close
Foods-high-in-inulin

What is Inulin

Inulin is a versatile carbohydrate with many diverse applications in foods and pharma 1. Inulin, depending on its chain length, is classified as either an oligosaccharide or polysaccharide and it belongs to the fructan carbohydrate subgroup. It is composed of β-d-fructosyl subgroups linked together by (2→1) glycosidic bonds and the molecule usually ends with a (1↔2) bonded α-d-glucosyl group 2. The length of these fructose chains varies and ranges from 2 to 60 monomers. Inulin containing maximally 10 fructose units is also referred to as oligofructose 3.

In food, iInulin has been used as a (low calorie) sweetener, to form gels, to increase viscosity, to improve organoleptic properties, and as a non-digestible fiber 4. Mostly inulin is used as a sugar (sweet-replacer) and longer chain inulin is used mostly as a fat replacer in dairy products and as a prebiotic 5 and and texture modifier 2. Examples of use in dairy are application in cheese, milk, yogurt and ice cream 5. Some examples of use of inulin in non-dairy food are use in bread, biscuits, cereal and meat products 6. Both inulin and oligofructose are used as dietary fiber and prebiotics in functional foods. Its longer chain length makes inulin more useful pharmaceutically than oligofructose.

Applications of inulin as pharmaceutical excipient are even more diverse and range from stabilization of protein-based pharmaceuticals 7, through solid dispersions to increase dissolution rate 8, to targeted colon delivery 9. Chemically modified inulin was also applied in controlled delivery to the colon. The mechanism behind colon targeting with inulin is based on the fact that inulin is only significantly hydrolyzed by inulinases produced by bifidobacteria in the colon and not by the digestive enzymes in the upper parts of the gastro intestinal tract 10. This means that gels and coatings of inulin are not hydrolyzed until these reach the colon where they are fermented, resulting in a colon-specific release of incorporated drug. Inulin is also used pharmaceutically, most notably as a diagnostic agent for the determination of kidney function 11. Inulin is injected intravenously, after which it is excreted renally. As inulin is not naturally present in the body and it is not metabolized in circulation, the amount of inulin secreted in the urine provides information on kidney function. Less widespread is the use of inulin for industrial and chemical purposes. Stevens, Meriggi, and Booten 12 reviewed the derivatization of inulin and applications of these chemically modified inulins for a wide range of applications, from inhibiting calcium carbonate crystallization industrially to use in hair gel.

Foods-high-in-inulin

Some examples of plants containing large quantities of inulin are Jerusalem artichoke, chicory root, garlic, asparagus root, salisfy and dandelion root 13. More commonly consumed vegetables and fruits containing inulin are onion, leek, garlic, banana, wheat, rye and barley. Daily intakes have been estimated to range from 1 to 10 g per day in the Western diet 14. The average American diet contains between 1.3 and 3.5 g of inulin per day, with an average of 2.6 g 14. The European consumption of inulin appears to be substantially higher at 3–11 g per day, which is below reported tolerances of at least 10–20 g per day 15. Inulin has also been used safely in infant nutrition 16. This has led to the American Food and Drug Administration (FDA) to issuing a Generally Recognized As Safe (GRAS) notification for inulin in 1992 17.

Inulin is predominately isolated from chicory root. The isolation process basically consists of three steps: (1) extraction of water-soluble components, including inulins, from chicory root (2) purification to remove impurities and optionally low degree of polymerization inulins and (3) finally spray drying. Inulin extracted from chicory root contains up to 10% of sugars (mono-, di- and small oligosaccharides) 14. Degree of polymerization is the number of repeat units in an oligomer or polymer chain.  Typically, extraction is done by boiling the cleaned and cut or ground up roots in water. Process conditions such as pH of the water, water–root ratio, boiling time, etc., may vary.

Apart from extraction from plants, inulin can also be produced enzymatically. Inulosucrase type fructosyltransferase can synthesize inulin from sucrose by catalyzing both transglycosylation and hydrolysis of sucrose 18. Several procedures to do so have been described, these mostly involve enzymes derived from bacteria. Enzymes from Bacillus species 217C–11have been used to produce inulin on a large scale 19 and Escherichia coli and Streptococcus mutans derived fructosyltransferase can produce very high molecular weight inulins 20. Both these studies reported remarkably low polydispersity (around 1.1) of the produced inulin. Inulin producing fructosyltransferases from several Lactobacillus strains have also been characterized 21. Inulosucrase from Leuconostoc citreum CW 28 was shown to produce different molecular weight inulin when it was cell associated compared to when it was free in solution. The cell associated enzyme predominately produced inulin with a molecular weight between 1.35–1.60 × 106 Da and the free enzyme produced more inulin with a molecular weight between 2600 and 3400 Da 22.

Inulin soluble fiber

The 5 basic attributes of a dietary fiber are 23:

  1. components of edible plant cell;
  2. carbohydrates (both oligosaccharides and polysaccharides);
  3. resistance to hydrolysis by human (mammal) alimentary enzymes;
  4. resistance to absorption in the small intestine; hydrolysis and
  5. fermentation (partial or total) by the bacteria in the large bowel.

Inulin-type fructans are plant carbohydrates that, because of the β–(2←1) configuration of the fructosyl-fructose glycosidic linkages, resist digestion in the upper gastrointestinal tract but are quantitatively fermented in the colon. They are thus undoubtedly part of the dietary fiber complex, and they must be labeled as dietary fiber on consumer food products.

However, because of their specific fermentative properties, inulin-type fructans do have characteristic features different from those of other dietary fibers.

Therefore, they may contribute in a significant way to a well-balanced diet by increasing the fiber content, by improving the diversity of the fiber sources, and by specifically affecting several gastrointestinal functions (composition of intestinal microflora, mucosal functions, endocrine activities, mineral absorption) and even systemic functions (especially lipid homeostasis and immune functions) as well as by reducing the risk of miscellaneous diseases. These effects are summarized in Table 1.

Table 1. Nutritional effects and potential health benefits of inulin-type fructans

Enhanced functions
    Composition and activities of the gut microflora
    Stool production
    Absorption of Ca and other minerals
    Production of gastrointestinal endocrine peptides
    Immunity and resistance to infections
    Lipid homeostasis
Reduction of disease risks
    Intestinal infections
    Irritable bowel diseases
    Colon cancer
    Osteoporosis
    Obesity
[Source 24]

The gastrointestinal functions are primary endpoints that benefit most from inulin-type fructans. One of the most promising effects is modulation of activities of the colon, an organ of the gastrointestinal tract that is recognized more and more as playing a variety of key roles in maintaining health and well-being as well as reducing the risk of diseases 25, 26, 27.

The concept of “colonic health” has thus emerged as a major target for functional food development in the area of enhanced function claims.

In addition to its important physiological and immunological functions, the colon is also involved in miscellaneous diseases from acute infections and diarrhea or constipation to chronic diseases such as inflammatory bowel diseases, irritable bowel syndrome, or cancer 25. Through modulation of the colonic functions, inulin-type fructans thus also have the potential to reduce the risk of some diseases.

Prebiotics are non-digestible selectively fermented dietary fibers that specifically promote the growth of one or more bacterial genera in the gastrointestinal tract and thus provide health benefit to the host. Prebiotics are usually dietary carbohydrates. The two major carbohydrates that fulfill the criteria for prebiotics are inulin-type fructans and the galacto-oligosaccharides, although many other classes are under investigation. Prebiotics are a promising dietary strategy by which the gastro-intestinal microbiota can be modified for health promotion.

The key characteristics of a prebiotic are that they are as follows 28:

  1. Non-digestible by endogenous enzymes in the human gut;
  2. Selectively fermented by specific genera/species of resident gut microbiota; and
  3. That this results in a targeted increase in specific bacteria that confers health benefits to the host.

These health benefits are diverse and can include immune modulation through increased intestinal-specific immunoglobulins and immuno-regulatory interleukins, and a reduction in pro-inflammatory interleukins. Health benefits also include the production of short-chain fatty acids—acetate, propionate, butyrate—and lactate that reduce luminal pH, which in turn, may be important in preventing colonization with acid-sensitive enteropathogens. Acetate production also contributes, through cross-feeding, to the production of butyrate, which is a primary substrate for colonocytes thus contributing to epithelial integrity 28.

Table 2. Prebiotics, the major bacterial groups that degrade them and the fermentation products

PrebioticsSourcesBacterial enzyme required to hydrolyse prebioticMajor bacterial groups with specificity for this prebiotic and their fermentation products
Inulin type fructansβ-fructofuranosidase (fructanase)Bifidobacteria (acetate, lactate); Lactobacilli (lactate); Bacteroides (acetate, propionate)
InulinChicory (inulin)
OligofructoseEnzymatic hydrolysis of inulin (oligofructose)
Fructo-oligosaccharidesEnzymatic synthesis from sucrose (FOS)
β-Galacto-oligosaccharidesEnzymatic trans-galactosylation of lactoseβ-GalactosidaseBifidobacteria (acetate, lactate)
[Source 29]

Prebiotics are selectively fermented food ingredients that promote specific changes in the composition and/or activity of bacteria already present within the gastro-intestinal tract, thus promoting host health and well-being 30. Bacterial fermentation of prebiotics results in production of short chain fatty acids, lactic acid, gases (hydrogen, methane, and carbon dioxide) and reduced luminal pH 31. Short chain fatty acids and particularly butyrate benefit host health by regulating fluid and electrolyte uptake, influencing epithelial cell cytokinetics and barrier function, and exerting anti-inflammatory effects 32, 33. Inulin and fructooligosaccharides were shown to promote the growth of bifidobacteria in infants and adults 34. Suggested health benefits of bifidobacteria include production of acetic and lactic acids, synthesizing B vitamins, excreting antimicrobial substances that reduce pathogenic bacteria, and influencing maturation of the immune system 35, 36. However, uncertainties in this field of research warrant further study.

Inulin and oligofructose are the most studied and well-established prebiotics. As previously mentioned, they escape digestion in the upper gastrointestinal tract and reach the large intestine virtually intact, where they are quantitatively fermented and act as prebiotics. In the studies 37, 38, 39 that investigated the effects of inulin and oligofructose on the human gut microbiota, a selective stimulation of growth of the beneficial flora, namely bifidobacteria, to a lesser extent lactobacilli, and possibly other species such as the Clostridium coccoides-Eubacterium rectale cluster known to be butyrate producers has been reported 40, 41.

Prebiotics in gastrointestinal disorders

A shift from a stable intestinal environment occurs when the gut microbiota community is temporarily or permanently altered and is termed “dysbiosis.” Factors that may lead to dysbiosis include antibiotics, diet, host immune system, inflammation, and infectious gastroenteritis 42. Dysbiosis is seen in numerous gastrointestinal disorders including irritable bowel syndrome, and Crohn’s disease, and may play a key role in their pathogenesis and possibly in management. Some of the common features of dysbiosis in IBS and Crohn’s disease are reduced microbial diversity, lower bifidobacteria, lower bacteroides to firmicutes ratio in irritable bowel syndrome and in Crohn’s disease, and decreased Faecalibacterium prausnitzii.

Prebiotics in irritable bowel syndrome

Irritable bowel syndrome is a chronic functional bowel disorder whose etiology is not entirely understood, although a role for the gut microbiota exists, including previous gastroenteritis, dysbiosis, alterations in colonic gas production, and low-grade inflammation have all been identified in some people with irritable bowel syndrome.

Luminal bifidobacteria are negatively associated with pain in both healthy controls and irritable bowel syndrome 43, 44 and bifidobacteria have been found to be lower in irritable bowel syndrome compared with healthy controls 28. Therefore, as a therapeutic target, specific prebiotic-stimulated growth of bifidobacteria is promising. However, to date, there have been few randomized control trials investigating the effect of prebiotics on IBS. Two studies in adults with IBS at doses of 6 g/d of oligofructose and 20 g/d of inulin showed no improvement in symptom or stool output measures. Another trial showed an improvement in composite symptom score with 5 g/d of short-chain fructo-oligosaccharides in the per-protocol population, but this was not analyzed intention to treat, with a high non-compliance rate and only 50/105 being included in the per protocol analysis 45. One single-center 12-week parallel crossover trial, which used a β-galactooligosaccharides, showed a dose-dependent stimulation of bifidobacteria at 3.5 and 7.0 g/d. Symptom relief measured as a global assessment was significantly improved compared with placebo in both groups receiving the prebiotic, but the lower dose resulted in lower symptoms scores flatulence, bloating, and stool consistency when individual symptoms were assessed and clustered 46. Therefore, the type and dose of prebiotics are likely to be important when considering prebiotic use in IBS.

Prebiotics in Crohn’s disease

Crohn’s disease is an inflammatory bowel disease caused by an inappropriate mucosal inflammatory response against commensal gut microbiota in genetically susceptibility individuals 45.

Animal studies demonstrate the effectiveness of inulin-type fructan prebiotics in treating Crohn’s disease via beneficial modulation of the gut microbiota 45, but the evidence in humans is less convincing. A pilot study of 10 patients showed promise using 15 g/d oligofructose/inulin for 3 weeks in active Crohn’s disease leading to increased bifidobacteria and reduction in disease activity (Harvey Bradshaw Index) compared with baseline 47. Two large randomized controlled trials using 15 g/d oligofructose/inulin in 103 patients with active Crohn’s disease 48 and 20 g/d oligofructose/inulin in 67 patients with mild/moderately active or inactive disease 49 did not confirm this finding. Furthermore, the prebiotic group in the first study had greater severity of symptoms and, in the second study, had a trend toward greater withdrawal rates due to symptoms 48, 49. Therefore, high-dose inulin-type fructans prebiotics may actually worsen symptoms in active Crohn’s disease, perhaps explaining why they also consume lower amounts of naturally occurring fructans and rarely use prebiotic supplements 50.

Contrasting approaches of prebiotics and the low FODMAP diet

While using prebiotics to enhance the growth of bifidobacteria seems logical to reduce symptoms of IBS, studies are limited by the type and dose of prebiotic used. On the other hand, there is growing evidence for the mechanisms and efficacy of the low FODMAP diet (diet low in Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols) for treating functional gastrointestinal symptoms in IBS 51 and possibly in inflammatory bowel disease 52, 53. Inulin, oligofructose, and galactooligosaccharides are all restricted in this diet. Two randomised controlled trials provide clinical evidence of the efficacy of the low FODMAP diet in the management of symptoms in IBS 54, 55. However, bifidobacteria were reduced in both studies (significantly so in the first), and stool pH was higher after the low FODMAP diet in the second study. These alterations in the gut microbiota may be of concern if raised colonic pH enables enteropathogenic colonization. Use of probiotics or prebiotics with narrow specificity for non-gas forming bifidobacteria concurrently with the low FODMAP diet may be able to modulate gut microbiota so long as this does not significantly reduce the efficacy of symptom improvement in IBS. There are limited studies of the use of the low FODMAP diet in managing functional gastrointestinal symptoms in Crohn’s disease, and only one has investigated the effect on the gut microbiota 56.

For Constipation

The composition of the symbiotic colonic microflora is a key player in maintaining the colon (and thus the whole body) health. That composition is largely determined by the flora that establishes at and immediately after birth, is mostly “individual,” can be modulated by specific compounds in the diet, and may change during the lifetime, becoming more and more complex as we age 57. Numerous studies have demonstrated that supplementation with inulin-type fructans increases the growth of bifidobacteria in healthy humans 28.

Inulin is widely used as a dietary fiber and prebiotic in so-called functional foods. Nutrition can be used to provide health benefits by modification of gut microbial flora 58. Inulin’s effect on gut flora and gut mobility has been linked to a variety of beneficial effects, both local and systemic 59. Inulin, in particular high molecular weight inulin, increased stool frequency and can thus be used against constipation 60. It has been shown to improve stool frequency in formula fed newborns, creating a gut microbiota closer to that associated with breastfeeding 61. Inulin was also able to relieve constipation in elderly patients, indicating that the stool promoting effect of inulin is present for all ages 62.

For Inflammatory bowel disease and colon cancer

Oral administration of inulin has been reported to achieve both local and systemic immune modulation. The immune-modulating effects of inulin-type fructans and galactooligosaccharides prebiotics have been investigated in humans. In healthy humans, inulin-type fructans modulate numerous gut immune markers in multiple studies 63. Faecal IgA is increased, Peyer’s patches express greater IL-10 and IFN-γ, and activity of immune cells in the spleen is elevated. A review of the supporting evidence concluded that the local immunomodulatory effect is apparent, but systemic effects are less substantiated 64. The local effect is possibly indirect through the prebiotic action of inulin, stimulating growth of beneficial bacteria in the gut 65. In a later in vitro study, it was found that inulin also possesses direct signaling capacity on human immune cells, mainly through the toll-like receptor 2 66. It is conceivable that these local effects could be beneficial to patients suffering from inflammatory bowel disease and irritable bowel syndrome 67. Thus far, investigations with small numbers of patients have reported mixed results in this regard and there is thus a need for further investigation of this potential application of inulin 68.

Inulin was able to reduce chemically induced pre-neoplastic lesions or tumors in the colon of mice and rats 69. Long chain inulins were more potent than smaller chain inulins in this sense. This reduction was associated with the gut flora-mediated fermentation of inulin 69. In a recent study in rats, inulin showed to have a bigger prophylactic potential to colon carcinogenesis compared to lactulose 70. Inulin reduced cytotoxicity and genotoxicity in vitro in human colon adenocarcinoma cells 71. Again these results seem promising, but further clinical research is required to see if these results can be reproduced in vivo in humans. Boutron-Ruault et al conducted an open multi-center pilot study of colon adenoma and adenoma-free subjects. All participants received 5 g inulin twice daily for three months; 74 subjects completed the study. Cell proliferation at the rectal crypts was not significantly modified by inulin ingestion in either group 72.

For Metabolic Syndrome

Metabolic syndrome is a complex disorder characterized by overweight/obesity, hypertension, and disturbances of lipid and carbohydrate metabolism 73. Each component of metabolic syndrome is a known risk factor for the development of type 2 diabetes, atherosclerosis, and coronary artery disease (coronary heart disease). Since, obesity is a precursor for metabolic syndrome, treating obesity with physical activities (exercises), behavioral modifications (counseling), calorie-restricted diets, weight-losing drugs, and finally with weight losing surgery will be the crucial factors in the management and control of metabolic syndrome.

Various dietary strategies based on probiotic and prebiotic interventions have been proposed by several investigators after establishing strong relationship between diet, gut microbiota, and pathophysiology of metabolic syndrome. The scientific data reveal that the gut microbiota is one of the important environmental factors co-evolved with the host since birth and maintains dynamic interactions with host throughout the life. The metabolic role of the gut microbiota is also essential for the biochemical activities of the human body, resulting in salvage of energy, generation of absorbable compounds, and production of vitamins and other essential nutrients 74.

The gut microbiota also regulates many aspects of innate and acquired immunity, protecting the host from pathogen invasion and chronic inflammation 75, 76. Recently, investigators related the imbalances in gut microbiota with susceptibility to infections, immune-based disorders and more importantly with obesity and insulin resistance 77, 78. These studies provided strong scientific evidence for using probiotics and prebiotics in formulation of dietary strategies in the management of metabolic syndrome 79, 80.

Probiotics are defined as live micro-organisms with Gebnerally regarded as safe (GRAS) status, which when administered in adequate amounts confer a health benefit on the host 81. The two key members of this group include lactobacilli and bifidobacteria. Prebiotics, on the other hand, are defined as specific indigestible substances such as inulin, oligofructose/galactose complex which selectively support the growth of probiotic bacteria and possibly other microorganisms in the intestine.

The putative role of prebiotics in the management of metabolic disorders has also been studied by different investigators. The scientific literature documents several favorable putative effects of prebiotics on food intake, body weight, glucose homeostasis, plasma lipid profile, and associated risk factors for cardiovascular disease 82. To support this hypothesis several mechanisms have been proposed: First, the modulation of gut flora microbiota supporting beneficial organisms 83. Second, induce enteroendocrine L cell proliferation and modulate gut peptide production and secretion (i.e., glucagon-like peptide-1 [GLP-1], peptide-YY, and ghrelin) 78, 82, 84. Third, modulate inflammation in obese individuals 76.

Recent studies have demonstrated that the distributions of free fatty acid receptor 2 (FFA2) positive, glucagon-like peptide-1 (GLP-1) containing enteroendocrine L-cells in human and rats were almost consistent. The supplementation of fructo-oligosaccharides (FOS) increased the densities of FFA2 – positive enteroendocrine L-cells over control in rats 84. This involves the fermentation of prebiotics by selective bacterial strains that increases the production of short-chain fatty acids in the gut lumen 85. These short-chain fatty acids stimulate intestinal proglucagon (precursor for GLP-1) mRNA expression and peptide-YY (PYY) secretion in rats 86, 87. The short chain fatty acids have recently been demonstrated to act as ligands for several G-protein-coupled receptors: FFA2 and FFA3 of enteroendocrine L cells and increases both the free fatty acid receptor 2-positive enteroendocrine L-cells proliferation with free fatty acid receptor 2 activation, which might be an important trigger to produce and release GLP-1 and PYY by enteroendocrine cells in the lower intestine 88. The released glucagon-like peptide-1 and peptide-YY cross the blood–brain barrier and associated with neural activation in areas of the hypothalamus and prefrontal cortex that are involved in the regulation of feeding behavior 89.

Recent data suggest the role of other important gut peptide, glucagon-like peptide-2 (GLP-2) in the regulation of gut permeability which could affect the plasma levels of microbial components that increase the inflammatory tone. The increased endogenous glucagon-like peptide-2 production was associated with improved mucosal barrier function via the restoration of tight junction protein expression and distribution. The role of glucagon-like peptide-2 in the protective effects of prebiotics was established recently 78. Pharmacological inhibition of GLP-2 signaling receptor abolished the effects of prebiotics, thus, established a direct link between the GLP-2 and gut permeability78. Hence, without a functional GLP-2 receptor, the prebiotic treatment failed to reduce metabolic endotoxaemia, hepatic inflammation, and oxidative stress markers. Collectively, these data support the concept that specific changes in the gut microbiota improve gut permeability and inflammatory tone via a GLP-2-dependent mechanism. Amongst the potential mechanisms involved, the gut microbial compositional change by prebiotics controls and increases endogenous production of the intestinotrophic proglucagon derived peptide GLP-2 not only in the colon but also in the jejunum and consequently improves gut barrier functions 78. In addition, short chain fatty acid from prebiotic fermentation down regulates inflammatory markers of insulin resistance by modulating gut hormone secretion was also studied 90, 91.

Agave inulin

Epidemiologic evidence suggests there are inverse associations between dietary fiber intake and obesity 92, diabetes 93, and coronary heart disease 94. Inadequate fiber consumption is a recognized problem in the United States 95, with average intakes barely surpassing 50% of the Adequate Intake recommendation (25–38 g/d) 96. Because inadequate fiber intake is also associated with increased risk of obesity, diabetes, and cardiovascular disease 97, 30, the role of fiber in GI microbial metabolism, function, and disease prevention is of particular interest.

Agave inulin, is composed of linear and branched fructose chains, connected with β-2,1 and β-2,6 linkages, and a degree of polymerization between 25 and 34 98. In comparison, chicory inulin is linear with β-2,1 linkages and a degree of polymerization that ranges from 2 to 60 99. In vitro experimentation has demonstrated that agave inulin is readily fermented by bifidobacteria and lactobacilli 100. In addition, rodent studies have provided evidence that the botanical origin and chemical structure of different inulin-type fibers (e.g., agave inulin and chicory inulin) induce variable effects on body composition, blood cholesterol, and blood glucose concentrations 101, 102.

The prebiotic effects of agave inulin supplementation in healthy adults, shifted the gastrointestinal microbiota composition and activity in healthy adults. However, further investigation is warranted to determine whether the observed changes translate into health benefits in human populations 103. Additional research is needed to define the relationship between the consumption of different prebiotics and improvement of human health.

Inulin side effects

The primary side effects of inulin-type prebiotics are gastrointestinal and can include osmotic diarrhea, abdominal rumbling, bloating, cramping, and excessive flatulence. These side effects are similar to the effects produced by lactose in people with lactose mal-digestion. Because of the configuration of the bond between fructose monomers, inulin-type prebiotics are not broken down by intestinal enzymes, which is the probable cause of osmotic diarrhea. Although daily doses of 40-50 g can cause an osmotic effect, doses greater than 50 g would be expected to produce osmotic diarrhea in a large percent of the population 104.

Based on available research, it appears inulin HP is the best prebiotic to reduce the likelihood of gastrointestinal side effects. Inulin HP is used to describe the exclusively long-chain, high-molecular weight mixes of inulin-type fructans (fructans with a DP <10 physically removed). Fructo-oligosaccharides and oligofructose are considered the forms most likely to produce side effects.

While osmotic diarrhea is typically expected only with high doses, in one study abdominal distension was reported at daily doses of 10.6 g fructooligosaccharides 105.

Although doses greater than 40 g/day are generally necessary to produce abdominal rumbling and bloating, and doses greater than 50 g/day to cause abdominal cramping 104, bloating has been reported with daily doses as low as 2.5-5 g 106 and abdominal pain has been reported at doses as low as 10 g daily 107.

Excessive flatulence is the most well-established side effect and can occur at daily doses as low as 1-2 g in sensitive individuals 104.

These data demonstrate that doses up to 7.5 g per day of agave inulin led to minimal GI upset, do not increase diarrhea, and improve laxation in healthy young adults 108. There were slight increases in bloating, flatulence, and rumbling frequency with agave inulin. Abdominal pain and rumbling intensity were marginally greater with 7.5 g. Bloating and flatulence intensity increased with 5.0 g and 7.5 g. Number of bowel movements per day increased, stools were softer, and stool dry matter percentage was lower with 7.5 g.

Inulin-type prebiotics contain free sugars (fructose, glucose, and sucrose) unless removed by additional processing. The potential contribution of free sugar content of inulin-type prebiotics to the incidence of abdominal side effects has not been explored.

Dosing Prebiotics

For the promotion of healthy bacterial flora, the usual recommendation for supplementation of inulin-type prebiotics is a daily dose of 2.5-10 g.

2.5-5 g daily is the low end for bifidogenic effects, which are typically dose-dependent. Bouhnik et al reported the optimal fructooligosaccharides dose for producing a bifidogenic effect, while remaining relatively well tolerated, is 10 g fructooligosaccharides daily 109.

In studies of inulin-type prebiotics, two supplementation approaches have been used to minimize gastrointestinal side effects: dose in two or more divided doses and start with a lower dose and increase after a week or more of supplementation. One or both strategies were used in many of the studies and are potential approaches for lessening the likelihood of side effects. In subjects who complain of side effects, several options exist. One option is to reduce the dose. A second option is to switch to a product consisting of either a lower percent of short-chain polymers or entirely of long-chain polymers (i.e., inulin or inulin HP instead of fructooligosaccharides or oligofructose). Inulin HP is used to describe the exclusively long-chain, high-molecular weight mixes of inulin-type fructans (fructans with a DP <10 physically removed). The galactose-based prebiotics/inulin HP mixture was designed to more closely mimic the oligosaccharide portfolios found in human breast milk than inulin-type prebiotics alone, since the oligosaccharides in breast milk contain relatively high amounts of galactose polymers 110. Studies in preterm and term infants have shown a formula supplemented with this galactose-based prebiotics/inulin HP prebiotic mixture results in an intestinal microbiota similar to that found in breast-fed infants 111, 112. In patients who complain of abdominal side effects it would be useful to inquire whether they are consuming foods or beverages that have added inulin-type prebiotics as ingredients. Since abdominal side effects tend to be dose dependent, dietary sources would be expected to contribute in an additive way to those prescribed as a supplement.

Summary

For non-infant nutrition applications, inulin-type prebiotics have been investigated for a range of clinical uses, with the majority falling into four categories:

  • Blood sugar regulation
  • Blood lipids
  • Gastrointestinal health
  • Mineral absorption and biomarkers of bone health

Because of a bifidogenic effect, the uses of inulin-type prebiotics for improvement of gastrointestinal function and as a potential therapy in gastrointestinal clinical conditions have been investigated. Several studies have investigated bowel transit time, stool consistency, and stool frequency. Existing evidence supports an effect in infants but not in adults without pre-existing functional problems. In subjects with existing constipation, one uncontrolled trial found supplementation with inulin improved stool frequency. In infants, inulin-type prebiotics as a stand-alone intervention do not appear to prevent diarrhea. Three studies report some degree of functional change subsequent to supplementation in persons with active inflammatory bowel disease. These studies were mixed with respect to the active intervention reducing disease activity compared to placebo. No trials have been conducted to determine whether chronic supplementation might help prevent disease reoccurrence or sustain periods of clinical remission. The two studies on IBS also had mixed findings.

A variety of studies have assessed biomarkers of blood sugar regulation in normo- and hyperglycemic subjects. To date, no form of inulin-type prebiotic has shown a consistent beneficial effect.

Lipids have been investigated in a variety of studies. In persons with normal lipid levels, the most consistent observation is inulin-type prebiotics have no statistically significant effect on lipid levels. In individuals with elevated lipids the results have been mixed, although the preponderance of studies report no benefit from supplementation. Positive results were reported in one trial of inulin. While definitive conclusions should wait for further research clarification, current evidence suggests that inulin, but not FOS or oligofructose, might positively influence lipids in hyperlipidemic individuals.

Mineral absorption and biomarkers of bone health have received a significant degree of research attention. Current evidence suggests inulin-type prebiotics do not have a significant impact on absorption of iron, selenium, and zinc, but might have a positive effect on copper and magnesium in some population subsets.

There appears to be significant individual variability in response to inulin-type prebiotics and effects on calcium absorption. Adolescents and perimenopausal women have responded positively more consistently than young adults. However, even in these population subsets, some individuals respond better than others. Baseline calcium absorption and possibly genetics appear to influence response. The best choice for prebiotic supplementation to positively impact calcium absorption and bone health biomarkers appears to be oligo-fructose-enriched inulin HP, which has produced the most consistent results.

Based on available research, it appears inulin HP is the best prebiotic to reduce the likelihood of gastrointestinal side effects. Fructo-oligosaccharides and oligofructose are considered the forms most likely to produce side effects.

References
  1. Inulin, a flexible oligosaccharide. II: Review of its pharmaceutical applications. Carbohydrate Polymers Volume 134, 10 December 2015, Pages 418-428. http://www.sciencedirect.com/science/article/pii/S0144861715007638
  2. Inulin-type prebiotics – A review: Part 1. Alternative Medicine Review, 13 (2008), pp. 315-329.
  3. Inulin and oligofructose as dietary fiber: A review of the evidence. Critical Reviews in Food Science and Nutrition, 41 (2001), pp. 353-362.
  4. Inulin, a flexible oligosaccharide I: Review of its physicochemical characteristics. Carbohydrate Polymers Volume 130, 5 October 2015, Pages 405-419. http://www.sciencedirect.com/science/article/pii/S0144861715004257
  5. Inulin as texture modifier in dairy products. Food Hydrocolloids, 25 (2011), pp. 1881-1890.
  6. Inulin in food products: Prebiotic and functional ingredient. British Food Journal, 117 (2015), pp. 371-387.
  7. Inulin glasses for the stabilization of therapeutic proteins. International Journal of Pharmaceutics, 215 (2001), pp. 163-174.
  8. Inulin solid dispersion technology to improve the absorption of the BCS Class IV drug TMC240. European Journal of Pharmaceutics and Biopharmaceutics, 74 (2010), pp. 233-238.
  9. Application and use of Inulin as a tool for therapeutic drug delivery. Biotechnology & Genetic Engineering Reviews, 28 (2012), pp. 33-45.
  10. Characterization of methacrylated inulin hydrogels designed for colon targeting: In vitro release of BSA. Pharmaceutical Research, 20 (2) (2003), pp. 303-307.
  11. Determination of inulin clearance by bolus intravenous injection in healthy subjects and ascitic patients: Equivalence of systemic and renal clearances as glomerular filtration markers. British Journal of Clinical Pharmacology, 46 (1998), pp. 605-609.
  12. Chemical modification of inulin, a valuable renewable resource, and its industrial applications. Biomacromolecules, 2 (2001), pp. 1-16.
  13. Applications of inulin and oligofructose in health and nutrition. Journal of Biosciences, 27 (2002), pp. 703-714.
  14. Nutritional and health benefits of inulin and oligofructose inulin and oligofructose: Safe intakes and legal status 1. The Journal of Nutrition, 129 (1999), pp. 1412-1417.
  15. Gastrointestinal tolerance of chicory inulin products. Journal of the American Dietetic Association, 110 (2010), pp. 865-868.
  16. Safety and efficacy of inulin and oligofructose supplementation in infant formula: Results from a randomized clinical trial. Clinical Nutrition, 32 (2013), pp. 918-927.
  17. Generally recognised as safe (GRAS) notification for Frutafit. U.S. Food and Drug Administration, Silver Spring, MD (2002).
  18. The levansucrase and inulosucrase enzymes of Lactobacillus reuteri 121 catalyse processive and non-processive transglycosylation reactions. Microbiology, 152 (2006), pp. 1187-1196.
  19. Physicochemical characterization and biological effects of inulin enzymatically synthesized from sucrose. Journal of Agricultural and Food Chemistry, 53 (2005), pp. 1246-1253.
  20. Structure of the enzymatically synthesized fructan inulin. Carbohydrate Research, 313 (1998), pp. 165-174.
  21. Inulin and levan synthesis by probiotic Lactobacillus gasseri strains: Characterization of three novel fructansucrase enzymes and their fructan products. Microbiology, 156 (2010), pp. 1264-1274.
  22. Biochemical properties of inulosucrase from Leuconostoc citreum CW28 used for inulin synthesis. Biocatalysis and Biotransformation, 22 (2004), pp. 275-281.
  23. Roberfroid M. Dietary fiber, inulin, and oligofructose: a review comparing their physiological effects. CRC Crit Rev Food Sci Nutr. 1993;33:103–48.
  24. Inulin-Type Fructans: Functional Food Ingredients. J. Nutr. November 2007 vol. 137 no. 11 2493S-2502S. http://jn.nutrition.org/content/137/11/2493S.full
  25. Cummings JH. The large intestine in nutrition and disease. Danone Chair Monograph. Brussels: Institut Danone; 1997.
  26. Berg RD. The indigenous gastrointestinal microflora. Trends Microbiol. 1996;4:430–5. https://www.ncbi.nlm.nih.gov/pubmed/8950812
  27. Rowland IR. Interactions of the gut microflora and the host in toxicology. Toxicol Pathol. 1988;16:147–53. https://www.ncbi.nlm.nih.gov/pubmed/3055224
  28. Roberfroid M, Gibson GR, Hoyles L et al. Prebiotic effects: metabolic and health benefits. Br. J. Nutr. 2010; 104: S1–S63. https://www.ncbi.nlm.nih.gov/pubmed/20920376
  29. Wilson, B., and Whelan, K. (2017) Prebiotic inulin-type fructans and galacto-oligosaccharides: definition, specificity, function, and application in gastrointestinal disorders. Journal of Gastroenterology and Hepatology, 32: 64–68. http://onlinelibrary.wiley.com/doi/10.1111/jgh.13700/full
  30. Gibson GR, Scott KP, Rastall RA, Tuohy KM, Hotchkiss A, Dubert-Ferrandon A, Gareau M, Murphy EF, Saulnier D, Loh G. Dietary prebiotics: current status and new definition. Food Sci Technol Bull Funct Foods. 2010;7:1–19.
  31. Cummings JH. Short chain fatty acids in the human colon. Gut 1981;22:763–79. http://gut.bmj.com/content/gutjnl/22/9/763.full.pdf
  32. Berni Canani R, Di Costanzo M, Leone L. The epigenetic effects of butyrate: potential therapeutic implications for clinical practice. Clin Epigenetics 2012;4:4. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3312834/
  33. Schaafsma G, Slavin JL. Significance of inulin fructans in the human diet. Compr Rev Food Sci Food Safety. 2015;14:37–47.
  34. Brownawell AM, Caers W, Gibson GR, Kendall CW, Lewis KD, Ringel Y, Slavin JL. Prebiotics and the health benefits of fiber: current regulatory status, future research, and goals. J Nutr 2012;142:962–74. http://jn.nutrition.org/content/142/5/962
  35. Belenguer A, Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ. Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol 2006;72:3593–9. http://aem.asm.org/content/72/5/3593
  36. Ventura M, Turroni F, Bottacini F, Giubellini V, van Sinderen D. Bifidobacterial ecology and comparative genomics: perspectives. Bifidobacteria Genomics Molbi Aspects 2010:31.
  37. Harmsen HJM, Raangs GC, Franks AH, Wildeboer-Veloo CM, Welling GW. The effect of the prebiotic inulin and the probiotic Bifidobacterium longum on the fecal microflora of healthy volunteers measured by FISH and DGGE. Microb Ecol Health Dis. 2002;14:211–9.
  38. Williams CH, Witherly SA, Buddington RK. Influence of dietary Neosugar on selected bacteria groups of the human fecal microbiota. Microb Ecol Health Dis. 1994;7:91–7.
  39. Menne E, Guggenbuhl N, Roberfroid M. Fn-type chicory inulin hydrolysate has a prebiotic effect in humans. J Nutr. 2000;130:1197–9.
  40. Kleessen B, Hartmann L, Blaut M. Oligofructose and long-chain inulin influence the gut microbial ecology of rats associated with a human faecal flora. Br J Nutr. 2001;86:291–300. https://www.ncbi.nlm.nih.gov/pubmed/11502244
  41. Apajalahti JHA, Kettunen H, Kettunen A, Holben WE, Nurminen PH, Rautanen N, Mutanen M. Culture-independent microbial community analysis reveals that inulin in the diet primarily affects previously unknown bacteria in the mouse cecum. Appl Environ Microbiol. 2002;68:4986–95. http://aem.asm.org/content/68/10/4986
  42. Bennet SM, Ohman L, Simren M. Gut microbiota as potential orchestrators of irritable bowel syndrome. Gut Lliver. 2015; 9: 318–331.
  43. Rajilic-Stojanovic M, Biagi E, Heilig HG et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011; 141: 1792–1801. https://www.ncbi.nlm.nih.gov/pubmed/21820992
  44. Jalanka-Tuovinen J, Salonen A, Nikkila J et al. Intestinal microbiota in healthy adults: temporal analysis reveals individual and common core and relation to intestinal symptoms. PLoS One 2011; 6: e23035. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3145776/
  45. Whelan K. Mechanisms and effectiveness of prebiotics in modifying the gastrointestinal microbiota for the management of digestive disorders. Proc. Nutr. Soc. 2013; 72: 288–298. https://www.ncbi.nlm.nih.gov/pubmed/23680358
  46. Silk DBA, Davis A, Vulevic J, Tzortzis G, Gibson GR. Clinical trial: The effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Aliment. Pharmacol. Ther. 2009; 29: 508–518. https://www.ncbi.nlm.nih.gov/pubmed/19053980
  47. Lindsay JO, Whelan K, Stagg AJ et al. Clinical, microbiological, and immunological effects of fructo-oligosaccharide in patients with Crohn’s disease. Gut 2006; 55: 348–355. www.ncbi.nlm.nih.gov/pubmed/16162680
  48. Benjamin JL, Hedin CR, Koutsoumpas A et al. Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s disease. Gut 2011; 60: 923–929. https://www.ncbi.nlm.nih.gov/pubmed/21262918
  49. Joossens M, De Preter V, Ballet V, Verbeke K, Rutgeerts P, Vermeire S. Effect of oligofructose-enriched inulin (OF-IN) on bacterial composition and disease activity of patients with Crohn’s disease: results from a double-blinded randomised controlled trial. Gut 2012; 61: 958. https://www.ncbi.nlm.nih.gov/pubmed/21749983
  50. Hedin CR, Mullard M, Sharratt E et al. Probiotic and prebiotic use in patients with inflammatory bowel disease: a case–control study. Inflamm. Bowel Dis. 2010; 16: 2099–2108. https://www.ncbi.nlm.nih.gov/pubmed/20848469
  51. Staudacher HM, Irving PM, Lomer MC, Whelan K. Mechanisms and efficacy of dietary FODMAP restriction in IBS. Nat. Rev. Gastroenterol. Hepatol. 2014; 11: 256–266. https://www.ncbi.nlm.nih.gov/pubmed/24445613
  52. Prince AC, Myers CE, Joyce T, Irving P, Lomer M, Whelan K. Fermentable carbohydrate restriction (low FODMAP diet) in clinical practice improves functional gastrointestinal symptoms in patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2016; 22: 1129–1136. https://www.ncbi.nlm.nih.gov/pubmed/26914438
  53. Gearry RB, Irving PM, Barrett JS, Nathan DM, Shepherd SJ, Gibson PR. Reduction of dietary poorly absorbed short-chain carbohydrates (FODMAPs) improves abdominal symptoms in patients with inflammatory bowel disease—a pilot study. J. Crohns Colitis 2009; 3: 8–14. https://www.ncbi.nlm.nih.gov/pubmed/21172242
  54. Staudacher HM, Lomer MC, Anderson JL et al. Fermentable carbohydrate restriction reduces luminal bifidobacteria and gastrointestinal symptoms in patients with irritable bowel syndrome. J. Nutr. 2012; 142: 1510–1518. https://www.ncbi.nlm.nih.gov/pubmed/22739368
  55. Halmos EP, Power VA, Shepherd SJ, Gibson PR, Muir JG. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology 2014; 146: 67–75 .e5. https://www.ncbi.nlm.nih.gov/pubmed/24076059
  56. Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Muir JG, Gibson PR. Consistent prebiotic effect on gut microbiota with altered FODMAP intake in patients with Crohn’s disease: a randomised, controlled cross-over trial of well-defined diets. Clin. Transl. Gastroenterol. 2016; 7: e164. https://www.ncbi.nlm.nih.gov/pubmed/27077959
  57. Blaut M, Collins MD, Welling GW, Doré J, Van Loo J, de Vos W. Molecular biological methods for studying the gut microbiota: the EU human gut flora project. Br J Nutr. 2002;87:S203–11. https://www.ncbi.nlm.nih.gov/pubmed/12088520
  58. The role of the gut microbiota in nutrition and health. Nature Reviews Gastroenterology & Hepatology, 9 (10) (2012), pp. 577-589.
  59. Inulin as texture modifier in dairy products. Food Hydrocolloids, 25 (8) (2011), pp. 1881-1890.
  60. Effect of high performance chicory inulin on constipation. Nutrition Research, 20 (5) (2000), pp. 731-736.
  61. Safety and efficacy of inulin and oligofructose supplementation in infant formula: Results from a randomized clinical trial. Clinical Nutrition, 32 (6) (2013), pp. 918-927.
  62. Effects of chicory inulin in constipated elderly people: A double-blind controlled trial. International Journal of Food Sciences and Nutrition, 62 (2) (2011), pp. 164-170. http://www.tandfonline.com/doi/abs/10.3109/09637486.2010.527323
  63. Vogt L, Meyer D, Pullens G et al. Immunological properties of inulin-type fructans. Crit. Rev. Food Sci. Nutr. 2015; 55: 414–436. https://www.ncbi.nlm.nih.gov/pubmed/24915372
  64. Inulin and oligofructose: Review of experimental data on immune modulation. The Journal of Nutrition, 137 (11 Suppl) (2007), pp. 2563S-2567S.
  65. Inulin and oligofructose: Impact on intestinal diseases and disorders. British Journal of Nutrition, 93 (S1) (2005), p. S61. https://doi.org/10.1079/BJN20041345
  66. Immune modulation by different types of β2 → 1-fructans is toll-like receptor dependent. PLoS ONE, 8 (7) (2013), p. e68367. http://journals.plos.org/plosone/article?id=10.1371/journal.pone.0068367
  67. Inulin and oligofructose in chronic inflammatory bowel disease. The Journal of Nutrition, 137 (11 Suppl) (2007), pp. 2572S-2575S.
  68. Inulin-type prebiotics: A review Part 2. Alternative Medicine Review, 4, March 1, 2009, pp. 36-55.
  69. Inulin-type fructans and reduction in colon cancer risk: Review of experimental and human data. British Journal of Nutrition, 93 (S1) (2005), p. S73. https://doi.org/10.1079/BJN20041349
  70. Administration of prebiotic inulin suppresses 1,2 dimethylhydrazine dihydrochloride induced procarcinogenic biomarkers fecal enzymes and preneoplastic lesions in early colon carcinogenesis in Sprague Dawley rats. Journal of Functional Foods, 5 (2) (2013), pp. 991-996. https://doi.org/10.1016/j.jff.2013.02.006
  71. Protective effects of prebiotics inulin and lactulose from cytotoxicity and genotoxicity in human colon adenocarcinoma cells. Food Research International, 52 (1) (2013), pp. 269-274. https://doi.org/10.1016/j.foodres.2013.03.024
  72. Boutron-Ruault MC, Marteau P, Lavergne-Slove A, et al. Effects of a 3-mo consumption of short-chain fructo-oligosaccharides on parameters of colorectal carcinogenesis in patients with or without small or large colorectal adenomas. Nutr Cancer 2005;53:160-168.
  73. Mallappa RH, Rokana N, Duary RK, Panwar H, Batish VK, Grover S. Management of metabolic syndrome through probiotic and prebiotic interventions. Indian Journal of Endocrinology and Metabolism. 2012;16(1):20-27. doi:10.4103/2230-8210.91178. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3263193/
  74. New scientific paradigms for probiotics and prebiotics. Reid G, Sanders ME, Gaskins HR, Gibson GR, Mercenier A, Rastall R, Roberfroid M, Rowland I, Cherbut C, Klaenhammer TR. J Clin Gastroenterol. 2003 Aug; 37(2):105-18. https://www.ncbi.nlm.nih.gov/pubmed/12869879/
  75. Probiotics as drugs against human gastrointestinal infections. Sanz Y, Nadal I, Sánchez E. Recent Pat Antiinfect Drug Discov. 2007 Jun; 2(2):148-56. https://www.ncbi.nlm.nih.gov/pubmed/18221171/
  76. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Furet JP, Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, Mariat D, Corthier G, Doré J, Henegar C, Rizkalla S, Clément K. Diabetes. 2010 Dec; 59(12):3049-57. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2992765/
  77. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Diabetes. 2008 Jun; 57(6):1470-81. http://diabetes.diabetesjournals.org/content/57/6/1470.long
  78. Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Cani PD, Lecourt E, Dewulf EM, Sohet FM, Pachikian BD, Naslain D, De Backer F, Neyrinck AM, Delzenne NM. Am J Clin Nutr. 2009 Nov; 90(5):1236-43. http://ajcn.nutrition.org/content/90/5/1236.long
  79. Modulation of glucagon-like peptide 1 and energy metabolism by inulin and oligofructose: experimental data. Delzenne NM, Cani PD, Neyrinck AM. J Nutr. 2007 Nov; 137(11 Suppl):2547S-2551S. http://jn.nutrition.org/content/137/11/2547S.long
  80. Oligofructose promotes satiety in healthy human: a pilot study. Cani PD, Joly E, Horsmans Y, Delzenne NM. Eur J Clin Nutr. 2006 May; 60(5):567-72. https://www.ncbi.nlm.nih.gov/pubmed/16340949/
  81. FAO/WHO. Working Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food London, Ontario, Canada. 2002.
  82. Colonic fermentation influences lower esophageal sphincter function in gastroesophageal reflux disease. Piche T, des Varannes SB, Sacher-Huvelin S, Holst JJ, Cuber JC, Galmiche JP. Gastroenterology. 2003 Apr; 124(4):894-902. https://www.ncbi.nlm.nih.gov/pubmed/12671885/
  83. Microbial ecology: human gut microbes associated with obesity. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Nature. 2006 Dec 21; 444(7122):1022-3. https://www.ncbi.nlm.nih.gov/pubmed/17183309/
  84. Density distribution of free fatty acid receptor 2 (FFA2)-expressing and GLP-1-producing enteroendocrine L cells in human and rat lower intestine, and increased cell numbers after ingestion of fructo-oligosaccharide. Kaji I, Karaki S, Tanaka R, Kuwahara A. J Mol Histol. 2011 Feb; 42(1):27-38. https://www.ncbi.nlm.nih.gov/pubmed/21113792/
  85. Comparison of different fibers for in vitro production of short chain fatty acids by intestinal microflora. Pylkas AM, Juneja LR, Slavin JL. J Med Food. 2005 Spring; 8(1):113-6. https://www.ncbi.nlm.nih.gov/pubmed/15857221/
  86. Dietary fiber modulates intestinal proglucagon messenger ribonucleic acid and postprandial secretion of glucagon-like peptide-1 and insulin in rats. Reimer RA, McBurney MI. Endocrinology. 1996 Sep; 137(9):3948-56. https://www.ncbi.nlm.nih.gov/pubmed/8756571/
  87. Peptide YY, glucagon-like peptide-1, and neurotensin responses to luminal factors in the isolated vascularly perfused rat ileum. Dumoulin V, Moro F, Barcelo A, Dakka T, Cuber JC. Endocrinology. 1998 Sep; 139(9):3780-6. https://www.ncbi.nlm.nih.gov/pubmed/9724030/
  88. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Parnell JA, Reimer RA. Am J Clin Nutr. 2009 Jun; 89(6):1751-9. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3827013/
  89. Interactions of glucagon-like peptide-1 (GLP-1) with the blood-brain barrier. Kastin AJ, Akerstrom V, Pan W. J Mol Neurosci. 2002 Feb-Apr; 18(1-2):7-14. https://www.ncbi.nlm.nih.gov/pubmed/11931352/
  90. Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-alpha. Freeland KR, Wolever TM. Br J Nutr. 2010 Feb; 103(3):460-6. https://www.ncbi.nlm.nih.gov/pubmed/19818198/
  91. Adaptation of colonic fermentation and glucagon-like peptide-1 secretion with increased wheat fibre intake for 1 year in hyperinsulinaemic human subjects. Freeland KR, Wilson C, Wolever TM. Br J Nutr. 2010 Jan; 103(1):82-90. https://www.ncbi.nlm.nih.gov/pubmed/19664300/
  92. Liu S, Willett WC, Manson JE, Hu FB, Rosner B, Colditz G. Relation between changes in intakes of dietary fiber and grain products and changes in weight and development of obesity among middle-aged women. Am J Clin Nutr 2003;78:920–7. http://ajcn.nutrition.org/content/78/5/920
  93. Schulze MB, Liu S, Rimm EB, Manson JE, Willett WC, Hu FB. Glycemic index, glycemic load, and dietary fiber intake and incidence of type 2 diabetes in younger and middle-aged women. Am J Clin Nutr 2004;80:348–56. http://ajcn.nutrition.org/content/80/2/348
  94. Streppel MT, Ocke MC, Boshuizen HC, Kok FJ, Kromhout D. Dietary fiber intake in relation to coronary heart disease and all-cause mortality over 40 y: the Zutphen Study. Am J Clin Nutr 2008;88:1119–25. http://ajcn.nutrition.org/content/88/4/1119
  95. US Department of Agriculture and US Department of Health and Human Services. Dietary guidelines for Americans. Washington (DC): US Government Printing Office; 2010.
  96. King DE, Mainous , III AG, Lambourne CA. Trends in dietary fiber intake in the United States, 1999–2008. J Acad Nutr Diet 2012;112:642–8. https://www.ncbi.nlm.nih.gov/pubmed/22709768
  97. Cho SS, Qi L, Fahey GC, Klurfeld DM. Consumption of cereal fiber, mixtures of whole grains and bran, and whole grains and risk reduction in type 2 diabetes, obesity, and cardiovascular disease. Am J Clin Nutr 2013;98:594–619. http://ajcn.nutrition.org/content/98/2/594
  98. Lopez MG, Mancilla-Margalli NA, Mendoza-Diaz G. Molecular structures of fructans from Agave tequilana Weber var. azul. J Agric Food Chem 2003;51:7835–40. https://www.ncbi.nlm.nih.gov/pubmed/14690361
  99. Roberfroid MB, Van Loo JA, Gibson GR. The bifidogenic nature of chicory inulin and its hydrolysis products. J Nutr 1998;128:11–9. http://jn.nutrition.org/content/128/1/11
  100. Allsopp P, Possemiers S, Campbell D, Oyarzábal IS, Gill C, Rowland I. An exploratory study into the putative prebiotic activity of fructans isolated from Agave angustifolia and the associated anticancer activity. Anaerobe 2013;22:38–44. https://www.ncbi.nlm.nih.gov/pubmed/23714623
  101. Rendón-Huerta JA, Juárez-Flores B, Pinos-Rodríguez JM, Aguirre-Rivera JR, Delgado-Portales RE. Effects of different sources of fructans on body weight, blood metabolites and fecal bacteria in normal and obese non-diabetic and diabetic rats. Plant Foods Hum Nutr 2012;67:64–70. https://www.ncbi.nlm.nih.gov/pubmed/22210166
  102. Urías-Silvas JE, Cani PD, Delmée E, Neyrinck A, López MG, Delzenne NM. Physiological effects of dietary fructans extracted from Agave tequilana Gto and Dasylirion spp. Br J Nutr 2008;99:254–61. https://www.ncbi.nlm.nih.gov/pubmed/17711612
  103. Agave Inulin Supplementation Affects the Fecal Microbiota of Healthy Adults Participating in a Randomized, Double-Blind, Placebo-Controlled, Crossover Trial. J. Nutr. September 1, 2015 vol. 145 no. 9 2025-2032. http://jn.nutrition.org/content/145/9/2025.long
  104. Briet F, Achour L, Flourie B, et al. Symptomatic response to varying levels of fructo-oligosaccharides consumed occasionally or regularly. Eur J Clin Nutr 1995;49:501-507.
  105. Giacco R, Clemente G, Luongo D, et al. Effects of short-chain fructo-oligosaccharides on glucose and lipid metabolism in mild hypercholesterolaemic individuals. Clin Nutr 2004;23:331-340.
  106. Bouhnik Y, Raskine L, Simoneau G, et al. The capacity of short-chain fructo-oligosaccharides to stimulate faecal Bifidobacteria: a dose-response relationship study in healthy humans. Nutr J 2006;5:8.
  107. Olesen M, Gudmand-Hoyer E. Efficacy, safety, and tolerability of fructooligosaccharides in the treatment of irritable bowel syndrome. Am J Clin Nutr 2000;72:1570-1575.
  108. Gastrointestinal tolerance and utilization of agave inulin by healthy adults. Food Funct. 2014 Jun;5(6):1142-9. doi: 10.1039/c3fo60666j. Epub 2014 Mar 25. https://www.ncbi.nlm.nih.gov/pubmed/24664349
  109. Bouhnik Y, Vahedi K, Achour L, et al. Short-chain fructo-oligosaccharide administration dose-dependently increases fecal Bifidobacteria in healthy humans. J Nutr 1999;129:113-116.
  110. Boehm G, Moro G. Structural and functional aspects of prebiotics used in infant nutrition. J Nutr 2008;138:1818S-1828S.
  111. Boehm G, Lidestri M, Casetta P, et al. Supplementation of a bovine milk formula with an oligosaccharide mixture increases counts of faecal Bifidobacteria in preterm infants. Arch Dis Child Fetal Neonatal Ed 2002;86:F178-F181.
  112. Moro G, Minoli I, Mosca M, et al. Dosage related bifidogenic effects of galacto- and fructooligosaccharides in formula-fed term infants. J Pediatr Gastroenterol Nutr 2002;34:291-295.
Health Jade Team

The author Health Jade Team

Health Jade