2,318
Views
6
CrossRef citations to date
0
Altmetric
Reviews

The role of cereal soluble fiber in the beneficial modulation of glycometabolic gastrointestinal hormones

ORCID Icon, ORCID Icon & ORCID Icon

Abstract

According to cohort studies, cereal fiber, and whole-grain products might decrease risk for type 2 diabetes (T2DM), inflammatory processes, cancer, and cardiovascular diseases. These associations, mainly affect insoluble, but not soluble cereal fiber. In intervention studies, soluble fiber elicit anti-hyperglycemic and anti-inflammatory short-term effects, partially explained by fermentation to short-chain fatty acids, which acutely counteract insulin resistance and inflammation. ß-glucans lower cholesterol levels and possibly reduce liver fat. Long-term benefits are not yet shown, maybe caused by T2DM heterogeneity, as insulin resistance and fatty liver disease – the glycometabolic points of action of soluble cereal fiber – are not present in every patient. Thus, only some patients might be susceptive to fiber. Also, incretin action in response to fiber could be a relevant factor for variable effects. Thus, this review aims to summarize the current knowledge from human studies on the impact of soluble cereal fiber on glycometabolic gastrointestinal hormones. Effects on GLP-1 appear to be highly contradictory, while these fibers might lower GIP and ghrelin, and increase PYY and CCK. Even though previous results of specific trials support a glycometabolic benefit of soluble fiber, larger acute, and long-term mechanistic studies are needed in order to corroborate the results.

Introduction

Case numbers for type 2 diabetes mellitus (T2DM) are rising steadily all over the world. Even though the apparent modifiable reasons of insufficient physical activity and poor quality of food are well known, recommendations for an improved lifestyle seem to fail in most patients. Given the behavioral inertia to initiate regular exercise, to reduce caloric intake and to increase food variability in our modern world, some impulses for a healthy living could be introduced from the food industry rather than their consumers. An important component of a healthy diet are insoluble and soluble dietary fibers, the latter of which are a heterogeneous, but well investigated group of indigestible carbohydrates found in vegetables, fruits, and also grains and cereals.

In our following review, we first highlight the overall and in particular metabolic health benefits of soluble cereal fiber, both from an epidemiological and interventional point of view. In general, soluble cereal fiber improves insulin resistance and glucose tolerance, accommodated by an amelioration of hypercholesterinemia and systemic inflammation. These effects are at least partially independent of weight loss, indicating specific mechanisms working beyond the gastrointestinal lumen.

We then also summarize the interaction of specific human genes with metabolic outcomes related to the intake of dietary fiber. Surprisingly, several of these nutrigenetic associations are linked to incretins and other gastrointestinal hormones, such as glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), peptide YY (PYY), ghrelin, and others. This group of endocrine players is well-known for its impact on insulin secretion, eating behavior and inflammation.

Therefore, we conduct a detailed review of randomized controlled trials (RCT) investigating the effect of soluble cereal fiber on glycemia and insulin response, followed by an overview over the reported effects on the release of incretins and similar endocrine signals. This is done for trials on acute effects, in preload studies and in long-term RCTs.

What is the general benefit of cereal fiber and whole-grain in cohort studies?

The type 2 diabetes mellitus epidemic is growing world-wide, with many countries still experiencing an uprise of the unhealthy lifestyle pattern, combining insufficient physical activity and hypercaloric diet. Despite higher standard of living, this leads to a rising prevalence of cardiovascular disease, cancer and other long-term complications, contributing to premature morbidity and death. As T2DM is the common precursor of these outcomes, large prevention trials have investigated, if it can be prevented by lifestyle modification. In randomized controlled trials (RCT) a 40–60% risk reduction by lifestyle treatment was reported (Pan et al. Citation1997; Ramachandran et al. Citation2006; Knowler et al. Citation2002; Tuomilehto et al. Citation2001). The Da Qing 30-years follow-up indicated a benefit in micro- and macrovascular morbidity and mortality (Gong et al. Citation2019).

Most patients with T2DM show insulin resistance, which is strongly associated with hypercaloric food intake and obesity as well as lacking physical activity. Besides these major pathomechanistic factors, specific dietary aspects, such as saturated fat, simple carbohydrates and excessive intake of alcohol contribute to the metabolic risk, even in the absence of weight gain. Some other nutritional factors have been identified as protective agents: moderate alcohol intake, coffee consumption and cereal fiber from whole-grain products (Ley et al. Citation2014; Weickert and Pfeiffer Citation2008; Aune et al. Citation2013). Consumption of whole-grain products is also associated with lower all-cause mortality (Ma et al. Citation2016) and mortality by cardiovascular (Li et al. Citation2016), neoplastic, infectious or respiratory diseases (Aune et al. Citation2016), but not by stroke or diabetes (Zhang et al. Citation2018; Benisi-Kohansal et al. Citation2016). Similar inverse associations besides mortality are found for morbidity: NAFLD (Xia et al. Citation2020), coronary heart disease (Tang et al. Citation2015), ischemic (but not hemorrhagic) stroke (Chen et al. Citation2016; Deng et al. Citation2018), colorectal, breast, and pancreatic cancer (Lei et al. Citation2016; Reynolds et al. Citation2019). These findings are supported by inverse relations between whole-grain intake and levels of LDL cholesterol (Hollaender, Ross, and Kristensen Citation2015) and obesity (Schlesinger et al. Citation2019). Surprisingly, soluble fruit and vegetable fiber has not been linked to a lower risk for T2DM (Aune et al. Citation2013; Ley et al. Citation2014; Weickert and Pfeiffer Citation2018). A discrimination of insoluble and soluble fiber within whole-grain products has not been done in cohort studies.

What is the interventional evidence on metabolic benefits of cereal fiber?

The strong benefit of cereal fiber, which was shown in epidemiological studies, has mainly been supported by interventional trials assessing whole grain of certain types and ß-glucans as a particular soluble fiber, which is found in whole grain. Interventional data for insoluble cereal fiber such as cellulose, hemicellulose are sparse and currently cannot mirror the effects of cohort studies on this topic.

Meta-analyses on RCTs testing whole-grain products are limited by study design: not a single study reported endpoint outcomes such as cardiovascular events or T2DM onset. Also, whole-grain treatment did not significantly affect BMI, blood pressure, triglycerides, cholesterol and fasting glucose (Maki et al. Citation2019; Kelly et al. Citation2017), but moderate effects for these surrogate parameters were seen in studies specifically addressing dietary fiber (Reynolds et al. Citation2019). There is also a trend for lower HbA1c and insulin resistance when investigating dietary fiber in RCTs (Maki et al. Citation2019). This inconsistency may result from the variety of grains. Barley and oat are rich in soluble fiber such as ß-glucans, which have been shown to be effective in lipid reduction, while other grains – and thus their predominantly insoluble fiber fraction – fail to demonstrate an effect on blood lipids (Hui et al. Citation2019).

However, whole-grain interventions do show benefits. RCT meta-analyses report significantly lower postprandial glucose and insulin excursions in acute trials (Marventano et al. Citation2017), and a trend for a similar benefit in medium- or long-term trials (Reynolds et al. Citation2019). There is also evidence for an anti-inflammatory benefit of whole-grain interventions on CRP and IL-6, but not TNF-alpha (Xu et al. Citation2018). Once again, one can only speculate, which grains or fibers are driving the effect, as not all means of application are tested consistently in the majority of trials. For example, long-term benefits of whole-grain on postprandial metabolism were seen with rice, but not wheat and rye, once again highlighting the heterogeneity of grains and their bioactive compounds (Musa-Veloso et al. Citation2018). Most certainly, this heterogeneity is explained by differences in proportions of certain types of dietary fiber, but most fiber trials are small and short, lacking statistical power (Maki et al. Citation2019).

Long-term trials with considerable statistical power are only available on insoluble fiber. The “Protein and Fiber in Metabolic Syndrome” study (ProFiMet; n = 111) demonstrated improvements of insulin sensitivity by supplementation with an insoluble oat fiber blend after 6, but not after 18 weeks. Insulin resistance, caused by a high-protein diet, was mitigated by fiber additive (Weickert et al. Citation2011). The Optimal Fiber Trial on Diabetes Prevention (OptiFiT, n = 180) was a 2-year diabetes prevention trial using the same fiber supplement. It showed moderate improvements in 2-h glucose, HbA1c and leucocyte count in the entire cohort, but especially in metabolic subgroups (IFG-IGT and obese subjects, respectively) (Honsek et al. Citation2018; Kabisch et al. Citation2019a; Kabisch et al. Citation2019b). Similar subtype-specific responses to fiber-rich diets had been reported previously, indicating that patients with higher fasting glucose levels are more susceptible to these interventions (Hjorth et al. Citation2018; Hjorth et al. Citation2019).

In the past few years, the clinical heterogeneity of T2DM has been investigated more thoroughly, indicating that obese, severely insulin-resistant subjects comprise a distinct phenotype among other groups of patients with a different risk profile (Stidsen et al. Citation2018; Ahlqvist et al. Citation2018). Cereal fiber might be beneficial for some of those phenotypes in particular, but not necessarily all of them. Inflammatory processes in obesity, insulin resistance, and NAFLD might be responsive to fiber intakes, while primary insulin deficiency may resist this kind of intervention. The metabolic amelioration in the “classical textbook phenotype” of T2DM could be achieved by changing the secretion pattern of gastrointestinal (GI) peptide hormones. Soluble cereal fiber might modulate incretin action and interact with their nutrigenetic background.

Nutrigenetic associations between cereal fiber and diabetes risk genes

Fiber intake is connected to obesity and T2DM risk genes by a nutrigenetic interaction, involving a variety of loci without clear pathomechanistic linkage. Obesity-related SNPs in FTO, SH2B1, and other genes modulate anthropometric and glycemic traits, parallel to differences in the degree of fiber intake (Steemburgo et al. Citation2013; Perez-Diaz-del-Campo et al. Citation2020; Vimaleswaran et al. Citation2016; Villegas et al. Citation2014).

Some other nutrigenetic associations are additionally related to the incretin system. The rs7903416 and the rs12255372 polymorphisms (TCF7L2, a strong diabetes risk gene) increase the risk for T2DM, partially by impairing incretin secretion (Lyssenko et al. Citation2007). rs10923931 and rs4457053 reside in the NOTCH2 and ZBED3 genes. Carriers of the T allele of rs10923931 (NOTCH2) show a higher T2DM risk and lower fasting insulin levels (Chen et al. Citation2014; Gupta et al. Citation2012). rs4457053 (ZBED3) has been shown to affect glycemic traits in children (Carayol et al. Citation2020). Both risk alleles in ZBED3 and NOTCH2 are also linked to higher glucagon levels, which might indicate involvement of GIP or CCK, the two nutrient-dependent hormones that increase postprandial glucagon levels (Jonsson et al. Citation2013). TCF7L2, ZBED3, and NOTCH2 are also involved in the WNT pathway, which is connected to insulin sensitivity and was shown to be activated by SCFAs (Abiola et al. Citation2009; Bordonaro, Lazarova, and Sartorelli Citation2007).

While subjects with the protective genotype of the TCF7L2 SNPs show an additional beneficial association of high fiber intake, carriers of the risk allele have a higher T2DM risk and HbA1c, potentiated by high fiber conditions (Hindy et al. Citation2012; Hindy et al. Citation2016). Contrary to that, subjects with the T2DM risk genotypes of NOTCH2 (T+) and ZBED3 (GG) benefit from high fiber intake, while those with generally lower genetic T2DM risk do not (Hindy et al. Citation2016).

Rs10923931 in NOTCH2 and rs780094 in GCKR are among the very few T2DM risk genes, sharing associations with both glycemic and lipid traits (Stančáková et al. Citation2011). Apart from that, rs780094 is additionally linked to NAFLD risk (Zain, Mohamed, and Mohamed Citation2015). Carriers of the TT genotype of rs780094 show lower insulin levels with higher fiber intake, while C allele carriers show higher fasting insulin, independent of fiber intake (Nettleton et al. Citation2010).

Gastrointestinal peptide hormones in the regulation of glucose metabolism

Apart from collecting and digesting food and absorbing nutrients, a major task of the gastrointestinal tract is the hormonal (self)-regulation of digestion and metabolic fate of nutrients. Specialized cells within the single-cell layer of stomach and intestine produce a variety of peptide hormones, which affect the activity of smooth muscle cells, exocrine glands and other endocrine signaling pathways. Among these hormones, the incretin group – represented by glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) – elicits the most relevant metabolic effects.

GLP-1 catalyzes insulin secretion in the presence of elevated or rising glucose levels (Meloni et al., Citation2013), but also suppresses glucagon release (Ramracheya et al. Citation2018). Furthermore, GLP-1 promotes satiety signals in the hypothalamus and reduces the rate of ß-cell apoptosis (Turton et al. Citation1996; Costes, Bertrand, and Ravier Citation2021; Cornu et al. Citation2009). In response to all passing digestible macronutrients, GLP-1 is released from L cells, which reside in the lower parts of the intestine (Lindgren et al. Citation2011). GLP-1 secretion is influenced by a multitude of other nutritional factors, among which fiber has been by far more intensively studied in rodents than humans (Bodnaruc et al. Citation2016). Other factors affecting GLP-1 levels are sex (Adam and Westerterp-Plantenga Citation2005a), body weight (Adam and Westerterp-Plantenga Citation2005b), weight loss (Adam Jocken, and Westerterp-Plantenga Citation2005), and glycemic state. For a decade now, GLP-1 analogues are an important part of antidiabetic pharmacotherapy.

Similar to GLP-1, GIP stimulates ß-cells to secrete insulin given supraphysiological glucose levels and inhibits ß-cell apoptosis (Widenmaier et al. Citation2009). Contrary to GLP-1, it does not possess the potential to induce satiety (Edholm et al. Citation2010; Hayes et al. Citation2021) and increases glucagon secretion (El and Campbell, Citation2020). GIP secretion derives from another type of enteroendocrine cells, the K cells, which are located in the upper parts of the gut and are exclusively stimulated during passage of saccharides, lipids and amino acids (Reimann et al. Citation2020). GIP secretion has an impact on postprandial nutrient distribution by modulating splanchnic blood circulation (Koffert et al., Citation2017). Especially in scenarios with elevated energy intake this may lead to an increase of inflammatory processes, ectopic lipid storage (liver fat and visceral fat) and insulin resistance (Keyhani-Nejad et al. Citation2015; Gögebakan et al. Citation2015; Pfeiffer and Keyhani-Nejad 2018; Keyhani-Nejad et al. Citation2020). In rodents, inhibition of GIP signaling counteracts obesity and NAFLD (Isken et al. Citation2008; Pfeiffer et al. Citation2010). GLP-1-GIP-glucagon co-agonists are tested as antidiabetic drugs (Knerr et al. Citation2020).

Some other GI peptide hormones are secreted in response to passing nutrients, but lack a direct metabolic effect. However, these endocrine players are strong satiety signals. Peptide YY1-36 (PYY) is released from the L cells in the ileum and colon (which are also the source of GLP-1), but also in other parts of the GI tract. PYY reduces gastric motility and emptying, which supports nutrient absorption in the gut (Savage et al. Citation1987). On the other hand, PYY inhibits the exocrine secretion from the gastric wall and the pancreatic acinus. Dietary fat and other chymus components are are the main stimuli (Onaga et al. Citation2002). Enzymatic cleavage of two amino acids leads to the antagonistic residue PYY3-36.

There are more hormones, which affect gut function and satiety but do not seem to affect insulin secretion: oxyntomodulin (OXM), glucagon-like peptide 2 (GLP-2), cholecystokinin (CCK), and Pancreatic Polypeptide (PP). OXM and GLP-2 are alternative splicing products of preproglucagon (PPG). They are secreted from the GLP-1 producing L cells, depending on carbohydrate or protein ingestion, and possess structural homologies to GLP-1 (Holst et al. Citation2018; Tang-Christensen et al. Citation2000; Baldassano, Amato, and Mulè Citation2016).

CCK is produced in so-called I cells, which are located in the duodenum and jejunum. This hormone is mainly released after meals containing fat and/or protein (Liou et al. Citation2011; Wang et al. Citation2011; Daly et al. Citation2013). Sex and eating behavior have an impact on circulating CCK levels (Burton-Freeman Citation2005). PP is a poorly understood hormone. It appears to improve hepatic insulin sensitivity (Śliwińska-Mossoń et al. Citation2017), and is secreted in response to rising levels of GIP (Veedfald et al. Citation2020).

Last, but not least, there is one unambiguous opponent of all satiety hormones: ghrelin. The ghrelin-producing X/A cells in the gastric wall are highly active during hunger but become dormant after meal initiation. Dietary triggers for ghrelin suppression are mainly proteins and carbohydrates (Tannous dit El Khoury et al. Citation2006; Overduin et al. Citation2005), but also fats (Heath et al. Citation2004). Ghrelin is only active as the acylated derivative of its precursor protein. Acylated ghrelin increases appetite, food-seeking behavior and craving, even exceeding energy balance. It also shows a glucoregulative effect, which is counteracted by the non-acylated ghrelin precursor (Heppner and Tong Citation2014; Gauna et al. Citation2005).

Relations between fiber fermentation, incretins, and glucose metabolism

Most soluble fibers, originating from cereals, fruits and vegetables, share a common pattern of viscosity and fermentability, inulin and fructanes being exceptions as non-viscous, but fermentable fibers. Both viscosity and fermentability contribute to the biological effects of soluble fibers. Viscosity is mainly acting by physics, as soluble fiber binds water, increases chyme and fecal mass and might therefore stimulate pressure-sensitive enteroendocrine cells in the GI tract. Viscous fiber was also proposed to produce a viscous “unstirred layer” which may delay the absorption of cabohydrates and thereby lower the GI (Lembcke et al. Citation1984).

Microbial fermentation in the lower parts of the gut leads to the production of short-chain fatty acids (SCFA), which are feeding both, other microbiota, and gut cells. After absorption into the blood-stream, acetate is circulating for a long time (Scheppach Citation1994), while most of the propionate is quickly metabolized in the liver to odd-chain fatty acids (Weitkunat et al. Citation2017). Butyrate is predominantly remaining in gut cells and serves as their metabolic fuel (Roediger Citation1980). Within the human body, these SCFA activate SCFA receptors: FFA2 (acetate and propionate), FFA3 (propionate and butyrate) and GPR109A (just butyrate). These receptors modulate immune responses (FFA2 and GPR109A) and incretin secretion (FFA2 and FFA3) (Kaji, Karaki, and Kuwahara Citation2014). In particular, L cells carry SCFA receptors, which highlights the role of SCFAs in the release of GLP-1 and PYY, the main signals derived from these cells. In-vitro stimulation with SCFA precursors stimulates L cell proliferation, while non-fermentable cereal fiber (cellulose) does not (Kaji et al. Citation2011; Mitsui et al. Citation2006). Upon administration of acetate or other SCFAs, FFA2 and to a smaller extent also FFA3 stimulate GLP-1 and PYY release (Freeland and Wolever Citation2010; Larraufie et al. Citation2018). After absorption, SCFAs from fermentable soluble fiber (including ß-glucans) stimulate insulin secretion and improve insulin sensitivity (Pingitore et al. Citation2017; Darzi et al. Citation2016; Hooda et al. Citation2010; Adam et al. Citation2014; Miyamoto et al. Citation2018). By this partially incretin-driven improvement of the insulin system SCFAs may also counteract the development of NAFLD (Weitkunat et al. Citation2016; Zhou et al. Citation2018). Alleviation of T2DM is also supported by SCFA-driven reinforcement of bacterial strains feeding on these fiber fermentation products (Zhao et al. Citation2018).

As SCFAs as fiber fermentation products contribute to the energy balance, excess intake of fermentable fiber may lead to a smaller metabolic benefit due to a different body weight trajectory. Mice being fed with high amounts of fermentable fiber gain weight, as the SCFAs provide excess energy and are metabolized to storable long-chain fatty acids (Isken et al. Citation2010).

Soluble cereal fiber – a distinct biochemical entity

As shown in cohort studies, whole-grain products and in particular insoluble total fiber (from all sources) contribute to reduced health risks. It is hard to determine, which fiber from which source is the active player. Whole-grain food, for example, contains both soluble and insoluble fiber, minerals, plant protein, vitamins, triterpenoids, and sterols, all of which might explain health benefits. While insoluble fibers in total (from cereals and other sources) are backed up by better evidence from cohort studies, they lack sufficient fiber-specific intervention studies. On the contrary, ß-glucans and other fermentable grain-based fibers have not been identified as strong predictors of lower health risks in epidemiological studies, but the active role of soluble fiber (in particular of cereal origin rather than fruits and vegetables) is well supported by mechanistic in-vitro and in-vivo studies. Additionally, many acute and short-term studies with clear and targeted intervention using ß-glucans confirm that at least some metabolic benefit is apparent. Soluble cereal fiber comprises two main chemical structures:

Cereal ß-glucans are alternating ß-1,3-/ß-1,4-linked glucose polymers. They are found predominantly in barley and oat, but only in minor fractions in rye, wheat and other grains. ß-glucans from yeasts are ß-1,3-/ß-1,6-linked, often water-insoluble glucose polymers, which possibly do not share the medical benefits of cereal ß-glucans (Kohl et al. Citation2009; Dalonso, Goldman, and Gern Citation2015). Other sources are algae, bacteria, lichens, yeasts, and specific edible fungi, which are not reviewed in this article.

Second, some members of the hemicellulose family are found to be water-soluble: pentosane-hemicelluloses from rye and oat, including some, but not all arabinoxylans. The third type – resistant starch type 2 (RS2) – comprises starch, which, despite being extracted from the cellular structure, resists digestion and acts as fermentable, soluble fiber. The main source is high-amylose maize.

Solubility depends on the proportion of arabinose monomers and their linkage to xylose (Kiszonas, Fuerst, and Morris Citation2013). Natural arabinoxylan occurs together with phenolic compounds (such as ferulic acid), binding to the fiber structure.

ß-glucans, soluble arabinoxylans and RS2 are both viscous and fermentable, sharing these properties with other kinds of soluble fiber: fructanes and inulin (non-viscous, but fermentable), polyuronides (pectin, alginates, agar, carrageen). These compounds are found in fruits, vegetables and legumes. As fruit and vegetable fiber has consistently shown not to be associated with lower risk for T2DM and the long-term risk profile (Aune et al. Citation2013; Ley et al. Citation2014; Weickert and Pfeiffer Citation2018), these types of fiber are not reviewed in this article.

Methodology of the review

The currently available literature on our topic was assessed via PubMed, using a search term, that covered whole grain (barley and oats, only), all types of cereal soluble fiber and all types of incretins or satiety-related gastrointestinal hormones: (((“fiber*” or “fiber*”) and soluble) or ß-glucan* or arabinoxylan* or pentosan* or (“whole grain" and (barley or oat)) OR “resistant starch”) and (glp-1 or “glucagon-like peptide” or gip or “glucose-dependent insulinotropic peptide” or glp-2 or ghrelin or “peptide yy” or pyy or cholecystokinin* OR cck or oxyntomodulin OR OXM OR incretin*). We selected all human intervention trials, using at least one specific soluble cereal high-fiber treatment in comparison to a low-fiber treatment or similar amount of insoluble fiber, assessing at least one of the hormones of interest, and being published until 30th June 2021.

We specifically included studies on clearly defined whole-grain foods, if they were derived from barley or oat. By this, we extend the view on possible effects of fiber-rich cereal-based diets. We are aware, that fiber-rich foods differ in their fiber composition (e.g. by containing both soluble and insoluble fiber) and in their overall food matrix, which might impair the generalizability of our review. For example, whole-grain foods and various kinds of grains can contain peptides inhibiting dipeptidyl peptidase IV, which may increase GLP-1 levels (Wang et al. Citation2015), while the sole fiber possibly would not alter GLP-1 release. Also, the magnitude of grounding affects dietary properties of whole-grain products, influencing the incretin profile and the glycemic response despite identical contents (Eelderink et al. Citation2017). However, by reviewing evidence for fiber and fiber-rich foods together, we can highlight the consistency of the results.

We excluded studies comparing complex fiber-rich diets to low-fiber diets, that were not based on a sole contrast between whole grain and refined grain, but rather used additional sources of fiber (e.g. fruits and vegetables) or other kinds of food modification (Zamaratskaia et al. Citation2017; Zhao et al. Citation2018; Hu et al. Citation2016; Silva et al. Citation2015; Bligh et al. Citation2015; Gonzalez-Anton et al. Citation2015; Dandona et al. Citation2015). We also excluded studies, which compared different kinds of grains or grain products, but not on the basis of fiber content (Bo et al. Citation2017; Stefoska-Needham et al. Citation2016; Bakhøj et al. Citation2003; Mano et al. Citation2018; Alyami et al. Citation2019; Stringer et al. Citation2013). Another study comparing glucose solution and breads with different types of soluble cereal fiber was also excluded (Binou et al. Citation2021).

The literature provides an enormous variety of treatments (fiber supplements, fiber-enriched foods and natural whole-grain foods), study designs (cross-over trials, parallel interventions, acute vs. preload treatment vs. long-term studies), cohorts (healthy subjects, patients with metabolic impairments), and assessments of outcomes (different ELISA or RIA kits, fasting and postprandial levels based on different oral stimulation tests). Therefore, a meta-analysis is not feasible at the time, even under consideration of subgroup analyses. Hence, we decide to provide a comprehensive, narrative review on the literature.

Included studies

Based on our selection criteria, we identified 38 eligible publications with 38 separate experiments or identical trials on separately reported study cohorts (e.g. a healthy normal weight and an obese sub-cohort). Out of these 38 experiments, 24 were reporting acute treatment effects only. Five papers reported a preload effect, in which the outcome was measured at a subsequent meal in the morning. Nine studies reported long-term outcomes after prolonged exposition to high- or low-fiber treatment. Most experiments on acute effects (n = 15) used an intervention with resistant starch or ß-glucans as food fortification, while nine studies investigated effects of specific types of soluble cereal fiber in other forms of application.

Most trials did not exceed 20 subjects per group and the majority of trials predominantly included female participants. Most trials were focused on young-to-middle-aged healthy and normal-weight subjects. Sex ratio is almost balanced for acute and preload trials, while women are overrepresented in the reviewed long-term studies on soluble cereal fiber. Details on cohort structure, reviewed interventions and assessed hormone parameters of all included trials can be found in .

Table 1. Reviewed acute studies with use of a specific fiber.

Table 2. Reviewed acute studies with use of fiber-enriched grain products.

Table 3. Reviewed preload studies.

Table 4. Reviewed cross-over and parallel-designed long-term studies.

Effects of soluble cereal fiber or whole-grain food on glucose and insulin levels

The reviewed studies on acute effects of soluble cereal fiber or whole-grain often showed lower postprandial glucose and insulin excursions in their fiber-rich interventions. In healthy subjects, an average reduction of 20% in glucose AUC or up to 60% in glucose iAUC could be seen in interventions with purely supplemented ß-glucans (Juvonen et al. Citation2009), but especially in studies investigating this kind of fiber in a starchy matrix (enriched to grain-based food products) (Vitaglione et al. Citation2009; Ames et al. Citation2015; Ekström, Björck, and Östman Citation2016). Some of those studies on fiber-fortified food reporting an improved glycemia also showed a marked effect on postprandial insulinemia (Ames et al. Citation2015; Ekström, Björck, and Östman Citation2016), while one trial failed to show a parallel finding on insulin (Vitaglione et al. Citation2009). Other studies on ß-glucans or resistant starch as cereal food additive reported 15–20% lower postprandial insulin levels, but no difference in glucose excursions (Juntunen et al. Citation2002; Beck, Tosh, et al. Citation2009; Beck, Tapsell, et al. Citation2009; Emilien, Hsu, and Hollis Citation2017).

Resistant starch as additive to grain-based foods such as breads or pancakes lowered postprandial glycemia by 20–40% (Gentile et al. Citation2015; Belobrajdic et al. Citation2019). Oat-bran fortified breakfast cereal also showed a 20–30% improved glycemia and insulinemia (Wolever et al. 2020). Smaller effects (10–20%) on postprandial glucose and insulin were reported after interventions with resistant starch supplements in comparison to control (Raben et al. Citation1994; Klosterbuer, Thomas, and Slavin Citation2012), and in one trial investigating oat ß-glucan-fortified breakfasts (Zaremba et al. Citation2018).

Unchanged glucose excursions were seen in studies on the acute effects of ß-glucans in non-starchy foods. Herein, insulin levels weren’t altered as well (Barone Lumaga et al. Citation2012; Doyon et al. Citation2015). Such non-significant results were also seen in some studies on ß-glucans in starchy foods (Bourdon et al. Citation1999), on resistant starch supplements (Bodinham et al. Citation2013) or RS-fortified foods (Emilien et al. Citation2018). For the specific soluble fiber Fibersol-2 no measurements on glycemia or insulinemia were reported (Ye et al. Citation2015).

In overweight or obese subjects, test meals with resistant starch (vs. non-fortified control) elicited small (10%) or no significant effects on glycemia and insulinemia (Rahat-Rozenbloom et al. Citation2017; Al-Mana and Robertson, Citation2018). Study participants with metabolic syndrome also achieved a glycemic benefit by ß-glucan enriched bread (vs. arabinoxylan-enriched bread), characterized by 20% lower early and late postprandial AUCs for both glucose and insulin (Hartvigsen et al. Citation2014). In patients with T2DM, resistant starch lowered late, but not early postprandial glycemia and insulinemia (MacNeil et al. Citation2013).

In preload studies with healthy subjects, administration of barley kernels or barley kernel bread in the evening before a test meal lowered post-peak glucose decrements and insulin iAUCs, while leaving fasting and early (0–15 min) postprandial levels unaffected (Nilsson et al. Citation2008; Johansson et al. Citation2013). Contrary to that, an arabinoxylan-fortified white bread evening preload did not change fasting glucose before next day’s breakfast, but lowered its postprandial glucose AUC by about 15%. On the other hand, in this study the arabinoxylan preload reduced fasting insulin by 16-24% and postprandial insulin excursions by only 6–10% (Boll et al. Citation2016). Whole-grain rye bread had a similar effect, especially when fortified with resistant starch. These preload interventions lowered both postprandial glucose and insulin levels by 15–25% during a standard breakfast in the next morning, but fasting glucose and insulin were not altered (Sandberg, Björck, and Nilsson Citation2017). In the only preload study in T2DM patients (comparing oat-fiber and conventional cereal flakes), the fiber intervention induced 20% (peak) to 50% (iAUC) lower glycemia in the morning standard test meal (Kim et al. Citation2016).

The long-term studies in this review also investigated glycemia and insulin response following their fiber and control interventions. Two cross-over trials investigated barley-kernel bread vs. white wheat bread in healthy persons over few days. Despite the short intervention, both studies noticed 20–25% lower postprandial glucose levels and 10–15% lower postprandial insulin concentrations (Nilsson et al. Citation2015, Citation2016). A 4-weeks cross-over intervention trial using resistant starch supplements found a 33% improvement in insulin sensitivity, a 44% increased insulin-adjusted glucose clearance, despite overall lower insulin levels (Robertson et al. Citation2005). In another rather similar study, glucose levels did not differ between intervention and control condition, but early postprandial (30 min) levels of c-peptide and insulin were 15% and 25% higher after 4 weeks (Zhang et al. Citation2019). A 10-day cross-over intervention with resistant starch 3 paralleling a weight-maintenance diet after weight loss did also not affect fasting or postprandial glucose levels, but significantly lowered pre- and postprandial insulin by about 20% (Johnstone et al. Citation2020). Prolonged supplementation with pullulan, corn fiber or soluble fiber dextrin over 2 weeks had no effect on fasting glucose or insulin levels (Stewart et al. Citation2010).

A few long-term RCTs on soluble fiber and GI peptide hormone release were conducted in parallel design, all of them in overweight or obese persons. One study, investigating ß-glucan-fortified food products (vs. non-fortified controls) over a period of 3 months did neither effect glucose nor insulin levels (Beck et al. Citation2010). Another trial examined the effect of resistant starch over 6 weeks. Similarly, fasting and postprandial glucose and insulin levels were not affected by the intervention (Maziarz et al. Citation2017). Major cardiometabolic outcomes of the study by White et al. (White et al. Citation2020) had been published earlier (Peterson et al. Citation2018). Apart from HbA1c, which increased in the control group and remained stable in the intervention, no other glycometabolic outcome (fasting or postprandial) showed a significant different result between the study arms (Peterson et al. Citation2018).

Effects of soluble cereal fiber or whole-grain food on GI peptide hormones

Glucagon-like peptide-1

In healthy subjects, several trials on the acute effects of soluble fiber found a weaker postprandial GLP-1 stimulation after ingestion compared to control. Reductions of 20–60% in AUC were reported in studies on resistant starch (Raben et al. Citation1994; Klosterbuer, Thomas, and Slavin Citation2012; Bodinham et al. Citation2013), where the impact of resistant starch was augmented by adding pullulan (Klosterbuer, Thomas, and Slavin Citation2012). In one study, investigating viscous vs. non-viscous ß-glucans, postprandial GLP-1 was found to be 60% lower in the viscous fiber condition (Juvonen et al. Citation2009). An acute 40% increase in postprandial GLP-1 has been shown solely for high doses of Fibersol-2 dissolved in icetea and compared to placebo icetea in healthy subjects (Ye et al. Citation2015). Unchanged postprandial GLP-1 secretion after acute fiber interventions was found in studies in healthy subjects, investigating wheat-based resistant starch or dextrin (Rahat-Rozenbloom et al. Citation2017; Emilien, Hsu, and Hollis Citation2017), ß-glucans (Ames et al. Citation2015; Zaremba et al. Citation2018) and grain products enriched with ß-glucans or resistant starch (Juntunen et al. Citation2002; Gentile et al. Citation2015; Belobrajdic et al. Citation2019). Non-significant differences between fiber intervention and control were found in two studies in overweight or obese patients testing the effects of resistant starch (Rahat-Rozenbloom et al. Citation2017; Al-Mana and Robertson Citation2018). Another study showed indifferent GLP-1 stimulation using bagels with different amounts of resistant starch in T2DM patients (MacNeil et al. Citation2013).

Evening preloads with barley kernels induced 50% higher fasting and postprandial GLP-1 levels at a following morning test meal in healthy subjects when compared to white wheat bread (Johansson et al. Citation2013). A similar study found higher postprandial (AUC +25%), but not preprandial GLP-1 levels (Nilsson et al. Citation2008). Preloads with arabinoxylans and/or resistant starch (in healthy subjects) and fiber-enriched flakes (in T2DM patients) did not significantly change pre- or postprandial GLP-1 compared to control (Boll et al. Citation2016; Kim et al. Citation2016).

Similarly, prolonged intake of soluble cereal fiber led to controversial results. In healthy subjects, barley-based bread (compared to white wheat bread) increased pre- and postprandial GLP-1 release by 56% and 13%, respectively (Nilsson et al. Citation2015). In a similar cross-over experiment of the same research group, the average postprandial, but not the fasting GLP-1 levels were significantly (+14%) higher under barley conditions compared to control (Nilsson et al. Citation2016). A Chinese study examined the effect of resistant starch supplementation over 4 weeks and found a significant increase of early postprandial GLP-1 levels (+30% after 30 min), while fasting and overall postprandial concentrations did not differ between the interventions (Zhang et al. Citation2019). In overweight subjects resistant starch compared to control elicited no change in fasting or stimulated GLP-1 levels after 6 weeks (Beck et al. Citation2010). Fiber-fortified food products (resistant starch type 2 or 3) did not change GLP-1 secretion in overweight or obese subjects after 10 days or 12 weeks (Maziarz et al. Citation2017; White et al. Citation2020). Also, ß-glucan supplementation during weight loss diet did not augment effects on fasting GLP-1 after 3 months in obese persons (Johnstone et al. Citation2020).

Glucose-dependent insulinotropic peptide

The literature on fiber effects on GIP release appears to be slightly more consistent, but most of the studies reporting GIP levels after acute stimulation with soluble cereal fiber used a starchy food matrix to deliver the intervention. Therefore, only sparse information is available about the action of soluble cereal fibers in a non-starchy meal or oral test solution. Overall, an increase in GIP levels has not been reported in any of the reviewed acute, preload, or long-term trials.

In healthy subjects, ingestion of ß-glucan supplemented rye bread, resistant starch and resistant dextrin in different applications, each compared to other cereal foods or suitable control, led to significantly lower postprandial GIP release. Those reductions achieved 20–30% in AUC and up to 50% in iAUC (Raben et al. Citation1994; Juntunen et al. Citation2002; Gentile et al. Citation2015; Ekström, Björck, and Östman Citation2016; Emilien et al. Citation2018). Acute reduction of GIP stimulation of similar magnitude was also reported in a trial with T2DM patients investigating the effect of resistant starch (MacNeil et al. Citation2013). FiberSol-2 and resistant starch did not have an impact on postprandial GIP levels in two other studies with healthy subjects (Ye et al. Citation2015; Belobrajdic et al. Citation2019). Also, ß-glucan-enriched bread did not induce significantly different postprandial GIP levels compared to white wheat or arabinoxylan-fortified bread, when tested in patients with metabolic syndrome (Hartvigsen et al. Citation2014).

Evening preloads with barley kernels or different kinds of breads, including barley bread, did not affect fasting and postprandial GIP levels in healthy subjects (Nilsson et al. Citation2008; Johansson et al. Citation2013). Fiber-enriched cereal flakes did not elicit a different GIP profile compared to conventional low-fiber flakes, when administered to T2DM patients (Boll et al. Citation2016).

Only one study investigated long-term effects of soluble cereal fiber on GIP release. In this trial, a 10-day low-fat maintenance diet, administered to overweight-to-obese patients after weight reduction, was fortified with resistant starch 3 (vs. placebo as cross-over condition). However, both groups showed similar AUCs for GIP after the intervention period (Johnstone et al. Citation2020).

Peptide YY

Acute fiber effects on PYY secretion have been investigated in more than a dozen cross-over trials, providing quite consistent evidence for some fiber species.

An increased secretion by about 15% was detected in healthy subjects undergoing an interventions with ß-glucan enriched bread (Vitaglione et al. Citation2009). A complete shift in PYY excursions from U-shaped decrease to U-shaped increase was reported in a study investigating ß-glucan-enriched cereals (Beck, Tapsell, et al. Citation2009). A massive increase of PYY AUC by 250% was found when examining the metabolic effects of FiberSol-2 as placebo-controlled additive to icetea (Ye et al. Citation2015). About 30–40% higher levels of late postprandial (120–180 min) PYY were also induced by enrichment of pancakes with resistant starch (Gentile et al. Citation2015).

An 80% decrease in PYY AUC was seen after ingestion of a viscous ß-glucan drinks (compared to a non-viscous ß-glucan drink) in healthy persons (Juvonen et al. Citation2009).

No significant differences in comparison to control were reported from studies in healthy subjects using resistant starch as plain substance or food-fortification (Rahat-Rozenbloom et al. Citation2017; Ekström, Björck, and Östman Citation2016; Emilien et al. Citation2018). A single study on ß-glucan barley tortillas (vs. non-fortified amylose tortillas) also found no impact of soluble cereal fiber on PYY excursions (Ames et al. Citation2015). Additionally, no PYY-modulating effect for sources of soluble cereal fiber – namely oat bran and resistant starch – was seen in overweight subjects (Rahat-Rozenbloom et al. Citation2017; Wolever et al. 2020).

Among preload studies, only one RCT examined effects of soluble cereal fiber on PYY. In healthy subjects, an evening preload of rye whole-grain bread with resistant starch (but not without it) induced 20% higher fasting and postprandial PYY levels compared to white wheat bread (Sandberg, Björck, and Nilsson Citation2017).

Long-term effects of fiber-rich food on PYY were studied in several trials. 18% higher postprandial PYY levels were reported after 3 days of intake of barley kernel bread in healthy subjects, while fasting levels were not significantly higher (Nilsson et al. Citation2015). A very similar study using 4 days of supplementation with barley kernel bread in healthy participants showed no significant change in fasting or postprandial PYY levels (Nilsson et al. Citation2016). Long-term trials with 2–12 weeks of supplementation with resistant starch also failed to show an impact of this soluble fiber on PYY secretion (Zhang et al. Citation2019; Johnstone et al. Citation2020; Maziarz et al. Citation2017; White et al. Citation2020). In a 3-months trial investigating the effects of ß-glucans on the metabolic and endocrine profile in obese subjects undergoing a weight-loss diet, only the moderate dose of fiber (5–6 g/d) led to a marked 30% reduction of fasting PYY1-36 levels, while the high dose (8–9 g/d) did not. The authors could not explain this U-shaped dose-response pattern of effect. Also, controversely, in this study, plasma concentrations of PYY3-36 did not change at all (Beck et al. Citation2010).

Other non-incretin satiety hormones

Very few studies investigated the impact of soluble cereal fiber on other satiety-related gastrointestinal hormones.

A three-day intervention comparing barley kernel bread with white wheat bread in healthy subjects did not show an impact on fasting or postprandial OXM secretion (Nilsson et al. Citation2015). This study is the only one on OXM.

GLP-2 has not been examined in any trial on immediate or long-term effects of soluble cereal fiber. However, one trial in healthy subjects assessed if an overnight preload with arabinoxylan and/or resistant starch modulates pre- or postprandial GLP-2 secretion, but did not find significant changes in comparison to white wheat bread (Kim et al. Citation2016).

After three days of ingestion of barley kernel bread, a trendwise 9% increase in fasting GLP-2 levels and a 13% significant rise in postprandial GLP-2 was seen in healthy subjects (Nilsson et al. Citation2015). A similar study from the same research group could not replicate these results in a four-day setting with the same intervention in once again healthy participants (Nilsson et al. Citation2016).

ß-glucans in starchy foods increased the immediate postprandial CCK release in healthy subjects by about 20–40% (Bourdon et al. Citation1999; Beck, Tosh, et al. Citation2009). Another study on non-viscous ß-glucans (vs. viscous ß-glucans) in a non-starchy food matrix found a 40% reduction of CCK AUC in healthy subjects (Juvonen et al. Citation2009). Other studies on resistant starch in muffins or wheat drinks (Emilien, Hsu, and Hollis Citation2017; Emilien et al. Citation2018) and Fibersol-2 as additive to icetea had no such effect on postprandial CCK levels (Ye et al. Citation2015). The only long-term study reporting CCK levels found no impact of ß-glucans on these hormone levels in obese subjects under caloric restriction (Beck et al. Citation2010).

Postprandial PP appeared to be strongly stimulated (+35%) by ß-glucan ingestion in a study on acute effects in healthy subjects, which is the only trial on this hormone (Barone Lumaga et al. Citation2012).

Ghrelin

Most studies about fiber effects on ghrelin levels show non-significant results. Very few cross-over RCTs reported a stronger suppression of postprandial ghrelin by ß-glucans, administered to healthy subjects. Ghrelin levels turned out 10–30% lower compared to control conditions (Juvonen et al. Citation2009; Barone Lumaga et al. Citation2012; Vitaglione et al. Citation2009). In another trial on various isocaloric and isoproteinemic yoghurt snacks, the combination of added inulin and ß-glucans, but neither of the single additives augmented the postprandial ghrelin suppression (Doyon et al. Citation2015). Lack of changes in postprandial ghrelin levels were described in acute trials investigating resistant starch, pullulan and Fibersol-2 as additives to non-starchy oral stimuli (Klosterbuer, Thomas, and Slavin Citation2012; Ye et al. Citation2015; Rahat-Rozenbloom et al. Citation2017), but also in four studies examining the effect of RS-fortified grain products (Gentile et al. Citation2015; Ekström, Björck, and Östman Citation2016; Emilien et al. Citation2018; Belobrajdic et al. Citation2019). A single trial on ß-glucans measured postprandial acylated ghrelin levels, but found no differences between the interventions (Beck, Tosh, et al. Citation2009). No effect on postprandial ghrelin suppression was also seen in an RCT comparing ß-glucan rich vs. arabinoxylan-fortified bread in patients with Metabolic Syndrome and in overweight subjects ingesting oat bran vs. control (Hartvigsen et al. Citation2014; Wolever et al. 2020).

The only preload study investigating effects of soluble cereal fiber on ghrelin compared barley kernels vs. wheat bread in healthy subjects, but found no effect on fasting ghrelin levels throughout a morning test meal (Johansson et al. Citation2013).

Several studies assessed the long-term effects of soluble fiber on ghrelin secretion. In a 4-weeks cross-over study on resistant starch in healthy subjects, a 34% increase in fasting ghrelin was reported, while the postprandial suppression was not affected (Robertson et al. Citation2005). Pre- and postprandial ghrelin levels did not change significantly after a three-day stimulation with barley kernel bread (vs. white wheat bread) in healthy participants (Nilsson et al. Citation2015). Postprandial ghrelin suppression was also not altered by interventions with pullulan, resistant starch type 3, soluble fiber dextrin or soluble corn fiber in a 2-weeks cross-over trial in healthy subjects (Stewart et al. Citation2010). Resistant starch type 2 and 3 also turned out to be ineffective on ghrelin levels during 10 days or 12 weeks of supplementation with fiber-fortified food products in overweight and obese patients (Johnstone et al. Citation2020; White et al. Citation2020).

Comprehension and outlook

Natural food items are affecting the incretin system in a very elaborate and complicated way (Ríos et al. Citation2019). The reviewed selection of well-controlled, randomized, but usually rather small studies in mostly healthy subjects shows somewhat conclusive results for some of the investigated parameters, but lacks the statistical power and the generalizability to clearly describe the role of soluble cereal fiber in the modulation of incretins and other GI peptide hormones. The existing literature includes studies using different types and dosages of fiber, most of the RCTs investigated acute, but not prolonged effects. The overall evidence has to be evaluated as low-to-moderate.

Our current work now provides insight for soluble fiber, which has been shown to acutely affect glucose and lipid metabolism. Up-to-date, there is only insufficient literature in order to conduct a systematic review with meta-analysis on incretin effects due to administration of soluble fiber. There are clearly many RCTs on various types of soluble fiber (ß-glucans, inulin, resistant starch), but most of them lack an analysis of gastrointestinal hormones. So far, only two meta-analyses have investigated the effects of prebiotics on ghrelin (da Silva Borges et al. Citation2020) and resistant starch on several GI peptides (Guo, Tan, and Kong Citation2020). Effects of all soluble cereal fibers on glucose metabolism and the involved GI peptides in humans can be summarized as follows:

  1. Epidemiological evidence points toward a strong antidiabetic association for whole grain and cereal insoluble fiber, but not soluble fiber of any kind. Whether whole grain effects are supported by soluble fiber is doubtful, as some sources of soluble cereal fiber are uncommon in the daily diet of most people. Oats are only consumed by a fraction of the population and barley is mainly used for livestock feeding and beer production, limiting the intake of ß-glucans by natural sources.

RCTs show consistent metabolic benefits for soluble fibers in acute trials, especially for ß-glucans, but there is insufficient data for long-term effects. Once again, studies on “whole-grain” fail to pinpoint their crucial component. Also, it is unclear, if certain effects of cereal fiber depend on a specific food matrix, even though ß-glucans are effectively reducing cholesterol levels as a supplement.

  • There are some nutrigenetic studies showing an interaction between (a) genes, which are involved in the regulation of incretin release, (b) whole-grain intake, and (c) diabetes risk or diabetes-associated traits.

  • More than 25 cross-over RCTs have assessed the immediate effects of soluble cereal fiber and fiber-enriched cereal products on glucose metabolism and relevant GI peptide hormones.

  • A glycometabolic benefit of ß-glucans and resistant starch can be consistently demonstrated, when these types of soluble cereal fiber are tested embedded in a starchy food matrix. Improvements in insulin levels were reported in several trials, but not consistently linked to lower glucose levels. Acute studies on non-starchy fiber interventions and trials in patients with metabolic impairment (obesity and T2DM) showed less impressive effects on glycemia and insulinemia. Both in acute and preload studies, fasting levels of glucose and insulin remained unaltered, while especially late postprandial metabolism was improved. In long-term studies, reduced insulinemia was more common than improved glycemia and generally improvements of metabolism were not consistently found over the spectrum of interventions.

  • Results of acute trials are completely inconclusive for GLP-1, as we cannot identify a consistently directed effect of any kind of soluble cereal fiber of fiber-rich food on this endocrine player. This is rather surprising, as this group of fiber is known to be fermented to SCFAs which increase GLP-1 levels. Increased GLP-1 levels could correspond to improved glycemia and insulin resistance, but higher GLP-1 was mostly reported in preload and long-term studies, but not after acute stimulation. Patients with overweight, obesity and/or T2DM did not show a change in GLP-1 levels to either acute, preload or prolonged fiber stimulation.

    Postprandial GIP seems to be either suppressed or unaffected by ß-glucans and resistant starch in acute feeding trials. Lack of effect was predominantly seen in subjects with metabolic syndrome.

    Consistently, ingestion of fiber-enriched foods (ß-glucans, resistant starch and other fiber species) in healthy subjects elicited either an increase or no changes in PYY levels. An increase would be expected, as SCFAs as fermentation product of soluble fibers increase PYY secretion. However, studies in patients with overweight mostly failed to show an effect on PYY.

    In particular ß-glucans seem to increase CCK and to suppress postprandial ghrelin. A reduction of GIP could contribute to the acute antihyperglycemic effects seen in soluble fiber treatment and might also indicate a therapeutic potential in NAFLD. Augmented PYY and CCK and suppression of ghrelin might explain weight loss effects, especially for ß-glucans and resistant starch (Rahmani et al. Citation2019; Snelson et al. Citation2019). Surprisingly, studies investigating fiber effects on ghrelin in patients with obesity or T2DM failed to show such a nutrient-driven modulation. For CCK, there is also some evidence for nutrigenetic mechanisms involving fiber intake (Jonsson et al. Citation2013).

    Overall, studies with insignificant effects on hormone secretion used comparable dosages of dietary fiber compared to RCTs with prominent interventional changes.

  • Only few trials investigated the effects of fiber-rich preloads on GLP-1, GIP, PYY, and ghrelin. They do not present any consistent effect for soluble fiber on any of the aforementioned hormones.

  • Long-term studies are sparse and most of the data does not show an impact of any kind of soluble cereal fiber on GI hormones.

  • Data on PP, CCK, OXM, and GLP-2 are available in only very few trials.

Type-2 resistant starch and ß-glucans are the most promising representatives of soluble cereal fiber and should be investigated in large, high-quality acute and long-term trials, addressing both the endocrine and the resulting metabolic outcome. As these fibers are prone to fermentation, identification of the respective individual SCFA proportions is warranted for each fiber species. Currently, most trials on GI hormones have been conducted in healthy subjects, while patients at metabolic risk (prediabetes, early T2DM) are rarely investigated. Further studies with a broader view on the metabolic potential and mechanisms of soluble cereal fiber (including inflammation and liver fat) are desired.

Authors’ contributions

SK, MOW and AFHP wrote the paper and serve as guarantors of this work.

Abbreviations
CCK=

cholecystokinin

GIP=

glucose-dependent insulinotropic peptide

GLP-1=

glucagon-like peptide 1

GLP-2=

glucagon-like peptide 2

IFG=

impaired fasting glucose

IGT=

impaired glucose tolerance

NAFLD=

nonalcoholic fatty liver disease

OXM=

oxyntomodulin

PP=

pancreatic polypeptide

PPG=

preproglucagon

PYY=

peptide YY

RCT=

randomized-controlled trial

RS=

resistant starch

T2DM=

type 2 diabetes mellitus

Disclosure statement

SK received a travel grant from Rettenmaier & Soehne, Holzmuehle, Germany, including conference fees and accomodation. All authors have conducted studies with non-financial support from Rettenmaier & Soehne, Holzmuehle, Germany. The authors declare no further conflicts of interest associated with this manuscript.

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Abiola, M., M. Favier, E. Christodoulou-Vafeiadou, A. L. Pichard, I. Martelly, and I. Guillet-Deniau. 2009. Activation of Wnt/beta-catenin signaling increases insulin sensitivity through a reciprocal regulation of Wnt10b and SREBP-1c in skeletal muscle cells. PloS One 4 (12):e8509. doi: 10.1371/journal.pone.0008509.
  • Adam, C. L., P. A. Williams, M. J. Dalby, K. Garden, L. M. Thomson, A. J. Richardson, S. W. Gratz, and A. W. Ross. 2014. Different types of soluble fermentable dietary fibre decrease food intake, body weight gain and adiposity in young adult male rats. Nutrition & Metabolism 11:36. doi: 10.1186/1743-7075-11-36.
  • Adam, T. C, and M. S. Westerterp-Plantenga. 2005a. Glucagon-like peptide-1 release and satiety after a nutrient challenge in normal-weight and obese subjects. The British Journal of Nutrition 93 (6):845–51. doi: 10.1079/bjn20041335.
  • Adam, T. C, and M. S. Westerterp-Plantenga. 2005b. Nutrient-stimulated GLP-1 release in normal-weight men and women. Hormone and metabolic research = Hormon- und Stoffwechselforschung = Hormones et metabolisme 37 (2):111–7. doi: 10.1055/s-2005-861160.
  • Adam, T. C., J. Jocken, and M. S. Westerterp-Plantenga. 2005. Decreased glucagon-like peptide 1 release after weight loss in overweight/obese subjects. Obesity Research 13 (4):710–6. doi: 10.1038/oby.2005.80.
  • Ahlqvist, E., P. Storm, A. Käräjämäki, M. Martinell, M. Dorkhan, A. Carlsson, P. Vikman, R. B. Prasad, D. M. Aly, P. Almgren, et al. 2018. Novel subgroups of adult-onset diabetes and their association with outcomes: A data-driven cluster analysis of six variables. The Lancet. Diabetes & Endocrinology 6 (5):361–9. doi: 10.1016/S2213-8587(18)30051-2.
  • Al-Mana, N. M, and M. D. Robertson. 2018. Acute effect of resistant starch on food intake, appetite and satiety in overweight/obese males. Nutrients 10 (12):1993. doi: 10.3390/nu10121993.
  • Alyami, J., E. Whitehouse, G. E. Yakubov, S. E. Pritchard, C. L. Hoad, E. Blackshaw, K. Heissam, S. M. Cordon, H. F. J. Bligh, R. C. Spiller, et al. 2019. Glycaemic, gastrointestinal, hormonal and appetitive responses to pearl millet or oats porridge breakfasts: A randomised, crossover trial in healthy humans. The British Journal of Nutrition 122 (10):1142–54. doi: 10.1017/S0007114519001880.
  • Ames, N., H. Blewett, J. Storsley, S. J. Thandapilly, P. Zahradka, and C. Taylor. 2015. A double-blind randomised controlled trial testing the effect of a barley product containing varying amounts and types of fibre on the postprandial glucose response of healthy volunteers. The British Journal of Nutrition 113 (9):1373–83. doi: 10.1017/S0007114515000367.
  • Aune, D., N. Keum, E. Giovannucci, L. T. Fadnes, P. Boffetta, D. C. Greenwood, S. Tonstad, L. J. Vatten, E. Riboli, and T. Norat. 2016. Whole grain consumption and risk of cardiovascular disease, cancer, and all cause and cause specific mortality: Systematic review and dose-response meta-analysis of prospective studies. BMJ (Clinical Research ed.) 353:i2716. doi: 10.1136/bmj.i2716.
  • Aune, D., T. Norat, P. Romundstad, and L. J. Vatten. 2013. Whole grain and refined grain consumption and the risk of type 2 ­diabetes: A systematic review and dose-response meta-analysis of cohort studies. European Journal of Epidemiology 28 (11):845–58. doi: 10.1007/s10654-013-9852-5.
  • Bakhøj, S., A. Flint, J. J. Holst, and I. Tetens. 2003. Lower glucose-dependent insulinotropic polypeptide (GIP) response but similar glucagon-like peptide 1 (GLP-1), glycaemic, and insulinaemic response to ancient wheat compared to modern wheat depends on processing. European Journal of Clinical Nutrition 57 (10):1254–61. doi: 10.1038/sj.ejcn.1601680.
  • Baldassano, S., A. Amato, and F. Mulè. 2016. Influence of glucagon-like peptide 2 on energy homeostasis. Peptides 86:1–5. doi: 10.1016/j.peptides.2016.09.010.
  • Barone Lumaga, R., D. Azzali, V. Fogliano, L. Scalfi, and P. Vitaglione. 2012. Sugar and dietary fibre composition influence, by different hormonal response, the satiating capacity of a fruit-based and a β-glucan-enriched beverage. Food & Function 3 (1):67–75. doi: 10.1039/c1fo10065c.
  • Beck, E. J., L. C. Tapsell, M. J. Batterham, S. M. Tosh, and X. F. Huang. 2009. Increases in peptide Y-Y levels following oat beta-glucan ingestion are dose-dependent in overweight adults. Nutrition Research (New York, N.Y.) 29 (10):705–9. doi: 10.1016/j.nutres.2009.09.012.
  • Beck, E. J., L. C. Tapsell, M. J. Batterham, S. M. Tosh, and X. F. Huang. 2010. Oat beta-glucan supplementation does not enhance the effectiveness of an energy-restricted diet in overweight women. The British Journal of Nutrition 103 (8):1212–22. doi: 10.1017/S0007114509992856.
  • Beck, E. J., S. M. Tosh, M. J. Batterham, L. C. Tapsell, and X. F. Huang. 2009. Oat beta-glucan increases postprandial cholecystokinin levels, decreases insulin response and extends subjective satiety in overweight subjects. Molecular Nutrition & Food Research 53 (10):1343–51. doi: 10.1002/mnfr.200800343.
  • Belobrajdic, D. P., A. Regina, B. Klingner, I. Zajac, S. Chapron, P. Berbezy, and A. R. Bird. 2019. High-amylose wheat lowers the postprandial glycemic response to bread in healthy adults: A randomized controlled crossover trial. The Journal of Nutrition 149 (8):1335–45. doi: 10.1093/jn/nxz067.
  • Benisi-Kohansal, S., P. Saneei, M. Salehi-Marzijarani, B. Larijani, and A. Esmaillzadeh. 2016. Whole-grain intake and mortality from all causes, cardiovascular disease, and cancer: A systematic review and dose-response meta-analysis of prospective cohort studies. Advances in Nutrition (Bethesda, Md.) 7 (6):1052–65. doi: 10.3945/an.115.011635.
  • Binou, P., A. E. Yanni, A. Stergiou, K. Karavasilis, P. Konstantopoulos, D. Perrea, N. Tentolouris, and V. T. Karathanos. 2021. Enrichment of bread with beta-glucans or resistant starch induces similar glucose, insulin and appetite hormone responses in healthy adults. European Journal of Nutrition 60 (1):455–64. doi: 10.1007/s00394-020-02265-6.
  • Bligh, H. F., I. F. Godsland, G. Frost, K. J. Hunter, P. Murray, K. MacAulay, D. Hyliands, D. C. Talbot, J. Casey, T. P. Mulder, et al. 2015. Plant-rich mixed meals based on Palaeolithic diet principles have a dramatic impact on incretin, peptide YY and satiety response, but show little effect on glucose and insulin homeostasis: An acute-effects randomised study. The British Journal of Nutrition 113 (4):574–84. doi: 10.1017/S0007114514004012.
  • Bo, S., M. Seletto, A. Choc, V. Ponzo, A. Lezo, A. Demagistris, A. Evangelista, G. Ciccone, M. Bertolino, M. Cassader, et al. 2017. The acute impact of the intake of four types of bread on satiety and blood concentrations of glucose, insulin, free fatty acids, triglyceride and acylated ghrelin. A randomized controlled cross-over trial. Food Research International (Ottawa, Ont.) 92:40–7. doi: 10.1016/j.foodres.2016.12.019.
  • Bodinham, C. L., N. M. Al-Mana, L. Smith, and M. D. Robertson. 2013. Endogenous plasma glucagon-like peptide-1 following acute dietary fibre consumption. The British Journal of Nutrition 110 (8):1429–33. doi: 10.1017/S0007114513000731.
  • Bodnaruc, A. M., D. Prud’homme, R. Blanchet, and I. Giroux. 2016. Nutritional modulation of endogenous glucagon-like peptide-1 secretion: A review. Nutrition & Metabolism 13:92. doi: 10.1186/s12986-016-0153-3.
  • Boll, E. V., L. M. Ekström, C. M. Courtin, J. A. Delcour, A. C. Nilsson, I. M. Björck, and E. M. Östman. 2016. Effects of wheat bran extract rich in arabinoxylan oligosaccharides and resistant starch on overnight glucose tolerance and markers of gut fermentation in healthy young adults. European Journal of Nutrition 55 (4):1661–70. doi: 10.1007/s00394-015-0985-z.
  • Bordonaro, M., D. L. Lazarova, and A. C. Sartorelli. 2007. Sartorelli AC The activation of beta-catenin by Wnt signaling mediates the effects of histone deacetylase inhibitors. Experimental Cell Research 313 (8):1652–66. doi: 10.1016/j.yexcr.2007.02.008.
  • Bourdon, I., W. Yokoyama, P. Davis, C. Hudson, R. Backus, D. Richter, B. Knuckles, and B. O. Schneeman. 1999. Postprandial lipid, glucose, insulin, and cholecystokinin responses in men fed barley pasta enriched with beta-glucan. The American Journal of Clinical Nutrition 69 (1):55–63. doi: 10.1093/ajcn/69.1.55.
  • Burton-Freeman, B. 2005. Sex and cognitive dietary restraint influence cholecystokinin release and satiety in response to preloads varying in fatty acid composition and content. The Journal of Nutrition 135 (6):1407–14. doi: 10.1093/jn/135.6.1407.
  • Carayol, J., J. Hosking, J. Pinkney, J. Marquis, A. Charpagne, S. Metairon, A. Jeffery, J. Hager, and F. P. Martin. 2020. Genetic susceptibility determines β-cell function and fasting glycemia trajectories throughout childhood: A 12-year cohort study (EarlyBird 76). Diabetes Care 43 (3):653–60. doi: 10.2337/dc19-0806.
  • Chen, J., Q. Huang, W. Shi, L. Yang, J. Chen, and Q. Lan. 2016. Meta-analysis of the association between whole and refined grain consumption and stroke risk based on prospective cohort studies. Asia-Pacific Journal of Public Health 28 (7):563–75. doi: 10.1177/1010539516650722.
  • Chen, Z., M. A. Pereira, M. Seielstad, W. P. Koh, E. S. Tai, Y. Y. Teo, J. Liu, C. Hsu, R. Wang, A. O. Odegaard, et al. 2014. Joint effects of known type 2 diabetes susceptibility loci in genome-wide association study of Singapore Chinese: The Singapore Chinese health study. PloS One 9 (2):e87762. doi: 10.1371/journal.pone.0087762.
  • Cornu, M., J. Y. Yang, E. Jaccard, C. Poussin, C. Widmann, and B. Thorens. 2009. Glucagon-like peptide-1 protects beta-cells against apoptosis by increasing the activity of an IGF-2/IGF-1 receptor autocrine loop. Diabetes 58 (8):1816–25. doi: 10.2337/db09-0063.
  • Costes, S., G. Bertrand, and M. A. Ravier. 2021. Mechanisms of beta-cell apoptosis in type 2 diabetes-prone situations and potential protection by GLP-1-based therapies. International Journal of Molecular Sciences 22 (10):5303. doi: 10.3390/ijms22105303.
  • da Silva Borges, D., R. Fernandes, A. Thives Mello, E. da Silva Fontoura, A. R. Soares Dos Santos, and E. B. Santos de Moraes Trindade. 2020. Prebiotics may reduce serum concentrations of C-reactive protein and ghrelin in overweight and obese adults: A systematic review and meta-analysis. Nutrition Reviews 78 (3):235–48. doi: 10.1093/nutrit/nuz045.
  • Dalonso, N., G. H. Goldman, and R. M. Gern. 2015. β-(1→3),(1→6)-Glucans: Medicinal activities, characterization, biosynthesis and new horizons. Applied Microbiology and Biotechnology 99 (19):7893–906. doi: 10.1007/s00253-015-6849-x.
  • Daly, K., M. Al-Rammahi, A. Moran, M. Marcello, Y. Ninomiya, and S. P. Shirazi-Beechey. 2013. Sensing of amino acids by the gut-expressed taste receptor T1R1-T1R3 stimulates CCK secretion. American Journal of Physiology-Gastrointestinal and Liver Physiology 304 (3):G271–G282. doi: 10.1152/ajpgi.00074.2012.
  • Dandona, P., H. Ghanim, S. Abuaysheh, K. Green, M. Batra, S. Dhindsa, A. Makdissi, R. Patel, and A. Chaudhuri. 2015. Decreased insulin secretion and incretin concentrations and increased glucagon concentrations after a high-fat meal when compared with a high-fruit and -fiber meal. American Journal of Physiology. Endocrinology and Metabolism 308 (3):E185–91. doi: 10.1152/ajpendo.00275.2014.
  • Darzi, J., G. S. Frost, J. R. Swann, A. Costabile, and M. D. Robertson. 2016. L-rhamnose as a source of colonic propionate inhibits insulin secretion but does not influence measures of appetite or food intake. Appetite 98:142–9. doi: 10.1016/j.appet.2015.12.011.
  • Deng, C., Q. Lu, B. Gong, L. Li, L. Chang, L. Fu, and Y. Zhao. 2018. Stroke and food groups: An overview of systematic reviews and meta-analyses. Public Health Nutrition 21 (4):766–76. doi: 10.1017/S1368980017003093.
  • Doyon, C. Y., A. Tremblay, L. E. Rioux, C. Rhéaume, K. Cianflone, P. Poursharifi, and S. L. Turgeon. 2015. Acute effects of protein composition and fibre enrichment of yogurt consumed as snacks on appetite sensations and subsequent ad libitum energy intake in healthy men. Applied Physiology, Nutrition, and Metabolism = Physiologie appliquee, nutrition et metabolisme 40 (10):980–9. doi: 10.1139/apnm-2014-0403.
  • Edholm, T., M. Degerblad, P. Grybäck, L. Hilsted, J. J. Holst, H. Jacobsson, S. Efendic, P. T. Schmidt, and P. M. Hellström. 2010. Differential incretin effects of GIP and GLP-1 on gastric emptying, appetite, and insulin-glucose homeostasis. Neurogastroenterology and Motility: The Official Journal of the European Gastrointestinal Motility Society 22 (11):1191–200, e315. doi: 10.1111/j.1365-2982.2010.01554.x.
  • Eelderink, C., M. W. J. Noort, N. Sozer, M. Koehorst, J. J. Holst, C. F. Deacon, J. F. Rehfeld, K. Poutanen, R. J. Vonk, L. Oudhuis, et al. 2017. Difference in postprandial GLP-1 response despite similar glucose kinetics after consumption of wheat breads with different particle size in healthy men. European Journal of Nutrition 56 (3):1063–76. doi: 10.1007/s00394-016-1156-6.
  • Ekström, L. M., I. M. Björck, and E. M. Östman. 2016. An improved course of glycaemia after a bread based breakfast is associated with beneficial effects on acute and semi-acute markers of appetite. Food & Function 7 (2):1040–7. doi: 10.1039/c5fo00969c.
  • El, K, and J. E. Campbell. 2020. The role of GIP in α-cells and glucagon secretion. Peptides 125:170213. doi: 10.1016/j.peptides.2019.170213.
  • Emilien, C. H., W. H. Hsu, and J. H. Hollis. 2017. Effect of resistant wheat starch on subjective appetite and food intake in healthy adults. Nutrition (Burbank, Los Angeles County, Calif.) 43-44:69–74. doi: 10.1016/j.nut.2017.06.020.
  • Emilien, C. H., Y. Zhu, W. H. Hsu, P. Williamson, and J. H. Hollis. 2018. The effect of soluble fiber dextrin on postprandial appetite and subsequent food intake in healthy adults. Nutrition (Burbank, Los Angeles County, Calif.) 47:6–12. doi: 10.1016/j.nut.2017.08.016.
  • Freeland, K. R, and T. M. Wolever. 2010. Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumor necrosis factor-α. The British Journal of Nutrition 103 (3):460–6. doi: 10.1017/S0007114509991863.
  • Gauna, C., P. J. Delhanty, L. J. Hofland, J. A. Janssen, F. Broglio, R. J. Ross, E. Ghigo, and A. J. van der Lely. 2005. Ghrelin stimulates, whereas des-octanoyl ghrelin inhibits, glucose output by primary hepatocytes. The Journal of Clinical Endocrinology and Metabolism 90 (2):1055–60. doi: 10.1210/jc.2004-1069.
  • Gentile, C. L., E. Ward, J. J. Holst, A. Astrup, M. J. Ormsbee, S. Connelly, and P. J. Arciero. 2015. Resistant starch and protein intake enhances fat oxidation and feelings of fullness in lean and overweight/obese women. Nutrition Journal 14:113. doi: 10.1186/s12937-015-0104-2.
  • Gögebakan, Ö., M. A. Osterhoff, R. Schüler, O. Pivovarova, M. Kruse, A. C. Seltmann, A. S. Mosig, N. Rudovich, M. Nauck, and A. F. Pfeiffer. 2015. GIP increases adipose tissue expression and blood levels of MCP-1 in humans and links high energy diets to inflammation: A randomised trial. Diabetologia 58 (8):1759–68. doi: 10.1007/s00125-015-3618-4.
  • Gong, Q., P. Zhang, J. Wang, J. Ma, Y. An, Y. Chen, B. Zhang, X. Feng, H. Li, X. Chen, et al. 2019. Da Qing Diabetes Prevention Study Group. Morbidity and mortality after lifestyle intervention for people with impaired glucose tolerance: 30-year results of the Da Qing Diabetes Prevention Outcome Study. Lancet Diabetes and Endocrinology. 7 (6):452–61. doi: 10.1016/S2213-8587(19)30093-2.
  • Gonzalez-Anton, C., B. Lopez-Millan, M. C. Rico, E. Sanchez-Rodriguez, M. D. Ruiz-Lopez, A. Gil, and M. D. Mesa. 2015. An enriched, cereal-based bread affects appetite ratings and glycemic, insulinemic, and gastrointestinal hormone responses in healthy adults in a randomized, controlled trial. The Journal of Nutrition 145 (2):231–8. doi: 10.3945/jn.114.200386.
  • Guo, J., L. Tan, and L. Kong. 2020. Impact of dietary intake of resistant starch on obesity and associated metabolic profiles in human: A systematic review of the literature. Critical Reviews in Food Science and Nutrition 23:1–17.
  • Gupta, V., D. G. Vinay, S. Rafiq, M. V. Kranthikumar, C. S. Janipalli, C. Giambartolomei, D. M. Evans, K. R. Mani, M. N. Sandeep, A. E. Taylor, et al. 2012. Association analysis of 31 common polymorphisms with type 2 diabetes and its related traits in Indian sib pairs. Diabetologia 55 (2):349–57. doi: 10.1007/s00125-011-2355-6.
  • Hartvigsen, M. L., S. Gregersen, H. N. Laerke, J. J. Holst, K. E. Bach Knudsen, and K. Hermansen. 2014. Effects of concentrated arabinoxylan and β-glucan compared with refined wheat and whole grain rye on glucose and appetite in subjects with the metabolic syndrome: A randomized study. European Journal of Clinical Nutrition 68 (1):84–90. doi: 10.1038/ejcn.2013.236.
  • Hayes, M. R., T. Borner, and B. C. De Jonghe. 2021. The role of GIP in the regulation of GLP-1 satiety and nausea. Diabetes 70 (9):1956–61. doi: 10.2337/dbi21-0004.
  • Heath, R. B., R. Jones, K. N. Frayn, and M. D. Robertson. 2004. Vagal stimulation exaggerates the inhibitory ghrelin response to oral fat in humans. The Journal of Endocrinology 180 (2):273–81. doi: 10.1677/joe.0.1800273.
  • Heppner, K. M, and J. Tong. 2014. Mechanisms in endocrinology: Regulation of glucose metabolism by the ghrelin system: Multiple players and multiple actions. European Journal of Endocrinology 171 (1):R21–R32. doi: 10.1530/EJE-14-0183.
  • Hindy, G., E. Sonestedt, U. Ericson, X. J. Jing, Y. Zhou, O. Hansson, E. Renström, E. Wirfält, and M. Orho-Melander. 2012. Role of TCF7L2 risk variant and dietary fibre intake on incident type 2 diabetes. Diabetologia 55 (10):2646–54. doi: 10.1007/s00125-012-2634-x.
  • Hindy, G., I. G. Mollet, G. Rukh, U. Ericson, and M. Orho-Melander. 2016. Several type 2 diabetes-associated variants in genes annotated to WNT signaling interact with dietary fiber in relation to incidence of type 2 diabetes. Genes & Nutrition 11:6. doi: 10.1186/s12263-016-0524-4.
  • Hjorth, M. F., G. A. Bray, Y. Zohar, L. Urban, D. C. Miketinas, D. A. Williamson, D. H. Ryan, J. Rood, C. M. Champagne, F. M. Sacks, et al. 2019. Pretreatment fasting glucose and insulin as determinants of weight loss on diets varying in macronutrients and dietary fibres-The POUNDS LOST Study. Nutrients 11 (3):586. doi: 10.3390/nu11030586.
  • Hjorth, M. F., Y. Zohar, J. O. Hill, and A. Astrup. 2018. Personalized dietary management of overweight and obesity based on measures of insulin and glucose. Annual Review of Nutrition 38:245–72. doi: 10.1146/annurev-nutr-082117-051606.
  • Hollaender, P. L., A. B. Ross, and M. Kristensen. 2015. Whole-grain and blood lipid changes in apparently healthy adults: A systematic review and meta-analysis of randomized controlled studies. The American Journal of Clinical Nutrition 102 (3):556–72. doi: 10.3945/ajcn.115.109165.
  • Holst, J. J., N. J. W. Albrechtsen, M. B. N. Gabe, and M. M. Rosenkilde. 2018. Oxyntomodulin: Actions and role in diabetes. Peptides 100:48–53. doi: 10.1016/j.peptides.2017.09.018.
  • Honsek, C., S. Kabisch, M. Kemper, C. Gerbracht, A. M. Arafat, A. L. Birkenfeld, U. Dambeck, M. A. Osterhoff, M. O. Weickert, and A. F. H. Pfeiffer. 2018. Fibre supplementation for the prevention of type 2 diabetes and improvement of glucose metabolism: The randomised controlled Optimal Fibre Trial (OptiFiT). Diabetologia 61 (6):1295–305. doi: 10.1007/s00125-018-4582-6.
  • Hooda, S., J. J. Matte, T. Vasanthan, and R. T. Zijlstra. 2010. Dietary oat beta-glucan reduces peak net glucose flux and insulin production and modulates plasma incretin in portal-vein catheterized grower pigs. The Journal of Nutrition 140 (9):1564–9. doi: 10.3945/jn.110.122721.
  • Hu, T., L. Yao, K. Reynolds, T. Niu, S. Li, P. Whelton, J. He, and L. Bazzano. 2016. The effects of a low-carbohydrate diet on appetite: A randomized controlled trial. Nutrition, Metabolism, and Cardiovascular Diseases: NMCD 26 (6):476–88. doi: 10.1016/j.numecd.2015.11.011.
  • Hui, S., K. Liu, H. Lang, Y. Liu, X. Wang, X. Zhu, S. Doucette, L. Yi, and M. Mi. 2019. Comparative effects of different whole grains and brans on blood lipid: A network meta-analysis. European Journal of Nutrition 58 (7):2779–87. doi: 10.1007/s00394-018-1827-6.
  • Isken, F., A. F. Pfeiffer, R. Nogueiras, M. A. Osterhoff, M. Ristow, B. Thorens, M. H. Tschöp, and M. O. Weickert. 2008. Deficiency of glucose-dependent insulinotropic polypeptide receptor prevents ovariectomy-induced obesity in mice. American Journal of Physiology. Endocrinology and Metabolism 295 (2):E350–5. doi: 10.1152/ajpendo.00008.2008.
  • Isken, F., S. Klaus, M. Osterhoff, A. F. Pfeiffer, and M. O. Weickert. 2010. Effects of long-term soluble vs. insoluble dietary fiber intake on high-fat diet-induced obesity in C57BL/6J mice. The Journal of Nutritional Biochemistry 21 (4):278–84. doi: 10.1016/j.jnutbio.2008.12.012.
  • Johansson, E. V., A. C. Nilsson, E. M. Östman, and I. M. Björck. 2013. Effects of indigestible carbohydrates in barley on glucose metabolism, appetite and voluntary food intake over 16 h in healthy adults. Nutrition Journal 12:46. doi: 10.1186/1475-2891-12-46.
  • Johnstone, A. M., J. Kelly, S. Ryan, R. Romero-Gonzalez, H. McKinnon, C. Fyfe, E. Naslund, R. Lopez-Nicolas, D. Bosscher, A. Bonnema, et al. 2020. Nondigestible carbohydrates affect metabolic health and gut microbiota in overweight adults after weight loss. The Journal of Nutrition 150 (7):1859–70. doi: 10.1093/jn/nxaa124.
  • Jonsson, A., C. Ladenvall, T. S. Ahluwalia, J. Kravic, U. Krus, J. Taneera, B. Isomaa, T. Tuomi, E. Renström, L. Groop, et al. 2013. Effects of common genetic variants associated with type 2 diabetes and glycemic traits on α- and β-cell function and insulin action in humans. Diabetes 62 (8):2978–83. doi: 10.2337/db12-1627.
  • Juntunen, K. S., L. K. Niskanen, K. H. Liukkonen, K. S. Poutanen, J. J. Holst, and H. M. Mykkänen. 2002. Postprandial glucose, insulin, and incretin responses to grain products in healthy subjects. The American Journal of Clinical Nutrition 75 (2):254–62. doi: 10.1093/ajcn/75.2.254.
  • Juvonen, K. R., A. K. Purhonen, M. Salmenkallio-Marttila, L. Lähteenmäki, D. E. Laaksonen, K. H. Herzig, M. I. Uusitupa, K. S. Poutanen, and L. J. Karhunen. 2009. Viscosity of oat bran-enriched beverages influences gastrointestinal hormonal responses in healthy humans. The Journal of Nutrition 139 (3):461–6. doi: 10.3945/jn.108.099945.
  • Kabisch, S., N. M. T. Meyer, C. Honsek, C. Gerbracht, U. Dambeck, M. Kemper, M. A. Osterhoff, A. L. Birkenfeld, A. M. Arafat, M. F. Hjorth, et al. 2019a. Fasting glucose state determines metabolic response to supplementation with insoluble cereal fibre: A secondary analysis of the optimal fibre trial (OptiFiT). Nutrients 11 (10):2385. doi: 10.3390/nu11102385.
  • Kabisch, S., N. M. T. Meyer, C. Honsek, C. Gerbracht, U. Dambeck, M. Kemper, M. A. Osterhoff, A. L. Birkenfeld, A. M. Arafat, M. O. Weickert, et al. 2019b. Obesity does not modulate the glycometabolic benefit of insoluble cereal fibre in subjects with prediabetes-A stratified post hoc analysis of the optimal fibre trial (OptiFiT). Nutrients 11;11 (11):2726. doi: 10.3390/nu11112726.
  • Kaji, I., S. Karaki, and A. Kuwahara. 2014. Short-chain fatty acid receptor and its contribution to glucagon-like peptide-1 release. Digestion 89 (1):31–6. doi: 10.1159/000356211.
  • Kaji, I., S. Karaki, R. Tanaka, and A. Kuwahara. 2011. 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. Journal of Molecular Histology 42 (1):27–38. doi: 10.1007/s10735-010-9304-4.
  • Kelly, S. A., L. Hartley, E. Loveman, J. L. Colquitt, H. M. Jones, L. Al-Khudairy, C. Clar, R. Germanò, H. R. Lunn, G. Frost, et al. 2017. Whole grain cereals for the primary or secondary prevention of cardiovascular disease. The Cochrane Database of Systematic Reviews 8:CD005051. doi: 10.1002/14651858.CD005051.pub3.
  • Keyhani-Nejad, F., M. Irmler, F. Isken, E. K. Wirth, J. Beckers, A. L. Birkenfeld, and A. F. Pfeiffer. 2015. Nutritional strategy to prevent fatty liver and insulin resistance independent of obesity by reducing glucose-dependent insulinotropic polypeptide responses in mice. Diabetologia 58 (2):374–83. doi: 10.1007/s00125-014-3423-5.
  • Keyhani-Nejad, F., R. L. Barbosa Yanez, M. Kemper, R. Schueler, O. Pivovarova-Ramich, N. Rudovich, and A. F. H. Pfeiffer. 2020. Endogenously released GIP reduces and GLP-1 increases hepatic insulin extraction. Peptides 125:170231. doi: 10.1016/j.peptides.2019.170231.
  • Kim, E. K., T. J. Oh, L. K. Kim, and Y. M. Cho. 2016. Improving effect of the acute administration of dietary fiber-enriched cereals on blood glucose levels and gut hormone secretion. Journal of Korean Medical Science 31 (2):222–30. doi: 10.3346/jkms.2016.31.2.222.
  • Kiszonas, A. M., E. P. Fuerst, and C. F. Morris. 2013. Wheat arabinoxylan structure provides insight into function. Cereal Chemistry 90 (4):387–95.
  • Klosterbuer, A. S., W. Thomas, and J. L. Slavin. 2012. Resistant starch and pullulan reduce postprandial glucose, insulin, and GLP-1, but have no effect on satiety in healthy humans. Journal of Agricultural and Food Chemistry 60 (48):11928–34. doi: 10.1021/jf303083r.
  • Knerr, P. J., S. A. Mowery, B. Finan, D. Perez-Tilve, M. H. Tschöp, and R. D. DiMarchi. 2020. Selection and progression of unimolecular agonists at the GIP, GLP-1, and glucagon receptors as drug candidates. Peptides 125:170225. doi: 10.1016/j.peptides.2019.170225.
  • Knowler, W. C., E. Barrett-Connor, S. E. Fowler, R. F. Hamman, J. M. Lachin, E. A. Walker, and D. M. Nathan. 2002. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. The New England Journal of Medicine 346 (6):393–403. doi: 10.1056/NEJMoa012512.
  • Koffert, J., H. Honka, J. Teuho, S. Kauhanen, S. Hurme, R. Parkkola, V. Oikonen, A. Mari, A. Lindqvist, N. Wierup, et al. 2017. Effects of meal and incretins in the regulation of splanchnic blood flow. Endocrine Connections 6 (3):179–87. doi: 10.1530/EC-17-0015.
  • Kohl, A., O. Gögebakan, M. Möhlig, M. Osterhoff, F. Isken, A. F. Pfeiffer, and M. O. Weickert. 2009. Increased interleukin-10 but unchanged insulin sensitivity after 4 weeks of (1, 3)(1, 6)-beta-glycan consumption in overweight humans. Nutrition Research (New York, N.Y.) 29 (4):248–54. doi: 10.1016/j.nutres.2009.03.002.
  • Larraufie, P., C. Martin-Gallausiaux, N. Lapaque, J. Dore, F. M. Gribble, F. Reimann, and H. M. Blottiere. 2018. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Scientific Reports 8 (1):74. doi: 10.1038/s41598-017-18259-0.
  • Lei, Q., H. Zheng, J. Bi, X. Wang, T. Jiang, X. Gao, F. Tian, M. Xu, C. Wu, L. Zhang, et al. 2016. Whole grain intake reduces pancreatic cancer risk: A meta-analysis of observational studies. Medicine 95 (9):e2747. doi: 10.1097/MD.0000000000002747.
  • Lembcke, B., R. Ebert, M. Ptok, W. F. Caspary, W. Creutzfeldt, H. Schicha, and D. Emrich. 1984. Role of gastrointestinal transit in the delay of absorption by viscous fibre (guar). Hepato-gastroenterology 31 (4):183–6.
  • Ley, S. H., O. Hamdy, V. Mohan, and F. B. Hu. 2014. Prevention and management of type 2 diabetes: Dietary components and nutritional strategies. Lancet (London, England) 383 (9933):1999–2007. doi: 10.1016/S0140-6736(14)60613-9.
  • Li, B., G. Zhang, M. Tan, L. Zhao, L. Jin, X. Tang, G. Jiang, and K. Zhong. 2016. Consumption of whole grains in relation to mortality from all causes, cardiovascular disease, and diabetes: Dose-response meta-analysis of prospective cohort studies. Medicine 95 (33):e4229. doi: 10.1097/MD.0000000000004229.
  • Lindgren, O., R. D. Carr, C. F. Deacon, J. J. Holst, G. Pacini, A. Mari, and B. Ahrén. 2011. Incretin hormone and insulin responses to oral versus intravenous lipid administration in humans. The Journal of Clinical Endocrinology and Metabolism 96 (8):2519–24. doi: 10.1210/jc.2011-0266.
  • Liou, A. P., X. Lu, Y. Sei, X. Zhao, S. Pechhold, R. J. Carrero, H. E. Raybould, and S. Wank. 2011. The G-protein-coupled receptor GPR40 directly mediates long-chain fatty acid-induced secretion of cholecystokinin. Gastroenterology 140 (3):903–12. doi: 10.1053/j.gastro.2010.10.012.
  • Lyssenko, V., R. Lupi, P. Marchetti, S. Del Guerra, M. Orho-Melander, P. Almgren, M. Sjögren, C. Ling, K.-F. Eriksson, A.-L. Lethagen, et al. 2007. Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. The Journal of Clinical Investigation 117 (8):2155–63. doi: 10.1172/JCI30706.
  • Ma, X., W. G. Tang, Y. Yang, Q. L. Zhang, J. L. Zheng, and Y. B. Xiang. 2016. Association between whole grain intake and all-cause mortality: A meta-analysis of cohort studies. Oncotarget 7 (38):61996–2005. doi: 10.18632/oncotarget.11491.
  • MacNeil, S., R. M. Rebry, I. J. Tetlow, M. J. Emes, B. McKeown, and T. E. Graham. 2013. Resistant starch intake at breakfast affects postprandial responses in type 2 diabetics and enhances the glucose-dependent insulinotropic polypeptide–insulin relationship following a second meal. Applied Physiology, Nutrition, and Metabolism = Physiologie appliquee, nutrition et metabolisme 38 (12):1187–95. doi: 10.1139/apnm-2013-0023.
  • Maki, K. C., O. M. Palacios, K. Koecher, C. M. Sawicki, K. A. Livingston, M. Bell, H. Nelson Cortes, and N. M. McKeown. 2019. The relationship between whole grain intake and body weight: Results of meta-analyses of observational studies and randomized controlled trials. Nutrients 11 (6):1245. doi: 10.3390/nu11061245.
  • Mano, F., K. Ikeda, E. Joo, Y. Fujita, S. Yamane, N. Harada, and N. Inagaki. 2018. The effect of white rice and white bread as staple foods on gut microbiota and host metabolism. Nutrients 10 (9):1323. doi: 10.3390/nu10091323.
  • Marventano, S., C. Vetrani, M. Vitale, J. Godos, G. Riccardi, and G. Grosso. 2017. whole grain intake and glycaemic control in healthy subjects: A systematic review and meta-analysis of randomized controlled trials. Nutrients 9 (7):769. doi: 10.3390/nu9070769.
  • Maziarz, M. P., S. Preisendanz, S. Juma, V. Imrhan, C. Prasad, and P. Vijayagopal. 2017. Resistant starch lowers postprandial glucose and leptin in overweight adults consuming a moderate-to-high-fat diet: A randomized-controlled trial. Nutrition Journal 16 (1):14. doi: 10.1186/s12937-017-0235-8.
  • Meloni, A. R., M. B. DeYoung, C. Lowe, and D. G. Parkes. 2013. GLP-1 receptor activated insulin secretion from pancreatic β-cells: Mechanism and glucose dependence. Diabetes, Obesity & Metabolism 15 (1):15–27. doi: 10.1111/j.1463-1326.2012.01663.x.
  • Mitsui, R., S. I. Karaki, Y. Kubo, Y. Sugiura, and A. Kuwahara. 2006. Fibre-free diet leads to impairment of neuronally mediated muscle contractile response in rat distal colon. Neurogastroenterology and Motility: The Official Journal of the European Gastrointestinal Motility Society 18 (12):1093–101. doi: 10.1111/j.1365-2982.2006.00847.x.
  • Miyamoto, J., K. Watanabe, S. Taira, M. Kasubuchi, X. Li, J. Irie, H. Itoh, and I. Kimura. 2018. Barley β-glucan improves metabolic condition via short-chain fatty acids produced by gut microbial fermentation in high fat diet fed mice. PloS One 13 (4):e0196579. doi: 10.1371/journal.pone.0196579.
  • Musa-Veloso, K., T. Poon, L. S. Harkness, M. O’Shea, and Y. Chu. 2018. The effects of whole-grain compared with refined wheat, rice, and rye on the postprandial blood glucose response: A systematic review and meta-analysis of randomized controlled trials. The American Journal of Clinical Nutrition 108 (4):759–74. doi: 10.1093/ajcn/nqy112.
  • Nettleton, J. A., N. M. McKeown, S. Kanoni, R. N. Lemaitre, M. F. Hivert, J. Ngwa, F. J. van Rooij, E. Sonestedt, M. K. Wojczynski, Z. Ye, et al. 2010. Interactions of dietary whole-grain intake with fasting glucose- and insulin-related genetic loci in individuals of European descent: A meta-analysis of 14 cohort studies. Diabetes Care 33 (12):2684–91. doi: 10.2337/dc10-1150.
  • Nilsson, A. C., E. M. Ostman, J. J. Holst, and I. M. Björck. 2008. Including indigestible carbohydrates in the evening meal of healthy subjects improves glucose tolerance, lowers inflammatory markers, and increases satiety after a subsequent standardized breakfast. The Journal of Nutrition 138 (4):732–9. doi: 10.1093/jn/138.4.732.
  • Nilsson, A. C., E. V. Johansson-Boll, and I. M. Björck. 2015. Increased gut hormones and insulin sensitivity index following a 3-d intervention with a barley kernel-based product: A randomised cross-over study in healthy middle-aged subjects. The British Journal of Nutrition 114 (6):899–907. doi: 10.1017/S0007114515002524.
  • Nilsson, A., E. Johansson-Boll, J. Sandberg, and I. Björck. 2016. Gut microbiota mediated benefits of barley kernel products on metabolism, gut hormones, and inflammatory markers as affected by co-ingestion of commercially available probiotics: A randomized controlled study in healthy subjects. Clinical Nutrition ESPEN 15:49–56. doi: 10.1016/j.clnesp.2016.06.006.
  • Onaga, T., R. Zabielski, and S. Kato. 2002. Multiple regulation of peptide YY secretion in the digestive tract. Peptides 23 (2):279–90. doi: 10.1016/S0196-9781(01)00609-X.
  • Overduin, J., R. S. Frayo, H. J. Grill, J. M. Kaplan, and D. E. Cummings. 2005. Role of the duodenum and macronutrient type in ghrelin regulation. Endocrinology 146 (2):845–50. doi: 10.1210/en.2004-0609.
  • Pan, X. R., G. W. Li, Y. H. Hu, J. X. Wang, W. Y. Yang, Z. X. An, Z. X. Hu, J. Lin, J. Z. Xiao, H. B. Cao, et al. 1997. Effects of diet and exercise in preventing NIDDM in people with impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care 20 (4):537–44. doi: 10.2337/diacare.20.4.537.
  • Perez-Diaz-del-Campo, N., I. Abete, I. Cantero, B. A. Marin-Alejandre, J. I. Monreal, M. Elorz, J. I. Herrero, A. Benito-Boillos, J. I. Riezu-Boj, F. I. Milagro, et al. 2020. Association of the SH2B1 rs7359397 gene polymorphism with steatosis severity in subjects with obesity and non-alcoholic fatty liver disease. Nutrients 12 (5):1260. doi: 10.3390/nu12051260.
  • Peterson, C. M., R. A. Beyl, K. L. Marlatt, C. K. Martin, K. J. Aryana, M. L. Marco, R. J. Martin, M. J. Keenan, and E. Ravussin. 2018. Effect of 12 wk of resistant starch supplementation on cardiometabolic risk factors in adults with prediabetes: A randomized controlled trial. The American Journal of Clinical Nutrition 108 (3):492–501. doi: 10.1093/ajcn/nqy121.
  • Pfeiffer, A. F. H, and F. Keyhani-Nejad. 2018. High glycemic index metabolic damage – A pivotal role of GIP and GLP-1. Trends in Endocrinology and Metabolism: TEM 29 (5):289–99. doi: 10.1016/j.tem.2018.03.003.
  • Pfeiffer, A. F., N. Rudovich, M. O. Weickert, and F. Isken. 2010. Role of the gut peptide glucose-induced insulinomimetic Peptide in energy balance. Results and Problems in Cell Differentiation 52:183–8. doi: 10.1007/978-3-642-14426-4_15.
  • Pingitore, A., E. S. Chambers, T. Hill, I. R. Maldonado, B. Liu, G. Bewick, D. J. Morrison, T. Preston, G. A. Wallis, C. Tedford, et al. 2017. The diet-derived short chain fatty acid propionate improves beta-cell function in humans and stimulates insulin secretion from human islets in vitro. Diabetes, Obesity & Metabolism 19 (2):257–65. doi: 10.1111/dom.12811.
  • Raben, A., A. Tagliabue, N. J. Christensen, J. Madsen, J. J. Holst, and A. Astrup. 1994. Resistant starch: The effect on postprandial glycemia, hormonal response, and satiety. The American Journal of Clinical Nutrition 60 (4):544–51. doi: 10.1093/ajcn/60.4.544.
  • Rahat-Rozenbloom, S., J. Fernandes, J. Cheng, and T. M. S. Wolever. 2017. Acute increases in serum colonic short-chain fatty acids elicited by inulin do not increase GLP-1 or PYY responses but may reduce ghrelin in lean and overweight humans. European Journal of Clinical Nutrition 71 (8):953–8. doi: 10.1038/ejcn.2016.249.
  • Rahmani, J., A. Miri, R. Černevičiūtė, J. Thompson, N. N. de Souza, R. Sultana, H. Kord Varkaneh, S. M. Mousavi, and A. Hekmatdoost. 2019. Effects of cereal beta-glucan consumption on body weight, body mass index, waist circumference and total energy intake: A meta-analysis of randomized controlled trials. Complementary Therapies in Medicine 43:131–9. doi: 10.1016/j.ctim.2019.01.018.
  • Ramachandran, A., C. Snehalatha, S. Mary, B. Mukesh, A. D. Bhaskar, and V. Vijay. 2006. The Indian Diabetes Prevention Programme shows that lifestyle modification and metformin prevent type 2 diabetes in Asian Indian subjects with impaired glucose tolerance (IDPP-1). Diabetologia 49 (2):289–97. doi: 10.1007/s00125-005-0097-z.
  • Ramracheya, R., C. Chapman, M. Chibalina, H. Dou, C. Miranda, A. González, Y. Moritoh, M. Shigeto, Q. Zhang, M. Braun, et al. 2018. GLP-1 suppresses glucagon secretion in human pancreatic alpha-cells by inhibition of P/Q-type Ca2+ channels. Physiological Reports 6 (17):e13852. doi: 10.14814/phy2.13852.
  • Reimann, F., E. Diakogiannaki, C. E. Moss, and F. M. Gribble. 2020. Cellular mechanisms governing glucose-dependent insulinotropic polypeptide secretion. Peptides 125:170206. doi: 10.1016/j.peptides.2019.170206.
  • Reynolds, A., J. Mann, J. Cummings, N. Winter, E. Mete, and L. Te Morenga. 2019. Te Morenga L. Carbohydrate quality and human health: A series of systematic reviews and meta-analyses. Lancet (London, England) 393 (10170):434–45. doi: 10.1016/S0140-6736(18)31809-9.
  • Ríos, J. L., I. Andújar, G. R. Schinella, and F. Francini. 2019. Modulation of diabetes by natural products and medicinal plants via incretins. Planta Medica 85 (11-12):825–39. doi: 10.1055/a-0897-7492.
  • Robertson, M. D., A. S. Bickerton, A. L. Dennis, H. Vidal, and K. N. Frayn. 2005. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. The American Journal of Clinical Nutrition 82 (3):559–67. doi: 10.1093/ajcn.82.3.559.
  • Roediger, W. E. 1980. Role of anaerobic bacteria in the metabolic welfare of the colonic mucosa in man. Gut 21 (9):793–8. doi: 10.1136/gut.21.9.793.
  • Sandberg, J. C., I. M. E. Björck, and A. C. Nilsson. 2017. Effects of whole grain rye, with and without resistant starch type 2 supplementation, on glucose tolerance, gut hormones, inflammation and appetite regulation in an 11-14.5 hour perspective; a randomized controlled study in healthy subjects. Nutrition Journal 16 (1):25. doi: 10.1186/s12937-017-0246-5.
  • Savage, A. P., T. E. Adrian, G. Carolan, V. K. Chatterjee, and S. R. Bloom. 1987. Effects of peptide YY (PYY) on mouth to caecum intestinal transit time and on the rate of gastric emptying in healthy volunteers. Gut 28 (2):166–70. doi: 10.1136/gut.28.2.166.
  • Scheppach, W. 1994. Effects of short chain fatty acids on gut morphology and function. Gut 35 (1 Suppl):S35–S8. doi: 10.1136/gut.35.1_suppl.s35.
  • Schlesinger, S., M. Neuenschwander, C. Schwedhelm, G. Hoffmann, A. Bechthold, H. Boeing, and L. Schwingshackl. 2019. Food groups and risk of overweight, obesity, and weight gain: A systematic review and dose-response meta-analysis of prospective studies. Advances in Nutrition (Bethesda, Md.) 10 (2):205–18. doi: 10.1093/advances/nmy092.
  • Silva, F. M., C. K. Kramer, D. Crispim, and M. J. Azevedo. 2015. A high-glycemic index, low-fiber breakfast affects the postprandial plasma glucose, insulin, and ghrelin responses of patients with type 2 diabetes in a randomized clinical trial. The Journal of Nutrition 145 (4):736–41. doi: 10.3945/jn.114.195339.
  • Śliwińska-Mossoń, M., G. Marek, and H. Milnerowicz. 2017. The role of pancreatic polypeptide in pancreatic diseases. Advances in Clinical and Experimental Medicine: Official Organ Wroclaw Medical University 26 (9):1447–55. doi: 10.17219/acem/65094.
  • Snelson, M., J. Jong, D. Manolas, S. Kok, A. Louise, R. Stern, and N. J. Kellow. 2019. Metabolic effects of resistant starch type 2: A systematic literature review and meta-analysis of randomized controlled trials. Nutrients 11 (8):1833. doi: 10.3390/nu11081833.
  • Stančáková, A., J. Paananen, P. Soininen, A. J. Kangas, L. L. Bonnycastle, M. A. Morken, F. S. Collins, A. U. Jackson, M. L. Boehnke, J. Kuusisto, et al. 2011. Effects of 34 risk loci for type 2 diabetes or hyperglycemia on lipoprotein subclasses and their composition in 6,580 nondiabetic Finnish men. Diabetes 60 (5):1608–16. doi: 10.2337/db10-1655.
  • Steemburgo, T., M. J. Azevedo, J. L. Gross, F. I. Milagro, J. Campión, and J. A. Martínez. 2013. The rs9939609 polymorphism in the FTO gene is associated with fat and fiber intakes in patients with type 2 diabetes. Journal of Nutrigenetics and Nutrigenomics 6 (2):97–106. doi: 10.1159/000350741.
  • Stefoska-Needham, A., E. J. Beck, S. K. Johnson, J. Chu, and L. C. Tapsell. 2016. Flaked sorghum biscuits increase postprandial GLP-1 and GIP levels and extend subjective satiety in healthy subjects. Molecular Nutrition & Food Research 60 (5):1118–28. doi: 10.1002/mnfr.201500672.
  • Stewart, M. L., S. D. Nikhanj, D. A. Timm, W. Thomas, and J. L. Slavin. 2010. Evaluation of the effect of four fibers on laxation, gastrointestinal tolerance and serum markers in healthy humans. Annals of Nutrition & Metabolism 56 (2):91–8. doi: 10.1159/000275962.
  • Stidsen, J. V., J. E. Henriksen, M. H. Olsen, R. W. Thomsen, J. S. Nielsen, J. Rungby, S. P. Ulrichsen, K. Berencsi, J. A. Kahlert, S. G. Friborg, et al. 2018. Pathophysiology-based phenotyping in type 2 diabetes: A clinical classification tool. Diabetes/Metabolism Research and Reviews 34 (5):e3005. doi: 10.1002/dmrr.3005.
  • Stringer, D. M., C. G. Taylor, P. Appah, H. Blewett, and P. Zahradka. 2013. Consumption of buckwheat modulates the post-prandial ­response of selected gastrointestinal satiety hormones in individuals with type 2 diabetes mellitus. Metabolism: clinical and Experimental 62 (7):1021–31. doi: 10.1016/j.metabol.2013.01.021.
  • Tang, G., D. Wang, J. Long, F. Yang, and L. Si. 2015. Meta-analysis of the association between whole grain intake and coronary heart disease risk. The American Journal of Cardiology 115 (5):625–9. doi: 10.1016/j.amjcard.2014.12.015.
  • Tang-Christensen, M., P. J. Larsen, J. Thulesen, J. Rømer, and N. Vrang. 2000. The proglucagon-derived peptide, glucagon-like peptide-2, is a neurotransmitter involved in the regulation of food intake. Nature Medicine 6 (7):802–7. doi: 10.1038/77535.
  • Tannous dit El Khoury, D., O. Obeid, S. T. Azar, and N. Hwalla. 2006. Variations in postprandial ghrelin status following ingestion of high-carbohydrate, high-fat, and high-protein meals in males. Annals of Nutrition & Metabolism 50 (3):260–9. doi: 10.1159/000091684.
  • Tuomilehto, J., J. Lindström, J. G. Eriksson, T. T. Valle, H. Hämäläinen, P. Ilanne-Parikka, S. Keinänen-Kiukaanniemi, M. Laakso, A. Louheranta, M. Rastas, et al. 2001. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. The New England Journal of Medicine 344 (18):1343–50. doi: 10.1056/NEJM200105033441801.
  • Turton, M. D., D. O’Shea, I. Gunn, S. A. Beak, C. M. Edwards, K. Meeran, S. J. Choi, G. M. Taylor, M. M. Heath, P. D. Lambert, et al. 1996. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 379 (6560):69–72. doi: 10.1038/379069a0.
  • Veedfald, S., L. Vedtofte, K. Skov-Jeppesen, C. F. Deacon, B. Hartmann, T. Vilsbøll, F. K. Knop, M. B. Christensen, and J. J. Holst. 2020. Glucose-dependent insulinotropic polypeptide is a pancreatic polypeptide secretagogue in humans. Journal of Clinical Endocrinology and Metabolism. (3):1;105.
  • Villegas, R., R. J. Goodloe, B. E. McClellan, Jr, J. Boston, and D. C. Crawford. 2014. Gene-carbohydrate and gene-fiber interactions and type 2 diabetes in diverse populations from the National Health and Nutrition Examination Surveys (NHANES) as part of the Epidemiologic Architecture for Genes Linked to Environment (EAGLE) study. BMC Genetics 15:69. doi: 10.1186/1471-2156-15-69.
  • Vimaleswaran, K. S., D. Bodhini, N. Lakshmipriya, K. Ramya, R. M. Anjana, V. Sudha, J. A. Lovegrove, S. Kinra, V. Mohan, V. Radha, et al. 2016. Interaction between FTO gene variants and lifestyle factors on metabolic traits in an Asian Indian population [published correction appears in Nutr Metab (Lond). 2016;13:41]. Nutrition & Metabolism 13:41. doi: 10.1186/s12986-016-0100-3.
  • Vitaglione, P., R. B. Lumaga, A. Stanzione, L. Scalfi and V. Fogliano. 2009. beta-Glucan-enriched bread reduces energy intake and modifies plasma ghrelin and peptide YY concentrations in the short term. Appetite 53 (3):338–44.
  • Wang, F., G. Yu, Y. Zhang, B. Zhang, and J. Fan. 2015. Dipeptidyl peptidase IV inhibitory peptides derived from oat (Avena sativa L.), buckwheat (Fagopyrum esculentum), and highland barley (Hordeum vulgare trifurcatum (L.) trofim) proteins. Journal of Agricultural and Food Chemistry 63 (43):9543–9. doi: 10.1021/acs.jafc.5b04016.
  • Wang, Y., R. Chandra, L. A. Samsa, B. Gooch, B. E. Fee, J. M. Cook, S. R. Vigna, A. O. Grant, and R. A. Liddle. 2011. Amino acids stimulate cholecystokinin release through the Ca2+-sensing receptor. American Journal of Physiology. Gastrointestinal and Liver Physiology 300 (4):G528–37. doi: 10.1152/ajpgi.00387.2010.
  • Weickert, M. O, and A. F. H. Pfeiffer. 2018. Impact of dietary fiber consumption on insulin resistance and the prevention of type 2 diabetes. The Journal of Nutrition 148 (1):7–12. doi: 10.1093/jn/nxx008.
  • Weickert, M. O, and A. F. Pfeiffer. 2008. Metabolic effects of dietary fibre consumption and prevention of diabetes. The Journal of Nutrition 138 (3):439–42. doi: 10.1093/jn/138.3.439.
  • Weickert, M. O., M. Roden, F. Isken, D. Hoffmann, P. Nowotny, M. Osterhoff, M. Blaut, C. Alpert, O. Gögebakan, C. Bumke-Vogt, et al. 2011. Effects of supplemented isoenergetic diets differing in cereal fiber and protein content on insulin sensitivity in overweight ­humans. The American Journal of Clinical Nutrition 94 (2):459–71. doi: 10.3945/ajcn.110.004374.
  • Weitkunat, K., S. Schumann, D. Nickel, K. A. Kappo, K. J. Petzke, A. P. Kipp, M. Blaut, and S. Klaus. 2016. Importance of propionate for the repression of hepatic lipogenesis and improvement of insulin sensitivity in high-fat diet-induced obesity. Molecular Nutrition & Food Research 60 (12):2611–21. doi: 10.1002/mnfr.201600305.
  • Weitkunat, K., S. Schumann, D. Nickel, S. Hornemann, K. J. Petzke, M. B. Schulze, A. F. Pfeiffer, and S. Klaus. 2017. Odd-chain fatty acids as a biomarker for dietary fiber intake: A novel pathway for endogenous production from propionate. The American Journal of Clinical Nutrition 105 (6):1544–51. doi: 10.3945/ajcn.117.152702.
  • White, U., C. M. Peterson, R. A. Beyl, C. K. Martin, and E. Ravussin. 2020. Resistant starch has no effect on appetite and food intake in individuals with prediabetes. Journal of the Academy of Nutrition and Dietetics 120 (6):1034–41. doi: 10.1016/j.jand.2020.01.017.
  • Widenmaier, S. B., Z. Ao, S. J. Kim, G. Warnock, and C. H. McIntosh. 2009. Suppression of p38 MAPK and JNK via Akt-mediated inhibition of apoptosis signal-regulating kinase 1 constitutes a core component of the beta-cell pro-survival effects of glucose-dependent insulinotropic polypeptide. The Journal of Biological Chemistry 284 (44):30372–82. doi: 10.1074/jbc.M109.060178.
  • Wolever, T. M. S., S. M. Tosh, S. E. Spruill, A. L. Jenkins, A. Ezatagha, R. Duss, J. Johnson, Y. Chu, and R. E. Steinert. 2020. Increasing oat β-glucan viscosity in a breakfast meal slows gastric emptying and reduces glycemic and insulinemic responses but has no effect on appetite, food intake, or plasma ghrelin and PYY responses in healthy humans: A randomized, placebo-controlled, crossover trial. The American Journal of Clinical Nutrition 111 (2):319–28. doi: 10.1093/ajcn/nqz285.
  • Xia, Y., S. Zhang, Q. Zhang, L. Liu, G. Meng, H. Wu, X. Bao, Y. Gu, S. Sun, X. Wang, et al. 2020. Insoluble dietary fibre intake is associated with lower prevalence of newly-diagnosed non-alcoholic fatty liver disease in Chinese men: A large population-based cross-sectional study. Nutrition & Metabolism 17:4. doi: 10.1186/s12986-019-0420-1.
  • Xu, Y., Q. Wan, J. Feng, L. Du, K. Li, and Y. Zhou. 2018. Whole grain diet reduces systemic inflammation: A meta-analysis of 9 randomized trials. Medicine 97 (43):e12995. doi: 10.1097/MD.0000000000012995.
  • Ye, Z., V. Arumugam, E. Haugabrooks, P. Williamson, and S. Hendrich. 2015. Soluble dietary fiber (Fibersol-2) decreased hunger and increased satiety hormones in humans when ingested with a meal. Nutrition Research (New York, N.Y.) 35 (5):393–400. doi: 10.1016/j.nutres.2015.03.004.
  • Zain, S. M., Z. Mohamed, and R. Mohamed. 2015. Common variant in the glucokinase regulatory gene rs780094 and risk of nonalcoholic fatty liver disease: A meta-analysis. Journal of Gastroenterology and Hepatology 30 (1):21–7. doi: 10.1111/jgh.12714.
  • Zamaratskaia, G., D. P. Johansson, M. A. Junqueira, L. Deissler, M. Langton, P. M. Hellström, and R. Landberg. 2017. Impact of sourdough fermentation on appetite and postprandial metabolic responses: A randomised cross-over trial with whole grain rye crispbread. The British Journal of Nutrition 118 (9):686–97. doi: 10.1017/S000711451700263X.
  • Zaremba, S. M. M., I. F. Gow, S. Drummond, J. T. McCluskey, and R. E. Steinert. 2018. Effects of oat β-glucan consumption at breakfast on ad libitum eating, appetite, glycemia, insulinemia and GLP-1 concentrations in healthy subjects. Appetite 128:197–204. doi: 10.1016/j.appet.2018.06.019.
  • Zhang, B., Q. Zhao, W. Guo, W. Bao, and X. Wang. 2018. Association of whole grain intake with all-cause, cardiovascular, and cancer mortality: A systematic review and dose-response meta-analysis from prospective cohort studies. European Journal of Clinical Nutrition 72 (1):57–65. doi: 10.1038/ejcn.2017.149.
  • Zhang, L., Y. Ouyang, H. Li, L. Shen, Y. Ni, Q. Fang, G. Wu, L. Qian, Y. Xiao, J. Zhang, et al. 2019. Metabolic phenotypes and the gut microbiota in response to dietary resistant starch type 2 in normal-weight subjects: A randomized crossover trial. Scientific Reports 9 (1):4736. doi: 10.1038/s41598-018-38216-9.
  • Zhao, L., F. Zhang, X. Ding, G. Wu, Y. Y. Lam, X. Wang, H. Fu, X. Xue, C. Lu, J. Ma, et al. 2018. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science (New York, N.Y.) 359 (6380):1151–6. doi: 10.1126/science.aao5774.
  • Zhou, D., Y.-W. Chen, Z.-H. Zhao, R.-X. Yang, F.-Z. Xin, X.-L. Liu, Q. Pan, H. Zhou, and J.-G. Fan. 2018. Sodium butyrate reduces high-fat diet-induced non-alcoholic steatohepatitis through upregulation of hepatic GLP-1R expression. Experimental & Molecular Medicine 50 (12):1–12. doi: 10.1038/s12276-018-0183-1.