosterols, glucosinolates, and many more. These compounds have aroused increasing interest in the area of nutrition and food science, due to their potential health benefits. Several epidemiological studies have suggested that their consumption or tissue concentration is associated with reduced risk of developing certain chronic diseases, namely, type 2 diabetes (T2D), cardiovascular diseases (CVD), and some types of cancer. For example, meta‐analyses have suggested that the consumption of flavonoids from fruits and vegetables improved flow‐mediated dilation, a marker of atherosclerosis 1, and suggested positive health effects of consuming carotenoids with respect to type 2 diabetes 2.
However, though a few examples of intervention studies with isolated compounds, e.g. resveratrol, or curcumin 3, 4, producing positive health effects exist, micronutrients and phytochemicals may not act efficiently in isolation but together with many other compounds in the food matrix, leading to synergistic effects 5. It is thus important to determine efficacy, safety and underlying mechanisms of these compounds, especially when taken in pharmaceutical doses/in combination with other drugs. For example, Prasain et al. 6 reviewed risks and benefits of dietary versus supplemented/isolated flavonoids, stating that flavonoids can be detrimental in some settings and therefore are not universally safe. Adverse effects were also found following the ingestion of beta‐carotene supplements administrated to CVD patients 7, 8, and vitamin C and E as well as beta‐carotene supplementation failed to show health benefits with respect to CVD 9. Supplementing individual flavonoids and isoflavonoids has also been met with criticism, e.g. due to endocrine disrupting properties for higher doses 10. The reasons for this discrepancy are not fully understood, but could involve “missing” synergistic effects with the food matrix, altered digestion and release of the compounds or changed degradation patterns, i.e. modified bioavailability (the fraction of a compound that is absorbed and can be used for physiological functions and/or storage). Indeed, many reviews on bioavailability and bioactivity of phytochemicals are available 11, 12, 13. Phytochemicals may have multiple functions in the human body, and depending on their dose they can exert both beneficial and deleterious effects 14.
This position paper attempts to identify the aspects of digestion, release, absorption and metabolism of food phytochemicals which are only poorly understood (Table 1) 15. Even prior to ingestion processing factors (food texture, e.g. heat, temperature, or pressure application 16) can impinge on the bioaccessibility (fraction of a compound that is released from the matrix and potentially available for further uptake and absorption) of bioactives 17. Similarly, the influence of consuming a mixed diet, i.e. “real” complex meals, on bioaccessibility and absorption is poorly comprehended, and biochemical and physico‐chemical aspects such as viscosity and surface tension surely play a role 18. Following ingestion, enzyme concentrations, pH, and time of digestion all play a role and influence release kinetics and degradation patterns 19, 20. A factor that is also poorly understood is the nature and bioactivity of metabolites formed during digestion, as following their fate is challenging. However, the bioavailability of some compounds and their metabolites may be higher than previously assumed, as shown, e.g. in isotope studies with anthocyanins 21. Some lipophilic compounds, such as carotenoids or triterpenes, require micellarization, as may certain polyphenol aglycones 22. Following release, pathways of absorption, i.e. active versus passive or paracellular routes remain largely marginally understood. The same is true for influx and efflux transporters in the epithelium. For example, several polyphenols were shown to be considerably better absorbed in the presence of additional polyphenols, blocking efflux transporters (to the gut), which normally reduce the intracellular concentration of such “xenobiotics” 19. On the other hand, transporters to the basolateral side are hardly understood. Finally, many native phytochemicals undergo considerable metabolism in the human body, e.g. deglycosylation and glucuronidation/sulfation for polyphenols in the gut, cleavage by beta‐carotene oxygenase 1 and beta‐carotene dioxygenase 2 for carotenoids 23, and many additional reactions may occur in other tissues such as in the liver or in the colon, where bacterial fermentation significantly alters the structure and profile and thus the potential bioactivity of many plant compounds that are not absorbed in the small intestine 24, 25, 26.
Table 1.
Summary of major gaps of knowledge around phytochemical bioavailability
| Stage | Knowledge gap | Examples | Reference examples |
|---|---|---|---|
| Food matrix | Physical state/compartmentalization of phytochemicals | Crystallinity of carotenoids in chromoplasts versus chloroplasts, polyphenols bound to cell wall (NEPP) | 31, 107 |
| Food preparation | Effect of cutting, mashing, grinding, peeling, trimming |
|
42, 43 |
| Food processing | Effect of cooking (heat, temperature, time, blanching) | Enhanced carotenoid content with mild conditions but decreased with severe treatments | 16, 52, 53 |
| Refining processes |
|
40, 41, 42 | |
| Nonthermal processing (HPP and PEF) | Both positive (polyphenols) and contradictory effects (carotenoids) | 67, 69 | |
| Food mixtures | Effect of mixed or real meals | Enhanced availability of polyphenols with presence of sugars, ascorbic acid, fat. Stabilizing effects of polyphenols on other phytochemicals? | 77, 81, 193 |
| Gastric digestion | Effect of pH, depolymerization of large polyphenols, binding effects | Hydrophobic interactions, hydrogen bonding of polyphenols to proteins | 19 |
| Small intestine |
|
|
22, 83 |
|
Efflux transporters such as P‐gp, BCRP blocked by several polyphenols, e.g. flavonoids | 124 | |
|
Effect of polyphenols on efflux transporters such as MRP3 | 194, 195, 196 | |
|
SRB‐1, CD36, NPC1L1, ABCG5/G8: affecting carotenoid uptake – influence of polyphenols | 19, 197, 198 | |
| Colon | Phase I/II interactions | Piperine increased curcumin absorption | 146 |
|
Metabolite formation, absorption of cleavage products? | 102, 172, 190 | |
| Metabolites and phase I/II products | Metabolite information limited by lack of standards | 160, 161 | |
| Tissues | Biotransformation, phase I and phase II metabolism | Need to develop/increased availability of more physiological cell models (liver stem cells, co‐culture cell models) to study metabolites | 125, 152, 153, 154, 155 |
| Interaction with transporters in certain tissues (blood–brain barrier, placenta, testis), or excretory organs (liver, kidney) | Role of MRPs, MCTs, CD36 | 140, 184, 185 |
ABCG5/G8 = ATP‐binding cassette sub‐family G, member 5/8; BCRP = Breast cancer resistance protein; CD36 = cluster of differentiation; HPP = high pressure processing; MCTs = monocarboxylic acid transporters; MRP = multidrug resistance protein; NEPP = non‐extractable polyphenols; NPC1L1 = Niemann‐Pick C1‐Like 1; PEF = pulsed electric fields; P‐gp = P‐glycoprotein; SRB‐1 = scavenger receptor class B member 1.
In this paper, we aim to highlight gaps that have received either limited attention or which are far from being understood, but which may play pivotal roles in the bioavailability of phytochemicals (Fig. 1). To allow for better focus, this review will concentrate on polyphenols and their metabolites as the most abundant water‐soluble compounds, and carotenoids as the most abundant lipophilic secondary plant compounds. For a more comprehensive overview on bioavailability aspects of specific compound classes, the reader is referred to other articles 22, 27, 28.
Figure 1.

Overview of important stages during the digestion of food items and open questions related to phytochemical bioavailability.
2. Food processing and matrix effects
Bioaccessibility, and further bioavailability, of phytochemicals starts with their content and composition of the raw plant material and how it is processed. The food matrix and structure and type of processing can have both positive and negative effects 29. Plant matrix disruption and cell cluster disintegration due to applied processing steps are the main prerequisites for phytochemical liberation and bioaccessibility, but they may also lead to oxidation and/or degradation, thus potentially counterbalancing each other.
To understand the release of bioactive compounds during digestion, it is also important to know where they are located in the tissue. Carotenoids are found either in the chloroplast membrane or in chromoplasts 30. The different physical forms of carotenoids in plant chromoplasts (crystals in tomato, lipid‐dissolved in papaya and liquid‐crystalline in mango) have major impacts on their liberation efficiency from the food matrix 31. Polyphenols are generally present in vacuoles and the apoplast of plant cells, in conjugated form with mono‐ and polysaccharides, and proteins. Although studies have focused mainly on the cellular localization of carotenoids, polyphenols, and more rarely on glucosinolates, more research on their localization in different foods is needed. Little is known about other phytochemicals.
2.1. Effects of physical and mechanical treatments
Food preparation (such as grating, chopping, cutting, slicing, trimming, mashing, and juicing) may have great influence on the bioaccessibility of nutrients and bioactive compounds 30, 32. The recovery of phytochemicals from intact, minimally processed fruits and vegetables can be different from juices as components must be released from the tissue matrix before being subject to degradation reactions, whereas those in the juices are subject to degradation from the offset. However, carotenoid bioacessibility was greater from juiced than from raw or cooked puréed tissue, as chopping and homogenization disrupted the plant matrix 32. The levels of carotenoids in sea buckthorn berries were lower if the berries were extracted in water or juiced than after direct acetone extraction. This appears to be due to the complexation of carotenoids with pectins 33, and similar interactions may also occur between pectins and polyphenols, such as anthocyanins, during digestion 34.
Other phytochemicals are subject to endogenous enzymatic digestion. Chopping or crushing raw garlic releases alliinase which produces allicin, which breaks down to diallyl sulfides and then other metabolites, which may influence chronic diseases and certain cancers 35, 36. Similarly, the glucosinolate glucobrassicin, which is found in cruciferous vegetables, is hydrolyzed when plant cells are damaged 37. On the other hand, flavonoids seem relatively stable to mechanical processing 38. Peeling and trimming steps may also influence phytochemical content. Trimming green leafy vegetables is known to be a main factor influencing carotenoid concentrations, since they are nearly exclusively present in leaves 39. Also industrial refining processes, e.g. as applied to vegetable oils and cereals, may lead to high losses in phytochemicals. Refining of palm oil decreased carotenoids by 99%, and polyphenols by 23% 40, 41.
Milling of cereals and grains has a great influence on phenolic acids 42. It is well known that milling of the whole grain results in higher amounts of phenolics than conventional milling resulting in white flour 43. Many studies have investigated the effects of refining on phytochemicals in edible oils and cereals but similar studies on vegetables and fruits (except juices) are largely absent.
In conclusion, although there are many studies reporting effects of food preparation on the bioaccessibility of polyphenols and carotenoids, understanding the mechanisms of chemical or enzymatic actions still requires extensive work. In addition, as novel methods are developed, further studies on how these processes impact bioaccessibility and bioavailability are warranted.
2.2. Effects of thermal and nonthermal treatments
Studies of thermal treatment on phytochemicals in foods have mainly focused on polyphenols, carotenoids and glucosinolates. Many reports indicate that thermal processing increases levels of free polyphenols 44, 45, 46, 47, possibly due to the release of bound phenolics due to the breakdown of cell constituents, perhaps through releasing nonextractable polyphenols (NEPP). However, also polymerization/oxidation reactions may be responsible for apparent increases 44, 45, and the nature of these released polyphenols requires investigation. Similarly, carotenoid availability often increases after heat treatment 48, 49, 50, 51, probably due to enhanced extractability following maceration of cells. However, severe heat treatment such as baking/sterilization of tomatoes 52, 53 or boiling of chili peppers 54 caused significant losses. Losses of glucosinolates in Brassica vegetables depended on processing time, type of vegetable, degree of cellular damage, and inactivation of myrosinase 55, 56, 57, 58, 59, 60. Overall, further research is required to better explain how thermal treatments impact bioaccessibility and bioavailability.
Nonthermal processing technologies have been revealed as useful tools to extend shelf‐life and to preserve nutritional and functional characteristics of fruit and vegetable products 61. However, there are scarce data on the effect of these emerging technologies on bioaccessibility and bioavailability of bioactive compounds. Considering bioaccessibility, studies have mainly focused on the effect of high pressure processing on carotenoids, with contradictory results. Some show improvements 16, 62, 63, 64, 65, 66, possibly related to the disintegration of cell clusters and disruption of cells containing carotenoids. Beneficial effects of a subsequent thermal treatment may weaken the physical barriers that enclose carotenoids and accelerate pectin degradation by β‐elimination, resulting in cell wall softening. Decreased carotenoid bioaccessibility has been noted in some vegetables and fruits and may be due to firmer texture and/or trapping of carotenoids within fiber networks 62, 63. Thus, processing variables (e.g. particle size, temperature, pressure applied) need to be carefully controlled, and there is a need for harmonization and validation (by comparison with in vivo assays) of the in vitro digestion models used.
Regarding human bioavailability, high pressure processing increased absorption of orange juice flavanones over conventionally pasteurized orange juice 67. No data are available for carotenoids. To our knowledge, the effect of pulsed electric fields has been investigated only on vitamin C bioavailability in humans from orange juice 68 and Mediterranean vegetable soup 69. No studies addressing the effect of pulsed electric field on carotenoid and polyphenol bioaccessibility and bioavailability are available. Other nonthermal processing technologies such as ultrasound 70, food irradiation, light pulses or oscillating magnetic fields, have received little or no attention in terms of their effect on bioaccessibility and bioavailability of phytochemicals. Thus, more studies in this field may be required especially as more novel processing techniques are being developed.
3. Mixtures of authentic food matrices
There has been comparatively little effort into assessing the effects of characteristic combinations of foodstuffs or realistic meals on the bioaccessability of phytochemicals. However, studies have shown that polyphenols from foods in combinations can have very different bioaccessibilities. Studies with raspberry juice showed that the addition of ice‐cream markedly reduced the recovery of total anthocyanins 71, whereas a wheat‐based breakfast cereal did not influence recovery. Co‐digestion with blueberries ± oat meal and/or milk revealed that milk reduced the recovery of total anthocyanins and total phenols 72 and co‐digestion of nuts and dried fruits yielded lower levels of total available phenols after simulated digestion than nuts or fruits alone.
The addition of bovine, soy and rice milks, ascorbic acid or citrus juices increased the bioaccessibility of galloylated green tea catechins (EGC, EGCG, EC, and ECG), by stabilization and protection from degradation at alkaline pH 73. The simultaneous presence of sucrose and ascorbic acid in green tea increased EGC and EGCG bioaccessibility, uptake in Caco‐2 cells and bioavailability in rats 74. Although the addition of skimmed milk reduced recovery of green tea catechins after in vitro digestion, the uptake of catechins by Caco‐2 cells was increased 75. This suggests that catechins bound to the milk proteins after in vitro digestion were available for uptake by the Caco‐2 cells. This may explain the lack of difference in serum/plasma bioavailability of tea catechins in studies where subjects were given tea with or without milk 76. Similarly, chocolate containing higher sucrose levels increased plasma concentrations of metabolites derived from catechins compared to dark and milk chocolate in rats 77. In humans, sucrose but also solid/beverage format influenced various aspects of the bioavailability of flavan‐3‐ols from commercial cocoa based products 78. Dietary fats can increase polyphenol bioavailability in humans by increasing absorption, possibly by enhancing micellarization in the small intestine 79, 80, as noted for carotenoids. For example, higher fat content increased the stability of cocoa procyanidins during in vitro digestion 81. This is particularly relevant as screening of different formulations (or different plant varieties) for the bioaccessibility of specific phytochemical components has become more common and it is possible that variations in other macronutrients influence the outcome. Using experience gained in pharmaceutical applications, strategies may also be designed to improve polyphenol bioavailability by co‐administering phenolics with compounds which modulate gut and/or liver metabolizing enzymes 80, 82, 83.
There is definitely more scope for the study of food mixtures that are more realistic of real meal choices. Complex models developed by the pharmaceutical industry to assess the digestion behavior of different drug formulations 84, 85 could be applied to estimate digestion of foods. Despite the increasing complexity achieved by in vitro models, they remain simple compared to in vivo models. Typical limitations of in vitro models include: absence of host response factors, poor stimulation of complex mechanical forces and gastric emptying, absence of microbial flora, low level of integration into an overall digestive process, general adherence to healthy/average conditions, and limited correlation to in vivo situations 86. Nevertheless, some examples have indicated a reasonable correlation between bioaccessibility studies and the situation in vivo. For lipophilic compounds such as α‐ and γ‐tocopherol, β‐carotene and lycopene 87, β‐cryptoxanthin 88 or phytosterols 89, high correlations have been found, indicating that estimating in vitro bioaccessibility (solubility/micellarization) can be indicative of the amount available for uptake in the GI tract in vivo. For polyphenols, comparisons to in vitro data have been allowed by studying ileostomists 15. In addition, combining in vitro digestion models and human intestinal cells (e.g. Caco‐2 cells with or without a layer of mucus‐producing cells such as HT‐29 MTX), carotenoid uptake 90, 91, 92 qualitatively and quantitatively correlated well with human data. Investigators using in vitro methods must consider how to adapt the digestion conditions according to the composition of the sample and/or to food components, seeking a balance between technical simplification and accuracy, considering the in vivo situation as reference.
However, the release of phytochemicals from different foods is complex and involves large numbers of variables 93, 94, 95. Furthermore, the transfer of phytochemicals from authentic food matrices will be even more complex due to a greater number of potentially rate‐determining processes. Digestion and absorption rates are limited by physical processes, operating within and between the different phases of the liquid and solid phases of the digesta 96. Using models of the human stomach 95, 97, distinct disintegration profiles and kinetics have been found for different food categories (e.g. meat products, nuts, fruits, baked and fried products, etc.). The digestion process in terms of secretion of gastric fluids and enzymatic degradation of macronutrients is well understood, but further detailed investigations are needed to understand lipid absorption and to predict responses in realistic meals. Further information on the release of bioactive compounds should provide a better understanding of how to combine foods and design meals to improve bioavailability and enhance potential bioactive effects.
Encapsulation may enhance the bio‐effectiveness of phytochemicals, and the choice of encapsulation agent can influence stability/recovery after digestion 98. For example, Haratifar et al. 99 found that casein micelles were effective encapsulation agents for EGCG. A novel approach involving biosorption of phenolics to Saccharomyces cerevisiae significantly improved bioaccessibility of total phenolics, presumably as the yeast provided a protective carrier against degradation at neutral pH 100.
The possibility that certain phytochemical classes could protect other phytochemicals is an interesting, but largely under‐researched area. The fact that the stability of individual phytochemical classes may be dependent on the overall phenolic composition of the sample has been raised in studies of berry polyphenol bioaccessibility 101, 102. This interdependence could be examined in studies where blends of juices are tested for bioaccessibility of a range of phytochemical components such as vitamin C, carotenoids, and polyphenols 103. The possibility that polyphenols may sacrificially protect carotenoids or vitamin C (or vice versa) could also be examined. For example, the effect of anthocyanins on carotenoid stability could be examined in comparative studies of purple tomatoes and related red tomatoes 104 or indeed in the recently genetically modified anthocyanin‐accumulating tomatoes over their non‐genetically modified counterparts 105. Combining foodstuffs, therefore, may provide another route to enhance the bioaccessibility of specific health beneficial components. Vice versa, it has been claimed that polyphenols may modulate the digestion of macronutrients (such as starches or fats) through inhibition of their digestive enzymes 106, which could possibly modulate their own stability in the gut. The possibility that the underestimated NEPP content of fruits/vegetables 107 could influence the activity of digestive enzymes or limit the access of certain phytonutrients to digestive processes could be examined in carefully designed studies where the effect of in vitro digestion on the phytochemical profile of whole fruit/vegetable purees was compared to extracts and cross‐compared to re‐combined samples containing NEPP plus extracts.
There has been a general shift away from measuring the levels of total phytochemical classes (e.g. total carotenoids and phenolics) to targeted analysis of compositional changes in specific components during digestion, as this is more relevant to the potential health benefits attributable to specific phytochemicals. However, greater value could be obtained by using an untargeted, metabolic profiling approach which could also identify potential breakdown products and other novel components.
4. Gastric phase and small intestine
The absorption of secondary plant components includes several phases (outlined below), and prominent gaps in our knowledge include factors influencing solubilization, micelle formation (of apolar compounds), diffusion to the unstirred water layer (including the influence of mucus), transporters involved in the uptake of phytochemicals, and factors affecting phase I/II metabolism and efflux pumps.
4.1. Release from the food matrix
Aspects impeding the release of phytochemicals include large particle size 108, high meal viscosity 18 reducing the transfer of lipophilic compounds to micelles and hindering interactions between lipase and oil droplets 109; or the presence of physically inaccessible forms such as NEPP 19, 110. As the majority of micronutrients and phytochemicals are presumably taken up in the small intestine (in their native form), and the epithelial “leakiness” decreases toward the colon 111, it is important that compounds are bioaccessible at this stage. Many polyphenols may not be detectable in the native matrix following chemical extraction, but may be released during digestion in the small intestine, such as those bound covalently or occluded by e.g. in accessible starch 112, though colonic fermentation may further result in the breakdown of NEPPs 113. Standardized methodologies which recognize and examine the potential contribution of NEPPs are required. It also has to be noted that polyphenols have the ability to reduce the activity of digestion enzymes (e.g. pepsin, lipase), thus high concentrations of polyphenols may reduce liberation of lipids and proteins, increasing the nondigestible bulk and in turn may result in increased amounts of polyphenols passed onto the colon 19.
4.2. Solubilization and micellarization
Solubility is not an issue for most polyphenols, but more lipophilic compounds such as carotenoids 114, triterpenes 115, and phytosterols 116 require emulsification/micellarization before uptake. It remains largely unknown to what extent digestion and food matrix influence micelle formation or size. Large micelles would compromise diffusion and subsequent release of apolar compounds. Micelle diameters of approximately 6–8 nm 117, 118 have been measured, but the relation of micelle size, shape, or constituents, and cellular uptake has never been studied in detail. Also, surprisingly little is known about the influence of dietary lipids and their digestion on polyphenol uptake 19, though there are indications that certain apolar polyphenols (e.g. curcumin, resveratrol, quercetin aglycones) are incorporated into micelles, and that lipid‐rich foods may enhance their bioavailability 79, 119, 120, 121.
A poorly understood factor in bioavailability is the role of brush border membrane enzymes, i.e. maltase, lactase‐phlorizin‐hydrolase, sucrase‐isomaltase, and peptidases 122. Lactase‐phlorizin‐hydrolase may play a crucial role in cleaving polyphenol glycosides, resulting in the uptake of free aglyones 123, 124. However, it is not certain whether cleavage occurs at this stage, by cytosolic‐beta‐glucosidase 19, or by colonic microbial action. The presence of esterases in the brush‐border, surely present in the enterocyte 125, has been speculated on, which would influence polyphenol esters such as chlorogenic acid, or xanthophyll esters, though cholesterol esterase may also act on the latter 126.
While proteins can exert negative effects on polyphenol bioavailability 127, the interaction of apolar phytochemcials and proteins during digestion has never been studied systematically, though certain proteins may aid in the emulsification of lipid soluble phytochemicals 128. Contrarily, a positive effect of sugars on polyphenol glucoside uptake [via stimulation of sodium‐glucose linked transporter 1 (SGLT‐1)] has been suggested 129. The effect of dietary fibers is assumed to be negative, due to gel formation, enhanced viscosity, or binding and entrapping of phytochemicals, but the effects of soluble versus insoluble, or prebiotic fiber are largely unknown. Other interactions with the food matrix exist during digestion, such as high concentrations of minerals impeding micelle formation 130, but their relevance in bioavailability remains unclear.
4.3. Cellular uptake
Prior to reaching the cellular surface, diffusion through the mucus 122 is required, though properties influencing diffusion are poorly understood. Viscosity and particle/micelle size of the digesta are expected to play a role. Porcine trials have shown that smaller particles diffuse more readily through the mucus layer 131; and it may be assumed that apolar compounds are likewise hindered 132. More sophisticated model studies including mucus‐producing cells, such as HT‐29‐MTX, are still rare but warranted.
Cellular uptake can occur by transcellular or paracellular routes. The latter is reserved for rather polar and small molecules <600 Da 133, i.e. ions, water, sugars, etc., as these pass through the tight junctions of the epithelium. Most phytochemicals are taken up via transcellular transport. Certain compounds (such as apple polyphenols) increase tight junction functionality 134, possibly via altered cellular signaling transduction pathways 135. On the other hand, certain fatty acids (such as caprylic acid) and high molecular weight polyphenols reduced tight junction barrier function, and may enhance uptake of other small compounds 134, 136, 137.
Transcellular uptake can occur by facilitated, active means or via passive diffusion. The latter is reserved for small and apolar molecules, as these readily pass through the cell membrane 138. Passive diffusion has been suggested for carotenoids 22, 23 and some apolar polyphenol aglycones 19. At low concentrations, presumably similar to levels of an average mixed diet, these compounds may also be taken up by carriers, actively or by facilitated diffusion. Over 400 transmembrane carrier proteins are known 139. Many polyphenols can inhibit these transporters, but stimulation was also described 140 and effects may depend on the polyphenol type, its concentration, exposure time, cell type, and substrate. For carotenoids, high concentrations of beta‐carotene reduced lutein uptake 141, which may be explained by higher affinity to transporters such as cluster of differentiation 36 (CD36) or unknown types 142. Enhanced uptake of polyphenol glucosides by sugars via SGLT‐1 activation 19, 143 is known, but the involvement of gut transporters other than SGLT‐1 and mono‐carboxyl‐transporter 1 (MCT1) for polyphenols is not certain, and most polyphenol aglycones are assumed to be taken up by passive diffusion 124.
4.4. First pass metabolism
A large number of metabolites can be formed following phase I (e.g. reduction/oxidation, methylation, hydroxylation, hydrolysis, e.g. via cytochrome P450‐dependent mixed‐function oxidases (CYPs) and catechol‐O‐methyl‐transferase) and phase II metabolism (e.g. glucuronidation by uridine‐5’‐diphosphate glucuronosyltransferase, and sulfation via sulfotransferases), in human enterocytes 144, 145. Certain phytochemicals can up‐ or downregulate these enzymes, influencing the availability of the native compounds. For example, uptake of curcumin was approximately 20‐fold enhanced when co‐administered with piperine in humans 146. High doses of certain polyphenols may override phase I/II metabolism, though not much is known on thresholds in this respect 19. Such interactions are certainly a gap in our knowledge. For carotenoids, cleavage by beta‐cartotene oxygenase 1/beta‐carotene dioxygenase 2 results in symmetric/asymmetric cleavage, producing apo‐carotenals. However, these reactions are far from quantitative, and appear to depend on genetic factors and the type of carotenoid 23. The bioactivities of apo‐carotenals (except retinols) are poorly comprehended, though they may be highly effective, e.g. in activating nuclear receptors 147. Detecting small amounts of these compounds is difficult, and suitable standards are often not commercially available.
4.5. Transport through the epithelium
Many phytochemicals are treated as xenobiotics, expelled from the cell, typically by increasing their polarity and via efflux transporters. These transmembrane proteins are typically ATP‐dependent efflux pumps. Little is known on the specificity of these transporters, and less on the potential of phytochemicals or nutrients to block them, which could result in enhanced bioavailability. It is assumed that P‐glycoprotein, MRP2 (multidrug resistance protein 2), and breast cancer resistance protein (BCRP) are the most important transporters, and that these may be competitively inhibited by polyphenols, perhaps by blocking ATPase 19. The transport from the cell to the basolateral side (i.e. bloodstream or lymph) is even more poorly understood. Compounds that could increase or decrease MRP3 (or MRP1, MRP4, and possibly several MCTs), could affect polyphenol uptake, as e.g. demonstrated by mice overexpressing MRP3 and showing high resveratrol bioavailability 148. It is assumed that carotenoids can be transported back into the lumen by scavenger receptor class B member 1 and possibly ATP‐binding cassette subfamily G, member 5/8, but more research is needed. Transport through the cells, e.g. by transporters such as fatty acid binding proteins is assumed, but not confirmed 23. It can be further assumed that proteins involved in chylomicron generation [i.e. microsomal triglyceride transfer protein, apoB48, apoAIV, and Sar1b 23, 149, also influence carotenoid transport. For polyphenols, transport through the cells is not understood, and may occur primarily by diffusion.
5. Metabolism in the colon and other organs
5.1. Metabolism of polyphenols
In general, polyphenol metabolism is fairly well known due to their xenobiotic nature. Polyphenols undergo metabolism in intestinal and liver tissues and by colon microbiota 15. In fact, it seems that the colon may be the major important site for polyphenol uptake, at least for orange juice rich in hesperitin and naringin 150, and studies with ileostomists suggested significant absorption from the colon 151. Phenolic compounds are glucuronidated and sulfated in the liver and intestinal tissues, and these metabolites are found in body fluids 125, 152, 153, 154, 155. Hepatic metabolites can be recycled back to the small intestine through biliary excretion 27, 156, 157, 158, 159 and end up in the colon, where they are deglucuronidated by microbial α, D‐glucuronidases before ring fission 160, 161.
Sophisticated methods for studying enterohepatic circulation, including humans, using a perfusion technique, have been published 162, 163, in addition to various articles on tissue metabolites of phenolic compounds 164, e.g. flavanol monomers and tea polyphenols. However, the enterohepatic circulation of colonic metabolites requires further investigation and hepatic metabolism of colonic metabolites of plant phenolic compounds should be addressed in the future 162. Furthermore, Monagas et al. 165 suggested that microbial metabolites may act as signal molecules, and their action should be taken into account in more detail in future investigations.
Absorbed microbial‐derived metabolites (such as enterolignans and dihydroxylated compounds such as methyl catechol) can be subjected to further glucuronidation, methylation, sulfation, or glycination in the liver, while phase I metabolism (oxidation/reduction reactions) seems to occur to a lesser extent 27. The interplay between the liver and the colon, the enterohepatic circulation, leads to a long residence time (up to 24–48 h) in the blood 157, 162, 166, 167, 168, which can go unobserved unless longer sampling of blood or urine is carried out 164, 169. Finally, the phenolic microbial metabolites are distributed to tissues and are excreted via urine, partly as free but mainly as hepatic conjugates, depending on the structure of the parent backbone 125, 157, 158.
One of the major gaps is the lack of knowledge concerning hepatic conversion of small colon‐derived phenolic acids, which may be missed in analyses of human body fluids or cell lines due to their hydrophilic nature. Extraction of samples may also cause bias, as Sawai et al. 157 elucidated the ratio of excreted conjugated and free phenolic acid colon‐derived metabolites from quercetin derivatives in urine, by using water saturated ethyl acetate to enhance the yield of polar conjugates.
Although human studies are more relevant, mechanisms of action can be studied using in vitro human‐based cell model systems designed to study properties of drugs 27. In recent years, novel cell lines and culture strategies have helped in overcoming the scarcity of human liver material and problems in maintaining the expression and function of metabolizing enzymes 170. The advent of human stem cell derived hepatocytes will potentially provide an unlimited source of human hepatocytes 171. Human hepatocyte three‐dimensional models, with complete hepatic metabolizing enzymes, transporters, and cofactors, may be applicable to metabolite profiling, pathway identification, CYP450 inhibition, CYP450 induction, and uptake and efflux transporter inhibition by polyphenols and their metabolites.
The lack of standards for metabolites causes limitations to study both polyphenol and phytosterol metabolism. Chemical procedures and biochemical labeling tools are available for the synthesis of many conjugated metabolites 172. However, the absence of standards limits the use of some analytical methodologies, and frequently enzyme treatment is used to overcome the problem of hepatic conjugative metabolism. For example, conjugated flavan‐3‐ol metabolites are rarely available commercially, and most studies have analyzed plasma and urine samples after treatment with glucuronidase/sulfatase, providing data only for the aglycones 164. However, some sulfate conjugates are resistant to enzyme hydrolysis, thus, this methodology may underestimate bioavailability 173, 174. This is the situation for estimating epicatechin bioavailability from cocoa products. Direct analysis of the individual epicatechin metabolites by LC–MS/MS may overcome this problem, however, the chirality of the aglycone will still be unknown, and it is plausible that the enantiomers could differ in their biological activity. Each flavan‐3‐ol and each associated phase II conjugate can, in theory, occur as four enantiomers, with the (+) and (–) forms resolvable only by chiral chromatography, and so far as we are aware there are no reports of chiral analysis of conjugated metabolites in urine or plasma 164.
However, targeted LC‐MS analytical approaches can be used for identifying many metabolites by exact mass, if a theoretical prediction of possible metabolites and conjugates based on expected metabolism is available 154. Nontargeted metabolomic profiling can find metabolites based on structural similarity to the parent compound, if the mass spectra can be coupled with a compound library. Profiling of phenolic metabolomes has been assessed by NMR, GC, LC and 2D‐GC with TOF mass detection coupled with a compound library 25, 175, 176, 177, 178.
5.2. Metabolism of lipophilic compounds
Regarding lipophilic bioactive compounds, serum phytosterol bioavailability is reported to be below 10%, suggesting that they may reach the colon and subjected to microbial metabolism 179. The pathway of microbial transformation from sterol to stanol form in humans was reported in the 1970's, but the kinetics of the reaction steps and metabolites in the large intestine have not been studied systematically 180. For carotenoids, the liver stores and distributes carotenoids to other tissues, but the mechanisms are not known accurately 181. However, intervention studies have shown that increased dietary intake of carotenoids influences serum concentrations more than colon concentrations 182, suggesting that absorption occurs mostly in the small intestine. In addition, metabolites of lycopene produced in vivo also occur naturally at low concentrations in tomato, which causes difficulties in the differentiation of the origin of the metabolites 183.
5.3. Transport, tissue distribution, and mechanism of action
Transport of metabolites and their parent compounds to tissues has not been studied adequately. This is important, since it is the key action to ensure the desired biological activity. There is a lack of information whether conjugated polyphenols can prolong their residence time in the body by modulating the activity of the transporters in the excretory organs. A recent study of transmembrane transport of flavonoids and some of their methylated and glucuronidated metabolites using human cerebral microvessel endothelial cells as a blood–brain barrier cell model 184 showed that the metabolites were transported in a time‐dependent manner and showed higher transport efficiency than the native flavonoids and were not further transformed by the cells. Polyphenols affect their own metabolism and tissue distribution, due to their capacity to modulate the activity of xenobiotic metabolizing enzymes and transporters, modulating their own concentration‐time profiles in the body and also promoting alterations in drug or toxin pharmacokinetics, the mechanisms of which are not known 140, 185. Therefore, additive, synergistic, or antagonistic effects of xenobiotics (dietary phytochemicals, drugs, and toxins) need to be addressed. For example, the downregulation of cytochrome P450 3A4 by grapefruit juice and its polyphenols, enhancing the concentration of several pharmaceuticals and increasing their effects is well known 186. Furthermore, the knowledge of pharmacodynamics and tissue distribution for most polyphenols and their metabolites is still lacking. It would also be relevant to study individual variation in the tissue distribution and the potential bioactivity caused by genetic variation in pathways related to bioavailability and transport 187.
In vitro models for preclinical research using stem cells, and patient‐specific induced pluripotent stem cells and reprogrammed somatic cells from patients are already applied in disease modeling and drug discovery, and may be applicable to test polyphenol metabolite health benefits. Microengineered physiological systems, also known as “organs‐on‐chips”, can reconstitute physiologically critical features of human tissues and their interactions 188. The nematode Caernohabditis elegans, with the presence of tissue and organ systems, is increasingly used as an in vivo model and has been employed to study the metabolism of methylated catechin derivatives and their biological effects on oxidative and thermal stress resistance 189. Thus, novel cell assays and nematode in vivo assays may be applied to study mechanisms of action of the parent compounds versus their liver/colon metabolites, although it should be noted that in vivo models such as C. elegans or Drosophila (like mice) may produce quite different metabolites from ingested phytochemicals than humans.
Only a few studies have combined the assay of biological effects of bioactive compounds or their colonic metabolites from whole foods or their extracts with an in vitro GI digestion process with/without colonic fermentation 102, 172, 190, or compared the biological activity of metabolites to their parent compound 178. Furthermore, the synergistic effects of the phytochemical metabolite pool, including the interplay between liver and colon microbiota should be studied. Finally, cell‐based assays measuring the bioactivity of the metabolites at relevant concentration are scarce 102, 191, 192. In the future, bioactivity assays to mimic the actual in vivo situation in the corresponding tissue, should use metabolite pools from whole foods, at tissue‐tolerant concentrations.
6. Conclusion
In this position paper, we have tried to highlight gaps of knowledge with respect to selected phytochemicals. It has to be noted that the data presented represent a somewhat simple and general paradigm for phytochemicals, and that, due to the very large variety and properties of secondary plant compounds, the statements cannot be generalized toward all groups of phytochemicals. In addition, some of the missing aspects discussed in this paper may currently be tackled, but have not been published.
However, while recent work has greatly enhanced our insight into the metabolism and bioavailability of a range of phytochemicals, many factors governing matrix release, solubilization, cellular uptake, and biotransformation remain poorly understood. Major aspects which deserve more attention when estimating bioavailability aspects include effects of innovative processing techniques, synergistic effects of mixed/whole diets, factors effecting micelle formation, co‐constituents influencing influx and efflux via transporter systems or altering phase I/II metabolism, as these have often been overlooked or excluded from consideration, in part due to difficulties to include their study in vivo or in vitro. In the future, enhanced availability of analytical possibilities to investigate these aspects such as through broader availability of instruments to measure food texture, visualization of micelles (TEM, Mastersizer), improved cell models of absorption and metabolism (mucus producing, liver cells, 3D models), ways to produce knock‐out variants (e.g. of certain transporters) in animal models (nematodes, mice, etc.) to study pathways of absorption and bioactivity of metabolites, and improved chromatographic techniques and commercial availability of metabolites (e.g. sulfates and glucuronides) will aid toward an improved understanding of these important aspects of bioavailability.
The authors have declared no conflict of interest.
Acknowledgement
The EU‐COST FA 1005 Infogest action is greatly acknowledged for providing funding for travel and meetings and allowing for open access.
Colour Online: See the article online to view Fig. 1 in colour.
7 References
- 1. Hooper, L. , Kroon, P. A. , Rimm, E. B. , Cohn, J. S. et al., Flavonoids, flavonoid‐rich foods, and cardiovascular risk: a meta‐analysis of randomized controlled trials. Am. J. Clin. Nutr. 2008, 88, 38–50. [DOI] [PubMed] [Google Scholar]
- 2. Hamer, M. , Chida, Y. , Intake of fruit, vegetables, and antioxidants and risk of type 2 diabetes: systematic review and meta‐analysis. J. Hypertens. 2007, 25, 2361–2369. [DOI] [PubMed] [Google Scholar]
- 3. Bar‐Sela, G. , Epelbaum, R. , Schaffer, M. , Curcumin as an anti‐cancer agent: review of the gap between basic and clinical applications. Curr. Med. Chem. 2010, 17, 190–197. [DOI] [PubMed] [Google Scholar]
- 4. Hausenblas, H. A. , Schoulda, J. A. , Smoliga, J. M. , Resveratrol treatment as an adjunct to pharmacological management in type 2 diabetes mellitus–systematic review and meta‐analysis. Mol. Nutr. Food Res. 2015, 59, 147–159. [DOI] [PubMed] [Google Scholar]
- 5. Jacobs, D. R. , Tapsell, L. C. , Food synergy: the key to a healthy diet. Proc. Nutr. Soc. 2013, 72, 200–206. [DOI] [PubMed] [Google Scholar]
- 6. Prasain, J. K. , Carlson, S. H. , Wyss, J. M. , Flavonoids and age‐related disease: risk, benefits and critical windows. Maturitas 2010, 66, 163–171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Bjelakovic, G. , Nikolova, D. , Gluud, L. L. , Simonetti, R. G. et al., Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta‐analysis. JAMA 2007, 297, 842–857. [DOI] [PubMed] [Google Scholar]
- 8. Bjelakovic, G. , Nikolova, D. , Gluud, C. , Antioxidant supplements and mortality. Curr. Opin. Clin. Nutr. Metab. Care 2014, 17, 40–44. [DOI] [PubMed] [Google Scholar]
- 9. Ye, Y. , Li, J. , Yuan, Z. , Effect of antioxidant vitamin supplementation on cardiovascular outcomes: a meta‐analysis of randomized controlled trials. PLoS One 2013, 8, e56803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Jordan, V. C. , Avoiding the bad and enhancing the good of soy supplements in breast cancer. J. Natl. Cancer Inst. 2014, 106, 1–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Del, R. D. , Rodriguez‐Mateos, A. , Spencer, J. P. , Tognolini, M. et al., Dietary (poly)phenolics in human health: structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Moran, N. E. , Erdman, J. W., Jr. , Clinton, S. K. , Complex interactions between dietary and genetic factors impact lycopene metabolism and distribution. Arch. Biochem. Biophys. 2013, 539, 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Rodriguez‐Mateos, A. , Vauzour, D. , Krueger, C. G. , Shanmuganayagam, D. et al., Bioavailability, bioactivity and impact on health of dietary flavonoids and related compounds: an update. Arch. Toxicol. 2014, 88, 1803–1853. [DOI] [PubMed] [Google Scholar]
- 14. Holst, B. , Williamson, G. , Nutrients and phytochemicals: from bioavailability to bioefficacy beyond antioxidants. Curr. Opin. Biotechnol. 2008, 19, 73–82. [DOI] [PubMed] [Google Scholar]
- 15. Alminger, M. , Aura, A.‐M. , Bohn, T. , Dufour, C. et al., In vitro models for studying secondary plant metabolite digestion and bioaccessibility. Compr. Rev. Fd. Technol. Fd. Saf. 2014, 13, 413–436. [DOI] [PubMed] [Google Scholar]
- 16. Svelander, C. A. , Lopez‐Sanchez, P. , Pudney, P. D. , Schumm, S. et al., High pressure homogenization increases the in vitro bioaccessibility of alpha‐ and beta‐carotene in carrot emulsions but not of lycopene in tomato emulsions. J. Food Sci. 2011, 76, H215–H225. [DOI] [PubMed] [Google Scholar]
- 17. Wang, L. , Bohn, T. , in: Bouayed J., Bohn T. (Eds.), Nutrition, Well‐Being and Health, Intech, Croatia: 2012, pp. 201–224. [Google Scholar]
- 18. Panozzo, A. , Lemmens, L. , Van, L. A. , Manzocco, L. et al., Microstructure and bioaccessibility of different carotenoid species as affected by high pressure homogenisation: a case study on differently coloured tomatoes. Food Chem. 2013, 141, 4094–4100. [DOI] [PubMed] [Google Scholar]
- 19. Bohn, T. , Dietary factors affecting polyphenol bioavailability. Nut. Rev. 2014, 72, 429–452. [DOI] [PubMed] [Google Scholar]
- 20. Minekus, M. , Alminger, M. , Alvito, P. , Ballance, S. et al., A standardised static in‐vitro digestion method suitable for food – an international consensus. Food Funct. 2014, 5, 1113–1124. [DOI] [PubMed] [Google Scholar]
- 21. Czank, C. , Cassidy, A. , Zhang, Q. , Morrison, D. J. et al., Human metabolism and elimination of the anthocyanin, cyanidin‐3‐glucoside: a (13)C‐tracer study. Am. J. Clin. Nutr. 2013, 97, 995–1003. [DOI] [PubMed] [Google Scholar]
- 22. Bohn, T. , Bioavailabilty of non‐provitamin A carotenoids. Curr. Nut. Food Sci. 2008, 4, 240–258. [Google Scholar]
- 23. Borel, P. , Genetic variations involved in interindividual variability in carotenoid status. Mol. Nutr. Food Res. 2012, 56, 228–240. [DOI] [PubMed] [Google Scholar]
- 24. Bazzocco, S. , Mattila, I. , Guyot, S. , Renard, C. M. et al., Factors affecting the conversion of apple polyphenols to phenolic acids and fruit matrix to short‐chain fatty acids by human faecal microbiota in vitro. Eur. J. Nutr. 2008, 47, 442–452. [DOI] [PubMed] [Google Scholar]
- 25. Aura, A. M. , Mattila, I. , Hyotylainen, T. , Gopalacharyulu, P. et al., Characterization of microbial metabolism of Syrah grape products in an in vitro colon model using targeted and non‐targeted analytical approaches. Eur. J. Nutr. 2013, 52, 833–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Aura, A. M. , Mattila, I. , Seppainen‐Laakso, T. , Miettinen, J. et al., Microbial metabolism of catechin stereoisomers by human faecal microbiota: comparison of targeted analysis and a non‐targeted metabolomics method. Phytochem. Lett. 2008, 1, 18–22. [Google Scholar]
- 27. del Rio, D. , Rodriguez‐Mateos, A. , Spencer, J. P. , Tognolini, M. et al., Dietary (poly)phenolics in human health: structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox. Signal. 2013, 18, 1818–1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Verkerk, R. , Schreiner, M. , Krumbein, A. , Ciska, E. et al., Glucosinolates in Brassica vegetables: the influence of the food supply chain on intake, bioavailability and human health. Mol. Nutr. Food Res. 2009, 53(Suppl 2), S219–S265. [DOI] [PubMed] [Google Scholar]
- 29. Van Buggenhout, S. , Alminger, M. , Lemmens, L. , Colle, I. et al., In vitro approaches to estimate the effect of food processing on carotenoid bioavailability need thorough understanding of process induced microstructural changes. Trends Food Sci. Technol. 2010, 21, 607–618. [Google Scholar]
- 30. Sensoy, I. , A review on the relationship between food structure, processing, and bioavailability. Crit. Rev. Food Sci. Nutr. 2014, 54, 902–909. [DOI] [PubMed] [Google Scholar]
- 31. Schweiggert, R. M. , Mezger, D. , Schimpf, F. , Steingass, C. B. et al., Influence of chromoplast morphology on carotenoid bioaccessibility of carrot, mango, papaya, and tomato. Food Chem. 2012, 135, 2736–2742. [DOI] [PubMed] [Google Scholar]
- 32. Tydeman, E. A. , Parker, M. L. , Faulks, R. M. , Cross, K. L. et al., Effect of carrot (Daucus carota) microstructure on carotene bioaccessibility in the upper gastrointestinal tract. 2. In vivo digestions. J. Agric. Food Chem. 2010, 58, 9855–9860. [DOI] [PubMed] [Google Scholar]
- 33. Palafox‐Carlos, H. , yala‐Zavala, J. F. , Gonzalez‐Aguilar, G. A. , The role of dietary fiber in the bioaccessibility and bioavailability of fruit and vegetable antioxidants. J. Food Sci. 2011, 76, R6–R15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Buchweitz, M. , Speth, M. , Kammerer, D. R. , Carle, R. , Stabilisation of strawberry (Fragaria x ananassa Duch.) anthocyanins by different pectins. Food Chem. 2013, 141, 2998–3006. [DOI] [PubMed] [Google Scholar]
- 35. Kodera, Y. , Suzuki, A. , Imada, O. , Kasuga, S. et al., Physical, chemical, and biological properties of s‐allylcysteine, an amino acid derived from garlic. J. Agric. Food Chem. 2002, 50, 622–632. [DOI] [PubMed] [Google Scholar]
- 36. Tapiero, H. , Townsend, D. M. , Tew, K. D. , Organosulfur compounds from alliaceae in the prevention of human pathologies. Biomed. Pharmacother. 2004, 58, 183–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Holst, B. , Williamson, G. , A critical review of the bioavailability of glucosinolates and related compounds. Nat. Prod. Rep. 2004, 21, 425–447. [DOI] [PubMed] [Google Scholar]
- 38. Ioannou, I. , Hafsa, I. , Hamdi, S. , Charbonnel, C. Ã. et al., Review of the effects of food processing and formulation on flavonol and anthocyanin behaviour. J. Food Engin. 2012, 111, 208–217. [Google Scholar]
- 39. Reif, C. , Arrigoni, E. , Berger, F. , Baumgartner, D. et al., Lutein and beta‐carotene content of green leafy Brassica species grown under different conditions. LWT ‐ Food Sci. Technol. 2013, 53, 378–381. [Google Scholar]
- 40. Szydlowska‐Czerniak, A. , Trokowski, K. , Karlovits, G. , Szlyk, E. , Effect of refining processes on antioxidant capacity, total contents of phenolics and carotenoids in palm oils. Food Chem. 2011, 129, 1187–1192. [DOI] [PubMed] [Google Scholar]
- 41. Szydlowska‐Czerniak, A. , Karlovits, G. , Dianoczki, C. , Recseg, K. et al., Comparison of two analytical methods for assessing antioxidant capacity of rapeseed and olive oils. J. Am. Oil Chem. Soc. 2008, 85, 141–149. [Google Scholar]
- 42. Wang, C. , Riedl, K. M. , Schwartz, S. J. , Fate of folates during vegetable juice processing — deglutamylation and interconversion. Food Res. Int. 2013, 53, 440–448. [Google Scholar]
- 43. Hung, P. V. , Maeda, T. , Miyatake, K. , Morita, N. , Total phenolic compounds and antioxidant capacity of wheat graded flours by polishing method. Food Res. Int. 2009, 42, 185–190. [Google Scholar]
- 44. Pradeep, S. R. , Guha, M. , Effect of processing methods on the nutraceutical and antioxidant properties of little millet (Panicum sumatrense) extracts. Food Chem. 2011, 126, 1643–1647. [DOI] [PubMed] [Google Scholar]
- 45. Kim, J. S. , Kang, O. J. , Gweon, O. C. , Comparison of phenolic acids and flavonoids in black garlic at different thermal processing steps. J. Funct. Foods 2013, 5, 80–86. [Google Scholar]
- 46. Wu, L. , Huang, Z. , Qin, P. , Ren, G. , Effects of processing on phytochemical profiles and biological activities for production of sorghum tea. Food Res. Int. 2013, 53, 678–685. [Google Scholar]
- 47. Leong, S. Y. , Oey, I. , Effects of processing on anthocyanins, carotenoids and vitamin C in summer fruits and vegetables. Food Chem. 2012, 133, 1577–1587. [Google Scholar]
- 48. Karakaya, S. , Yilmaz, N. , Lycopene content and antioxidant activity of fresh and processed tomatoes and in vitro bioavailability of lycopene. J. Sci. Food Agric. 2007, 87, 2342–2347. [Google Scholar]
- 49. Sommano, S. , Caffin, N. , McDonald, J. , Cocksedge, R. , The impact of thermal processing on bioactive compounds in Australian native food products (bush tomato and Kakadu plum). Food Res. Int. 2013, 50, 557–561. [Google Scholar]
- 50. Zhang, D. , Hamauzu, Y. , Phenolics, ascorbic acid, carotenoids and antioxidant activity of broccoli and their changes during conventional and microwave cooking. Food Chem. 2004, 88, 503–509. [Google Scholar]
- 51. Mayer‐Miebach, E. , Behsnilian, D. , Regier, M. , Schuchmann, H. P. , Thermal processing of carrots: lycopene stability and isomerisation with regard to antioxidant potential. Food Res. Int. 2005, 38, 1103–1108. [Google Scholar]
- 52. Mayeaux, M. , Xu, Z. , King, J. M. , Prinyawiwatkul, W. , Effects of cooking conditions on the lycopene content in tomatoes. J. Food Sci. 2006, 71, C461–C464. [Google Scholar]
- 53. Seybold, C. , Frohlich, K. , Bitsch, R. , Otto, K. et al., Changes in contents of carotenoids and vitamin E during tomato processing. J. Agric. Food Chem. 2004, 52, 7005–7010. [DOI] [PubMed] [Google Scholar]
- 54. Pugliese, A. , Loizzo, M. R. , Tundis, R. , O'Callaghan, Y. et al., The effect of domestic processing on the content and bioaccessibility of carotenoids from chili peppers (Capsicum species). Food Chem. 2013, 141, 2606–2613. [DOI] [PubMed] [Google Scholar]
- 55. Ciska, E. , Kozlowska, H. , The effect of cooking on the glucosinolates content in white cabbage. Eur. Food Res. Technol. 2001, 212, 582–587. [Google Scholar]
- 56. Wennberg, M. , Ekvall, J. , Olsson, K. , Nyman, M. , Changes in carbohydrate and glucosinolate composition in white cabbage (Brassica oleracea var. capitata) during blanching and treatment with acetic acid. Food Chem. 2006, 95, 226–236. [Google Scholar]
- 57. Volden, J. , Borge, G. I. , Bengtsson, G. B. , Hansen, M. et al., Effect of thermal treatment on glucosinolates and antioxidant‐related parameters in red cabbage (Brassica oleracea L. ssp. capitata f. rubra). Food Chem. 2008, 109, 595–605. [Google Scholar]
- 58. Francisco, M. , Velasco, P. , Moreno, D. A. , Garcia‐Viguera, C. et al., Cooking methods of Brassica rapa affect the preservation of glucosinolates, phenolics and vitamin C. Food Res. Int. 2010, 43, 1455–1463. [Google Scholar]
- 59. Hanschen, F. S. , Rohn, S. , Mewis, I. , Schreiner, M. et al., Influence of the chemical structure on the thermal degradation of the glucosinolates in broccoli sprouts. Food Chem. 2012, 130, 1–8. [Google Scholar]
- 60. Girgin, N. , El, S. N. , Effects of cooking on in vitro sinigrin bioaccessibility, total phenols, antioxidant and antimutagenic activity of cauliflower (Brassica oleraceae L. var. Botrytis). J. Food Compos. Anal. 2015, 37, 119–127. [Google Scholar]
- 61. Sanchez‐Moreno, C. , de, A. B. , Plaza, L. , Elez‐Martinez, P. et al., Nutritional approaches and health‐related properties of plant foods processed by high pressure and pulsed electric fields. Crit. Rev. Food Sci. Nutr. 2009, 49, 552–576. [DOI] [PubMed] [Google Scholar]
- 62. McInerney, J. K. , Seccafien, C. A. , Stewart, C. M. , Bird, A. R. , Effects of high pressure processing on antioxidant activity, and total carotenoid content and availability, in vegetables. Innov. Food Sci. Emerg. Technol. 2007, 8, 543–548. [Google Scholar]
- 63. Colle, I. , Van Buggenhout, S. , Van Loey, A. , Hendrickx, M. , High pressure homogenization followed by thermal processing of tomato pulp: influence on microstructure and lycopene in vitro bioaccessibility. Food Res. Int. 2010, 43, 2193–2200. [Google Scholar]
- 64. Knockaert, G. , Pulissery, S. K. , Colle, I. , Van, B. S. et al., Lycopene degradation, isomerization and in vitro bioaccessibility in high pressure homogenized tomato puree containing oil: effect of additional thermal and high pressure processing. Food Chem. 2012, 135, 1290–1297. [DOI] [PubMed] [Google Scholar]
- 65. Cilla, A. , Alegria, A. , de, A. B. , Sanchez‐Moreno, C. et al., Bioaccessibility of tocopherols, carotenoids, and ascorbic acid from milk‐ and soy‐based fruit beverages: influence of food matrix and processing. J. Agric. Food Chem. 2012, 60, 7282–7290. [DOI] [PubMed] [Google Scholar]
- 66. Gupta, R. , Kopec, R. E. , Schwartz, S. J. , Balasubramaniam, V. M. , Combined pressure‐temperature effects on carotenoid retention and bioaccessibility in tomato juice. J. Agric. Food Chem. 2011, 59, 7808–7817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Tomas‐Navarro, M. , Vallejo, F. , Sentandreu, E. , Navarro, J. L. et al., Volunteer stratification is more relevant than technological treatment in orange juice flavanone bioavailability. J. Agric. Food Chem. 2013, 62, 24–27. [DOI] [PubMed] [Google Scholar]
- 68. Sanchez‐Moreno, C. , Cano, M. P. , de, A. B. , Plaza, L. et al., Pulsed electric fields‐processed orange juice consumption increases plasma vitamin C and decreases F2‐isoprostanes in healthy humans. J. Nutr. Biochem. 2004, 15, 601–607. [DOI] [PubMed] [Google Scholar]
- 69. Sanchez‐Moreno, C. , Pilar, C. M. , de, A. B. , Plaza, L. et al., Intake of Mediterranean vegetable soup treated by pulsed electric fields affects plasma vitamin C and antioxidant biomarkers in humans. Int. J. Food Sci. Nutr. 2005, 56, 115–124. [DOI] [PubMed] [Google Scholar]
- 70. Anese, M. , Mirolo, G. , Beraldo, P. , Lippe, G. , Effect of ultrasound treatments of tomato pulp on microstructure and lycopene in vitro bioaccessibility. Food Chem. 2013, 136, 458–463. [DOI] [PubMed] [Google Scholar]
- 71. McDougall, G. J. , Dobson, P. , Smith, P. , Blake, A. et al., Assessing potential bioavailability of raspberry anthocyanins using an in vitro digestion system. J. Agric. Food Chem. 2005, 53, 5896–5904. [DOI] [PubMed] [Google Scholar]
- 72. Cebeci, F. , Sahin‐Yesilcubuk, N. , The matrix effect of blueberry, oat meal and milk on polyphenols, antioxidant activity and potential bioavailability. Int. J. Food Sci. Nutr. 2014, 65, 69–78. [DOI] [PubMed] [Google Scholar]
- 73. Green, R. J. , Murphy, A. S. , Schulz, B. , Watkins, B. A. et al., Common tea formulations modulate in vitro digestive recovery of green tea catechins. Mol. Nutr. Food Res. 2007, 51, 1152–1162. [DOI] [PubMed] [Google Scholar]
- 74. Peters, C. M. , Green, R. J. , Janle, E. M. , Ferruzzi, M. G. , Formulation with ascorbic acid and sucrose modulates catechin bioavailability from green tea. Food Res. Int. 2010, 43, 95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Xie, Y. , Kosinska, A. , Xu, H. , Andlauer, W. , Milk enhances intestinal absorption of green tea catechins in in vitro digestion/Caco‐2 cells model. Food Res. Int. 2013, 53, 793–800. [Google Scholar]
- 76. van het Hof, K. H. , Kivits, G. A. , Weststrate, J. A. , Tijburg, L. B. , Bioavailability of catechins from tea: the effect of milk. Eur. J. Clin. Nutr. 1998, 52, 356–359. [DOI] [PubMed] [Google Scholar]
- 77. Neilson, A. P. , Sapper, T. N. , Janle, E. M. , Rudolph, R. et al., Chocolate matrix factors modulate the pharmacokinetic behavior of cocoa flavan‐3‐ol phase II metabolites following oral consumption by Sprague‐Dawley rats. J. Agric. Food Chem. 2010, 58, 6685–6691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Neilson, A. P. , George, J. C. , Janle, E. M. , Mattes, R. D. et al., Influence of chocolate matrix composition on cocoa flavan‐3‐ol bioaccessibility in vitro and bioavailability in humans. J. Agric. Food Chem. 2009, 57, 9418–9426. [DOI] [PubMed] [Google Scholar]
- 79. Guo, Y. , Mah, E. , Davis, C. G. , Jalili, T. et al., Dietary fat increases quercetin bioavailability in overweight adults. Mol. Nutr. Food Res. 2013, 57, 896–905. [DOI] [PubMed] [Google Scholar]
- 80. Rein, M. J. , Renouf, M. , Cruz‐Hernandez, C. , Actis‐Goretta, L. et al., Bioavailability of bioactive food compounds: a challenging journey to bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Ortega, N. , Reguant, J. , Romero, M. P. , Macia, A. et al., Effect of fat content on the digestibility and bioaccessibility of cocoa polyphenol by an in vitro digestion model. J. Agric. Food Chem. 2009, 57, 5743–5749. [DOI] [PubMed] [Google Scholar]
- 82. Henning, S. M. , Choo, J. J. , Heber, D. , Nongallated compared with gallated flavan‐3‐ols in green and black tea are more bioavailable. J. Nutr. 2008, 138, 1529S‐1534S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Scheepens, A. , Tan, K. , Paxton, J. W. , Improving the oral bioavailability of beneficial polyphenols through designed synergies. Genes Nutr. 2010, 5, 75–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Huang, W. , Lee, S. L. , Yu, L. X. , Mechanistic approaches to predicting oral drug absorption. AAPS J. 2009, 11, 217–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Parrott, N. , Lave, T. , Applications of physiologically based absorption models in drug discovery and development. Mol. Pharm. 2008, 5, 760–775. [DOI] [PubMed] [Google Scholar]
- 86. Guerra, A. , Etienne‐Mesmin, L. , Livrelli, V. , Denis, S. et al., Relevance and challenges in modeling human gastric and small intestinal digestion. Trends Biotechnol. 2012, 30, 591–600. [DOI] [PubMed] [Google Scholar]
- 87. Reboul, E. , Richelle, M. , Perrot, E. , smoulins‐Malezet, C. et al., Bioaccessibility of carotenoids and vitamin E from their main dietary sources. J. Agric Food Chem. 2006, 54, 8749–8755. [DOI] [PubMed] [Google Scholar]
- 88. Granado‐Lorencio, F. , Donoso‐Navarro, E. , Sanchez‐Siles, L. M. , Blanco‐Navarro, I. et al., Bioavailability of beta‐cryptoxanthin in the presence of phytosterols: in vitro and in vivo studies. J. Agric. Food Chem. 2011, 59, 11819–11824. [DOI] [PubMed] [Google Scholar]
- 89. Garcia‐Llatas, G. , Cilla, A. , Alegria, A. , Lagarda, M. A. J. , Bioavailability of plant sterol‐enriched milk‐based fruit beverages: in vivo and in vitro studies. J. Funct. Foods 2015, 14, 44–50. [Google Scholar]
- 90. Garrett, D. A. , Failla, M. L. , Sarama, R. J. , Development of an in vitro digestion method to assess carotenoid bioavailability from meals. J. Agric. Food Chem. 1999, 47, 4301–4309. [DOI] [PubMed] [Google Scholar]
- 91. Garrett, D. A. , Failla, M. L. , Sarama, R. J. , Estimation of carotenoid bioavailability from fresh stir‐fried vegetables using an in vitro digestion/Caco‐2 cell culture model. J. Nutr. Biochem. 2000, 11, 574–580. [DOI] [PubMed] [Google Scholar]
- 92. Alminger, M. , Svelander, C. , Wellner, A. , Martinez‐Tomas, R. et al., Applicability of in vitro models in predicting the in vivo bioavailability of lycopene and beta‐carotene from differently processed soups. Food Nutr. Sci. 2012, 3, 477–489. [Google Scholar]
- 93. Van Buggenhout, S. , Sila, D. N. , Duvetter, T. , Van Loey, A. et al., Pectins in processed fruits and vegetables: Part III “Texture Engineering”. Comp. Rev. Food Sci. Food Saf. 2009, 8, 105–117. [Google Scholar]
- 94. Kong, F. , Singh, R. P. , A model stomach system to investigate disintegration kinetics of solid foods during gastric digestion. J. Food Sci. 2008, 73, E202–E210. [DOI] [PubMed] [Google Scholar]
- 95. Kong, F. , Singh, R. P. , Modes of disintegration of solid foods in simulated gastric environment. Food Biophys. 2009, 4, 180–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Lentle, R. G. , Janssen, P. W. M. , The Physical Processes of Digestion; Springer, New York: 2011. [Google Scholar]
- 97. Kong, F. , Oztop, M. H. , Paul Singh, R. , McCarthy, M. J. , Effect of boiling, roasting and frying on disintegration of peanuts in simulated gastric environment. LWT ‐ Food Sci. Technol. 2013, 50, 32–38. [Google Scholar]
- 98. Flores, F. P. , Singh, R. K. , Kerr, W. L. , Pegg, R. B. et al., Total phenolics content and antioxidant capacities of microencapsulated blueberry anthocyanins during in vitro digestion. Food Chem. 2014, 153, 272–278. [DOI] [PubMed] [Google Scholar]
- 99. Haratifar, S. , Meckling, K. A. , Corredig, M. , Bioefficacy of tea catechins encapsulated in casein micelles tested on a normal mouse cell line (4D/WT) and its cancerous counterpart (D/v‐src) before and after in vitro digestion. Food Funct. 2014, 5, 1160–1166. [DOI] [PubMed] [Google Scholar]
- 100. Jilani, H. , Cilla A., Barbera R., Hamdi, M. , Impact of biosorption into Saccharomyces cerevisiae and in vitro gastrointestinal digestion on total polyphenols and antioxidant capacity of tea and olive leaves infusions. Proceedings of the 2nd International Conference on Food Digestion, 137. Proceedings of the 2nd International Conference on Food Digestion, 137 2013. [Google Scholar]
- 101. McDougall, G. , Dobson, P. , Shapiro, F. , Smith, P. et al., Assessing bioavailability of soft fruit polyphenols in vitro. Acta Hort. 2007, 744, 135–148. [Google Scholar]
- 102. Brown, E. M. , McDougall, G. J. , Stewart, D. , Pereira‐Caro, G. et al., Persistence of anticancer activity in berry extracts after simulated gastrointestinal digestion and colonic fermentation. PLoS One 2012, 7, e49740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Rodriguez‐Roque, M. J. , Rojas‐Grau, M. A. , Elez‐Martinez, P. , Martin‐Belloso, O. , Changes in vitamin C, phenolic, and carotenoid profiles throughout in vitro gastrointestinal digestion of a blended fruit juice. J. Agric. Food Chem. 2013, 61, 1859–1867. [DOI] [PubMed] [Google Scholar]
- 104. Li, H. , Deng, Z. , Liu, R. , Loewen, S. et al., Bioaccessibility, in vitro antioxidant activities and in vivo anti‐inflammatory activities of a purple tomato (Solanum lycopersicum L.). Food Chem. 2014, 159, 353–360. [DOI] [PubMed] [Google Scholar]
- 105. Bassolino, L. , Zhang, Y. , Schoonbeek, H. J. , Kiferle, C. et al., Accumulation of anthocyanins in tomato skin extends shelf life. New Phytol. 2013, 200, 650–655. [DOI] [PubMed] [Google Scholar]
- 106. McDougall, G. J. , Kulkarni, N. N. , Stewart, D. , Current developments on the inhibitory effects of berry polyphenols on digestive enzymes. Biofactors 2008, 34, 73–80. [DOI] [PubMed] [Google Scholar]
- 107. Arranz, S. , Silvan, J. M. , Saura‐Calixto, F. , Nonextractable polyphenols, usually ignored, are the major part of dietary polyphenols: a study on the Spanish diet. Mol. Nutr. Food Res. 2010, 54, 1646–1658. [DOI] [PubMed] [Google Scholar]
- 108. Maeda‐Yamamoto, M. , Ema, K. , Tokuda, Y. , Monobe, M. et al., Effect of green tea powder (Camellia sinensis L. cv. Benifuuki) particle size on O‐methylated EGCG absorption in rats; The Kakegawa Study. Cytotechnology 2011, 63, 171–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. McClements, D. J. , Decker, E. A. , Park, Y. , Controlling lipid bioavailability through physicochemical and structural approaches. Crit. Rev. Food Sci. Nutr. 2009, 49, 48–67. [DOI] [PubMed] [Google Scholar]
- 110. Bravo, L. , Polyphenols: chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998, 56, 317–333. [DOI] [PubMed] [Google Scholar]
- 111. Anderson, J. M. , Van Itallie, C. M. , Physiology and function of the tight junction. Cold Spring Harb. Perspect. Biol. 2009, 1, a002584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Miranda, L. , Deusser, H. , Evers, D. , The impact of in vitro digestion on bioaccessibility of polyphenols from potatoes and sweet potatoes and their influence on iron absorption by human intestinal cells. Food Funct. 2013, 4, 1595–1601. [DOI] [PubMed] [Google Scholar]
- 113. Perez‐Jimenez, J. , AZ‐Rubio, M. E. , Saura‐Calixto, F. , Non‐extractable polyphenols, a major dietary antioxidant: occurrence, metabolic fate and health effects. Nutr. Res. Rev. 2013, 26, 118–129. [DOI] [PubMed] [Google Scholar]
- 114. Chitchumroonchokchai, C. , Failla, M. L. , Hydrolysis of zeaxanthin esters by carboxyl ester lipase during digestion facilitates micellarization and uptake of the xanthophyll by Caco‐2 human intestinal cells. J. Nutr. 2006, 136, 588–594. [DOI] [PubMed] [Google Scholar]
- 115. Andre, C. M. , Greenwood, J. M. , Walker, E. G. , Rassam, M. et al., Anti‐inflammatory procyanidins and triterpenes in 109 apple varieties. J. Agric. Food Chem. 2012, 60, 10546–10554. [DOI] [PubMed] [Google Scholar]
- 116. Bohn, T. , Tian, Q. , Chitchumroonchokchai, C. , Failla, M. L. et al., Supplementation of test meals with fat‐free phytosterol products can reduce cholesterol micellarization during simulated digestion and cholesterol accumulation by Caco‐2 cells. J. Agric. Food Chem. 2007, 55, 267–272. [DOI] [PubMed] [Google Scholar]
- 117. Parker, R. S. , Absorption, metabolism, and transport of carotenoids. FASEB J. 1996, 10, 542–551. [PubMed] [Google Scholar]
- 118. Sy, C. , Gleize, B. , Dangles, O. , Landrier, J. F. et al., Effects of physicochemical properties of carotenoids on their bioaccessibility, intestinal cell uptake, and blood and tissue concentrations. Mol. Nutr. Food Res. 2012, 56, 1385–1397. [DOI] [PubMed] [Google Scholar]
- 119. Lesser, S. , Cermak, R. , Wolffram, S. , Bioavailability of quercetin in pigs is influenced by the dietary fat content. J. Nutr. 2004, 134, 1508–1511. [DOI] [PubMed] [Google Scholar]
- 120. Chitchumroonchokchai, C. , Riedl, K. M. , Suksumrarn, S. , Clinton, S. K. et al., Xanthones in mangosteen juice are absorbed and partially conjugated by healthy adults. J. Nutr. 2012, 142, 675–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Bohn, T. , Blackwood, M. , Francis, D. , Tian, Q. et al., Bioavailability of phytochemical constituents from a novel soy fortified lycopene rich tomato juice developed for targeted cancer prevention trials. Nutr. Canc. 2013, 65, 919–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Thomson, A. B. , Jarocka‐Cyrta, E. , Faria, J. , Wild, G. E. , Small bowel review–Part II. Can. J. Gastroenterol. 1997, 11, 159–165. [DOI] [PubMed] [Google Scholar]
- 123. Han, X. , Shenemail, T. , Hongxiang L., Dietary polyphenols and their biological significance. Int. J. Mol. Sci. 2007, 8, 950–988. [Google Scholar]
- 124. Manach, C. , Scalbert, A. , Morand, C. , Remesy, C. et al., Polyphenols: food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [DOI] [PubMed] [Google Scholar]
- 125. Kern, S. M. , Bennett, R. N. , Needs, P. W. , Mellon, F. A. et al., Characterization of metabolites of hydroxycinnamates in the in vitro model of human small intestinal epithelium caco‐2 cells. J. Agric. Food Chem. 2003, 51, 7884–7891. [DOI] [PubMed] [Google Scholar]
- 126. Granado‐Lorencio, F. , Olmedilla‐Alonso, B. , Herrero‐Barbudo, C. , Blanco‐Navarro, I. et al., In vitro bioaccessibility of carotenoids and tocopherols from fruits and vegetables. Food Chem. 2007, 102, 641–648. [DOI] [PubMed] [Google Scholar]
- 127. Cai, K. , Bennick, A. , Effect of salivary proteins on the transport of tannin and quercetin across intestinal epithelial cells in culture. Biochem. Pharmacol. 2006, 72, 974–980. [DOI] [PubMed] [Google Scholar]
- 128. Singh, H. , Ye, A. , Structural and biochemical factors affecting the digestion of protein‐stabilized emulsions. Curr. Opin. Colloid Interface Sci. 2013, 18, 360–370. [Google Scholar]
- 129. Bitsch, R. , Netzel, M. , Frank, T. , Strass, G. et al., Bioavailability and biokinetics of anthocyanins from red grape juice and red wine. J. Biomed. Biotechnol. 2004, 2004, 293–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Biehler, E. , Hoffmann, L. , Krause, E. , Bohn, T. , Divalent minerals decrease micellarization and uptake of carotenoids and digestion products into Caco‐2 cells. J. Nutr. 2011, 141, 1769–1776. [DOI] [PubMed] [Google Scholar]
- 131. Lai, S. K. , Wang, Y. Y. , Wirtz, D. , Hanes, J. , Micro‐ and macrorheology of mucus. Adv. Drug Deliv. Rev. 2009, 61, 86–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Dunnhaupt, S. , Barthelmes, J. , Hombach, J. , Sakloetsakun, D. et al., Distribution of thiolated mucoadhesive nanoparticles on intestinal mucosa. Int. J. Pharm. 2011, 408, 191–199. [DOI] [PubMed] [Google Scholar]
- 133. Menard, S. , Cerf‐Bensussan, N. , Heyman, M. , Multiple facets of intestinal permeability and epithelial handling of dietary antigens. Mucosal. Immunol 2010, 3, 247–259. [DOI] [PubMed] [Google Scholar]
- 134. Rogoll, D. , Bergmann, H. , Hellenschmidt, D. , Heinze, J. et al., Influence of apple polyphenols on the intestinal barrier in a colonic cell model. J. Appl. Bot. Food Qual. 2010, 83, 110–117. [Google Scholar]
- 135. Ulluwishewa, D. , Anderson, R. C. , McNabb, W. C. , Moughan, P. J. et al., Regulation of tight junction permeability by intestinal bacteria and dietary components. J. Nutr. 2011, 141, 769–776. [DOI] [PubMed] [Google Scholar]
- 136. Appeldoorn, M. M. , Vincken, J. P. , Gruppen, H. , Hollman, P. C. , Procyanidin dimers A1, A2, and B2 are absorbed without conjugation or methylation from the small intestine of rats. J. Nutr. 2009, 139, 1469–1473. [DOI] [PubMed] [Google Scholar]
- 137. Shoji, T. , Masumoto, S. , Moriichi, N. , Akiyama, H. et al., Apple procyanidin oligomers absorption in rats after oral administration: analysis of procyanidins in plasma using the porter method and high‐performance liquid chromatography/tandem mass spectrometry. J. Agric. Food Chem. 2006, 54, 884–892. [DOI] [PubMed] [Google Scholar]
- 138. Chhabra, R. S. , Intestinal absorption and metabolism of xenobiotics. Environ. Health Perspect. 1979, 33, 61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Estudante, M. , Morais, J. G. , Soveral, G. , Benet, L. Z. , Intestinal drug transporters: an overview. Adv. Drug Deliv. Rev. 2013, 65, 1340–1356. [DOI] [PubMed] [Google Scholar]
- 140. Li, Y. , Paxton, J. W. , The effects of flavonoids on the ABC transporters: consequences for the pharmacokinetics of substrate drugs. Exp. Opin. Drug Metab. Toxicol. 2013, 9, 267–285. [DOI] [PubMed] [Google Scholar]
- 141. Reboul, E. , Thap, S. , Tourniaire, F. , Andre, M. et al., Differential effect of dietary antioxidant classes (carotenoids, polyphenols, vitamins C and E) on lutein absorption. Br. J. Nutr. 2007, 97, 440–446. [DOI] [PubMed] [Google Scholar]
- 142. Reboul, E. , Absorption of vitamin A and carotenoids by the enterocyte: focus on transport proteins. Nutrients 2013, 5, 3563–3581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Schramm, D. D. , Karim, M. , Schrader, H. R. , Holt, R. R. et al., Food effects on the absorption and pharmacokinetics of cocoa flavanols. Life Sci. 2003, 73, 857–869. [DOI] [PubMed] [Google Scholar]
- 144. Paine, M. F. , Fisher, M. B. , Immunochemical identification of UGT isoforms in human small bowel and in caco‐2 cell monolayers. Biochem. Biophys. Res. Commun. 2000, 273, 1053–1057. [DOI] [PubMed] [Google Scholar]
- 145. Chen, G. , Zhang, D. , Jing, N. , Yin, S. et al., Human gastrointestinal sulfotransferases: identification and distribution. Toxicol. Appl. Pharmacol. 2003, 187, 186–197. [DOI] [PubMed] [Google Scholar]
- 146. Shoba, G. , Joy, D. , Joseph, T. , Majeed, M. et al., Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med. 1998, 64, 353–356. [DOI] [PubMed] [Google Scholar]
- 147. Kaulmann, A. , Bohn, T. , Carotenoids, inflammation and oxidative stress – implications in cellular signalling pathways. Nutr. Res. 2014, 34, 907–929. [DOI] [PubMed] [Google Scholar]
- 148. van de Wetering, K. , Burkon, A. , Feddema, W. , Bot, A. et al., Intestinal breast cancer resistance protein (BCRP)/Bcrp1 and multidrug resistance protein 3 (MRP3)/Mrp3 are involved in the pharmacokinetics of resveratrol. Mol. Pharmacol. 2009, 75, 876–885. [DOI] [PubMed] [Google Scholar]
- 149. Hussain, M. M. , Fatma, S. , Pan, X. , Iqbal, J. , Intestinal lipoprotein assembly. Curr. Opin. Lipidol. 2005, 16, 281–285. [DOI] [PubMed] [Google Scholar]
- 150. Escudero‐Lopez, B. , Calani, L. , Fernandez‐Pachon, M. S. , Ortega, A. et al., Absorption, metabolism, and excretion of fermented orange juice (poly)phenols in rats. Biofactors 2014, 40, 327–335. [DOI] [PubMed] [Google Scholar]
- 151. Borges, G. , Lean, M. E. , Roberts, S. A. , Crozier, A. , Bioavailability of dietary (poly)phenols: a study with ileostomists to discriminate between absorption in small and large intestine. Food Funct. 2013, 4, 754–762. [DOI] [PubMed] [Google Scholar]
- 152. Nardini, M. , Cirillo, E. , Natella, F. , Scaccini, C. , Absorption of phenolic acids in humans after coffee consumption. J. Agric. Food Chem. 2002, 50, 5735–5741. [DOI] [PubMed] [Google Scholar]
- 153. Natsume, M. , Osakabe, N. , Oyama, M. , Sasaki, M. et al., Structures of (‐)‐epicatechin glucuronide identified from plasma and urine after oral ingestion of (‐)‐epicatechin: differences between human and rat. Free Radic. Biol. Med. 2003, 34, 840–849. [DOI] [PubMed] [Google Scholar]
- 154. Pimpao, R. C. , Dew, T. , Figueira, M. E. , McDougall, G. J. et al., Urinary metabolite profiling identifies novel colonic metabolites and conjugates of phenolics in healthy volunteers. Mol. Nutr. Food Res. 2014, 58, 1414–1425. [DOI] [PubMed] [Google Scholar]
- 155. Felgines, C. , Talavera, S. , Gonthier, M. P. , Texier, O. et al., Strawberry anthocyanins are recovered in urine as glucuro‐ and sulfoconjugates in humans. J. Nutr. 2003, 133, 1296–1301. [DOI] [PubMed] [Google Scholar]
- 156. Lampe, J. W. , Isoflavonoid and lignan phytoestrogens as dietary biomarkers. J. Nutr. 2003, 133, 956S–964S. [DOI] [PubMed] [Google Scholar]
- 157. Sawai, Y. , Kohsaka, K. , Nishiyama, Y. , Ando, K. , Serum concentrations of rutoside metabolites after oral administration of a rutoside formulation to humans. Arzneimittelforschung 1987, 37, 729–732. [PubMed] [Google Scholar]
- 158. Adlercreutz, H. , van der, W. J. , Kinzel, J. , Attalla, H. et al., Lignan and isoflavonoid conjugates in human urine. J. Steroid Biochem. Mol. Biol. 1995, 52, 97–103. [DOI] [PubMed] [Google Scholar]
- 159. Axelson, M. , Setchell, K. D. , The excretion of lignans in rats – evidence for an intestinal bacterial source for this new group of compounds. FEBS Lett. 1981, 123, 337–342. [DOI] [PubMed] [Google Scholar]
- 160. Duenas, M. , Surco‐Laos, F. , Gonzalez‐Manzano, S. , Gonzalez‐Paramas, A. M. et al., Deglycosylation is a key step in biotransformation and lifespan effects of quercetin‐3‐O‐glucoside in Caenorhabditis elegans . Pharmacol. Res. 2013, 76, 41–48. [DOI] [PubMed] [Google Scholar]
- 161. Aura, A. M. , O'Leary, K. A. , Williamson, G. , Ojala, M. et al., Quercetin derivatives are deconjugated and converted to hydroxyphenylacetic acids but not methylated by human fecal flora in vitro. J. Agric. Food Chem. 2002, 50, 1725–1730. [DOI] [PubMed] [Google Scholar]
- 162. Crozier, A. , Absorption, metabolism, and excretion of (‐)‐epicatechin in humans: an evaluation of recent findings. Am. J. Clin. Nutr. 2013, 98, 861–862. [DOI] [PubMed] [Google Scholar]
- 163. Actis‐Goretta, L. , Leveques, A. , Rein, M. , Teml, A. et al., Intestinal absorption, metabolism, and excretion of (‐)‐epicatechin in healthy humans assessed by using an intestinal perfusion technique. Am. J. Clin. Nutr. 2013, 98, 924–933. [DOI] [PubMed] [Google Scholar]
- 164. Clifford, M. N. , van der Hooft, J. J. , Crozier, A. , Human studies on the absorption, distribution, metabolism, and excretion of tea polyphenols. Am. J. Clin. Nutr. 2013, 98, 1619S–1630S. [DOI] [PubMed] [Google Scholar]
- 165. Monagas, M. , Urpi‐Sarda, M. , Sanchez‐Patan, F. , Llorach, R. et al., Insights into the metabolism and microbial biotransformation of dietary flavan‐3‐ols and the bioactivity of their metabolites. Food Funct. 2010, 1, 233–253 [DOI] [PubMed] [Google Scholar]
- 166. Kuijsten, A. , Arts, I. C. , Vree, T. B. , Hollman, P. C. , Pharmacokinetics of enterolignans in healthy men and women consuming a single dose of secoisolariciresinol diglucoside. J. Nutr. 2005, 135, 795–801. [DOI] [PubMed] [Google Scholar]
- 167. Seeram, N. P. , Henning, S. M. , Zhang, Y. , Suchard, M. et al., Pomegranate juice ellagitannin metabolites are present in human plasma and some persist in urine for up to 48 hours. J. Nutr. 2006, 136, 2481–2485. [DOI] [PubMed] [Google Scholar]
- 168. Gross, M. , Pfeiffer, M. , Martini, M. , Campbell, D. et al., The quantitation of metabolites of quercetin flavonols in human urine. Cancer Epidemiol. Biomarkers Prev. 1996, 5, 711–720. [PubMed] [Google Scholar]
- 169. Vetrani, C. , Rivellese, A. A. , Annuzzi, G. , Mattila, I. et al., Phenolic metabolites as compliance biomarker for polyphenol intake in a randomized controlled human intervention. Food Res. Int. 2014, 63, 233–238. [Google Scholar]
- 170. Griffith, L. G. , Wells, A. , Stolz, D. B. , Engineering liver. Hepatology 2014, 60, 1426–1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Mandenius, C. F. , Andersson, T. B. , Alves, P. M. , Batzl‐Hartmann, C. et al., Toward preclinical predictive drug testing for metabolism and hepatotoxicity by using in vitro models derived from human embryonic stem cells and human cell lines – a report on the Vitrocellomics EU‐project. Altern. Lab. Anim. 2011, 39, 147–171. [DOI] [PubMed] [Google Scholar]
- 172. Barron, D. , Smarrito‐Menozzi, C. , Viton, F. , (Bio)chemical labelling tools for studying absorption & metabolism of dietary phenols – an overview. Curr. Org. Chem. 2012, 16, 663–690. [Google Scholar]
- 173. Saha, S. , Hollands, W. , Needs, P. W. , Ostertag, L. M. et al., Human O‐sulfated metabolites of (‐)‐epicatechin and methyl‐(‐)‐epicatechin are poor substrates for commercial aryl‐sulfatases: implications for studies concerned with quantifying epicatechin bioavailability. Pharmacol. Res. 2012, 65, 592–602. [DOI] [PubMed] [Google Scholar]
- 174. Actis‐Goretta, L. , Leveques, A. , Giuffrida, F. , Romanov‐Michailidis, F. et al., Elucidation of (‐)‐epicatechin metabolites after ingestion of chocolate by healthy humans. Free Radic. Biol. Med. 2012, 53, 787–795. [DOI] [PubMed] [Google Scholar]
- 175. Jacobs, D. M. , Deltimple, N. , van, V. E. , van Dorsten, F. A. et al., (1)H NMR metabolite profiling of feces as a tool to assess the impact of nutrition on the human microbiome. NMR Biomed. 2008, 21, 615–626. [DOI] [PubMed] [Google Scholar]
- 176. Hanhineva, K. , Aura, A. M. , Rogachev, I. , Matero, S. et al., In vitro microbiotic fermentation causes an extensive metabolite turnover of rye bran phytochemicals. PLoS One 2012, 7, e39322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Grun, C. H. , van Dorsten, F. A. , Jacobs, D. M. , Le, B. M. et al., GC‐MS methods for metabolic profiling of microbial fermentation products of dietary polyphenols in human and in vitro intervention studies. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2008, 871, 212–219. [DOI] [PubMed] [Google Scholar]
- 178. Oleaga, C. , Ciudad, C. J. , Izquierdo‐Pulido, M. , Noe, V. , Cocoa flavanol metabolites activate HNF‐3beta, Sp1, and NFY‐mediated transcription of apolipoprotein AI in human cells. Mol. Nutr. Food Res. 2013, 57, 986–995. [DOI] [PubMed] [Google Scholar]
- 179. Awad, A. B. , Fink, C. S. , Phytosterols as anticancer dietary components: evidence and mechanism of action. J. Nutr. 2000, 130, 2127–2130. [DOI] [PubMed] [Google Scholar]
- 180. Wong, A. , Chemical and microbiological considerations of phytosterols and their relative efficacies in functional foods for the lowering of serum cholesterol levels in humans: a review. J. Funct. Foods 2014, 6, 60–72. [Google Scholar]
- 181. Moussa, M. , Landrier, J. F. , Reboul, E. , Ghiringhelli, O. et al., Lycopene absorption in human intestinal cells and in mice involves scavenger receptor class B type I but not Niemann‐Pick C1‐like 1. J. Nutr. 2008, 138, 1432–1436. [DOI] [PubMed] [Google Scholar]
- 182. Sen, A. , Ren, J. , Ruffin, M. T. , Turgeon, D. K. et al., Relationships between serum and colon concentrations of carotenoids and fatty acids in randomized dietary intervention trial. Cancer Prev. Res. (Phila) 2013, 6, 558–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Khachik, F. , Carvalho, L. , Bernstein, P. S. , Muir, G. J. et al., Chemistry, distribution, and metabolism of tomato carotenoids and their impact on human health. Exp. Biol. Med. (Maywood) 2002, 227, 845–851. [DOI] [PubMed] [Google Scholar]
- 184. Faria, A. , Meireles, M. , Fernandes, I. , Santos‐Buelga, C. et al., Flavonoid metabolites transport across a human BBB model. Food Chem. 2014, 149, 190–196. [DOI] [PubMed] [Google Scholar]
- 185. Sergent, T. , Garsou, S. , Schaut, A. , De, S. S. et al., Differential modulation of ochratoxin A absorption across Caco‐2 cells by dietary polyphenols, used at realistic intestinal concentrations. Toxicol. Lett. 2005, 159, 60–70. [DOI] [PubMed] [Google Scholar]
- 186. Bailey, D. G. , Malcolm, J. , Arnold, O. , Spence, J. D. , Grapefruit juice–drug interactions. Br. J. Clin. Pharmacol. 1998, 46, 101–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Lampe, J. W. , Interindividual differences in response to plant‐based diets: implications for cancer risk. Am. J. Clin. Nutr. 2009, 89, 1553S–1557S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Yum, K. , Hong, S. G. , Healy, K. E. , Lee, L. P. , Physiologically relevant organs on chips. Biotechnol. J. 2014, 9, 16–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Surco‐Laos, F. , Cabello, J. , Gomez‐Orte, E. , Gonzalez‐Manzano, S. et al., Effects of O‐methylated metabolites of quercetin on oxidative stress, thermotolerance, lifespan and bioavailability on Caenorhabditis elegans . Food Funct. 2011, 2, 445–456. [DOI] [PubMed] [Google Scholar]
- 190. Cilla, A. , Alegria, A. , Barbera, R. , in: Morales‐Gonzales J. A. (Ed.), Oxidative Stress and Chronic Degenerative Diseases‐a Role for Antioxidants Rijeka, InTech, Croatia: 2013, 131–151. [Google Scholar]
- 191. Tavares, L. , Figueira, I. , Macedo, D. , McDougall, G. J. et al., Neuroprotective effect of blackberry (Rubus sp.) polyphenols is potentiated after simulated gastrointestinal digestion. Food Chem. 2012, 131, 1443–1452. [Google Scholar]
- 192. Tavares, L. , Figueira, I. , McDougall, G. J. , Vieira, H. L. et al., Neuroprotective effects of digested polyphenols from wild blackberry species. Eur. J. Nutr. 2013, 52, 225–236. [DOI] [PubMed] [Google Scholar]
- 193. Ferruzzi, M. G. , The influence of beverage composition on delivery of phenolic compounds from coffee and tea. Physiol. Behav. 2010, 100, 33–41. [DOI] [PubMed] [Google Scholar]
- 194. Konishi, Y. , Shimizu, M. , Transepithelial transport of ferulic acid by monocarboxylic acid transporter in Caco‐2 cell monolayers. Biosci. Biotechnol. Biochem. 2003, 67, 856–862. [DOI] [PubMed] [Google Scholar]
- 195. Watanabe, H. , Yashiro, T. , Tohjo, Y. , Konishi, Y. , Non‐involvement of the human monocarboxylic acid transporter 1 (MCT1) in the transport of phenolic acid. Biosci. Biotechnol. Biochem. 2006, 70, 1928–1933. [DOI] [PubMed] [Google Scholar]
- 196. Gill, R. K. , Saksena, S. , Alrefai, W. A. , Sarwar, Z. et al., Expression and membrane localization of MCT isoforms along the length of the human intestine. Am. J. Physiol. Cell Physiol. 2005, 289, C846–C852. [DOI] [PubMed] [Google Scholar]
- 197. Wolffram, S. , Block, M. , Ader, P. , Quercetin‐3‐glucoside is transported by the glucose carrier SGLT1 across the brush border membrane of rat small intestine. J. Nutr. 2002, 132, 630–635. [DOI] [PubMed] [Google Scholar]
- 198. Reboul, E. , Borel, P. , Proteins involved in uptake, intracellular transport and basolateral secretion of fat‐soluble vitamins and carotenoids by mammalian enterocytes. Prog. Lipid Res. 2011, 50, 388–402. [DOI] [PubMed] [Google Scholar]
