Open access peer-reviewed chapter

Models to Evaluate the Prebiotic Potential of Foods

Written By

Jailane de Souza Aquino, Kamila Sabino Batista, Francisca Nayara Dantas Duarte Menezes, Priscilla Paulo Lins, Jessyca Alencar de Sousa Gomes and Laiane Alves da Silva

Submitted: 08 November 2016 Reviewed: 12 April 2017 Published: 02 August 2017

DOI: 10.5772/intechopen.69174

From the Edited Volume

Functional Food - Improve Health through Adequate Food

Edited by Maria Chavarri Hueda

Chapter metrics overview

2,558 Chapter Downloads

View Full Metrics

Abstract

The interest in studying the prebiotic effect of foods is increasing due to the way in which the consumption of these foods influences the gut microbiota and how the metabolic activity of the microbiota affects the health and well‐being of the host. Several in vitro and in vivo studies have been developed to elucidate the prebiotic effect of foods, and particularly in in vivo studies, the physiological dynamics of this effect has been studied in healthy or diseased individuals. In this chapter, the main in vitro and in vivo models developed for the study of the prebiotic potential of foods will be approached, which can be used by those planning to advance in this field of research.

Keywords

  • functional foods
  • prebiotics
  • chronic diseases
  • animal models
  • intestinal microbiota

1. Introduction

Modern society has changed its standard of living every decade and today, health is becoming an increasingly important personal and social value. Prevention of health problems is prioritized due to the costs associated with curative medicine, especially chronic diseases, which can be prevented by a healthier lifestyle [1]. In addition to the practice of physical activity, adequate nutrition is an essential aspect influencing a person’s health status. Consumers are more aware that their food choices can have consequences for their health and maintenance of a healthy lifestyle [2, 3].

Food matrixes are composed of several nutrient or non‐nutrient substances that interact in a complex way. In this perspective, foods have the basic function of feeding, some of which present health benefits that go beyond nutrition, such as functional foods. Functional foods may exert physiological benefits and/or reduce the risk of chronic diseases, in addition to basic nutritional functions, and may be similar in appearance to conventional foods and consumed as part of a regular diet [4].

Prebiotics are among functional foods, which are defined as a component of the edible product, in which its health benefit must be measurable and not due to its absorption in the blood stream or due to the sole action of the component, but it should be evidenced that the simple presence of the prebiotic component and the formulation in which it is inserted alter the composition or activity of the microbial flora in the target host by modulating it [4], for stimulating the proliferation of a select group of beneficial colon bacteria and suppressing the proliferation of micro‐organisms harmful to health [5].

To be considered prebiotic, food or its components must: (i) resist the processes of host digestion, absorption, and adsorption; (ii) be fermented by the microbiota that colonize the gastrointestinal tract (GI); and (iii) selectively stimulate the growth and/or activity of one or a limited number of bacteria within the gastrointestinal tract, altering the colonic microbiota in favor of a healthier composition [3, 4].

Prebiotics found in natural sources such as vegetables, roots, fruits, milk, and honey are non‐digestible carbohydrates such as resistant starch (RS), galacto‐oligosaccharides (GOS), fructooligosaccharides (FOS), xylooligosaccharides (XOS), pectic oligosaccharides (POS), and various oligosaccharides that provide carbohydrates fermentable by the beneficial colon micro‐organisms [6, 7]. Among these, probiotic micro‐organisms such as bacteria belonging to the genus Lactobacillus and Bifidobacterium, as well as Streptococcus, Saccharomyces cerevisiae, Escherichia coli, and Bacillus spp. stand out, which have been studied on a smaller scale. These bacteria are fermentative, obligatory, or facultative anaerobes, and their inherent biological characteristics allow them to prevail over potential pathogenic micro‐organisms in the digestive tract [8].

Probiotic micro‐organisms are currently defined as live micro‐organisms, which when consumed in adequate amounts provide a positive health effect on the host [9]. Butel [10] suggests three modes of action of probiotics, which influence the host’s health. One of the first suggested modes of action is called “barrier” effect or resistance to colonization against pathogenic bacteria due to the production of broad‐spectrum inhibition bacteriocins, metabolites such as acid lactic and short‐chain fatty acids—SCFA (e.g., acetate, butyrate, propionate)—which induce a decrease in pH, being favorable for bacterial growth, or biosurfactants with antimicrobial activity. The improvement of the barrier function in the gut mucosa may be due to the increase of the mucus layer or to the production of defensins and proteins of tight junctions.

In addition to prebiotic and probiotic foods, symbiotic foods, in which probiotic and prebiotic are combined, have been increasingly developed due to the favorable adaptation of the probiotic to the prebiotic substrate before consumption, which may increase the beneficial effects of each of them [11, 12].

In this context, the modulation of the gut microbiota by diet has been studied [13, 14]. The composition and metabolism of the colonic microbiota can be influenced by the type of diet, nutrient balance (mainly carbohydrates, proteins, and fats), and the amount of diet ingested [15]. The impact of diet on microbiota composition is determined by tolerance of gut conditions and by the competition for substrates among microbial species, which demonstrate different capabilities to utilize dietary substrates, promoting the competition for substrates available in the large intestine, playing an important role in defining microbiota composition [16]. The healthy microbiota can be defined as the normal microbiota that maintains and promotes well‐being and absence of diseases, especially of the gastrointestinal tract. The colon is the most densely populated part of the gastrointestinal tract and houses about 500 different bacterial species. These bacteria, each with its own spectrum of metabolic activities, make the colon the most metabolically active organ in the human body [17].

The gut microbiota influences the metabolic processes, preventing and modulating chronic diseases such as obesity, diabetes, insulin resistance, and cardiovascular diseases [18] because it interferes in several systems such as cardiovascular [19], nervous [20, 21], immune [22], endocrine [23], and the gastrointestinal system itself.

From this perspective, the prebiotic effect of foods can be studied from in vitro systems or from in vivo models using healthy and diseased animals or humans. Each model has advantages and disadvantages, which will be discussed in the next sections of this chapter.

Advertisement

2. Types of prebiotics

Dietary fibers (DF) are bioactive components, which may have prebiotic activity, present in plants, defined as the edible part of plants or analogous carbohydrates resistant to digestion and absorption in the small intestine of humans, with complete or partial fermentation in the large intestine [24, 25]. Regarding water solubility, DFs are classified as soluble (SDF) and insoluble (IDF). IDFs include cellulose, lignin, and some hemicelluloses and pectins [26, 27]. SDFs, however, comprise the majority of pectins, gums, mucilages, and hemicelluloses [28, 29].

The concept of DF has been expanded to include functionally similar substances such as RS, inulin, FOS, and GOS. GOS or FOS may have beneficial effects such as anti‐adhesion or direct immunomodulation that do not require fermentation and are therefore called additional biological activities not related to their effects on the gut microbiota [30]. There are several prebiotics with various origin and chemical properties. Inulin, FOS, GOS, lactulose, and polydextose are recognized as established prebiotics, whereas isomaltooligosaccharides (IMO), XOS, and lactitol are categorized as emerging prebiotics. In addition, resistant starch‐rich whole grains are considered prebiotic in nature, and it is assumed that their consumption leads to many health benefits [31]. The fermentability of dietary fibers such as oat b‐glucan, flaxseed gum, and fenugreek gum suggests their potential prebiotic application in promoting human health [31]. The main technological applications of prebiotics and the potential beneficial health effects on consumers of these foods are described in Figure 1.

Figure 1.

Degradation of dietary fibers and phenolic compounds by the gut microbiota. Dietary fibers () and phenolic compounds () reach the colon (mainly in the proximal part) and suffer a primary degradation by bacteria () to oligosaccharides and monosaccharides () and small phenolic acids (), respectively. Then, these compounds are used by the gut microbiota for the production of SCFA (), which increase the number of beneficial intestinal bacteria.

Plant‐derived polysaccharides arrive unchanged in the colon, being degraded by micro‐organisms living in the human GI tract to SCFA (Figure 2). The degradation of complex oligosaccharides (pectin, cellulose, hemicellulose, and resistant starches) involves a strong metabolic alignment among diverse micro‐organisms that makes up the intestinal microbiota, but these mechanisms are still not fully understood [24, 32].

Figure 2.

Some technological applications of prebiotics and health benefits from consumption.

In addition to DF, phenolic compounds (PC) or polyphenols may also benefit the gut microbiota, as up to 90% of plant PCs reach the colon and are used as substrates for the microbial production of small phenolic acids [33]. In turn, these biotransformed compounds modulate the microbial population in the gastrointestinal tract and are used as substrates for the production of SCFA [33, 34]. Results have reported that there is a possible interference of PC in the increase of viable Bifidobacterium and Lactobacillus cells in the intestine (in vivo model) and feces of animals or humans (in vitro model) [35, 36].

PC are secondary metabolites derived from pentoses‐phosphate, shikimic acid, and phenylpropanoid pathways in plants. They are divided into four main classes according to their chemical structure: flavonoids (including flavonols, flavanols, flavanones, flavones, anthocyanidins, chalcones, dihydrochalcones, dihydroflavonols, and isoflavones), lignans, stilbenes, and tannins. They have numerous reported physiological properties, such as vasodilators, anti‐thrombotic, anti‐inflammatory, anti‐apoptotic, hypolipemic, or anti‐atherogenic properties [37].

Prebiotics should be ingested daily as a way of ensuring continuous effect on the intestinal microbiota. However, recommendations for daily doses will depend on the type of food containing the prebiotic compound (naturally or added) or the isolated prebiotic compound consumed as a nutraceutical or prebiotic administrated by gavage (orogastric) or added to diet. The consumption of 5–8 g per day of inulin, FOS, or RS has been shown to significantly increase fecal bifidobacteria [38, 39]. In another study, rats received daily oral administration (gavage) of FOS (3 g/kg) or GOS (4 g/kg) for 5 weeks [40].

Other studies have added prebiotics to diets for rodents such as Sprague‐Dawley rats that consumed a high‐fat diet and diet added of 10% oligofructose [41] or rats that consumed AIN‐G diet added with 10% inulin or oligofructose [42]. Healthy or diabetic Wistar rats consumed basal diet supplemented with XOS (10%) or FOS (10%) or a combination of XOS (5%) and FOS (5%) [43].

Advertisement

3. Use of in vitro models in the study of the prebiotic potential of foods

In vitro modeling is useful for investigating the prebiotic potential of foods as it is less expensive, does not require sophisticated handling techniques, and allows simulating fermentation processes that occur along the large intestine and have few ethical limitations. However, they present limitations such as absence of interaction between neuroendocrine and immunological systems with the microbiota; absorptive processes, secretions, and defense systems are not incorporated into the models, as well as difficulty in controlling changes in the structures of microbial communities after inoculation. In these studies, it is possible to use pure microbial populations, known mixtures or fecal material [44].

The groups of colon bacteria present selective characteristics regarding the substrates available, and it is recommended that the studies use the mixed microbial culture, which simulates the microbial ecology of the human intestinal tract. Fermentation in anaerobic batches inoculated with fecal suspensions provides an excellent mode for small‐scale screening of new substrates. Until recently, the growth of specific bacteria in such fermentations was measured by counting colonies on selective agar. This approach, however, has several disadvantages (time‐consuming, labor intensive, and non‐recovery of uncultivable organisms). As a result, molecular techniques such as fluorescence in situ hybridization (FISH) were developed to study microbial communities [13, 45]. FISH involves the use of genus‐specific and in some cases species‐specific fluorescently labeled oligonucleotide probes. Hybridization of the probe that has its own specificity to recognize a particular group of bacteria to the complementary target sequence within bacterial cells results in fluorescently labeled cells that can be visualized and enumerated using fluorescence microscopy [45].

Generally, food or a substrate prebiotic extracted from the test food itself is lyophilized and supplemented in different concentrations to Man, Rogosa and Sharpe (MRS) medium; the negative control is represented by the MRS medium without the addition of the test food or substrate, and the positive control is represented by inulin [46, 47] or fructooligosaccharide [17, 48], which are recognized prebiotics. Frequently, experiments include the MRS medium with addition of glucose as the carbon source, which also serves as a control. After media are defined, probiotic micro‐organism strains such as Lactobacillus or Bifidobacterium are incubated and the samples are incubated under ideal conditions for the selected micro‐organisms. Thereafter, viable cell counts and metabolism monitoring of these micro‐organisms (quantification of short‐chain fatty acids and pH, among other parameters) are performed to confirm the prebiotic property of the food [47, 49]. SCFAs are saturated aliphatic organic acids that have from one to six carbon atoms, such as acetate (C2), propionate (C3), and butyrate (C4), and are the final products of bacterial fermentation processes.

Recently, many byproducts of the food industry have been studied as cheap and alternative sources of prebiotics [6, 49, 50]. The prebiotic effect of cashew apple (Anacardium occidentale L.) agro‐industrial byproduct powder on different potentially probiotic Lactobacillus strains (L. acidophilus LA‐05 and L. casei L‐26 and L. paracasei L‐10) was cultivated in broth containing cashew apple powder (20 or 30 g.L−1), glucose (20 g.L−1), or FOS (20 g.L−1). The cell viability of Lactobacillus strains (counts of viable cells) and changes in pH values, production of organic acids, and consumption of sugars in growth media were monitored for 48 h. The cultivation of Lactobacillus strains in broth containing glucose, FOS, or cashew apple powder resulted in high counts of viable cells, decreased pH, production of organic acids, and consumption of sugars over time, revealing intense bacterial metabolic activity and prebiotic activity [50]. Thuaytong and Anprung [51] used 1% (v/v) of prepared L. acidophilus LA‐5, and Bifidobacterium lactis BB‐12 was transferred into MRS broth, which was composed of 1% (w/v) glucose or 1% (w/v) inulin or 1% (w/v) prebiotic (guava samples), and demonstrated that both red guava and white pulp induced similar growth of prebiotic bacteria in glucose‐containing medium.

The study by Gómez et al. [49] confirmed the prebiotic effects caused by a refined product containing POS that promoted the growth of beneficial bacteria and the increase of SCFA concentrations. In a study carried out by Sousa et al. [52], yacon flour revealed a potential prebiotic activity in the growth of probiotic strains Enterococcus faecium 32, Bifidobacterium animalis Bo, L. acidophilus Ki, and L. casei L26, probably due to its content in FOS. Teixeira et al. [47] evaluated the influence of Amazonian tubers Dioscorea trifida, Calathea allouia, and Dioscorea altissima on the growth of Lactobacillus acidophillus bacteria and observed that the best in vitro result was for D. trifida fiber, which stimulated the bacterial growth without significant difference from commercial inulin.

Another in vitro model that is being used to evaluate the prebiotic activity of foods is the fermentation of animal or human feces added to the test food or extract [13, 53] and it is also used for the purpose of evaluating the metabolism of fecal micro‐organisms.

The beneficial health effects of prebiotics are related to their influence on the gut microbiota composition, stimulation of growth, metabolism, and activities of lactic acid bacteria, bifidobacteria, and other emergent strains such as Roseburia intestinales and Faecalibacterium prausnitzii) [7].

Quinoa (Chenopodium quinoa W.) and amaranth (Amaranthus caudatus L.) submitted to in vitro digestion and together with a control (without external carbon source) were used as carbon sources in batch cultures with fecal human inocula. After 48 h of incubation, both substrates stimulated in a similar proportion the growth of certain numerically predominant bacterial groups in the human gut microbiota, including Bifidobacterium spp., Lactobacillus‐Enterococcus, Atopobium, Bacteroides‐Prevotella, Clostridium coccoides‐Eubacterium rectale, F. prausnitzii, and Roseburia intestinalis assessed by FISH, in addition to total SCFAs (acetate, propionate, and butyrate) with a decrease in pH, suggesting that these pseudocereals can have prebiotic potential [13].

Broad beans (Vicia faba) and lupin seeds (Lupinus albus) were submitted to in vitro digestion used as carbon sources in anaerobic batch cultures to evaluate their impact on the gut microbiota composition (by FISH) and on their metabolic products (lactate and SCFAs). The fermentation of the lupine seeds resulted in a higher total amount of SCFA than the bean fermentation, and in both, there was a decrease in the pH of the fermentation medium. In addition, legume fermentation increased microbial fecal batch cultures, such as Bifidobacterium spp., Lactobacillus‐Enterococcus, Atopobium, Bacteroides‐Pretovella, C. coccoides‐E. rectale, F. prausnitzii, and R. intestinalis [54].

The prebiotic potential of POS obtained by orange peel wastes was assessed by in vitro fermentation using human fecal inocula. For comparative purposes, similar experiments were performed using orange pectin and commercial FOS as substrates for fermentation. POS particularly increased the amount of bifidobacteria and lactobacilli (assessed by FISH) so that the ratio between the counts of both genera and the total cell number increased from 17 in the inocula to 27% after fermentation. SCFA generation from POS fermentation was similar to that observed with FOS [49].

Sugar beet pulp (Beta vulgaris L.) and lemon peel wastes (Citrus limon L.) were used to obtain two mixtures of POS and in comparison, FOS and commercial pectins were assessed by in vitro fermentation and FISH using human fecal inocula. The joint populations of bifidobacteria and lactobacilli increased from 19 up to 29, 34, and 32% in cultures with pectic oligosaccharides from lemon peel wastes, beet pulp, and FOS, respectively. Faecalibacterium and Roseburia also increased their counts with all substrates (especially with pectic oligosaccharides from lemon peel wastes). The highest concentrations of organic acids were observed in media containing oligosaccharides, and these results confirm that pectic oligosaccharides present better prebiotic properties than pectins and are similar or better than FOS [6].

The prebiotic effect of oligosaccharides recovered and purified from caprine whey was evaluated by in vitro fermentation under anaerobic conditions using batch cultures at 37°C with human feces (by FISH). In this research, growth of Bifidobacterium spp. was significantly higher with purified oligosaccharides compared to the negative control. Lactic and propionic acids were the main SCFAs produced. These findings indicate that oligosaccharides naturally extracted from caprine whey or cheese whey (byproduct) could be used as new and valuable sources of prebiotics naturally produced in the lactating mammary gland of domestic species and not obtained by enzymatic reaction (trans‐galactosylation) from lactose, although numerous papers and patents mostly refer to specific GOS [55].

Other studies evaluating the prebiotic potential of food using in vitro models are described in Table 1.

FoodMain resultsReferences
Oligosaccharides from Pitaya (Hylocereus undatus (Haw.))↑ Resistance to gastric acidity[56]
↑ Growth of Bifidobacterium and Lactobacillus
Byproducts of date pits (Phoenix dactylifera L. var.Medjoul) and apple bagasse (Malus domestica var. rayada)Fermentation by colonic bacteria produced AGCC (formate, succinate, acetate, propionate, and butyrate)[57]
Pomegranate peel (Punica granatum)Fermentation of pomegranate peel flour by colonic bacteria generated acetic, propionic, and butyric acids[36]
water‐soluble xylan from wheat bran (XOS extraction)↑ Growth of L. brevis, B. adolescentis, and the Weissella spp. on XOS[58]
↑ Lactic acid and acetic acid production after 48‐h incubation.
raw and roasted almonds (Prunus amygdalus)Predigested raw and roasted almonds promoted the growth of Lactobacillus acidophilus (La‐14) and Bifidobacterium breve (JCM 1192), and no significant differences were found between these two nuts[59]
Apple pectin (Malus domestica)Bifidobacterium, Lactobacillus, and Streptococcus (including Enterococcus) in feces; ↓ C. perfringens, enterobacteria and Pseudomonas; ↑ Fecal concentrations of SFCA[60]

Table 1.

In vitro studies on the prebiotic potential of foods.

Advertisement

4. Use of in vivo models to study the prebiotic potential of foods

It has been well established that the colon microbiota has a deep influence on health. The study of the prebiotic potential in humans would be considered as a gold standard in case of absence of ethical and practical limitations, which may make the research unreliable or limited, in addition to the high dropout rates of study participants. Thus, animal models become an alternative to study the prebiotic potential of foods, since they allow direct access to intestinal contents as well as to organs and tissues [61].

Usually, the animal models used for the study of gut microbiota are swine [62], zebrafish [63], and more widely in rodents such as rats [47], hamsters [64], and mice [53], especially when the potential prebiotic of foods is evaluated.

Teixeira et al. [47] confirmed the prebiotic potential of Amazonian tubers by adding them to the diet of Wistar rats for 28 days, evaluating the pH and microbiota present in feces collected from the animals’ caecum. Samal et al. [65] evaluated the prebiotic potential of Jerusalem artichoke (Helianthus tuberosus L.) added at different concentrations to the diet of rats for 12 weeks and observed that the consumption promoted beneficial effects on immunity, intestinal morphometry, and hindgut fermentation of rats. Supplementation with 2.5% of insoluble fibers from pineapple peel decreased the daily production of fecal ammonia, shortened gastrointestinal transit time, and increased the total amounts of SCFA in the caecal content as well as the growth of gut microflora such as Lactobacillus spp. and Bifidobacterium spp. in hamsters [64].

Not only should the gut microbiota be evaluated in in vivo studies but also other variables such as pH, feces humidity, and SCFAs production, which is directly related to the selective bacterial fermentation of prebiotics [66, 67]. In the large intestine, 95% of SCFA produced are rapidly absorbed by colonocytes, whereas the remaining 5% are expelled in the feces [68]. These microbial metabolites can be used as sources of energy by the host and can also act as regulators of energy consumption and metabolism [69]. pH acidification can also be an indicator of fermentation of prebiotic components of foods in the colon by endogenous bacteria and production of organic acids directly responsible for this process [70, 71]. In addition, the preservation of the intestinal epithelium in healthy rats or its recovery in diseased rats may provide evidence of the prebiotic potential, as observed by Hu et al. [72] and Moura et al. [73].

Bränning et al. [74] evaluated the potential prebiotic of blueberry husks added in diet as a substitute for digestible starch. The consumption of diet containing blueberry husk by rats for 5 days resulted in higher amounts of propionic acid and butyric acid in the distal colon and feces, respectively, when compared to rats that were fed a control diet without fibers. Both acids are essential substrates for colonic epithelial cells, improving gut health, and a surplus of substrates which also have metabolic effects. However, blueberry husk has antimicrobial effects, as observed by the decreased counts of lactobacilli, bifidobacteria, and enterobacteriaceae, and the larger pool of succinic acid may be a consequence of these antimicrobial effects. In this model, blueberry husks do not demonstrate prebiotic properties.

Rodríguez‐Cabezas et al. [39] evaluated the synergistic effect of two dietary fibers with different fermentation patterns, FOS (Beneo Ò‐95) and RS (FibersolÒ‐2), administrated to healthy rats or in trinitrobenzenesulphonic acid (TNBS) colitic rats. Treatment groups (n = 20) received FOS (2 g/rat/day), RS (2 g/rat/day), or the mixture of both (37.5 FOS and 62.5% RS) (2 g/rat/day) incorporated in drinking water during 2 weeks. In healthy rats, the administration of the combination of FOS and RS induced changes in the intestinal microbiota and increased lactobacilli and bifidobacteria in caecum and colonic contents. In addition, treatment increased the moisture content and decreased the pH of caecum and colon. Furthermore, its administration upregulated the expression of trefoil factor‐3 and mucin 2 (MUC‐2) in comparison with untreated rats, thus improving the intestinal barrier function and increasing the propionate, butyrate, and total SCFA colonic contents. The beneficial effects observed with this combination were confirmed in the healthy or colitis rats.

Young adult male rats were fed ad libitum with purified control diet (CONT) containing 5% w/w cellulose (insoluble fiber) or diet containing 10% w/w cellulose (CELL), FOS, oat beta‐glucan (GLUC), or apple pectin (PECT) for 4 weeks. Comparing CONT and CELL, caecal concentrations of fermentation products increased from 1.4 to 2.2 times in GLUC, FOS, and PECT, and colonic concentrations increased from 1.9 to 2.5 times in GLUC and FOS; however, no consistent changes in SCFA receptor gene expression were detected. The main fermentation products detected were acetate, propionate, butyrate, and succinate, and the differences in amounts of fermentation products among soluble fibers may reflect different fermentation patterns and/or different fermentation rates and turnover. This research concluded that the presence of soluble fermentable fiber appears to be more important than its source [75].

Other studies evaluating the prebiotic potential of foods using in vivo models are described in Table 2.

Study modelFoodsMain resultsReferences
Female rats WistarCocoa fibers (Theobroma cacao L.)↑Bifidobacterium and Lactobacillus; ↑ SCFA production and ↓ cecal and fecal pH[76]
Male golden Syrian hamstersPineapple peel (Ananas comosus L. Merr.)Modulation of the activities of fecal bacterial enzymes; ↓ ammonia contents in caecum and feces; ↑ concentration in the caecum of SCFA[64]
Male rats WistarPassion fruit peel (Passiflora edulis)Positive effect on SCFA production, but no change in gut microbiota was observed[77]
Male rats WistarFOS and PC of strawberry (Fragaria ananassa)↓ Cecal pH and ↓ production of putrefactive SCFA (sum of isobutyric, isovaleric, and valeric acids)[78]
Male guinea pigsFOS of Yacon (Smallanthus sonchifolius Poepp. & Endl)↑ Cecal SFCA concentration[79]
Male BALB/c miceGOS of Chinese roots (Deshipu stachyose granules)Growth of beneficial intestinal bacteria (Lactobacilli and Bifidobacteria) and inhibition of pathogenic bacteria (Clostridium perfringens)[80]
Effects on intestinal peristalsis promotion and bowel function improvement (constipation treatment)

Table 2.

In vivo models for prebiotic food assessment.

Advertisement

5. Prebiotics and other beneficial effects on health

The modulations of the intestinal microbiota and SCFA production are associated with many beneficial effects about the ingestion of prebiotics and isolated or added to foods, such as regulation of various physiological processes (e.g., inflammation) and metabolic processes (e.g., lipid and glucose metabolism), thus contributing to the treatment or prevention of chronic non‐degenerative diseases [38].

Rats treated with prebiotics had a reduction of plasma pro‐inflammatory cytokines, reduction of hepatic inflammatory expression, and oxidative stress markers [81]. Everard et al. [82] showed that diet enriched with prebiotics led to an improvement in glucose tolerance, increase in amount of L‐cells, and associated parameters (expression of intestinal pro‐glucagon mRNA and plasma glucagon‐like peptide‐1 levels or GLP‐1) in addition to reduction in body fat accumulation, oxidative stress, and level of inflammation in obese rats.

Salazar et al. [69] supplemented 15 obese women with a mixture of inulin and oligofructose for 3 months and observed that prebiotics had a bifidogenic effect, but the elimination of SCFA in feces did not show a significant correlation with the serum concentration of lipids.

A prospective longitudinal cohort study with 1592 workers with metabolic syndrome found that there was an inverse association between consumption of insoluble fibers and increase in systolic and diastolic blood pressure, total cholesterol (TC), triglycerides (TG), apolipoprotein B100, and TG/high‐density lipoprotein (HDL) ratio; however, the ingestion of soluble fibers was inversely associated only with triglycerides and apolipoprotein B100. Thus, the prevalence of metabolic syndrome was lower in participants who ingested larger amounts of insoluble fibers [83]. In contrast, a meta‐analysis by Wu et al. [84] that included 18 cohort studies with 672,408 participants confirmed that dietary intake of soluble or insoluble fibers (especially from cereals and fruits) has a similar inverse effect associated with the risk of coronary heart disease.

Barbalho et al. [85] reported that the supplementation of passion fruit peels to healthy Wistar rats contributed to the elevation of HDL levels and the decrease in glycemia, TG, and TC levels of these animals compared to the control group. Such results would be associated with the soluble dietary fiber present in passion fruit peels, such as mucilage and pectins, which form a viscous gel that retains water and reduce the sensation of hunger, body weight, plasma levels of TC, TG, and low‐density lipoprotein (LDL) and increase the excretion of cholesterol and bile salts in feces and HDL levels.

Obese rats fed with hyperlipid diet and diet added of lyophilized jabuticaba peel (rich in anthocyanins) exhibited increased HDL and improved insulin resistance, suggesting that the diet added of this byproduct may have a protective effect against cardiovascular diseases by increasing HDL levels [86].

Amaya‐Cruz et al. [87] evaluated the effect of dietary fibers and polyphenols from guava (Pisidium guajava), peach (Prunus persica), and mango (Mangifera indica) byproducts on obesity‐related hyperglycemia and hepatic steatosis in Wistar rats. Mango and peach byproducts presented better soluble/insoluble fiber ratio and high amount of polyphenols, which may have attenuated the development of hepatic steatosis and hyperglycemia in rats. In guava byproducts, they found great amount of soluble dietary fibers and condensed tannins, which may be related to the greater anti‐obesogenic effect on animals, when compared to control rats and to those treated with other byproducts.

Changes in the intestinal microbiota may also influence the homeostasis of the immune [35], renal [88], and nervous systems [89], as well as the development and progression of pathophysiological processes such as hypertension [90] and colorectal cancer [91]. A mixture of non‐digestible GOS ingested by mice for 3 weeks prior to induction of inflammatory neuropathology and anxiety improved anxiety and inflammation through decreased expression of IL‐1b cytokine and 5‐HT2AR serotonin receptor in the frontal cortex compared to the control group [92]. Healthy men and women daily supplied with FOS or GOS for 3 weeks showed decreased response to cortisol awakening, protecting against the risk of depression [93]. Rats with chronic kidney disease (CKD) fed for 3 weeks with RS diets had a delay in CKD progression and increased creatinine clearance when compared to CKD mice that received amylopectin [94].

Advertisement

6. Innovations in food processing with added prebiotics

The inclusion of prebiotics in industrialized foods has become a viable and healthy alternative, since there is a great demand of consumers for functional foods that can help in maintaining health. Moreover, the food industry can obtain numerous advantages from the addition of prebiotics in food products, such as improvement of sensory characteristics, better balance of the nutritional composition, and longer shelf‐life [67]. In general, prebiotics are added to bakery products, breakfast cereals, beverages (e.g., fruit juices, coffee, cocoa, and tea), dairy products, table spreads, butter‐based products, and desserts (ice cream, puddings, jellies, and chocolates) [67, 95]. Prebiotics also have gelling properties (e.g., inulin), which maintain the emulsion stability, provide spreadable texture, and water retention (e.g., inulin and FOS), thus allowing the development of processed foods with low fat content, with pleasant taste and texture [67, 96].

However, some important characteristics of the manufacturing process, such as low pH, high temperatures, and conditions favoring the Maillard reaction must be taken into account when choosing the prebiotic to be added to foods in order to avoid the formation of anti‐nutritional compounds detrimental to the sensory quality of the final product and consumer health as well as the partial or total reduction of their action. Among prebiotics commonly used in the food industry, GOS are more stable at high temperatures and low pH mainly due to the beta bonds of their structure, which provide greater hydrolysis stability compared to FOS and inulin [96]. A type of RS known as RS3 can be added to fried battered products to increase the content of dietary fibers and avoid reducing moisture and the absorption of fats, since RS3 is very resistant to frying temperatures [97].

Advertisement

7. Concluding remarks

The importance of the consumption of prebiotics is unquestionable and they should be part of healthy diet. Prebiotics exert various technological functions in food and many health benefits not only related to the modulation of the intestinal microbiota but also to other beneficial physiological actions in various organs and systems of healthy or diseased men/animals. In this sense, the development of foods added due to prebiotics by the industry can be advantageous due to the demand and profitability of this market, as well as for consumers who will have healthy foods available that can be readily consumed for the prevention or treatment of diseases, thus reducing public health costs. However, there is no consensus on the recommended quantity of specific prebiotics for consumption in the diet, and this limitation is a major challenge regarding the different in vitro and in vivo models used to test the prebiotic potential of foods.

Both in vivo and in vitro models have helped advances of researches aimed at evaluating the prebiotic potential of foods through the composition and metabolism of the intestinal microbiota and their interactions. However, it is noteworthy that there are no ideal models, and the most adequate are those based on the study objectives and using association of complementary techniques.

References

  1. 1. Goetzke B, Nitzko S, Spiller A. Consumption of organic and functional food. A matter of well‐being and health? Appetite. 2014;77:96-105
  2. 2. Bachl T. Wellness trend benefits markets. In: Consumers’ Choice ‘07. Berlin: BVE; 2007. pp. 9-12
  3. 3. Chrysochou P. Food health branding: The role of marketing mix elements and public discourse in conveying a healthy brand image. Journal of Marketing Communications. 2010;16(1-2):69-85
  4. 4. Food Quality and Standards Service, Food and Agriculture Organization. FAO Technical Meeting on Prebiotics. Food Quality and Standards Service (AGNIS), Food and Agriculture Organization of the United Nations; 2007
  5. 5. Sreenivas KM, Lele SS. Prebiotic activity of gourd family vegetable fibres using in vitro fermentation. Food Bioscience. 2013;1:26-30
  6. 6. Gómez B, Gullón B, Yáñez R, Schols H, Alonso JL. Prebiotic potential of pectins and pectic oligosaccharides derived from lemon peel wastes and sugar beet pulp: A comparative evaluation. Journal of Functional Foods [Internet]. 2016;20:108-121. DOI: 10.1016/j.jff.2015.10.029
  7. 7. Scott KP, Martin JC, Duncan SH, Flint HJ. Prebiotic stimulation of human colonic butyrate‐producing bacteria and bifidobacteria, in vitro. FEMS Microbiology Ecology. 2014;87(1):30-40
  8. 8. Donelli G, Vuotto C, Mastromarino P. Phenotyping and genotyping are both essential to identify and classify a probiotic microorganism. Microbial Ecology in Health and Disease [Internet]. 2013;24:1-8. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3758930&tool=pmcentrez&rendertype=abstract
  9. 9. Joint FAO/WHO Working Group Report. Guidelines for the Evaluation of Probiotics in Food. London, Ontario, Canada: World Health Organization, Food and Agriculture Organization; 2002
  10. 10. Butel MJ. Probiotics, gut microbiota and health. Médecine et Maladies Infectieuses [Internet]. 2014;44(1):1-8. DOI: 10.1016/j.medmal.2013.10.002
  11. 11. He Z, Wang X, Li G, Zhao Y, Zhang J, Niu C, et al. Antioxidant activity of prebiotic ginseng polysaccharides combined with potential probiotic Lactobacillus plantarum C88. International Journal of Food Science & Technology. 2015;50:1673-1682
  12. 12. Rodrigues FC, Castro ASB, Rodrigues VC, Fernandes SA, Fontes EAF, de Oliveira TT, et al. Yacon flour and Bifidobacterium longum modulate bone health in rats. Journal of Medicinal Food [Internet]. 2012;15(7):664-670. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22510044
  13. 13. Estévez BG, Gullón P, Tavaria F, Yáñez R. Assessment of the prebiotic effect of quinoa and amaranth in the human intestinal ecosystem. Food & Function [Internet]. 2016;7:3782-3788. Available from: http://pubs.rsc.org/en/Content/ArticleLanding/2016/FO/C6FO00924G
  14. 14. Maurer AD, Eller LK, Hallam MC, Taylor K, Reimer RA. Consumption of diets high in prebiotic fiber or protein during growth influences the response to a high fat and sucrose diet in adulthood in rats. Nutrition and Metabolism (London). 2010;7:77
  15. 15. Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH. The influence of diet on the gut microbiota. Pharmacological Research [Internet]. 2013;69(1):52-60. DOI: 10.1016/j.phrs.2012.10.020
  16. 16. Flint HJ, Duncan SH, Scott KP, Louis P. Links between diet, gut microbiota composition and gut metabolism. Proceedings of the Nutrition Society [Internet]. 2015;74(1):13-22. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25268552
  17. 17. Gibson GR, Probert HM, Van Loo J, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutrition Research Reviews. 2004;17(2):259-275
  18. 18. Marques TM, Cryan JF, Shanahan F, Fitzgerald GF, Ross RP, Dinan TG, et al. Gut microbiota modulation and implications for host health: Dietary strategies to influence the gut‐brain axis. Innovative Food Science and Emerging Technologies. 2014;22:239-247
  19. 19. Ufnal M, Pham K. The gut‐blood barrier permeability—a new marker in cardiovascular and metabolic diseases? Medical Hypotheses [Internet]. 2017;98:35-37. DOI: 10.1016/j.mehy.2016.11.012
  20. 20. Principi N, Esposito S. Gut microbiota and central nervous system development. Journal of Infection [Internet]. 2016;73(6):536-546. DOI: 10.1016/j.jinf.2016.09.010
  21. 21. Luna RA, Foster JA. Gut brain axis: Diet microbiota interactions and implications for modulation of anxiety and depression. Current Opinion in Biotechnology [Internet]. 2015;32:35-41. DOI: 10.1016/j.copbio.2014.10.007
  22. 22. Kabouridis PS, Pachnis V. Emerging roles of gut microbiota and the immune system in the development of the enteric nervous system. Journal of Clinical Investigation. 2015;125(3):956-964
  23. 23. Sandrini S, Aldriwesh M, Alruways M, Freestone P. Microbial endocrinology: Host‐bacteria communication within the gut microbiome. Journal of Endocrinology [Internet]. 2015;225(2):R21‐R34. Available from: http://joe.endocrinology‐journals.org/cgi/doi/10.1530/JOE‐14-0615
  24. 24. Candela M, Maccaferri S, Turroni S, Carnevali P, Brigidi P. Functional intestinal microbiome, new frontiers in prebiotic design. International Journal of Food Microbiology [Internet]. 2010;140(2-3):93-101. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0168160510002485
  25. 25. American Dietetic Association. Position of the American Dietetic Association: Health implications of dietary fiber. Journal of the American Dietetic Association [Internet]. 2008;108(10):1716-1731. Available from: http://www.sciencedirect.com/science/article/pii/S0002822308015666
  26. 26. Cupersmid L, Fraga APR, de Abreu ES, Pereira IRO. Linhaça: Composição química e efeitos biológicos. e‐Scientia. 2012;5(2):33-40
  27. 27. Porfírio E, Henrique VSM, de Abreu Reis MJ. Elaboração de farofa de grãos, sementes oleaginosas e castanha de caju: composição de fi bras, ácidos graxos e aceitação [Development of a “farofa”(*) containing grains, oilseeds and cashew nut: Fatty acid & fibre composition and acceptance]. Brazilian Journal of Food Technology. 2014;17(3):185
  28. 28. Ayala‐Zavala JF, Vega‐Vega V, Rosas‐Domínguez C, Palafox‐Carlos H, Villa‐Rodriguez JA, Siddiqui MW, et al. Agro‐industrial potential of exotic fruit byproducts as a source of food additives. Food Research International [Internet]. 2011;44(7):1866-1874. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0963996911001086
  29. 29. Kosmala M, Zduńczyk Z, Jus¨kiewicz J, Jurgoński A, Karlińska E, Macierzyński J, et al. Chemical composition of defatted strawberry and raspberry seeds and the effect of these dietary ingredients on polyphenol metabolites, intestinal function, and selected serum parameters in rats. Journal of Agricultural and Food Chemistry. 2015;63(11):2989-2996
  30. 30. Roberfroid MB, Delzenne NM. Dietary fructans. Annual Review of Nutrition. 1998;18(1):117-143
  31. 31. Patel S, Goyal A. The current trends and future perspectives of prebiotics research: A review. 3 Biotech. 2012;2(2):115-125
  32. 32. Ganapathy V, Thangaraju M, Prasad PD, Martin PM, Singh N. Transporters and receptors for short‐chain fatty acids as the molecular link between colonic bacteria and the host. Current Opinion in Pharmacology. 2013;13(6):869-874
  33. 33. Parkar SG, Trower TM, Stevenson DE. Fecal microbial metabolism of polyphenols and its effects on human gut microbiota. Anaerobe [Internet]. 2013;23:12-19. DOI: 10.1016/j.anaerobe.2013.07.009
  34. 34. da Silva JK, Cazarin CBB, Colomeu TC, Batista ÂG, Meletti LMM, Paschoal JAR, et al. Antioxidant activity of aqueous extract of passion fruit (Passiflora edulis) leaves: In vitro and in vivo study. Food Research International. 2013;53(2):882-890
  35. 35. Valdés L, Cuervo A, Salazar N, Ruas‐Madiedo P, Gueimonde M, González S. The relationship between phenolic compounds from diet and microbiota: Impact on human health. Food &Function [Internet]. 2015;6:2424-2439. Available from: www.rsc.org/foodfunction
  36. 36. Gullon B, Pintado ME, Fernández‐López J, Pérez‐Álvarez JA, Viuda‐Martos M. In vitro gastrointestinal digestion of pomegranate peel (Punica granatum) flour obtained from co‐products: Changes in the antioxidant potential and bioactive compounds stability. Journal of Functional Foods [Internet]. 2015;19:617-628. DOI: 10.1016/j.jff.2015.09.056
  37. 37. Quiñones M, Miguel M, Aleixandre A. Beneficial effects of polyphenols on cardiovascular disease. Pharmacological Research. 2013;68(1):125-131
  38. 38. Slavin J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients [Internet]. 2013;5(4):1417-1435. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3705355&tool=pmcentrez&rendertype=abstract
  39. 39. Rodríguez‐Cabezas ME, Camuesco D, Arribas B, Garrido‐Mesa N, Comalada M, Bailón E, et al. The combination of fructooligosaccharides and resistant starch shows prebiotic additive effects in rats. Clinical Nutrition [Internet]. 2010;29(6):832-839. DOI: 10.1016/j.clnu.2010.05.005
  40. 40. Savignac HM, Corona G, Mills H, Chen L, Spencer JPE, Tzortzis G, et al. Prebiotic feeding elevates central brain derived neurotrophic factor, N‐methyl‐d‐aspartate receptor subunits and d‐serine. Neurochemistry International [Internet]. 2013;63(8):756-764. DOI: 10.1016/j.neuint.2013.10.006
  41. 41. Pyra KA, Saha DC, Reimer RA. Prebiotic fiber increases hepatic acetyl CoA carboxylase phosphorylation and suppresses glucose‐dependent insulinotropic polypeptide secretion more effectively when used with metformin in obese rats. Journal of Nutrition. 2012;142(2):213-220
  42. 42. Freitas KDC, Amancio OMS, Morais MB. High‐performance inulin and oligofructose prebiotics increase the intestinal absorption of iron in rats with iron deficiency anaemia during the growth phase. British Journal of Nutrition [Internet]. 2012;108(6):1008-1016. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22172251
  43. 43. Gobinath D, Madhu AN, Prashant G, Srinivasan K, Prapulla SG. Beneficial effect of xylo‐oligosaccharides and fructo‐oligosaccharides in streptozotocin‐induced diabetic rats. British Journal of Nutrition. 2010;104(1):40-47
  44. 44. Macfarlane GT, Gibson GR, Cummings JH. Comparison of fermentation reactions in different regions of the human colon. Journal of Applied Bacteriology. 1992;72(1):57-64
  45. 45. Moter A, Göbel UB. Fluorescence in situ hybridization (FISH) for direct visualization of microorganisms. Journal of Microbiological Methods. 2000;41(2):85-112
  46. 46. Olson DW, Aryana KJ. Effect of prebiotics on Lactobacillus acidophilus growth and resulting pH changes in skim milk and a model peptone system. Journal of Microbial & Biochemical Technology. 2012;4:121-125
  47. 47. Teixeira LS, Martim SR, Silva LSC, Kinupp VF, Teixeira MFS, Porto ALF. Efficiency of Amazonian tubers flours in modulating gut microbiota of male rats. Innovative Food Science and Emerging Technologies. 2016;38:1-6
  48. 48. Tuohy KM, Probert HM, Smejkal CW, Gibson GR. Using probiotics and prebiotics to improve gut health. Drug Discovery Today. 2003;8(15):692-700
  49. 49. Gómez B, Gullón B, Remoroza C, Schols HA, Parajó JC, Alonso JL. Purification, characterization, and prebiotic properties of pectic oligosaccharides from orange peel wastes. Journal of Agricultural and Food Chemistry. 2014;62(40):9769-9782
  50. 50. Duarte FND, Rodrigues JB, da Costa Lima M, Lima MD, Pacheco MT, Pintado MM, de Souza Aquino J, de Souza EL. Potential prebiotic properties of cashew apple (Anacardium occidentale L.) agro‐industrial byproduct on Lactobacillus species. Journal of Organizational Behavior. 2017;31:1-8
  51. 51. Thuaytong W, Anprung P. Bioactive compounds and prebiotic activity in Thailand‐grown red and white guava fruit (Psidium guajava L.). Food Science and Technology International. 2011;17(3):205-212
  52. 52. Sousa S, Pinto J, Pereira C, Malcata FX, Pacheco MTB, Gomes AM, et al. In vitro evaluation of yacon (Smallanthus sonchifolius) tuber flour prebiotic potential. Food and Bioproducts Processing. 2015;95:96-105
  53. 53. Condezo‐Hoyos L, Mohanty IP, Noratto GD. Assessing non‐digestible compounds in apple cultivars and their potential as modulators of obese faecal microbiota in vitro. Food Chemistry. 2014;161:208-215
  54. 54. Gullon P, Gullon B, Tavaria F, Vasconcelos M, Gomes AM. In vitro fermentation of lupin seeds (Lupinus albus) and broad beans (Vicia faba): Dynamic modulation of the intestinal microbiota and metabolomic output. Food & Function [Internet]. 2015;6(10):3316-3322. Available from: http://www.ncbi.nlm.nih.gov/pubmed/26252418
  55. 55. Oliveira DL, Costabile A, Wilbey RA, Grandison AS, Duarte LC, Roseiro LB. In vitro evaluation of the fermentation properties and potential prebiotic activity of caprine cheese whey oligosaccharides in batch culture systems. BioFactors. 2012;38(6):440-449
  56. 56. Wichienchot S, Jatupornpipat M, Rastall RA. Oligosaccharides of pitaya (dragon fruit) flesh and their prebiotic properties. Food Chemistry [Internet]. 2010;120(3):850-857. DOI: 10.1016/j.foodchem.2009.11.026
  57. 57. Gullon B, Pintado ME, Barber X, Fernández‐López J, Pérez‐Álvarez JA, Viuda‐Martos M. Bioaccessibility, changes in the antioxidant potential and colonic fermentation of date pits and apple bagasse flours obtained from co‐products during simulated in vitro gastrointestinal digestion. Food Research International [Internet]. 2015;78:169-176. DOI: 10.1016/j.foodres.2015.10.021
  58. 58. Immerzeel P, Falck P, Galbe M, Adlercreutz P, Nordberg Karlsson E, Stålbrand H. Extraction of water‐soluble xylan from wheat bran and utilization of enzymatically produced xylooligosaccharides by Lactobacillus, Bifidobacterium and Weissella spp. LWT—Food Science and Technology [Internet]. 2014;56(2):321-327. DOI: 10.1016/j.lwt.2013.12.013
  59. 59. Liu Z, Wang W, Huang G, Zhang W, Ni L. In vitro and in vivo evaluation of the prebiotic effect of raw and roasted almonds (Prunus amygdalus). Journal of the Science of Food and Agriculture. 2016;96(5):1836-1843
  60. 60. Shinohara K, Ohashi Y, Kawasumi K, Terada A, Fujisawa T. Anaerobe effect of apple intake on fecal microbiota and metabolites in humans. Anaerobe [Internet]. 2010;16(5):510-515. DOI: 10.1016/j.anaerobe.2010.03.005
  61. 61. Macfarlane GT, Macfarlane S. Models for intestinal fermentation: Association between food components, delivery systems, bioavailability and functional interactions in the gut. Current Opinion in Biotechnology. 2007;18(2):156-162
  62. 62. Han KS, Balan P, Molist Gasa F, Boland M. Green kiwifruit modulates the colonic microbiota in growing pigs. Letters in Applied Microbiology. 2011;52(4):379-385
  63. 63. Cheesman SE, Neal JT, Mittge E, Seredick BM, Guillemin K. Epithelial cell proliferation in the developing zebrafish intestine is regulated by the Wnt pathway and microbial signaling via Myd88. Proceedings of the National Academy of Sciences. 2011;108(Suppl. 1):4570-4577
  64. 64. Huang YL, Tsai YH, Chow CJ. Water‐insoluble fiber‐rich fraction from pineapple peel improves intestinal function in hamsters: Evidence from cecal and fecal indicators. Nutrition Research [Internet]. 2014;34(4):346-354. DOI: 10.1016/j.nutres.2014.03.001
  65. 65. Samal L, Chaturvedi VB, Saikumar G, Somvanshi R, Pattanaik AK. Prebiotic potential of Jerusalem artichoke (Helianthus tuberosus L.) in Wistar rats: Effects of levels of supplementation on hindgut fermentation, intestinal morphology, blood metabolites and immune response. Journal of the Science of Food and Agriculture. 2015;95(8):1689-1696
  66. 66. Yasmin A, Butt MS, Afzaal M, van Baak M, Nadeem MT, Shahid MZ. Prebiotics, gut microbiota and metabolic risks : Unveiling the relationship. Journal of Functional Foods [Internet]. 2015;17:189-201. DOI: 10.1016/j.jff.2015.05.004
  67. 67. Al‐Sheraji SH, Ismail A, Manap MY, Mustafa S, Yusof RM, Hassan FA. Prebiotics as functional foods: A review. Journal of Functional Foods [Internet]. 2013;5(4):1542-1553. Available from: http://linkinghub.elsevier.com/retrieve/pii/S175646461300193X
  68. 68. den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud D‐J, Bakker BM. The role of short‐chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. Journal of Lipid Research [Internet]. 2013;54(9):2325-2340. Available from: http://www.jlr.org/cgi/doi/10.1194/jlr.R036012
  69. 69. Salazar N, Dewulf EM, Neyrinck AM, Bindels LB, Cani PD, Mahillon J, et al. Inulin‐type fructans modulate intestinal Bifidobacterium species populations and decrease fecal short‐chain fatty acids in obese women. Clinical Nutrition [Internet]. 2015;34(3):501-517. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0261561414001599
  70. 70. McOrist AL, Miller RB, Bird AR, Keogh JB, Noakes M, Topping DL, et al. Fecal butyrate levels vary widely among individuals but are usually increased by a diet high in resistant starch. Journal of Nutrition. 2011;141(5):883-889
  71. 71. Paturi G, Butts CA, Monro JA, Hedderley D, Stoklosinski H, Roy NC, et al. Evaluation of gastrointestinal transit in rats fed dietary fibres differing in their susceptibility to large intestine fermentation. Journal of Functional Foods [Internet]. 2012;4(1):107-115. DOI: 10.1016/j.jff.2011.08.005
  72. 72. Hu Y, Le Leu RK, Christophersen CT, Somashekar R, Conlon MA, Meng XQ, et al. Manipulation of the gut microbiota using resistant starch is associated with protection against colitis‐associated colorectal cancer in rats. Carcinogenesis. 2016;37(4):366-375
  73. 73. de Moura NA, Caetano BFR, Sivieri K, Urbano LH, Cabello C, Rodrigues MAM, et al. Protective effects of yacon (Smallanthus sonchifolius) intake on experimental colon carcinogenesis. Food and Chemical Toxicology [Internet]. 2012;50(8):2902-2910. DOI: 10.1016/j.fct.2012.05.006
  74. 74. Bränning C, Håkansson A, Ahrné S, Jeppsson B, Molin G, Nyman M. Blueberry husks and multi‐strain probiotics affect colonic fermentation in rats. British Journal of Nutrition. 2009;101(6):859-870
  75. 75. Adam CL, Williams PA, Dalby MJ, Garden K, Thomson LM, Richardson AJ, et al. Different types of soluble fermentable dietary fibre decrease food intake, body weight gain and adiposity in young adult male rats. Nutrition and Metabolism (London) [Internet]. 2014;11(1):36. Available from: http://nutritionandmetabolism.biomedcentral.com/articles/10.1186/1743‐7075‐11-36
  76. 76. Massot‐Cladera M, Costabile A, Childs CE, Yaqoob P, Franch À, Castell M, et al. Prebiotic effects of cocoa fibre on rats. Journal of Functional Foods [Internet]. 2015;19:341-352. DOI: 10.1016/j.jff.2015.09.021
  77. 77. da Silva JK, Cazarin CBB, Bogusz Junior S, Augusto F, Maróstica Junior MR. Passion fruit (Passiflora edulis) peel increases colonic production of short‐chain fatty acids in Wistar rats. LWT—Food Science and Technology [Internet]. 2014;59(2):1252-1257. Available from: http://www.sciencedirect.com/science/article/pii/S0023643814003193
  78. 78. Fotschki B, Juśkiewicz J, Jurgoński A, Kołodziejczyk K, Milala J, Kosmala M, et al. Anthocyanins in strawberry polyphenolic extract enhance the beneficial effects of diets with fructooligosaccharides in the rat cecal environment. PLoS One. 2016;11(2):1-18
  79. 79. Campos D, Betalleluz‐Pallardel I, Chirinos R, Aguilar‐Galvez A, Noratto G, Pedreschi R. Prebiotic effects of yacon (Smallanthus sonchifolius Poepp. & Endl), a source of fructooligosaccharides and phenolic compounds with antioxidant activity. Food Chemistry [Internet]. 2012;135(3):1592-1599. DOI: 10.1016/j.foodchem.2012.05.088
  80. 80. Li T, Lu X, Yang X. Stachyose‐enriched α‐galacto‐oligosaccharides regulate gut microbiota and relieve constipation in mice. Journal of Agricultural and Food Chemistry. 2013;61(48):11825-11831
  81. 81. Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, et al. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP‐2‐driven improvement of gut permeability. Gut [Internet]. 2009;58(8):1091-1103. Available from: http://gut.bmj.com/cgi/doi/10.1136/gut.2008.165886
  82. 82. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GG, Neyrinck AM, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet‐induced leptin‐resistant mice. Diabetes [Internet]. 2011;60(11):2775-2786. Available from: http://diabetes.diabetesjournals.org/cgi/doi/10.2337/db11-0227
  83. 83. Franco BM, Latre ML, Esteban EMA, Ordovás JM, Casasnovas JA, Peñalvo JL. Soluble and insoluble dietary fibre intake and risk factors for metabolic syndrome and cardiovascular disease in middle‐aged adults: The AWHS Cohort. Nutrición Hospitalaria. 2014;30(6):1279-1288
  84. 84. Wu Y, Qian Y, Pan Y, Li P, Yang J, Ye X, et al. Association between dietary fiber intake and risk of coronary heart disease: A meta‐analysis. Clinical Nutrition. 2015;34(4):603-611
  85. 85. Barbalho SM, da Silva Soares de Souza M, de Paula e Silva JC, Mendes CG, de Oliveira GA, Costa T, et al. Yellow passion fruit rind (Passiflora edulis): An industrial waste or an adjuvant in the maintenance of glycemia and prevention of dyslipidemia? Journal of Diabetes Research and Clinical Metabolism. 2012;1(1):5
  86. 86. Lenquiste SA, Batista AG, Marineli RDS, Dragano NRV, Maróstica MR. Freeze‐dried jaboticaba peel added to high‐fat diet increases HDL‐cholesterol and improves insulin resistance in obese rats. Food Research International. 2012;49(1):153-160
  87. 87. Amaya‐Cruz DM, Rodríguez‐González S, Pérez‐Ramírez IF, Loarca‐Piña G, Amaya‐Llano S, Gallegos‐Corona MA, et al. Juice by‐products as a source of dietary fibre and antioxidants and their effect on hepatic steatosis. Journal of Functional Foods [Internet]. 2015;17:93-102. Available from: http://linkinghub.elsevier.com/retrieve/pii/S1756464615002340
  88. 88. Wanchai K, Pongchaidecha A, Chatsudthipong V, Chattipakorn SC, Chattipakorn N, Lungkaphin A. Role of gastrointestinal microbiota on kidney injury and the obese condition. American Journal of the Medical Sciences [Internet]. 2017;353(1):59-69. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28104104;http://linkinghub.elsevier.com/retrieve/pii/S0002962916306292
  89. 89. Oriach CS, Robertson RC, Stanton C, Cryan JF, Dinan TG. Food for thought: The role of nutrition in the microbiota‐gut‐brain axis. Clinical Nutrition Experimental [Internet]. 2016;6:25-38. DOI: 10.1016/j.yclnex.2016.01.003
  90. 90. Jose PA, Raj D. Gut microbiota in hypertension. Current Opinion in Nephrology and Hypertension. 2015;24(5):403-409
  91. 91. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nature Reviews Microbiology [Internet]. 2014;12(10):661-672. DOI: 10.1038/nrmicro3344
  92. 92. Savignac HM, Couch Y, Stratford M, Bannerman DM, Tzortzis G, Anthony DC, et al. Prebiotic administration normalizes lipopolysaccharide (LPS)‐induced anxiety and cortical 5‐HT2A receptor and IL1‐β levels in male mice. Brain, Behavior, and Immunity [Internet]. 2016;52:120-131. DOI: 10.1016/j.bbi.2015.10.007
  93. 93. Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PWJ. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology (Berlin). 2015;232(10):1793-1801
  94. 94. Vaziri ND, Liu SM, Lau WL, Khazaeli M, Nazertehrani S, Farzaneh SH, et al. High amylose resistant starch diet ameliorates oxidative stress, inflammation, and progression of chronic kidney disease. PLoS One. 2014;9(12):1-15
  95. 95. de Sousa C, dos Santos EF, Sgarbieri VC. The importance of prebiotics in functional foods and clinical practice. Food and Nutrition Sciences [Internet]. 2011;2(2):133-144. Available from: http://www.scirp.org/journal/PaperInformation.aspx?PaperID=4536&#abstract
  96. 96. Charalampopoulos D, Rastall RA. Prebiotics in foods. Current Opinion in Biotechnology [Internet]. 2012;23(2):187-191. DOI: 10.1016/j.copbio.2011.12.028
  97. 97. Homayouni A, Amini A, Keshtiban AK, Mortazavian AM, Esazadeh K, Pourmoradian S. Resistant starch in food industry: A changing outlook for consumer and producer. Starch/Staerke. 2014;66(1-2):102-114

Written By

Jailane de Souza Aquino, Kamila Sabino Batista, Francisca Nayara Dantas Duarte Menezes, Priscilla Paulo Lins, Jessyca Alencar de Sousa Gomes and Laiane Alves da Silva

Submitted: 08 November 2016 Reviewed: 12 April 2017 Published: 02 August 2017