Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-20T10:11:15.243Z Has data issue: false hasContentIssue false

Dietary fibre and the gut–brain axis: microbiota-dependent and independent mechanisms of action

Published online by Cambridge University Press:  08 September 2021

Danique La Torre
Affiliation:
Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium Leuven Brain Institute, KU Leuven, Leuven, Belgium
Kristin Verbeke
Affiliation:
Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium Leuven Brain Institute, KU Leuven, Leuven, Belgium
Boushra Dalile*
Affiliation:
Translational Research Center in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism, Faculty of Medicine, KU Leuven, Leuven, Belgium Leuven Brain Institute, KU Leuven, Leuven, Belgium
*
*Corresponding author. Email: boushra.dalile@kuleuven.be

Abstract

Dietary fibre is an umbrella term comprising various types of carbohydrate polymers that cannot be digested nor absorbed by the human small intestine. Consumption of dietary fibre is linked to beneficial effects on cognitive and affective processes, although not all fibres produce the same effects. Fibres that increase short-chain fatty acid (SCFA) production following modulation of the gut microbiota are thought to be the most potent fibres to induce effects on cognitive and affective processes. SCFAs can exert their effects by improving central, peripheral and systemic immunity, lowering hypertension and enhancing intestinal barrier integrity. Here, we propose additional mechanisms by which dietary fibres may contribute to improvements in affective and cognitive processes. Fibre-induced modulation of the gut microbiota may influence affective processes and cognition by increasing brain-derived neurotrophic factor levels. Depending on the physicochemical properties of dietary fibre, additional effects on affect and cognition may occur via non-microbiota-related routes, such as enhancement of the immune system and lowering cholesterol levels and subsequently lowering blood pressure. Mechanistic randomised placebo-controlled trials are needed to establish the effects of dietary fibre consumption and the magnitude of explained variance in affect and cognition when incorporating measurements of microbiota-dependent and microbiota-independent mechanisms in humans.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2021. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

Dietary fibre, which is derived from fruits and vegetables, legumes, whole-grain breads, and cereals, is an umbrella term comprising various types of carbohydrate polymers that cannot be digested nor absorbed by the human small intestine. Consequently, dietary fibre ends up in the colon, where it is fermented by the gut microbiota. Consumption of dietary fibre has been linked to various beneficial physiological effects, including effects on affective (Lawton et al., Reference Lawton, Walton, Hoyland, Howarth, Allan, Chesters and Dye2013) and cognitive (Sandberg et al., Reference Sandberg, Björck and Nilsson2018) processes. Notably, dietary fibre consumption can aid in prevention or treatment of symptoms of depression, anxiety, and stress (Jacka et al., Reference Jacka, O’Neil, Opie, Itsiopoulos, Cotton, Mohebbi, Castle, Dash, Mihalopoulos, Chatterton, Brazionis, Dean and Hodge2017; Silk et al., Reference Silk, Davis, Vulevic, Tzortzis and Gibson2009), which are psychological states characterised by negative mood (e.g., worry, fear, sadness, loss of interest, or emotional tension) that are often accompanied by impaired facets of cognition. Moreover, preclinical data suggests that impaired facets of cognition, such as attention, mental flexibility, and executive functioning, can be restored by dietary fibre (Chunchai et al., Reference Chunchai, Thunapong, Yasom, Wanchai, Eaimworawuthikul, Metzler, Lungkaphin, Pongchaidecha, Sirilun, Chaiyasut, Pratchayasakul, Thiennimitr, Chattipakorn and Chattipakorn2018).

Since dietary fibres vary in origin, chemical composition, and physicochemical properties (comprising solubility, fermentability, and viscosity), they can exert different effects on the body. These differences in physicochemical properties might differentially affect the communicative pathways to the brain, and consequently variably influence affective and cognitive processes. Commonly accepted mechanisms by which dietary fibres are thought to affect cognitive and affective processes are the gut microbiome and short chain fatty acids (SCFAs) (Dalile et al., Reference Dalile, Van Oudenhove, Vervliet and Verbeke2019). Here, we shed light on additional potential mechanisms that may contribute to the positive effects of dietary fibres on these processes, such as the immune system, cholesterol, the intestinal barrier, brain-derived neurotrophic factor (BDNF), and blood pressure, and would help explain additional variance that is not explained by SCFAs. Since in human studies most evidence is associative, we also included animal studies that assess the impact of dietary fibre intake on these various mechanisms. To this end, the current review assesses microbiota-dependent and microbiota-independent biological mechanisms that may underlie the effects of dietary fibre on affective processes and cognition and outlines which physicochemical properties may predict such effects.

Dietary fibres: a definition

Since dietary fibres are characteristically heterogeneous, different classifications have been used to define them, including origin, chemical composition, and physicochemical properties with additional subcategorisation based on the degree of polymerisation. The most commonly used definition of dietary fibre is according to the Codex Alimentarius, which states that dietary fibres include (1) edible carbohydrate polymers occurring in foods as consumed, (2) edible carbohydrates obtained from food raw materials by physical, enzymatic or chemical means and (3) synthetic carbohydrate polymers, which have beneficial physiological effect demonstrated by generally accepted scientific evidence (Joint FAO/WHO Food Standards Programme Secretariat of the CODEX Alimentarius Commission, 2010). In 2016, the Food and Drug Administration (FDA) defined dietary fibre as either (1) non-digestible soluble and insoluble carbohydrates (with three or more monomeric units), and lignin that are intrinsic and intact in plants, or (2) isolated or synthetic non-digestible carbohydrates (with three or more monomeric units), and induce physiological effects that are beneficial to human health (Food and Drug Administration, 2020). At that time, only seven non-digestible carbohydrates were included in the latter category (β-glucan soluble fibre, psyllium husk, cellulose, guar gum, pectin, locust bean gum, and hydroxypropylmethylcellulose). Since then, the list has been extended with ten additional types of dietary fibre [mixed plant cell wall fibres, arabinoxylan (AX), alginate, inulin, and inulin-type fructans (ITFs), high amylose starch (resistant starch 2), galactooligosaccharides (GOS), polydextrose, resistant maltodextrin/dextrin, cross linked phosphorylated RS4, and glucomannan]. Certain types of fibres, such as ITFs and GOS fulfil the criteria of a prebiotic. Prebiotics are defined as “a substrate that is selectively utilised by host microorganisms conferring a health benefit” (Gibson et al., Reference Gibson, Hutkins, Ellen Sanders, Prescott, Reimer, Salminen, Scott, Stanton, Swanson, Cani, Verbeke and Reid2017).

Properties of dietary fibres

The physicochemical characteristics of dietary fibres are subtyped by solubility, viscosity and fermentability. Table 1 provides an overview of the most frequently investigated types of fibres and their concomitant physicochemical properties.

Table 1. Overview of different types of dietary fibres with their concomitant origin, structure and degree of solubility, viscosity and fermentability.

Solubility

Dietary fibres can be water-soluble or water-insoluble, with most plant foods containing a mixture of both. Approximately 20 per cent of consumed dietary fibre is soluble, whereas 80 per cent is insoluble. Insoluble fibres consist mainly of cellulose, hemicellulose, and lignin, and are primarily present in wheat bran, most grain products and vegetables (Cho and Dreher, Reference Cho and Dreher2001). Soluble fibres consist of polysaccharides such as pectin, mucilage, and gum, and are predominantly found in some fruits (e.g., dried prunes, oranges, and grapefruit), oats, barley, dried beans, and legumes (e.g., lentils and pinto beans) (Cho and Dreher, Reference Cho and Dreher2001). Although soluble fibres are less prevalent in foods than insoluble fibres, they have an important influence on digestive and absorptive processes, such as delaying gastric emptying, decreasing glucose absorption, enhancing immune function, and lowering total and low-density lipoprotein (LDL) cholesterol levels (Cho and Dreher, Reference Cho and Dreher2001; Jenkins et al., Reference Jenkins, Kendall, Axelsen, Augustin and Vuksan2000). Insoluble fibres are important for shortening bowel transit time, increasing faecal bulk, and softening stool (Cho and Dreher, Reference Cho and Dreher2001).

Viscosity

Viscosity, the capacity to gel with water, depends on the solubility of the fibre with soluble fibres having higher viscosity. Viscous fibres reduce postprandial glucose response after carbohydrate-rich meals, lower total and LDL cholesterol levels (Jenkins et al., Reference Jenkins, Kendall, Axelsen, Augustin and Vuksan2000; McRorie and McKeown, Reference McRorie and McKeown2017), and slow gastric emptying and macronutrient absorption from the gut (McRorie and McKeown, Reference McRorie and McKeown2017).

Fermentability

The fermentability of dietary fibre varies greatly, ranging from not at all (e.g., lignin) to almost complete fermentation (e.g., pectin) (Mudgil and Barak, Reference Mudgil and Barak2013). Soluble fibres are fermented in the colon, whereas insoluble fibres are less and more slowly fermented. The fermentation of soluble fibres results in the production of short-chain fatty acids (SCFAs) which, in turn, exhibit anti-inflammatory effects in the gut, maintain intestinal barrier integrity (Lewis et al., Reference Lewis, Lutgendorff, Phan, Söderholm, Sherman and McKay2010), induce mucin secretion in the gastrointestinal tract (Monk et al., Reference Monk, Lepp, Wu, Pauls, Robinson and Power2017), and promote gastrointestinal motility (Cherbut et al., Reference Cherbut, Ferrier, Rozé, Anini, Blottière, Lecannu and Galmiche1998). In addition to solubility, chain length and particle size determine fermentability with shorter polymers and small particles being more readily fermented. In in vitro incubation experiments, neutral pectin fractions produced more SCFAs and more acetate than acidic fractions, whereas rhamnose and arabinan produced high proportions of propionate (Onumpai et al., Reference Onumpai, Kolida, Bonnin and Rastall2011). Wood derived xylan and mannan derivatives produced similar total amounts of SCFAs as fructooligosaccharides (FOS) but proportionally more propionate and butyrate, respectively (La Rosa et al., Reference La Rosa, Kachrimanidou, Buffetto, Pope, Pudlo, Martens, Rastall, Gibson and Westereng2019). In contrast, the glycosidic bond configuration has little impact on SCFA production (Harris et al., Reference Harris, Edwards and Morrison2017).

Dietary fibre studies on cognitive and affective processes

Here, we provide a brief overview of the state of the literature on the effects of dietary fibre on cognitive and affective processes for the sake of completeness. Cross-sectional and prospective cohort studies across the lifespan continue to show positive associations between higher dietary fibre intake and better cognitive and affective processes (Muth and Park, Reference Muth and Park2021; Swann et al., Reference Swann, Kilpatrick, Breslin and Oddy2020). Affective processes are measured by, for instance, better mental health quality of life and lower incidents of depression and anxiety. Moreover, dietary source of fibres appears to be differentially associated with the incidence of depression (Kim et al., Reference Kim, Byeon and Shin2020). Despite assessing effects on the short-term, human interventional studies enable controlled administration of specific fibres and exclusion of confounding effects. These studies are displayed in Table 2. Briefly, 18 studies were found, with 7 of them (Azpiroz et al., Reference Azpiroz, Dubray, Bernalier-Donadille, Cardot, Accarino, Serra, Wagner, Respondek and Dapoigny2017; Buigues et al., Reference Buigues, Fernández-Garrido, Pruimboom, Hoogland, Navarro-Martínez, Martínez-Martínez, Verdejo, Carmen Mascarós, Peris and Cauli2016; Dehhaghi et al., Reference Dehhaghi, Kazemi, Panahi and Guillemin2019; Farhangi et al., Reference Farhangi, Zare Javid, Sarmadi, Karimi and Dehghan2018; Grimaldi et al., Reference Grimaldi, Gibson, Vulevic, Giallourou, Castro-Mejía, Hansen, Leigh Gibson, Nielsen and Costabile2018; Kao et al., Reference Kao, Safarikova, Marquardt, Mullins and Lennox2019; Silk et al., Reference Silk, Davis, Vulevic, Tzortzis and Gibson2009) conducted in populations with various mental, physical, or gastrointestinal disorders, and the remaining 11 conducted in healthy populations (Best et al., Reference Best, Bryan and Burns2008, Reference Best, Kemps and Bryan2009, Reference Best, Howe, Bryan, Buckley and Scholey2015; Childs et al., Reference Childs, Röytiö, Alhoniemi, Fekete, Forssten, Hudjec, Lim, Steger, Yaqoob, Tuohy, Rastall, Ouwehand and Gibson2014; Lawton et al., Reference Lawton, Walton, Hoyland, Howarth, Allan, Chesters and Dye2013; Pasman et al., Reference Pasman, Blokdijk, Bertina, Hopman and Hendriks2003; Ramnani et al., Reference Ramnani, Costabile, Bustillo and Gibson2015; Schmidt et al., Reference Schmidt, Cowen, Harmer, Tzortzis, Errington and Burnet2015; Smith, Reference Smith2005; Smith et al., Reference Smith, Sutherland and Hewlett2015; Talbott and Talbott, Reference Talbott and Talbott2009). The duration of the intervention periods varied, with three studies carrying out single administration (Best et al., Reference Best, Bryan and Burns2008, Reference Best, Howe, Bryan, Buckley and Scholey2015; Smith et al., Reference Smith, Sutherland and Hewlett2015), one study comprising 2 days (Pasman et al., Reference Pasman, Blokdijk, Bertina, Hopman and Hendriks2003), and the others comprising 2–13 weeks (Azpiroz et al., Reference Azpiroz, Dubray, Bernalier-Donadille, Cardot, Accarino, Serra, Wagner, Respondek and Dapoigny2017; Best et al., Reference Best, Kemps and Bryan2009; Buigues et al., Reference Buigues, Fernández-Garrido, Pruimboom, Hoogland, Navarro-Martínez, Martínez-Martínez, Verdejo, Carmen Mascarós, Peris and Cauli2016; Childs et al., Reference Childs, Röytiö, Alhoniemi, Fekete, Forssten, Hudjec, Lim, Steger, Yaqoob, Tuohy, Rastall, Ouwehand and Gibson2014; Farhangi et al., Reference Farhangi, Zare Javid, Sarmadi, Karimi and Dehghan2018; Grimaldi et al., Reference Grimaldi, Gibson, Vulevic, Giallourou, Castro-Mejía, Hansen, Leigh Gibson, Nielsen and Costabile2018; Kao et al., Reference Kao, Safarikova, Marquardt, Mullins and Lennox2019; Kazemi et al., Reference Kazemi, Noorbala, Azam, Eskandari and Djafarian2019; Lawton et al., Reference Lawton, Walton, Hoyland, Howarth, Allan, Chesters and Dye2013; Ramnani et al., Reference Ramnani, Costabile, Bustillo and Gibson2015; Schmidt et al., Reference Schmidt, Cowen, Harmer, Tzortzis, Errington and Burnet2015; Silk et al., Reference Silk, Davis, Vulevic, Tzortzis and Gibson2009; Smith, Reference Smith2005; Talbott and Talbott, Reference Talbott and Talbott2009). Eleven studies utilised ITFs, with the majority of them (nine studies) showing some positive effects on affective and cognitive indices. Five studies utilised mixtures of fibre, mostly revealing positive effects of interventions, unless it was administered only once. One study administrated β-glucans and one study administered resistant starch. Not all studies are of equal quality, with some lacking proper control conditions. Notably, most human intervention studies administered dietary fibre supplements rather than whole foods. Whole foods are not only rich in dietary fibre but also contain minerals and antioxidants that may improve mood and cognition (Gomez-Pinilla, Reference Gomez-Pinilla2008). Moreover, it cannot be excluded that some placebos (e.g., maltodextrin) may have had an effect on mood and/or cognition as well, possibly via gut-independent mechanisms (Kendig et al., Reference Kendig, Lin, Beilharz, Rooney and Boakes2014). Future intervention studies should take such effects into account. Some fibres such as pectin, hemicelluloses (e.g., AX), and gums (e.g., guar, xanthan, and Arabic gum) are yet to be explored in the context of mood and cognition. As these fibres are highly viscous, they may affect mood and cognition through microbiota-independent mechanisms as discussed below. Overall, the majority of studies point to beneficial effects on mood and cognition, but none of these studies causally addresses potential mechanisms of action. In the next section, we will discuss potential mechanisms, both microbiota-dependent as well as microbiota-independent, that may be involved in dietary fibre’s beneficial effects on affective and cognitive processes.

Table 2. Human studies assessing the effect of dietary fibre interventions on mood and cognition.

Abbreviations: GOS, galactooligosaccharides; FOS, fructooligosaccharides; XOS, xylooligosaccharides.

a CAR a measure of hypothalamic–pituitary–adrenal (HPA) axis function (Clow et al., Reference Clow, Hucklebridge, Stalder, Evans and Thorn2010). CAR has been previously associated with anticipation of daily stress (Fries et al., Reference Fries, Dettenborn and Kirschbaum2009) and executive function (Butler et al., Reference Butler, Klaus, Edwards and Pennington2017).

Biological mechanisms

Dietary fibre may affect mood and cognition via various biological mechanisms, both microbiota-dependent and microbiota-independent, which are reviewed in this section. Figure 1 provides an overview of the different biological mechanisms that may underlie dietary fibres’ effect on affective and cognitive processes according to their physicochemical properties.

Figure 1 Overview of potential biological mechanisms underlying dietary fibres’ effects on mood and cognition according to physicochemical properties. The degree of viscosity, fermentability, and solubility of dietary fibres influences gut microbiota composition and function, the immune system, as well as cholesterol levels, through which mood and cognition can be modulated. Via the direct immune route, viscous, fermentable, and soluble fibres can reduce pro-inflammatory cytokines, lower numbers and activation of splenic macrophages and DCs, and increase neutrophils in the blood. Cholesterol levels can be lowered by viscous, fermentable, and soluble fibres and impact the brain, but can also indirectly affect it by reducing hypertension. Fermentability and solubility of dietary fibre modulate the gut microbiota, by which it can influence affective and cognitive processes via SCFA-dependent mechanisms or BDNF levels. SCFAs can lower hypertension as well as improve immune function and intestinal barrier integrity. Via the indirect immune route, SCFAs suppress pro-inflammatory reactions by reducing pro-inflammatory cytokines, increasing anti-inflammatory cytokines and Treg cells, as well as restoring microglial cell morphology and reversing microglial immaturity. All these mechanisms have been associated with modulation of mood and cognition in health and disease. The likelihood that the different biological mechanisms underlie dietary fibres’ effect on mood and cognition are displayed in proportion, with larger cubicle areas reflecting increased potential. The green plus sign indicates beneficial effects. BDNF, brain-derived neurotrophic factor; DCs, dendritic cells; Treg cells, regulatory T-cells; SCFAs, short chain fatty acids.

Gut microbiome

Dietary fibre’s ability to influence affective and cognitive processes is most likely through its interactions with the gut microbiome. The gut microbiome refers to the 1 × 1013 to 1 × 1014 microorganisms that reside in the human large intestine, which is one of the most densely and diversely colonised organs in the human body. Pre-clinical studies suggest that the gut microbiome plays an important modulating role in affective and cognitive processes. Studies in germ-free (GF) mice suggested a causal role of the gut microbiota in developing brain function and behaviour, as those mice exhibit compromised social, cognitive, and anxiety-like behaviours (Desbonnet et al., Reference Desbonnet, Clarke, Shanahan, Dinan and Cryan2014), such as reduced non-spatial memory, lower social motivation, and symptoms of anxiety compared to their conventionally raised specific pathogen-free (SPF) counterparts (Clarke et al., Reference Clarke, Grenham, Scully, Fitzgerald, Moloney, Shanahan, Dinan and Cryan2013; Desbonnet et al., Reference Desbonnet, Clarke, Shanahan, Dinan and Cryan2014). Also, faecal microbiota transplantation of “depressive microbiota” derived from patients with major depressive disorder (MDD; a disorder specifically known for its symptoms of negative mood and impaired cognition) to GF mice (Zheng et al., Reference Zheng, Zeng, Zhou, Liu, Fang, Xu, Zeng, Chen, Fan, Du, Zhang, Yang, Yang, Meng, Li, Melgiri, Licinio, Wei and Xie2016) resulted in depression-like behaviours relative to colonisation with “healthy microbiota” derived from healthy donors.

Human studies also suggest that differences in the gut microbiome composition are associated with mood and cognition. For instance, gut microbiota composition of healthy subjects differs from that of patients suffering from MDD. Specifically, the relative abundance of Firmicutes (Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin, Wang, Tang, Tan, Shi, Li and Ruan2015), Bifidobacteria and Lactobacilli (Aizawa et al., Reference Aizawa, Tsuji, Asahara, Takahashi, Teraishi, Yoshida, Ota, Koga, Hattori and Kunugi2016) appears lower in MDD patients compared to healthy controls, whereas Actinobacteria, Proteobacteria, and Bacteroidetes appear to be higher (Jiang et al., Reference Jiang, Ling, Zhang, Mao, Ma, Yin, Wang, Tang, Tan, Shi, Li and Ruan2015). Increases in the relative abundance of Bifidobacteria and Lactobacilli in particular have been associated with lower anxiety and better memory and learning in both rodents (Bravo et al., Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock, Cryan and Designed2011) and humans (Messaoudi et al., Reference Messaoudi, Lalonde, Violle, Javelot, Desor, Nejdi, Bisson, Rougeot, Pichelin, Cazaubiel and Cazaubiel2011). Nevertheless, it seems unlikely that bacterial strains are directly responsible for these effects. Rather, it is speculated that bacterial metabolites like SCFAs and BDNF mediate the interaction between the microbiota and psychological functioning.

Short-chain fatty acids

Soluble fibres (e.g., pectin and gums) provide substrates for bacterial fermentation, which in turn leads to the production of SCFAs in the colon. Acetate, propionate, and butyrate are the most abundant SCFAs, and they are present in the colon in an approximate molar ratio of 60:20:20, respectively. Type and availability of substrate, as well as gut transit time and composition of the gut microbiota influence the levels and relative proportions of SCFAs in the colon. Following their production by microbial fermentation of dietary fibre, SCFAs are rapidly absorbed by colonic cells via monocarboxylate transporters and produce energy for the cells. Those that are not absorbed travel via the basolateral membrane into the portal circulation. In the liver, SCFAs are incorporated in glucose, cholesterol, and fatty acids, thus only small amounts actually reach systemic circulation, and subsequently the brain (Boets et al., Reference Boets, Gomand, Deroover, Preston, Vermeulen, De Preter, Hamer, Van Den Mooter, De Vuyst, Courtin, Annaert, Delcour and Verbeke2017).

SCFAs and cognitive and affective processes

While numerous animal studies have relied on intracerebroventricular (ICV), intraperitoneal, subcutaneous, and oral administrations to deliver SCFAs and investigate their impact on brain and behaviour (see Dalile et al., Reference Dalile, Van Oudenhove, Vervliet and Verbeke2019), few studies explored whether the effects of dietary fibre/prebiotics on affective and cognitive functioning are mediated by or correlated with the observed increases in SCFA levels. One of these studies showed that FOS- and GOS-induced increases in cecal SCFAs correlated with effects on depressive and anxious behaviour and stress responses in mice (Burokas et al., Reference Burokas, Arboleya, Moloney, Peterson, Murphy, Clarke, Stanton, Dinan and Cryan2017). Moreover, drinking water supplemented with a GOS mixture (B-GOS; 3 per cent) for 3 weeks increased plasma acetate levels and upregulated subunits of the N-methyl-d-aspartate receptor (NMDAR) in the brain (Gronier et al., Reference Gronier, Savignac, Di Miceli, Idriss, Tzortzis, Anthony and Burnet2018; Kao et al., Reference Kao, Spitzer, Anthony, Lennox and Burnet2018), which is implicated in synaptic plasticity and memory formation, as well as Acetyl Co-A Carboxylase mRNA, an enzyme in the brain that plays an important role in supplying fatty acids for myelination. Indeed, in one of these studies, mice fed with the GOS mixture gained greater cognitive flexibility (Gronier et al., Reference Gronier, Savignac, Di Miceli, Idriss, Tzortzis, Anthony and Burnet2018). Unfortunately, the other did not test whether the GOS mixture induced changes in cognition (Kao et al., Reference Kao, Spitzer, Anthony, Lennox and Burnet2018). Direct administration of acetate also induced similar neurochemical changes as that of GOS (Gronier et al., Reference Gronier, Savignac, Di Miceli, Idriss, Tzortzis, Anthony and Burnet2018), suggesting that acetate may play a mechanistic role in the observed effects of GOS administration.

Unfortunately, studies in humans administering prebiotics and assessing effects on affective and cognitive processes do not quantify SCFAs and do not explore the extent to which circulating SCFAs mediate the observed effects (Dalile et al., Reference Dalile, Van Oudenhove, Vervliet and Verbeke2019). However, one study found that increasing colonic propionate by consumption of 10 g of an inulin–propionate ester (thereby delivering of 2.36 g propionate to the colon, which is 2.5 times habitual daily propionate production) influenced brain anticipatory reward responses during a functional magnetic resonance imaging (fMRI) food picture evaluation task in non-obese men (Byrne et al., Reference Byrne, Chambers, Alhabeeb, Chhina, Morrison, Preston, Tedford, Fitzpatrick, Irani, Busza, Garcia-Perez, Fountana, Holmes, Goldstone and Frost2016). In parallel, the subjective appeal of high-energy food pictures decreased and energy intake during an ad libitum meal was reduced. These results support the mediating role of SCFAs in microbiota-gut-brain axis communication. Moreover, our group recently showed that daily administration of known amounts of a SCFA mixture equivalent to 20 or 10 g of AX (174.2 mmol acetate, 13.3 mmol propionate, and 52.4 mmol butyrate, or half the dose, respectively) to the colon of healthy men for 1 week significantly reduced the cortisol response to an acute psychosocial stress challenge (Dalile et al., Reference Dalile, Vervliet, Bergonzelli, Verbeke and Van Oudenhove2020). Furthermore, we found that the increase in circulating SCFAs was associated with a decrease in cortisol response to stress. This preliminary study indicates that SCFAs are clearly an important mechanism through which fermentable fibres can affect HPA axis (a major neuroendocrine system that regulates stress responses as well as affective processes) reactivity to stress.

Conceptually, SCFAs may influence affective and cognitive processes by influencing multiple pathways that have been reviewed elsewhere (Dalile et al., Reference Dalile, Van Oudenhove, Vervliet and Verbeke2019). However, the extent to which increasing the substrate of SCFAs (soluble fibres) increases SCFA concentrations and consequently influences affective and cognitive processes, remains understudied. In the following sections, we focus on potential mechanisms by which fibre consumption may alter affective and cognitive processes via interactions with SCFAs and other microbiota-gut-brain axis mediators, namely, the immune system, the intestinal barrier, and hypertension. As studies that directly investigate the effect of dietary fibre on SCFAs and these mediators and subsequent changes in affective and cognitive processes are currently lacking, we summarise the available evidence for an effect of SCFAs on these intermediate systems and for the involvement of these systems in the regulation of affective and cognitive processes.

Immune system (indirect effects)

Immune system and cognitive and affective processes

The immune system is important for both affective and cognitive processes. Challenging the immune system activates cytokines which are involved in the repair of damaged tissue and the restoration of homeostasis. The inflammatory response may also elicit deleterious effects, such as alterations in mood and cognition. This became evident from, for instance, a study vaccinating healthy volunteers with Salmonella typhi which increased IL-6, IL-1Ra, and tumour necrosis factor (TNF)-α, but also decreased mood without any signs of physical sickness (Wright et al., Reference Wright, Strike, Brydon and Steptoe2005). Other studies using acute inflammatory stimulation showed similar results (Brydon et al., Reference Brydon, Walker, Wawrzyniak, Whitehead, Okamura, Yajima, Tsuda and Steptoe2009; Strike et al., Reference Strike, Wardle and Steptoe2004). Also, endotoxin-induced inflammation showed a dose-dependent elevation in pro-inflammatory cytokines IL-6 and TNF-α, but also anti-inflammatory cytokine IL-10 (Grigoleit et al., Reference Grigoleit, Kullmann, Wolf, Hammes and Wegner2011). These elevations were accompanied by a dose-dependent increase in anxiety, negative mood, and poor long-term memory. Involvement of inflammation in negative mood and poor cognitive function is supported by numerous reviews that repeatedly showed an increased incidence of negative mood symptoms with elevated levels of pro-inflammatory markers (CRP, TNF-α, IL-1β, IL-2, and IL6) in peripheral blood and cerebrospinal fluid (CSF) in patients with MDD (Dowlati et al., Reference Dowlati, Herrmann, Swardfager, Liu, Sham, Reim and Lanctôt2010; Hannestad et al., Reference Hannestad, Dellagioia and Bloch2011; Hiles et al., Reference Hiles, Baker, De Malmanche and Attia2012). Regarding cognition, higher levels of CRP are associated with impairment in several cognitive domains such as with lower psychomotor speed and poorer executive function (Krogh et al., Reference Krogh, Benros, Jørgensen, Vesterager, Elfving and Nordentoft2014).

Reducing inflammation has been shown to improve mood (Allison and Ditor, Reference Allison and Ditor2015). Moreover, mood stabilisers such as lithium and valproate suppressed IL-6 levels in patients with bipolar disorder compared to untreated patients (Kim et al., Reference Kim, Jung, Myint, Kim and Park2007). Atypical antipsychotics, which are used as a treatment for both MDD and bipolar disorder, have also been shown to decrease levels of TNF-α and IL-6 in animal models (Bian et al., Reference Bian, Kato, Monji, Hashioka, Mizoguchi, Horikawa and Kanba2008; Kato et al., Reference Kato, Monji, Hashioka and Kanba2007). Furthermore, meta-analyses assessing various cytokine levels after anti-depressant treatment in people with depression show significant decreases in IL-6, IL-1β, and CRP (Hannestad et al., Reference Hannestad, Dellagioia and Bloch2011; Hiles et al., Reference Hiles, Baker, De Malmanche and Attia2012). It seems therefore likely that inflammation plays a significant role in mood. To this end, reducing inflammation may aid negative mood symptoms in some individuals.

SCFAs and immune system

SCFAs are able to modulate the immune system. SCFAs may reduce inflammation by modulating molecular signalling pathways, including free fatty acid receptor (FFAR) 2 and 3 activation (Vinolo et al., Reference Vinolo, Rodrigues, Nachbar and Curi2011), and histone deacetylation (HDAC) inhibition (Waldecker et al., Reference Waldecker, Kautenburger, Daumann, Busch and Schrenk2008). As SCFAs directly interact with FFAR2 on immune cells, which is suggested to regulate regulatory T-cells (Treg cells) in the colon (Smith et al., Reference Smith, Howitt, Panikov, Michaud, Gallini, Bohlooly-Y, Glickman and Garrett2013), Treg cells may then suppress immune responses, thereby reducing inflammation.

Of all SCFAs, butyrate seems the most potent SCFA in modulating the immune system. Butyrate increases the expression of anti-inflammatory molecules in dendritic cells (DCs) and macrophages, thereby supporting Treg differentiation (Singh et al., Reference Singh, Gurav, Sivaprakasam, Brady, Padia, Shi, Thangaraju, Prasad, Manicassamy, Munn, Lee, Offermanns and Ganapathy2014), and hence suppress inflammation. Furthermore, due to its ability to inhibit HDAC activity, butyrate decreases the secretion of proinflammatory cytokines interleukin (IL)-12 and IL-6 in dendritic cells and allows dendritic cells to promote anti-inflammatory cytokine IL-10-secreting T cells (Chang et al., Reference Chang, Hao, Offermanns and Medzhitov2014). Moreover, butyrate inhibits the production of pro-inflammatory cytokines interferon (IFN)-y and IL-2 (Looijer-Van Langen and Dieleman, Reference Looijer-Van Langen and Dieleman2009). Propionate may also modulate inflammation by inducing forkhead box P3 (FOXP3) expression, a protein that regulates the development and function of Tregs (Arpaia et al., Reference Arpaia, Campbell, Fan, Dikiy, Van Der Veeken, Deroos, Liu, Cross, Pfeffer, Coffer and Rudensky2013). Furthermore, both propionate and acetate have been found to increase the production of anti-inflammatory cytokine IL-10 (Cavaglieri et al., Reference Cavaglieri, Nishiyama, Claudio Fernandes, Curi, Miles and Calder2003).

Indeed, most animal studies using prebiotics indicate beneficial effects on the immune system by lowering proinflammatory cytokine expression. For instance, gene expression of pro-inflammatory cytokines IL-1β and (TNF)-α decreased in the brain of mice fed with a high fibre (5 per cent inulin) diet but not in mice fed a low fibre (1 per cent cellulose) diet (Matt et al., Reference Matt, Allen, Lawson, Mailing, Woods and Johnson2018). The decrease in IL-β and TNF-α correlated significantly with faecal butyrate levels, with higher butyrate levels corresponding with lower pro-inflammatory gene expression. Furthermore, 3-week administration of a FOS + GOS mixture (dissolved in drinking water for 0.3–0.4 g/mouse/day) reduced the elevations in proinflammatory cytokine levels (IL-6 and TNF-α) caused by chronic stress induction in C57BL/6J male mice (Burokas et al., Reference Burokas, Arboleya, Moloney, Peterson, Murphy, Clarke, Stanton, Dinan and Cryan2017). Moreover, mice fed with a high fermentable fibre (10 per cent pectin) diet became less sick and recovered faster from lipopolysaccharide (LPS)-induced sickness compared to mice fed with non-fermentable fibre (5 per cent cellulose) (Sherry et al., Reference Sherry, Kim, Dilger, Bauer, Moon, Tapping, Fahey, Tappenden and Freund2010). LPS-stimulated macrophages from the mice fed with pectin showed decreased pro-inflammatory cytokines IL-1β, TNF-α, interferon (IFN)-γ, IL-12, and nitrate, and increased anti-inflammatory cytokine IL-1RA compared to mice fed with cellulose. With regards to brain-based inflammation levels, increased levels of anti-inflammatory IL-1RA and IL-4 mRNA, and a decrease in pro-inflammatory IL-1β and TNF-α were observed in mice fed with pectin compared to mice fed with cellulose (Sherry et al., Reference Sherry, Kim, Dilger, Bauer, Moon, Tapping, Fahey, Tappenden and Freund2010). Since IL-4 expression is stimulated by histone acetylation, the authors hypothesised that the increased butyrate concentrations observed as a result from dietary fibre fermentation of pectin stimulated IL-4, and hence may aid immune response regulation (McLoughlin et al., Reference McLoughlin, Berthon, Jensen, Baines and Wood2017).

In humans, few studies have administered SCFAs and found an effect on inflammation as shown by a systematic review where only two of five studies revealed statistically significant decreases in serum inflammatory markers, namely, decreases in IL-1β following colonic infusion of SCFA mixture, and decreases in TNF levels following rectal acetate administration. Some studies using prebiotics support the hypothesis that SCFAs induce immunomodulatory effects SCFAs. For instance, in healthy elderly, consumption of a mixture of GOS (B-GOS; 5.5 g/day) for 10 weeks increased levels of anti-inflammatory cytokine IL-10, and lowered levels of pro-inflammatory cytokines IL-1, IL-6, and TNF-α (Vulevic et al., Reference Vulevic, Drakoularakou, Yaqoob, Tzortzis and Gibson2008). Furthermore, supplementation with inulin and xylooligosaccharides (XOS; 6.64 g/day) for 4 weeks lowered the expression of pro-inflammatory IL-1b, IL-8, IL-12, and TNF-α, whereas it increased anti-inflammatory IL-10 and IL-13 in the blood of healthy subjects (Lecerf et al., Reference Lecerf, Dépeint, Clerc, Dugenet, Niamba, Rhazi, Cayzeele, Abdelnour, Jaruga, Younes, Jacobs, Lambrey, Abdelnour and Pouillart2012). In a recent meta-analysis, 9 out of 13 studies reported a significant decrease in one or more systemic pro-inflammatory cytokine (primarily TNF-α, IL-6, c-reactive protein (CRP), or IFN-γ) after consumption of prebiotic oligosaccharides compared with control (McLoughlin et al., Reference McLoughlin, Berthon, Jensen, Baines and Wood2017). In contrast, in two studies that were conducted in healthy participants, CRP, TNF-α, and IL-6 increased following oligosaccharide supplementation. Another systematic review, conducted in 10 prebiotic and synbiotic (supplements combining probiotics and prebiotics) trials, of which 7 prebiotic and 3 synbiotic, representing 534 obese/overweight subjects, also found inconsistent effects of prebiotic treatment on immunomodulation. They found that only 6 out of 10 trials (two with GOS, one with inulin, and three with different synbiotics) reduced CRP levels, four out of four trials (one with oligofructose-enriched inulin, one with inulin, and two with different synbiotics) reduced levels of IL-6 and/or TNF, and four out of the five trials (one with oligofructose-enriched inulin, one with inulin, one with GOS, and one with different synbiotics) lowered endotoxin levels (Fernandes et al., Reference Fernandes, do, Mocellin, Kuntz and Trindade2017). Taken together, it might still be too early to promote the use of prebiotics for reduction of low-grade inflammation. However, the evidence seems promising and more studies are needed to conclude whether prebiotics can lower inflammation via increased SCFA production in humans, and in turn promote beneficial effects on mood and cognition.

Intestinal barrier

Intestinal barrier and cognitive and affective processes

The intestinal barrier is mainly responsible for regulating the absorption of nutrients, electrolytes, and water from the lumen, and to prevent pathogenic microorganisms and toxic luminal substances to enter the host. A dysfunctional intestinal barrier may lead to increased intestinal permeability, and hence to the translocation of luminal antigens, bacteria, and toxins through the intestinal wall into the blood stream. In turn, this may cause low-grade inflammation (Caspani and Swann, Reference Caspani and Swann2019) and hence may affect affective and cognitive processes. Indeed, some studies report associations between depressive symptoms and increased levels of indirect markers of intestinal permeability such as intestinal-type fatty acid-binding protein (I-FABP), fatty acid-binding protein 2 (FABP2), LPS, and zonulin (Ohlsson et al., Reference Ohlsson, Gustafsson, Lavant, Suneson, Brundin, Westrin, Ljunggren and Lindqvist2019; Stevens et al., Reference Stevens, Goel, Seungbum, Richards, Holbert, Pepine and Raizada2018). Moreover, a study using a direct measure of intestinal permeability (lactulose to mannitol ratio) also found a positive association with depression severity (Calarge et al., Reference Calarge, Devaraj and Shulman2019).

SCFAs and intestinal barrier

SCFAs are the preferred energy substrates for the colonic epithelium (Cushing et al., Reference Cushing, Alvarado and Ciorba2015) and can contribute to enhanced intestinal barrier function. This has been shown in in vitro and in in vivo experiments in which the application of individual and combinations of SCFAs increased transepithelial electrical resistance (Feng et al., Reference Feng, Wang, Wang, Huang and Wang2018; Suzuki et al., Reference Suzuki, Yoshida and Hara2008), decreased paracellular transport markers (Suzuki et al., Reference Suzuki, Yoshida and Hara2008), and stimulated the formation of tight-junctions (Feng et al., Reference Feng, Wang, Wang, Huang and Wang2018). Furthermore, SCFAs may protect the intestinal barrier from disruption induced by LPS through the inhibition of the NLPR3 inflammasome and autophagy (Feng et al., Reference Feng, Wang, Wang, Huang and Wang2018).

Interestingly, an experimental study showed that 1-week oral administration of a SCFA mixture (67.5 mM acetate, 25 mM propionate, and 40 mM butyrate) alleviated stress-induced increases in intestinal permeability and decreased depressive-like behaviour in mice (Van De Wouw et al., Reference Van De Wouw, Boehme, Lyte, Wiley, Strain, O’sullivan, Clarke, Stanton, Dinan, Cryan, Barrett, Grundy and Cryan2018). Moreover, animal studies show that inulin supplementation (0.5 per cent as well as 1 per cent) increased expression of intestinal barrier function-related genes after Salmonella enterica infection in SPF chickens (Song et al., Reference Song, Li, Everaert, Liu, Zheng, Zhao and Wen2020). In obese mice, oligofructose increased the relative abundance of Bifidobacterium spp., leading to increases in GLP-2 and tight-junctions, lower levels of inflammation, and a significant improvement of intestinal permeability (Cani et al., Reference Cani, Possemiers, Van De Wiele, Guiot, Everard, Rottier, Geurts, Naslain, Neyrinck, Lambert, Muccioli and Delzenne2009). As ITFs stimulate SCFA production, the effect is likely mediated by SCFAs, although this is speculative as SCFA concentrations were not measured.

Human studies that evaluated the impact of dietary fibre on intestinal permeability yielded inconsistent results. Administration of green bananas and pectin for 1 week significantly reduced intestinal permeability and stool output in children with diarrhoea (Rabbani et al., Reference Rabbani, Teka, Zaman, Majid, Khatun and Fuchs2001). Another study in healthy subjects found a significant reduction in intestinal permeability markers zonulin and GLP-2 after consumption of inulin-pasta (8 weeks) compared to control pasta (Russo et al., Reference Russo, Linsalata, Clemente, Chiloiro, Orlando, Marconi, Chimienti and Riezzo2012). In contrast, a high dose of inulin (30–35 g/day) for 1 week in patients on liquid enteral nutrition did not affect intestinal permeability (Sobotka et al., Reference Sobotka, Brátova, Šlemrová, Maňák, Vižd’a and Zadák1997) neither did supplementation with GOS (5.5 g/day) for 12 weeks in men with well-controlled type 2 diabetes (Pedersen et al., Reference Pedersen, Gallgher, Horton, Ellis, Ijaz, Wu, Jaiyeola, Diribe, Duparc, Cani, Gibson, Hinton, Wright, La Ragione and Robertson2016).

These results suggest that the beneficial effects of SCFA on intestinal permeability are not always evident from human studies. Fermentable fibres may induce beneficial effects on affective and cognitive processes via stimulating SCFA production. However, there is a need for human studies that assess whether improvements in intestinal barrier function by means of SCFA stimulation are also associated with improvements in mood and cognition, albeit by attenuating stress-induced negative effects, to confirm this speculation.

Hypertension

Hypertension and cognitive and affective processes

Hypertension, also known as high blood pressure, is a well-established risk factor for cognitive impairment and dementia (Forte et al., Reference Forte, De Pascalis, Favieri and Casagrande2020). Cognitive impairment is more frequent in patients with hypertension, and this is positively associated with the severity (stage) of hypertension (Muela et al., Reference Muela, Costa-Hong, Yassuda, Moraes, Memória, Machado, Macedo, Shu, Massaro, Nitrini, Mansur and Bortolotto2017). Moreover, the prevalence of anxiety disorders is higher in patients with essential hypertension (Vetere et al., Reference Vetere, Ripaldi, Ais, Korob, Kes and Villamil2007). However, it remains to be established to what extent a reduction in blood pressure improves affective processes and cognition. A systematic review and meta-analysis of randomised controlled trials (RCTs) indicated that pharmacological treatment of hypertension reduces cognitive decline in older adults (Gupta et al., Reference Gupta, Perdomo, Billinger, Beddhu, Burns and Gronseth2020). On the other hand, recent large-scale associative studies report mixed findings. In 14,195 hypertensive older adults from the United States and Australia, the use of beta blockers alone or combined with angiotensin-receptor blockers was associated with depressive symptoms, but this was not the case when using angiotensin-receptor blockers on its own, angiotensin-converting enzyme inhibitors, calcium channel blockers, or any other possible combination of drugs among them (Agustini et al., Reference Agustini, Mohebbi, Woods, McNeil, Nelson, Shah, Murray, Ernst, Reid, Tonkin, Lockery and Berk2020). However, another cross-sectional study comprising 1.8 million patients from Scotland reported that angiotensin antagonists and calcium channel blockers were associated with episodes of MDD in hypertensive patients with or without history of mood disorders (Shaw et al., Reference Shaw, Mackay, Pell, Padmanabhan, Bailey and Smith2020). Another nation-wide population-based study of 5.4 million people in Denmark found that no drugs belonging to angiotensin agents, calcium antagonists, β-blockers, or diuretics were associated with an increased risk of depression. In contrast, angiotensin agents, calcium antagonists, and β-blockers were associated with decreased rates of depression. Moreover, non-clinical evidence indicates that mood and blood pressure are related, with increasing intensity of negative moods (stress, anxiety, and anger) being associated with increased blood pressure, with feeling tired (low energy) being associated with the opposite pattern, while changes in intensity in positive mood (happy) showing little changes in blood pressure (Shapiro et al., Reference Shapiro, Jamner, Goldstein and Delfino2001).

SCFAs and hypertension

Meta-analyses and reviews of RCTs reveal that higher dietary fibre consumption reduces blood pressure, an effect that is most pronounced in patients with hypertension, regardless of the type of fibre (Aleixandre and Miguel, Reference Aleixandre and Miguel2016). Some studies report differences in gut microbiota composition, particularly in SCFA-producers, as well as in SCFA profiles in spontaneously hypertensive rats (SHR) and in hypertensive patients (Yang et al., Reference Yang, Santisteban, Rodriguez, Li, Ahmari, Carvajal, Zadeh, Gong, Qi, Zubcevic, Sahay, Pepine, Raizada and Mohamadzadeh2015). For example, patients with high blood pressure exhibit significantly less butyrate-producers such as Roseburia and Eubacterium but also lower plasma butyrate concentrations compared to normotensive subjects (Kim et al., Reference Kim, Goel, Kumar, Qi, Lobaton, Hosaka, Mohammed, Handberg, Richards, Pepine and Raizada2018). Furthermore, intraperitoneal or intramedullary butyrate administration (Kim et al., Reference Kim, Goel, Kumar, Qi, Lobaton, Hosaka, Mohammed, Handberg, Richards, Pepine and Raizada2018; Wang et al., Reference Wang, Zhu, Lu, Liu, Zhang, Xu, Liu, Li and Yang2017), acetate (Marcques et al., Reference Marcques, Nelson, Chu, Horlock, Fiedler, Ziemann, Tan, Kuruppu, Rajapakse, El-Osta, Mackay and Kaye2017), and propionate (Bartolomaeus et al., Reference Bartolomaeus, Balogh, Yakoub, Homann, Markó, Höges, Tsvetkov, Krannich, Wundersitz, Avery, Haase, Kräker, Hering, Maase, Kusche-Vihrog, Grandoch, Fielitz, Kempa, Gollasch, Zhumadilov, Kozhakhmetov, Kushugulova, Eckardt, Dechend, Rump, Forslund, Müller, Stegbauer and Wilck2019) administration in drinking water resulted in anti-hypertensive effects in rodents. Most recently, expression levels of butyrate‐sensing receptors FFAR3 and olfactory receptor (OLFR) 59 in the hypothalamus of SHR were found to be lower than in control rats, likely underlying the reduced effects of centrally administered butyrate on blood pressure in the SHR. Finally, functional magnetic resonance imaging revealed reduced activation of cardio regulatory brain regions of SHR compared to controls following ICV injection of butyrate to reach physiological concentrations (3–5 μM/L) in the CSF (Yang et al., Reference Yang, Magee, Colon-Perez, Larkin, Liao, Balazic, Cowart, Arocha, Redler, Febo, Vickroy, Martyniuk, Reznikov and Zubcevic2019). Only correlational evidence exists in humans, which indicate that higher SCFA-producing bacteria are associated with lower blood pressure and high faecal SCFA levels (De La Cuesta-Zuluaga et al., Reference De La Cuesta-Zuluaga, Mueller, Álvarez-Quintero, Velásquez-Mejía, Sierra, Corrales-Agudelo, Carmona, Abad and Escobar2019; Verhaar et al., Reference Verhaar, Collard, Prodan, Levels, Zwinderman, Bäckhed, Vogt, Peters, Muller, Nieuwdorp and van den Born2020). Studies that aim to reduce hypertension in humans by supplementing prebiotics or other dietary fibres would benefit from quantifying SCFAs to determine the extent to which they mediate the effect of these dietary interventions on blood pressure. These studies suggest that more research is needed to understand the role of hypertension in mood and cognition, and non-pharmacological interventions such as dietary fibre or SCFA supplementation may be ripe targets for such research.

Brain-derived neurotrophic factor

BDNF is a nerve growth factor found in the brain and in the periphery with an important function in normal neural development and in long-term memory. It is considered as a potential marker for neuronal integrity and brain functions. As central and peripheral BDNF are highly correlated in rats (r = 0.86) (Harris et al., Reference Harris, Lennen, Brydges, Jansen, Pernet, Whalley, Marshall, Baker, Basso, Day, Holmes and Hall2016), circulating levels of BDNF may be used as an appropriate measure for brain levels of BDNF. Dietary fibre may indirectly increase circulating BDNF levels via modification of the microbiota, in particular levels of Bifidobacterium and Lactobacillus. Increases in the relative abundance of these strains and consequent increases in BDNF levels were accompanied by a reduction in depressive symptoms as well as improvements in cognitive performance in both animal (Bercik et al., Reference Bercik, Verdu, Foster, Macri, Potter, Huang, Malinowski, Jackson, Blennerhassett, Neufeld, Lu, Khan, Corthesy-Theulaz, Cherbut, Bergonzelli and Collins2010, Reference Bercik, Park, Sinclair, Khoshdel, Lu, Huang, Deng, Blennerhassett, Fahnestock, Moine, Berger, Huizinga, Kunze, Mclean, Bergonzelli, Collins and Verdu2011) and human studies (Haghighat et al., Reference Haghighat, Rajabi and Mohammadshahi2019).

Dietary fibres have indeed shown to increase BDNF levels in both animals and humans, and some already found associations with improvements in cognition. For instance, in a rat study administering placebo, inulin, E. faecium, or E. faecium + inulin, BDNF levels only increased in the probiotic and synbiotic group, but not in the prebiotic group, with only the synbiotic group showing improved memory (Romo-Araiza et al., Reference Romo-Araiza, Gutiérrez-Salmeán, Galván, Hernández-Frausto, Herrera-López, Romo-Parra, García-Contreras, María Fernández-Presas, Jasso-Chávez, Borlongan, Ibarra, Kumar, Smulders and Yegla2018). Furthermore, in healthy subjects, an evening meal consisting of rye kernel bread increased plasma BDNF levels by 27 per cent at fasting on the next morning compared to white wheat bread (Sandberg et al., Reference Sandberg, Björck and Nilsson2018). Consumption of the same bread for three consecutive days was associated with a higher abundance of Prevotella compared to the white wheat bread (Prykhodko et al., Reference Prykhodko, Sandberg, Burleigh, Björck, Nilsson and Fåk Hållenius2018), and the Prevotella genus was positively associated with plasma levels of BDNF. Unfortunately, no plasma SCFAs were measured which prevented evaluating the relationship between SCFAs and BDNF. Despite evidence that butyrate increases acetylation around the promoters of BDNF, thereby increasing its transcription (Intlekofer et al., Reference Intlekofer, Berchtold, Malvaez, Carlos, Mcquown, Cunningham, Wood and Cotman2013) and the myriad pre-clinical studies showing that it increases following butyrate administration (Stilling et al., Reference Stilling, de Wouw, Clarke, Stanton, Dinan and Cryan2016), our recent study in healthy subjects showed that colonic administration of SCFAs did not alter serum BDNF levels (Dalile et al., Reference Dalile, Vervliet, Bergonzelli, Verbeke and Van Oudenhove2020). Together, the evidence thus far seems to indicate that the relationship between dietary fibre consumption and BDNF levels is mediated by changes in gut microbiota composition, rather than increases in colonic or circulating SCFAs.

Direct effects of dietary fibre on the immune system

Dietary fibre and immune system

Few studies explored immune effects of dietary fibres that are independent of the gut microbiota. A recent study demonstrated that dietary cellulose supplementation modulates the immune response. Mice were fed either a normal diet or a high-fibre cellulose diet for 2 weeks and were, subsequently, injected with endotoxin (LPS). Mice fed the high-fibre cellulose diet showed lower levels of proinflammatory cytokines (IL-1α, IL-1β, and IFN-α), lower numbers and activation of splenic macrophages and DCs, and hyporesponsiveness of T cells (Di Caro et al., Reference Di Caro, Cummings, Alcamo, Piganelli, Clark, Morowitz and Aneja2019). Splenic macrophages and DCs are innate effector cells that are crucial for the host defence to protect against bacterial infections (Di Caro et al., Reference Di Caro, Cummings, Alcamo, Piganelli, Clark, Morowitz and Aneja2019). Since cellulose is hardly fermentable, the observed effects likely arose from a direct interaction of the fibre with the immune system, rather than from SCFA production.

Also, other glucans, more specifically β-1,3/1,6-glucans, modulate the innate and acquired immune system. Those compounds bind to pattern recognition receptors including complement receptor 3, scavenger receptors, lactosylceramide, and dectin-1, which are expressed on cells of myeloid origin, including macrophages, dendritic cells, and neutrophils (Murphy et al., Reference Murphy, Davis and Carmichael2010). Although small amounts of β-glucan may be absorbed after oral administration, it is most likely that β-glucan primarily act in the gut epithelium. β-Glucan has been shown useful in the prevention of treatment of allergic disease. No studies to date have evaluated whether the impact of β-glucan on the immune system results in improvements in cognitive or affective processes.

Cholesterol

Cholesterol and cognitive and affective processes

Cholesterol is important for normal brain function and our mental well-being as well as cognition. However, its involvement is complex. Cholesterol demand by neurons in the brain is very high, since it is implicated in a variety of neuronal processes such as neurite formation and synaptic activity. Several studies found that higher levels of LDL cholesterol were associated with depressive mood in men (Kim et al., Reference Kim, Hong and Hwang2019; Tedders et al., Reference Tedders, Fokong, Mckenzie, Wesley, Yu and Zhang2011) whereas low levels of high-density lipoprotein (HDL) cholesterol were significantly associated with depression symptoms in women (Tedders et al., Reference Tedders, Fokong, Mckenzie, Wesley, Yu and Zhang2011) and men (Lehto et al., Reference Lehto, Niskanen, Tolmunen, Hintikka, Viinamäki, Heiskanen, Honkalampi, Kokkonen and Koivumaa-Honkanen2010), and is a potential risk factor for developing a mood disorder in females (Kim et al., Reference Kim, Han, Jang and Park2018). Up to 44 per cent of the subjects who were diagnosed with a mood disorder suffered from mild hypercholesterolemia (>5.2 and 6.2 mmol/L) and 21 per cent from hypercholesterolemia (>6.2 mmol/L) (Davison and Kaplan, Reference Davison and Kaplan2012). Importantly, dietary cholesterol cannot cross the blood brain barrier. Therefore, the mechanisms by which cholesterol influences learning and memory are thought to combine peripheral (atherogenic and proinflammatory) and central (accumulation of intracellular beta amyloid) effects.

There is some evidence that pharmacological reduction of plasma cholesterol with statins, the most common cholesterol-lowering drugs, may improve cognitive function in subjects without dementia (Schreurs, Reference Schreurs2010). However, the effects of statins on mood modulation remain controversial. For instance, chronic statin treatment in rats (10 mg/kg atorvastatin; 10 mg/kg simvastatin; 30 mg/kg pravastatin) reduced anxious behaviour in the open-field task (Citraro et al., Reference Citraro, Chimirri, Aiello, Gallelli, Trimboli, Britti, De Sarro and Russo2014; Wang and Gao, Reference Wang and Gao2009). Preclinical studies using the forced swimming test also support the anti-depressant effects of statin treatment (Can et al., Reference Can, Ulupınar, Özkay, Yegin and Öztürk2012). Another study demonstrated that statin treatment decreased the risk of depression (Yang et al., Reference Yang, Jick and Jick2003). In contrast, low dose statin treatment (simvastatin 1 mg/kg; atorvastatin 0.5 mg/kg) did not improve spatial memory and learning in guinea pigs, but instead increased levels of anxiety (Maggo et al., Reference Maggo, Clark and Ashton2012). Furthermore, some studies in humans found that depressive symptoms increased rather than attenuated after statin treatment (Lechleitner et al., Reference Lechleitner, Hoppichler, Konwalinka, Patsch and Braunsteiner1992; Morales et al., Reference Morales, Wittink, Datto, Difilippo, Cary, Tenhave and Katz2006). It is possible that lowering cholesterol induces positive effects on cognitive and affective processes by lowering blood pressure (see section “Hypertension”).

Dietary fibre and cholesterol

Soluble rather than insoluble fibres possess cholesterol lowering abilities in humans. The mechanism is most likely related to the viscous properties of soluble fibre. By increasing the viscosity in the small intestine, reabsorption of bile acids is reduced, in turn leading to increased de novo synthesis of bile acids from cholesterol in the liver and lower circulating cholesterol levels (Fuller et al., Reference Fuller, Beck, Salman and Tapsell2016). The role of β-glucan in lowering cholesterol has been extensively documented and led the U.S. Food and Drug Administration (FDA) to authorise the use of health claims on oat-containing products, stating that consumption of β-glucans at least 3 g per day lowers the risk of cardiovascular disease (Food and Drug Administration, 2019). These health claims have also been approved by the European Commission, as well as in various other jurisdictions (European Commision, 2011, 2012). A meta-analysis showed that also pectin, soluble fibre from psyllium, and guar gum have cholesterol-lowering abilities (Brown et al., Reference Brown, Rosner, Willett and Sacks1999). Viscous fibres should be utilised in intervention studies to test their effects of affective and cognitive processes, and if supported, investigate whether their cholesterol-lowering ability drives the effects on brain function in humans.

Conclusion

Collectively, studies suggest that dietary fibres may be promising in inducing psychological changes in cognitive and affective processes. The evidence base indicates that fermentable fibres, such as ITFs, β-glucans, pectin, and gums, are the strongest candidates to modulate these psychological functions. This is mainly due to their ability to modulate the microbiota and increase SCFA production. However, their ability to increase BDNF levels may also reinforce their effects on cognitive and affective processes. On the other hand, viscous fibres such as β-glucan and non-fermentable fibres such as cellulose may exert beneficial effects on cognitive and affective processes via microbiota-independent mechanisms such as lowering cholesterol and inflammation, respectively. Notably, a research gap remains, where the effects of different types of fibres on mood and cognition via the proposed mechanisms still needs to be investigated. In addition, while preclinical studies of dietary fibres on gastrointestinal mechanisms are ample, well-designed studies of dietary fibre effects on mood and cognition with simultaneous assessment of putative mechanisms of actions in humans are needed. The aim with this review was to outline potential additional microbiota-dependent and independent mechanism through which different fibre properties can act to affect affective and cognitive processes in order to give a more complete picture of the effects of dietary fibres and to help explain additional variance in inter-individual psychological responses to fibre consumption. The resolution of various mechanistic and functional links outlined here may better facilitate personalised nutrition for maintaining optimal psychological functioning in states of health or disease.

Acknowledgement

The authors would like to thank Francesca Zoppas for her helpful contribution to content of the review.

Data availability statement

Data sharing is not applicable to this article as no new data were created or analysed.

Notes on contributors

Danique La Torre is a PhD student in the Translational Research Center in Gastrointestinal Disorders (TARGID) at KU Leuven. Her research focuses on the interaction between stress and intestinal permeability, and dietary and physical interventions that could modulate this interaction.

Kristin Verbeke is a Pharmacist by training, and now a Full Professor and head of the Translational Research Center in Gastrointestinal Disorders (TARGID) at KU Leuven. She has published widely on the bacterial colonic metabolism in humans, including on the development of analytical methods to quantify bacterial metabolism as well as dietary intervention studies that aim to modulate the gut microbiome.

Boushra Dalile is a Junior Postdoctoral researcher at the Translational Research Center in Gastrointestinal Disorders (TARGID), KU Leuven. She conducts research within the gut-brain axis, mainly revolving around the role of the bacterial metabolites short-chain fatty acids in human stress sensitivity and anxiety.

Funding

The PhD of B.D. is financially supported by an unrestricted grant from Société des Produits Nestlé S.A.

Disclosure

The authors declare none.

Author contributions

Conceptualization, D.L.T., K.V., B.D.; Supervision, K.V., B.D.; Visualisation, B.D., D.L.T.; Writing – original draft, D.L.T., K.V., B.D.; Writing – review and editing, D.L.T., K.V., B.D. All authors have read an approved the final version of this manuscript.

References

Agustini, B, Mohebbi, M, Woods, RL, McNeil, JJ, Nelson, MR, Shah, RC, Murray, AM, Ernst, ME, Reid, CM, Tonkin, A, Lockery, JE and Berk, M (2020) The association of antihypertensive use and depressive symptoms in a large older population with hypertension living in Australia and the United States: A cross-sectional study. Journal of Human Hypertension 34(11), 787794. https://doi.org/10.1038/s41371-020-0303-yCrossRefGoogle Scholar
Aizawa, E, Tsuji, H, Asahara, T, Takahashi, T, Teraishi, T, Yoshida, S, Ota, M, Koga, N, Hattori, K and Kunugi, H (2016) Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. Journal of Affective Disorders 202, 254257. https://doi.org/10.1016/j.jad.2016.05.038CrossRefGoogle ScholarPubMed
Aleixandre, A and Miguel, M (2016) Dietary fiber and blood pressure control. Food & Function 7, 1864. https://doi.org/10.1039/c5fo00950bCrossRefGoogle ScholarPubMed
Ali, BH, Ziada, A and Blunden, G (2009) Biological effects of gum arabic: A review of some recent research. Food and Chemical Toxicology 47, 18. https://doi.org/10.1016/j.fct.2008.07.001CrossRefGoogle ScholarPubMed
Allison, DJ and Ditor, DS (2015) Targeting inflammation to influence mood following spinal cord injury: A randomized clinical trial. Journal of Neuroinflammation 12, 204. https://doi.org/10.1186/s12974-015-0425-2CrossRefGoogle ScholarPubMed
Arpaia, N, Campbell, C, Fan, X, Dikiy, S, Van Der Veeken, J, Deroos, P, Liu, H, Cross, JR, Pfeffer, K, Coffer, PJ and Rudensky, AY (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 504(7480), 451455. https://doi.org/10.1038/nature12726CrossRefGoogle ScholarPubMed
Azpiroz, F, Dubray, C, Bernalier-Donadille, A, Cardot, J-M, Accarino, A, Serra, J, Wagner, A, Respondek, F and Dapoigny, M (2017) Effects of scFOS on the composition of fecal microbiota and anxiety in patients with irritable bowel syndrome: A randomized, double blind, placebo controlled study. Neurogastroenterology & Motility 29(2), e12911. https://doi.org/10.1111/nmo.12911CrossRefGoogle ScholarPubMed
Bartolomaeus, H, Balogh, A, Yakoub, M, Homann, S, Markó, L, Höges, S, Tsvetkov, D, Krannich, A, Wundersitz, S, Avery, EG, Haase, N, Kräker, K, Hering, L, Maase, M, Kusche-Vihrog, K, Grandoch, M, Fielitz, J, Kempa, S, Gollasch, M, Zhumadilov, Z, Kozhakhmetov, S, Kushugulova, A, Eckardt, KU, Dechend, R, Rump, LC, Forslund, SK, Müller, DN, Stegbauer, J and Wilck, N (2019) Short-chain fatty acid propionate protects from hypertensive cardiovascular damage. Circulation 139(11), 14071421. https://doi.org/10.1161/CIRCULATIONAHA.118.036652CrossRefGoogle ScholarPubMed
Bercik, P, Park, AJ, Sinclair, D, Khoshdel, A, Lu, J, Huang, X, Deng, Y, Blennerhassett, PA, Fahnestock, M, Moine, D, Berger, B, Huizinga, JD, Kunze, W, Mclean, PG, Bergonzelli, GE, Collins, SM and Verdu, EF (2011) The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication. Neurogastroenterology & Motility 23, 1132e544. https://doi.org/10.1111/j.1365-2982.2011.01796.xCrossRefGoogle ScholarPubMed
Bercik, P, Verdu, EF, Foster, JA, Macri, J, Potter, M, Huang, X, Malinowski, P, Jackson, W, Blennerhassett, P, Neufeld, KA, Lu, J, Khan, WI, Corthesy-Theulaz, I, Cherbut, C, Bergonzelli, GE and Collins, SM (2010) Chronic gastrointestinal inflammation induces anxiety-like behavior and alters central nervous system biochemistry in mice. Gastroenterology 139, 21022112. https://doi.org/10.1053/j.gastro.2010.06.063CrossRefGoogle ScholarPubMed
Best, T, Bryan, J and Burns, N (2008) An investigation of the effects of saccharides on the memory performance of middle-aged adults. Journal of Nutrition, Health and Aging 12(9), 657662. https://doi.org/10.1007/bf03008278Google ScholarPubMed
Best, T, Howe, P, Bryan, J, Buckley, J and Scholey, A (2015) Acute effects of a dietary non-starch polysaccharide supplement on cognitive performance in healthy middle-aged adults acute effects of a dietary non-starch polysaccharide supplement on cognitive performance in healthy middle-aged adults. Nutritional Neuroscience 18(2), 7686. https://doi.org/10.1179/1476830513Y.0000000101CrossRefGoogle ScholarPubMed
Best, T, Kemps, E and Bryan, J (2009) Saccharide effects on cognition and well-being in middle-aged adults: A randomized controlled trial. Developmental Neuropsychology 35(1), 6680. https://doi.org/10.1080/87565640903325709.CrossRefGoogle Scholar
Bian, Q, Kato, T, Monji, A, Hashioka, S, Mizoguchi, Y, Horikawa, H and Kanba, S (2008) The effect of atypical antipsychotics, perospirone, ziprasidone and quetiapine on microglial activation induced by interferon-γ. Progress in Neuro-Psychopharmacology & Biological Psychiatry 32, 4248. https://doi.org/10.1016/j.pnpbp.2007.06.031CrossRefGoogle ScholarPubMed
Boets, E, Gomand, SV, Deroover, L, Preston, T, Vermeulen, K, De Preter, V, Hamer, HM, Van Den Mooter, G, De Vuyst, L, Courtin, CM, Annaert, P, Delcour, JA and Verbeke, KA (2017) Systemic availability and metabolism of colonic-derived short-chain fatty acids in healthy subjects: A stable isotope study. The Journal of Physiology 2, 541555. https://doi.org/10.1113/JP272613CrossRefGoogle Scholar
Bravo, JA, Forsythe, P, Chew, MV, Escaravage, E, Savignac, HM, Dinan, TG, Bienenstock, J, Cryan, JF and Designed, JFC (2011) Ingestion of lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. PNAS 108(38), 1605016055. https://doi.org/10.1073/pnas.1102999108CrossRefGoogle Scholar
Brown, L, Rosner, B, Willett, WW and Sacks, FM (1999) Cholesterol-lowering effects of dietary fiber: A meta-analysis. American Journal of Clinical Nutrition 69, 3042. https://doi.org/10.1093/ajcn/69.1.30.CrossRefGoogle ScholarPubMed
Brydon, L, Walker, C, Wawrzyniak, A, Whitehead, D, Okamura, H, Yajima, J, Tsuda, A and Steptoe, A (2009) Synergistic effects of psychological and immune stressors on inflammatory cytokine and sickness responses in humans. Brain, Behavior, and Immunity 23(2), 217224. https://doi.org/10.1016/j.bbi.2008.09.007CrossRefGoogle ScholarPubMed
Buigues, C, Fernández-Garrido, J, Pruimboom, L, Hoogland, AJ, Navarro-Martínez, R, Martínez-Martínez, M, Verdejo, Y, Carmen Mascarós, M, Peris, C and Cauli, O (2016) Effect of a prebiotic formulation on frailty syndrome: A randomized, double-blind clinical trial. International Journal of Molecular Sciences 17(6), 932. https://doi.org/10.3390/ijms17060932CrossRefGoogle ScholarPubMed
Burokas, A, Arboleya, S, Moloney, RD, Peterson, VL, Murphy, K, Clarke, G, Stanton, C, Dinan, TG and Cryan, JF (2017) Targeting the microbiota-gut–brain axis: Prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biological Psychiatry 82(1), 472487. https://doi.org/10.1016/j.biopsych.2016.12.031CrossRefGoogle ScholarPubMed
Butler, K, Klaus, K, Edwards, L and Pennington, K (2017) Elevated cortisol awakening response associated with early life stress and impaired executive function in healthy adult males. Hormones and Behavior 95, 1321. https://doi.org/10.1016/j.yhbeh.2017.07.013CrossRefGoogle ScholarPubMed
Byrne, CS, Chambers, ES, Alhabeeb, H, Chhina, N, Morrison, DJ, Preston, T, Tedford, C, Fitzpatrick, J, Irani, C, Busza, A, Garcia-Perez, I, Fountana, S, Holmes, E, Goldstone, AP and Frost, GS (2016) Increased colonic propionate reduces anticipatory reward responses in the human striatum to high-energy foods. The American Journal of Clinical Nutrition 104, 514. https://doi.org/10.3945/ajcn.115.126706CrossRefGoogle ScholarPubMed
Calarge, CA, Devaraj, S and Shulman, RJ (2019) Gut permeability and depressive symptom severity in unmedicated adolescents. Journal of Affective Disorders 246, 586594. https://doi.org/10.1016/j.jad.2018.12.077CrossRefGoogle ScholarPubMed
Can, ÖD, Ulupınar, E, Özkay, ÜD, Yegin, B and Öztürk, Y (2012) The effect of simvastatin treatment on behavioral parameters, cognitive performance, and hippocampal morphology in rats fed a standard or a high-fat diet. Behavioural Pharmacology 23, 582592. https://doi.org/10.1097/FBP.0b013e328356c3f2CrossRefGoogle ScholarPubMed
Cani, PD, Possemiers, S, Van De Wiele, T, Guiot, Y, Everard, A, Rottier, O, Geurts, L, Naslain, D, Neyrinck, A, Lambert, DM, Muccioli, GG and Delzenne, NM (2009) Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 58(8), 10911103. https://doi.org/10.1136/gut.2008.165886CrossRefGoogle ScholarPubMed
Caspani, G and Swann, J (2019) Small talk: Microbial metabolites involved in the signaling from microbiota to brain. Current Opinion in Pharmacology 48, 99106. https://doi.org/10.1016/j.coph.2019.08.001CrossRefGoogle Scholar
Cavaglieri, CR, Nishiyama, A, Claudio Fernandes, L, Curi, R, Miles, EA and Calder, PC (2003) Differential effects of short-chain fatty acids on proliferation and production of pro-and anti-inflammatory cytokines by cultured lymphocytes. Life Sciences 73, 16831690. https://doi.org/10.1016/S0024-3205(03)00490-9CrossRefGoogle ScholarPubMed
Chang, PV, Hao, L, Offermanns, S and Medzhitov, R (2014) The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. PNAS 111(6), 22472252. https://doi.org/10.1073/pnas.1322269111CrossRefGoogle ScholarPubMed
Cherbut, C, Ferrier, L, Rozé, C, Anini, Y, Blottière, H, Lecannu, G and Galmiche, J-P (1998) Short-chain fatty acids modify colonic motility through nerves and polypeptide YY release in the rat. American Journal of Physiology 275, G1415G1422.Google ScholarPubMed
Childs, CE, Röytiö, H, Alhoniemi, E, Fekete, AA, Forssten, SD, Hudjec, N, Lim, YN, Steger, CJ, Yaqoob, P, Tuohy, KM, Rastall, RA, Ouwehand, AC and Gibson, GR (2014) Xylo-oligosaccharides alone or in synbiotic combination with Bifidobacterium animalis subsp. lactis induce bifidogenesis and modulate markers of immune function in healthy adults: A double-blind, placebo-controlled, randomised, factorial cross-over study. British Journal of Nutrition 111, 19451956. https://doi.org/10.1017/S0007114513004261CrossRefGoogle ScholarPubMed
Cho, SS and Dreher, ML (2001) Handbook of Dietary Fiber. New York: Taylor & Francis.CrossRefGoogle Scholar
Chunchai, T, Thunapong, W, Yasom, S, Wanchai, K, Eaimworawuthikul, S, Metzler, G, Lungkaphin, A, Pongchaidecha, A, Sirilun, S, Chaiyasut, C, Pratchayasakul, W, Thiennimitr, P, Chattipakorn, N and Chattipakorn, SC (2018) Decreased microglial activation through gut-brain axis by prebiotics, probiotics, or synbiotics effectively restored cognitive function in obese-insulin resistant rats. Journal of Neuroinflammation 15, 11. https://doi.org/10.1186/s12974-018-1055-2CrossRefGoogle ScholarPubMed
Citraro, R, Chimirri, S, Aiello, R, Gallelli, L, Trimboli, F, Britti, D, De Sarro, G and Russo, E (2014) Protective effects of some statins on epileptogenesis and depressive-like behavior in WAG/Rij rats, a genetic animal model of absence epilepsy. Epilepsia 55(8), 12841291. https://doi.org/10.1111/epi.12686CrossRefGoogle ScholarPubMed
Clarke, G, Grenham, S, Scully, P, Fitzgerald, P, Moloney, RD, Shanahan, F, Dinan, TG and Cryan, JF (2013) The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular Psychiatry 18(6), 666673. https://doi.org/10.1038/mp.2012.77CrossRefGoogle Scholar
Clow, A, Hucklebridge, F, Stalder, T, Evans, P and Thorn, L (2010) The cortisol awakening response: More than a measure of HPA axis function. Neuroscience and Biobehavioral Reviews 35, 97103. https://doi.org/10.1016/j.neubiorev.2009.12.011CrossRefGoogle ScholarPubMed
Courtin, CM and Delcour, JA (2001) Relative activity of endoxylanases towards water-extractable and water-unextractable arabinoxylan. Journal of Cereal Science 33, 301312. https://doi.org/10.1006/jcrs.2000.0354CrossRefGoogle Scholar
Cushing, K, Alvarado, DM and Ciorba, MA (2015) Butyrate and mucosal inflammation: New scientific evidence supports clinical observation. Clinical and Translational Gastroenterology 6, 108. https://doi.org/10.1038/ctg.2015.34CrossRefGoogle ScholarPubMed
Dalile, B, Van Oudenhove, L, Vervliet, B and Verbeke, K (2019) The role of short-chain fatty acids in microbiota–gut–brain communication. Nature Reviews Gastroenterology and Hepatology 16(8), 461478. https://doi.org/10.1038/s41575-019-0157-3CrossRefGoogle ScholarPubMed
Dalile, B, Vervliet, B, Bergonzelli, G, Verbeke, K and Van Oudenhove, L (2020) Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: A randomized, placebo-controlled trial. Neuropsychopharmacology 45(13), 22572266. https://doi.org/10.1038/s41386-020-0732-xCrossRefGoogle ScholarPubMed
Davison, KM and Kaplan, BJ (2012) Food intake and blood cholesterol levels of community-based adults with mood disorders. BMC Psychiatry 12, 10. https://doi.org/10.1186/1471-244X-12-10CrossRefGoogle ScholarPubMed
De La Cuesta-Zuluaga, J., Mueller, N. T., Álvarez-Quintero, R., Velásquez-Mejía, E. P., Sierra, J. A., Corrales-Agudelo, V., Carmona, J. A., Abad, J. M., & Escobar, J. S. (2019). Higher Fecal Short-Chain Fatty Acid Levels Are Associated with Gut Microbiome Dysbiosis, Obesity, Hypertension and Cardiometabolic Disease Risk Factors. Nutrients, 11, 51. https://doi.org/10.3390/nu11010051CrossRefGoogle Scholar
Dehhaghi, M, Kazemi, H, Panahi, S and Guillemin, GJ (2019) Microorganisms, tryptophan metabolism, and kynurenine pathway: A complex interconnected loop influencing human health status. International Journal of Tryptophan Research 12, 110. https://doi.org/10.1177/1178646919852996CrossRefGoogle ScholarPubMed
Delzenne, NM (2003) Oligosaccharides: State of the art. Proceedings of the Nutrition Society 62, 177182. https://doi.org/10.1079/PNS2002225CrossRefGoogle ScholarPubMed
Desbonnet, L, Clarke, G, Shanahan, F, Dinan, TG and Cryan, JF (2014) Microbiota is essential for social development in the mouse. Molecular Psychiatry 19, 146148. https://doi.org/10.1038/mp.2013.65CrossRefGoogle ScholarPubMed
Di Caro, V, Cummings, JL, Alcamo, AM, Piganelli, JD, Clark, RSB, Morowitz, MJ and Aneja, RK (2019) Dietary cellulose supplementation modulates the immune response in a murine endotoxemia model. Shock 51(4), 526534. https://doi.org/10.1097/SHK.0000000000001180CrossRefGoogle Scholar
Dowlati, Y, Herrmann, N, Swardfager, W, Liu, H, Sham, L, Reim, EK and Lanctôt, KL (2010) A meta-analysis of cytokines in major depression. Journal of Biological Psychiatry 67, 446457. https://doi.org/10.1016/j.biopsych.2009.09.033CrossRefGoogle ScholarPubMed
European Commision (2011) Commission Regulation (EU) No 1160/2011 of 14 November 2011 on the Authorisation and Refusal of Authorisation of Certain Health Claims Made on Foods and Referring to the Reduction of Disease Risk. 20 September 2021 Available at https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2011:296:0026:0028:EN:PDF.Google Scholar
European Commision (2012) Commission Regulation (EU) NO 432/2012 of 16 May 2012 Establishing a List of Permitted Health Claims Made on Foods, Other Than Those Referring to the Reduction of Disease Risk and to Children’s Development and Health.Google Scholar
Farhangi, MA, Zare Javid, A, Sarmadi, B, Karimi, P and Dehghan, P (2018) A randomized controlled trial on the efficacy of resistant dextrin, as functional food, in women with type 2 diabetes: Targeting the hypothalamicepituitaryeadrenal axis and immune system. Clinical Nutrition 37, 12161223. https://doi.org/10.1016/j.clnu.2017.06.005CrossRefGoogle ScholarPubMed
Feng, Y, Wang, Y, Wang, P, Huang, Y and Wang, F (2018) Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cellular Physiology and Biochemistry 49, 190205. https://doi.org/10.1159/000492853CrossRefGoogle ScholarPubMed
Fernandes, R, do, VA, Mocellin, MC, Kuntz, MGF and Trindade, EBSM (2017) Effects of inulin-type fructans, galacto-oligosaccharides and related synbiotics on inflammatory markers in adult patients with overweight or obesity: A systematic review. Clinical Nutrition 36(5), 11971206. https://doi.org/10.1016/j.clnu.2016.10.003CrossRefGoogle ScholarPubMed
Food and Drug Administration (2019) Food and Drug Administration: Food for Human Consumption. 05 November 2020 Available at https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/cfrsearch.cfm?fr=101.81.Google Scholar
Food and Drug Administration (2020) Food Labeling and Nutrition: Questions and Answers on Dietary Fiber. Available at https://www.fda.gov/food/food-labeling-nutrition/questions-and-answers-dietary-fiber#synthetic_fibers.Google Scholar
Forte, G, De Pascalis, V, Favieri, F and Casagrande, M (2020) Effects of blood pressure on cognitive performance: A systematic review. Journal of Clinical Medicine 9(1), 34. https://doi.org/10.3390/jcm9010034CrossRefGoogle Scholar
Fries, E, Dettenborn, L and Kirschbaum, C (2009) The cortisol awakening response (CAR): Facts and future directions. International Journal of Psychophysiology 72, 6773. https://doi.org/10.1016/j.ijpsycho.2008.03.014CrossRefGoogle ScholarPubMed
Fuller, S, Beck, E, Salman, H and Tapsell, L (2016) New horizons for the study of dietary fiber and health: A review. Plant Foods for Human Nutrition 71(1), 112. https://doi.org/10.1007/s11130-016-0529-6CrossRefGoogle Scholar
Gibson, GR, Hutkins, R, Ellen Sanders, M, Prescott, SL, Reimer, RA, Salminen, SJ, Scott, K, Stanton, C, Swanson, KS, Cani, PD, Verbeke, K and Reid, G (2017) Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nature Reviews Gastroenterology & Hepatology 14, 491502. https://doi.org/10.1038/nrgastro.2017.75CrossRefGoogle ScholarPubMed
Gill, SK, Rossi, M, Bajka, B and Whelan, K (2020) Dietary fibre in gastrointestinal health and disease. Nature Reviews Gastroenterology & Hepatology 18(2), 101116. https://doi.org/10.1038/s41575-020-00375-4CrossRefGoogle ScholarPubMed
Gomez-Pinilla, F (2008) Brain foods: The effects of nutrients on brain function. Nature Reviews Neuroscience 9, 568578. https://doi.org/10.1038/nrn2421Talbott.CrossRefGoogle ScholarPubMed
Grigoleit, J-S, Kullmann, JS, Wolf, OT, Hammes, F and Wegner, A (2011) Dose-dependent effects of endotoxin on neurobehavioral functions in humans. PLoS One 6(12), 28330. https://doi.org/10.1371/journal.pone.0028330CrossRefGoogle ScholarPubMed
Grimaldi, R, Gibson, GR, Vulevic, J, Giallourou, N, Castro-Mejía, JL, Hansen, LH, Leigh Gibson, E, Nielsen, DS and Costabile, A (2018) A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome 6(1), 113. https://doi.org/10.1186/s40168-018-0523-3CrossRefGoogle Scholar
Gronier, B, Savignac, HM, Di Miceli, M, Idriss, SM, Tzortzis, G, Anthony, D and Burnet, PWJ (2018) Increased cortical neuronal responses to NMDA and improved attentional set-shifting performance in rats following prebiotic (B-GOS®) ingestion. European Neuropsychopharmacology 28(1), 211224. https://doi.org/10.1016/j.euroneuro.2017.11.001CrossRefGoogle ScholarPubMed
Gupta, A, Perdomo, S, Billinger, S, Beddhu, S, Burns, J and Gronseth, G (2020) Treatment of hypertension reduces cognitive decline in older adults: A systematic review and meta-analysis. BMJ Open 10, 38971. https://doi.org/10.1136/bmjopen-2020-038971CrossRefGoogle ScholarPubMed
Haghighat, N, Rajabi, S and Mohammadshahi, M (2019) Effect of synbiotic and probiotic supplementation on serum brain-derived neurotrophic factor level, depression and anxiety symptoms in hemodialysis patients: A randomized, double-blinded, clinical trial. Nutritional Neuroscience 24(6), 490499. https://doi.org/10.1080/1028415X.2019.1646975CrossRefGoogle ScholarPubMed
Hannestad, J, Dellagioia, N and Bloch, M (2011) The effect of antidepressant medication treatment on serum levels of inflammatory cytokines: A meta-analysis. Neuropsychopharmacology 36, 24522459. https://doi.org/10.1038/npp.2011.132CrossRefGoogle ScholarPubMed
Harris, AP, Lennen, RJ, Brydges, NM, Jansen, MA, Pernet, CR, Whalley, HC, Marshall, I, Baker, S, Basso, AM, Day, M, Holmes, MC and Hall, J (2016) The role of brain-derived neurotrophic factor in learned fear processing: An awake rat fMRI study. Genes, Brain and Behavior 15, 221230. https://doi.org/10.1111/gbb.12277CrossRefGoogle ScholarPubMed
Harris, HC, Edwards, CA and Morrison, DJ (2017) Impact of glycosidic bond configuration on short chain fatty acid production from model fermentable carbohydrates by the human gut microbiota. Nutrients 9(1), 26. https://doi.org/10.3390/nu9010026CrossRefGoogle ScholarPubMed
Hiles, SA, Baker, AL, De Malmanche, T and Attia, J (2012) Interleukin-6, C-reactive protein and interleukin-10 after antidepressant treatment in people with depression: A meta-analysis. Psychological Medicine 42(10), 20152026. https://doi.org/10.1017/S0033291712000128CrossRefGoogle ScholarPubMed
Intlekofer, KA, Berchtold, NC, Malvaez, M, Carlos, AJ, Mcquown, SC, Cunningham, MJ, Wood, MA and Cotman, CW (2013) Exercise and sodium butyrate transform a subthreshold learning event into long-term memory via a brain-derived neurotrophic factor-dependent mechanism. Neuropsychopharmacology 38, 20272034. https://doi.org/10.1038/npp.2013.104CrossRefGoogle Scholar
Jacka, FN, O’Neil, A, Opie, R, Itsiopoulos, C, Cotton, S, Mohebbi, M, Castle, D, Dash, S, Mihalopoulos, C, Chatterton, ML, Brazionis, L, Dean, OM, Hodge, AM and Berk (2017) A randomised controlled trial of dietary improvement for adults with major depression (the “SMILES” trial). BMC Medicine 15(1), 114. https://doi.org/10.1186/s12916-017-0791-yCrossRefGoogle Scholar
Jenkins, DJA, Kendall, CWC, Axelsen, M, Augustin, LSA and Vuksan, V (2000) Viscous and nonviscous fibres, nonabsorbable and low glycaemic index carbohydrates, blood lipids and coronary heart disease. Current Opinion in Lipidology 11(1), 4956. https://doi.org/10.1097/00041433-200002000-00008CrossRefGoogle ScholarPubMed
Jiang, H, Ling, Z, Zhang, Y, Mao, H, Ma, Z, Yin, Y, Wang, W, Tang, W, Tan, Z, Shi, J, Li, L and Ruan, B (2015) Altered fecal microbiota composition in patients with major depressive disorder. Brain, Behaviour, and Immunity 48, 186194. https://doi.org/10.1016/j.bbi.2015.03.016CrossRefGoogle ScholarPubMed
Joint FAO/WHO Food Standards Programme Secretariat of the CODEX Alimentarius Commission (2010) CODEX Alimentarius (CODEX) Guidelines on Nutrition Labeling CAC/GL 2-1985 as Last Amended 2010. Rome: FAO.Google Scholar
Kao, ACC, Safarikova, J, Marquardt, T, Mullins, B and Lennox, B (2019) Pro-cognitive effect of a prebiotic in psychosis: A double blind placebo controlled cross-over study. Schizophrenia Research 208, 460461.CrossRefGoogle ScholarPubMed
Kao, ACC, Spitzer, S, Anthony, DC, Lennox, B and Burnet, PWJ (2018) Prebiotic attenuation of olanzapine-induced weight gain in rats: Analysis of central and peripheral biomarkers and gut microbiota. Translational Psychiatry 8(1), 66. https://doi.org/10.1038/s41398-018-0116-8CrossRefGoogle ScholarPubMed
Kato, T, Monji, A, Hashioka, S and Kanba, S (2007) Risperidone significantly inhibits interferon-γ-induced microglial activation in vitro. Schizophrenia Research 92, 108115. https://doi.org/10.1016/j.schres.2007.01.019CrossRefGoogle ScholarPubMed
Katzbauer, B (1998) Properties and applications of xanthan gum. Polymer Degradation and Solubility 59, 8184.CrossRefGoogle Scholar
Kazemi, A, Noorbala, AA, Azam, K, Eskandari, MH and Djafarian, K (2019) Effect of probiotic and prebiotic vs placebo on psychological outcomes in patients with major depressive disorder: A randomized clinical trial. Clinical Nutrition 38(2), 522528. https://doi.org/10.1016/j.clnu.2018.04.010CrossRefGoogle ScholarPubMed
Kendig, MD, Lin, CS, Beilharz, JE, Rooney, KB and Boakes, RA (2014) Maltodextrin can produce similar metabolic and cognitive effects to those of sucrose in the rat. Appetite 77, 112. https://doi.org/10.1016/j.appet.2014.02.011CrossRefGoogle ScholarPubMed
Kim, C-S, Byeon, S and Shin, D-M (2020) Sources of dietary fiber are differently associated with prevalence of depression. Nutrients 12, 2813. https://doi.org/10.3390/nu12092813CrossRefGoogle ScholarPubMed
Kim, EJ, Hong, J and Hwang, J-W (2019) The association between depressive mood and cholesterol levels in korean adolescents. Korean Neuropsychiatric Association 16(10), 737744. https://doi.org/10.30773/pi.2019.03.24Google ScholarPubMed
Kim, S, Goel, R, Kumar, A, Qi, Y, Lobaton, G, Hosaka, K, Mohammed, M, Handberg, EM, Richards, EM, Pepine, CJ and Raizada, MK (2018) Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clinical Science 132, 701718. https://doi.org/10.1042/CS20180087Google ScholarPubMed
Kim, SJ, Han, K-T, Jang, S-Y and Park, E-C (2018) The association between low level of high-density lipoprotein cholesterol and mood disorder using time-dependent analysis. Journal of Affective Disorders 225, 317325. https://doi.org/10.1016/j.jad.2017.08.016CrossRefGoogle ScholarPubMed
Kim, Y-K, Jung, H-G, Myint, A-M, Kim, H and Park, S-H (2007) Imbalance between pro-inflammatory and anti-inflammatory cytokines in bipolar disorder. Journal of Affective Disorders 104, 9195. https://doi.org/10.1016/j.jad.2007.02.018CrossRefGoogle ScholarPubMed
Krogh, J, Benros, ME, Jørgensen, MB, Vesterager, L, Elfving, B and Nordentoft, M (2014) The association between depressive symptoms, cognitive function, and inflammation in major depression. Brain, Behavior, and Immunity 35, 7076. https://doi.org/10.1016/j.bbi.2013.08.014CrossRefGoogle ScholarPubMed
La Rosa, SL, Kachrimanidou, V, Buffetto, F, Pope, PB, Pudlo, NA, Martens, EC, Rastall, RA, Gibson, GR and Westereng, B (2019) Wood-derived dietary fibers promote beneficial human gut microbiota. Applied and Environmental Science 4(1), e0055418. https://doi.org/10.1128/mSphere.00554-18Google ScholarPubMed
Lattimer, JM and Haub, MD (2010) Effects of dietary fiber and its components on metabolic health. Nutrients 2(12), 12661289. https://doi.org/10.3390/nu2121266CrossRefGoogle ScholarPubMed
Lawton, CL, Walton, J, Hoyland, A, Howarth, E, Allan, P, Chesters, D and Dye, L (2013) Short term (14 days) consumption of insoluble wheat bran fibre-containing breakfast cereals improves subjective digestive feelings, general wellbeing and bowel function in a dose dependent manner. Nutrients 5(4), 14361455. https://doi.org/10.3390/nu5041436CrossRefGoogle ScholarPubMed
Lecerf, J-M, Dépeint, F, Clerc, E, Dugenet, Y, Niamba, CN, Rhazi, L, Cayzeele, A, Abdelnour, G, Jaruga, A, Younes, H, Jacobs, H, Lambrey, G, Abdelnour, AM and Pouillart, PR (2012) Xylo-oligosaccharide (XOS) in combination with inulin modulates both the intestinal environment and immune status in healthy subjects, while XOS alone only shows prebiotic properties. British Journal of Nutrition 108, 18471858. https://doi.org/10.1017/S0007114511007252CrossRefGoogle ScholarPubMed
Lechleitner, M, Hoppichler, F, Konwalinka, G, Patsch, JR and Braunsteiner, H (1992) Depressive symptoms in hypercholesterolaemic patients treated with pravastatin. The Lancet 340(8824), 910.Google ScholarPubMed
Lehto, SM, Niskanen, L, Tolmunen, T, Hintikka, J, Viinamäki, H, Heiskanen, T, Honkalampi, K, Kokkonen, M and Koivumaa-Honkanen, H (2010) Low serum HDL-cholesterol levels are associated with long symptom duration in patients with major depressive disorder. Psychiatry and Clinical Neurosciences 64(3), 279283. https://doi.org/10.1111/j.1440-1819.2010.02079.xCrossRefGoogle ScholarPubMed
Lewis, K, Lutgendorff, F, Phan, V, Söderholm, JD, Sherman, PM and McKay, D (2010) Enhanced translocation of bacteria across metabolically stressed epithelia is reduced by butyrate. Inflammatory Bowel Diseases 16(7), 11381148.CrossRefGoogle ScholarPubMed
Looijer-Van Langen, MAC and Dieleman, LA (2009) Prebiotics in chronic intestinal inflammation. Inflammatory Bowel Diseases 15(3), 454462. https://doi.org/10.1002/ibd.20737Google ScholarPubMed
Maggo, S, Clark, D and Ashton, JC (2012) The effect of statins on performance in the Morris water maze in Guinea pig. European Journal of Pharmacology 674, 287293. https://doi.org/10.1016/j.ejphar.2011.11.006CrossRefGoogle ScholarPubMed
Marcques, FZ, Nelson, E, Chu, P-Y, Horlock, D, Fiedler, A, Ziemann, M, Tan, JK, Kuruppu, S, Rajapakse, NW, El-Osta, A, Mackay, CR and Kaye, DM (2017) High-fiber diet and acetate supplementation change the gut microbiota and prevent the development of hypertension and heart failure in hypertensive mice. Ciculation 135, 964977. https://doi.org/10.1161/CIRCULATIONAHA.116.024545CrossRefGoogle Scholar
Matt, SM, Allen, JM, Lawson, MA, Mailing, LJ, Woods, JA and Johnson, RW (2018) Butyrate and dietary soluble fiber improve neuroinflammation associated with aging in mice. Frontiers in Immunology 9, 14. https://doi.org/10.3389/fimmu.2018.01832CrossRefGoogle ScholarPubMed
McLoughlin, RF, Berthon, BS, Jensen, ME, Baines, KJ and Wood, LG (2017) Short-chain fatty acids, prebiotics, synbiotics, and systemic inflammation: A systematic review and meta-analysis. The American Journal of Clinical Nutrition 106, 930945. https://doi.org/10.3945/ajcnGoogle ScholarPubMed
McRorie, JW and McKeown, NM (2017) Understanding the physics of functional fibers in the gastrointestinal tract: An evidence-based approach to resolving enduring misconceptions about insoluble and soluble fiber. Journal of the Academy of Nutrition and Dietetics 117(2), 251264. https://doi.org/10.1016/j.jand.2016.09.021CrossRefGoogle ScholarPubMed
Messaoudi, M, Lalonde, R, Violle, N, Javelot, H, Desor, D, Nejdi, A, Bisson, J-F, Rougeot, C, Pichelin, M, Cazaubiel, M and Cazaubiel, J-M (2011) Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. British Journal of Nutrition 105, 755764. https://doi.org/10.1017/S0007114510004319CrossRefGoogle ScholarPubMed
Monk, JM, Lepp, D, Wu, W, Pauls, KP, Robinson, LE and Power, KA (2017) Navy and black bean supplementation primes the colonic mucosal microenvironment to improve gut health. Journal of Nutritional Biochemistry 49, 89100. https://doi.org/10.1016/j.jnutbio.2017.08.002CrossRefGoogle ScholarPubMed
Morales, K, Wittink, M, Datto, C, Difilippo, S, Cary, M, Tenhave, T and Katz, IR (2006) Simvastatin causes changes in affective processes in elderly volunteers. Journal of the American Geriatrics Society 54, 7076. https://doi.org/10.1111/j.1532-5415.2005.00542.xCrossRefGoogle ScholarPubMed
Mudgil, D and Barak, S (2013) Composition, properties and health benefits of indigestible carbohydrate polymers as dietary fiber: A review. International Journal of Biological Macromolecules 61, 16. https://doi.org/10.1016/j.ijbiomac.2013.06.044CrossRefGoogle ScholarPubMed
Mudgil, D, Barak, S and Khatkar, BS (2014) Optimization of enzymatic hydrolysis of guar gum using response surface methodology. Journal of Food Science and Technology 51(8), 16001605. https://doi.org/10.1007/s13197-012-0678-zCrossRefGoogle ScholarPubMed
Muela, HCS, Costa-Hong, VA, Yassuda, MS, Moraes, NC, Memória, CM, Machado, MF, Macedo, TA, Shu, EBS, Massaro, AR, Nitrini, R, Mansur, AJ and Bortolotto, LA (2017) Hypertension severity is associated with impaired cognitive performance. Journal of the American Heart Association 6(1), e004579. https://doi.org/10.1161/JAHA.116.004579CrossRefGoogle ScholarPubMed
Murphy, EA, Davis, JM and Carmichael, MD (2010) Immune modulating effects of β-glucan. Current Opinion in Clinical Nutrition and Metabolic Care 13(6), 656661. https://doi.org/10.1097/MCO.0b013e32833f1afbCrossRefGoogle ScholarPubMed
Muth, AK and Park, SQ (2021) The impact of dietary macronutrient intake on cognitive function and the brain. Clinical Nutrition 40(6), 39994010. https://doi.org/10.1016/j.clnu.2021.04.043CrossRefGoogle ScholarPubMed
Ohlsson, L, Gustafsson, A, Lavant, E, Suneson, K, Brundin, L, Westrin, Ljunggren, L and Lindqvist, D (2019) Leaky gut biomarkers in depression and suicidal behavior. Acta Psychiatrica Scandinavica 139(2), 185193. https://doi.org/10.1111/acps.12978CrossRefGoogle ScholarPubMed
Onumpai, C, Kolida, S, Bonnin, E and Rastall, RA (2011) Microbial utilization and selectivity of pectin fractions with various structures. Applied and Environmental Microbiology 77(16), 57475754. https://doi.org/10.1128/AEM.00179-11CrossRefGoogle ScholarPubMed
Pasman, WJ, Blokdijk, VM, Bertina, FM, Hopman, W and Hendriks, H (2003) Effect of two breakfasts, different in carbohydrate composition, on hunger and satiety and mood in healthy men. International Journal of Obesity 27, 663668. https://doi.org/10.1038/sj.ijo.0802284CrossRefGoogle ScholarPubMed
Pedersen, C, Gallgher, E, Horton, F, Ellis, RJ, Ijaz, UZ, Wu, H, Jaiyeola, E, Diribe, O, Duparc, T, Cani, PD, Gibson, GR, Hinton, P, Wright, J, La Ragione, R and Robertson, MD (2016) Host-microbiome interactionsin human type 2 diabetes following prebiotic fibre (galactooligosaccharide) intake. British Journal of Nutrition 116(11), 18691877. https://doi.org/10.1017/S0007114516004086CrossRefGoogle Scholar
Prykhodko, O, Sandberg, J, Burleigh, S, Björck, I, Nilsson, A and Fåk Hållenius, F (2018) Impact of Rye kernel-based evening meal on microbiota composition of young healthy lean volunteers with an emphasis on their hormonal and appetite regulations, and blood levels of brain-derived neurotrophic factor. Frontiers in Nutrition 5, 45. https://doi.org/10.3389/fnut.2018.00045CrossRefGoogle ScholarPubMed
Qing, Q, Li, H, Kumar, R and Wyman, CE (2013) Xylooligosaccharides production, quantification, and characterization in context of lignocellulosic biomass pretreatment. In Aqueous Pretreatment of Plant Biomass for Biological and Chemical Conversion to Fuels and Chemicals, 1st edn. Hoboken, NJ: Wiley, pp. 391415. https://doi.org/10.1002/9780470975831.ch19CrossRefGoogle Scholar
Rabbani, GH, Teka, T, Zaman, B, Majid, N, Khatun, M and Fuchs, GJ (2001) Clinical studies in persistent diarrhea: Dietary management with green Banana or pectin in Bangladeshi children. Gastroenterology 121(3), 554560. https://doi.org/10.1053/gast.2001.27178CrossRefGoogle ScholarPubMed
Ramnani, P, Costabile, A, Bustillo, AGR and Gibson, GR (2015) A randomised, double-blind, cross-over study investigating the prebiotic effect of agave fructans in healthy human subjects. Journal of Nutritional Science 4, e10. https://doi.org/10.1017/jns.2014.68CrossRefGoogle ScholarPubMed
Romo-Araiza, A, Gutiérrez-Salmeán, G, Galván, EJ, Hernández-Frausto, M, Herrera-López, G, Romo-Parra, H, García-Contreras, V, María Fernández-Presas, A, Jasso-Chávez, R, Borlongan, CV, Ibarra, A, Kumar, A, Smulders, TV and Yegla, B (2018) Probiotics and prebiotics as a therapeutic strategy to improve memory in a model of middle-aged rats. Frontiers in Aging Neuroscience 10, 416. https://doi.org/10.3389/fnagi.2018.00416CrossRefGoogle Scholar
Russo, F, Linsalata, M, Clemente, C, Chiloiro, M, Orlando, A, Marconi, E, Chimienti, G and Riezzo, G (2012) Inulin-enriched pasta improves intestinal permeability and modifies the circulating levels of zonulin and glucagon-like peptide 2 in healthy young volunteers. Nutrition Research 32(12), 940946. https://doi.org/10.1016/j.nutres.2012.09.010CrossRefGoogle ScholarPubMed
Sajilata, MG, Singhal, RS and Kulkarni, PR (2006) Resistant starch – A review. Comprehensive Reviews in Food Science and Food Safety 5, 117.CrossRefGoogle ScholarPubMed
Sandberg, JC, Björck, IME and Nilsson, AC (2018) Impact of rye-based evening meals on cognitive functions, mood and cardiometabolic risk factors: A randomized controlled study in healthy middle-aged subjects. Nutrition Journal 17, 102. https://doi.org/10.1186/s12937-018-0412-4CrossRefGoogle ScholarPubMed
Schmidt, K, Cowen, PJ, Harmer, CJ, Tzortzis, G, Errington, S and Burnet, PWJ (2015) Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 232, 17931801. https://doi.org/10.1007/s00213-014-3810-0CrossRefGoogle ScholarPubMed
Schreurs, BG (2010) The effects of cholesterol on learning and memory. Neuroscience and Biobehavioral Reviews 34, 13661379. https://doi.org/10.1016/j.neubiorev.2010.04.010CrossRefGoogle ScholarPubMed
Shapiro, D, Jamner, LD, Goldstein, IB and Delfino, RJ (2001) Striking a chord: Moods, blood pressure, and heart rate in everyday life. Psychophysiology 38(2), 197204. https://doi.org/10.1111/1469-8986.3820197CrossRefGoogle Scholar
Shaw, RJ, Mackay, D, Pell, JP, Padmanabhan, S, Bailey, DS and Smith, DJ (2020) The relationship between antihypertensive medications and mood disorders: Analysis of linked healthcare data for 1.8 million patients. Psychological Medicine 51, 11831191. https://doi.org/10.1017/S0033291719004094CrossRefGoogle ScholarPubMed
Sherry, CL, Kim, SS, Dilger, RN, Bauer, LL, Moon, ML, Tapping, RI, Fahey, GC, Tappenden, KA and Freund, GG (2010) Sickness behavior induced by endotoxin can be mitigated by the dietary soluble fiber, pectin, through up-regulation of IL-4 and Th2 polarization. Brain Behavior and Immunity 24, 631640. https://doi.org/10.1016/j.bbi.2010.01.015CrossRefGoogle ScholarPubMed
Silk, DBA, Davis, A, Vulevic, J, Tzortzis, G and Gibson, GR (2009) Clinical trial: The effects of a trans-galactooligosaccharide prebiotic on faecal microbiota and symptoms in irritable bowel syndrome. Alimentary Pharmacology & Therapeutics 29(5), 508518. https://doi.org/10.1111/j.1365-2036.2008.03911.xCrossRefGoogle ScholarPubMed
Singh, N, Gurav, A, Sivaprakasam, S, Brady, E, Padia, R, Shi, H, Thangaraju, M, Prasad, PD, Manicassamy, S, Munn, DH, Lee, JR, Offermanns, S and Ganapathy, V (2014) Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40(1), 128139. https://doi.org/10.1016/j.immuni.2013.12.007CrossRefGoogle ScholarPubMed
Smith, AP (2005) The concept of well-being: Relevance to nutrition research. British Journal of Nutrition 93(1), S1S5.CrossRefGoogle ScholarPubMed
Smith, AP, Sutherland, D and Hewlett, P (2015) An investigation of the acute effects of oligofructose-enriched inulin on subjective wellbeing, mood and cognitive performance. Nutrients 7(11), 88878896. https://doi.org/10.3390/nu7115441Google ScholarPubMed
Smith, PM, Howitt, MR, Panikov, N, Michaud, M, Gallini, CA, Bohlooly-Y, M, Glickman, JN and Garrett, WS (2013) The microbial metabolites, short-chain fatty acids, regulate colonig Treg cell homeostasis. Science 341, 569573. https://doi.org/10.1126/science.1237947CrossRefGoogle ScholarPubMed
Sobotka, L, Brátova, M, Šlemrová, M, Maňák, J, Vižd’a, J and Zadák, Z (1997) Inulin as the soluble fiber in liquid enteral nutrition. Nutrition 13(1), 2125. https://doi.org/10.1016/S0899-9007(97)90874-1CrossRefGoogle ScholarPubMed
Song, J, Li, Q, Everaert, N, Liu, R, Zheng, M, Zhao, G and Wen, J (2020) Effects of inulin supplementation on intestinal barrier function and immunity in specific pathogen-free chickens with salmonella infection. Journal of Animal Science 2020(1), 110. https://doi.org/10.1093/jas/skz396Google Scholar
Stevens, BR, Goel, R, Seungbum, K, Richards, EM, Holbert, RC, Pepine, CJ and Raizada, MK (2018) Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut 67(8), 15551557. https://doi.org/10.1136/gutjnl-2017-314759CrossRefGoogle ScholarPubMed
Stilling, RM, de Wouw, M, Clarke, G, Stanton, C, Dinan, TG and Cryan, JF (2016) The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochemistry International 99, 110132. https://doi.org/10.1016/j.neuint.2016.06.011CrossRefGoogle ScholarPubMed
Strike, PC, Wardle, J and Steptoe, A (2004) Mild acute inflammatory stimulation induces transient negative mood. Journal of Psychosomatic Research 57, 189194. https://doi.org/10.1016/S0022-3999(03)00569-5CrossRefGoogle ScholarPubMed
Suzuki, T, Yoshida, S and Hara, H (2008) Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability. British Journal of Nutrition 100, 297305. https://doi.org/10.1017/S0007114508888733CrossRefGoogle ScholarPubMed
Swann, OG, Kilpatrick, M, Breslin, M and Oddy, WH (2020) Dietary fiber and its associations with depression and inflammation. Nutrition Reviews 78(5), 394411. https://doi.org/10.1093/nutrit/nuz072CrossRefGoogle ScholarPubMed
Talbott, S and Talbott, J (2009) Effect of BETA 1, 3/1, 6 GLUCAN on upper respiratory tract infection symptoms and mood state in marathon athletes. Journal of Sports Science and Medicine 8, 509515. 04 August 2020. Available at http://www.jssm.org.Google Scholar
Tedders, SH, Fokong, KD, Mckenzie, LE, Wesley, C, Yu, L, and Zhang, J (2011) Low cholesterol is associated with depression among US household population. Journal of Affective Disorders 135, 115121. https://doi.org/10.1016/j.jad.2011.06.045CrossRefGoogle ScholarPubMed
Van De Wouw, M, Boehme, M, Lyte, JM, Wiley, N, Strain, C, O’sullivan, O, Clarke, G, Stanton, C, Dinan, TG, Cryan, JF, Barrett, K, Grundy, D and Cryan, JF (2018) Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. The Journal of Physiology C 596, 49234944. https://doi.org/10.1113/JP276431CrossRefGoogle ScholarPubMed
Verhaar, BJ, Collard, D, Prodan, A, Levels, JH, Zwinderman, AH, Bäckhed, F, Vogt, L, Peters, MJ, Muller, M, Nieuwdorp, M and van den Born, B-JH (2020) Associations between gut microbiota, faecal short-chain fatty acids, and blood pressure across ethnic groups: The HELIUS study. European Heart Journal 41, 42594267. https://doi.org/10.1093/eurheartj/ehaa760CrossRefGoogle ScholarPubMed
Vetere, G, Ripaldi, L, Ais, E, Korob, G, Kes, M and Villamil, A (2007) Prevalence of anxiety disorders in patients with essential hypertension. Vertex 18(71), 2025.Google ScholarPubMed
Vinolo, MAR, Rodrigues, HG, Nachbar, RT and Curi, R (2011) Regulation of inflammation by short chain fatty acids. Nutrients 3(10), 858876. https://doi.org/10.3390/nu3100858CrossRefGoogle Scholar
Vulevic, J, Drakoularakou, A, Yaqoob, P, Tzortzis, G and Gibson, GR (2008) Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers 1-3. The American Journal of Clinical Nutrition 88, 14381484. https://doi.org/10.3945/ajcn.2008.26242Google Scholar
Waldecker, M, Kautenburger, T, Daumann, H, Busch, C and Schrenk, D (2008) Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. Journal of Nutritional Biochemistry 19, 587593. https://doi.org/10.1016/j.jnutbio.2007.08.002Google ScholarPubMed
Wang, H-X and Gao, W-J (2009) Cell type-specific development of NMDA receptors in the interneurons of rat prefrontal cortex. Neuropsychopharmacology 34, 20282040. https://doi.org/10.1038/npp.2009.20CrossRefGoogle ScholarPubMed
Wang, L, Zhu, Q, Lu, A, Liu, X, Zhang, L, Xu, C, Liu, X, Li, H and Yang, T (2017) Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin-angiotensin system. Journal of Hypertension 35(9), 18991908. https://doi.org/10.1097/HJH.0000000000001378CrossRefGoogle ScholarPubMed
Wright, CE, Strike, PC, Brydon, L and Steptoe, A (2005) Acute infammation and negative mood: Mediation by cytokine activation. Brain, Behavior, and Immunity 19, 345350. https://doi.org/10.1016/j.bbi.2004.10.003CrossRefGoogle Scholar
Yang, CC, Jick, SS and Jick, H (2003) Lipid-lowering drugs and the risk of depression and suicidal behavior. Archives of Internal Medicine 163(16), 19261932. https://doi.org/10.1001/archinte.163.16.1926CrossRefGoogle ScholarPubMed
Yang, T, Magee, KL, Colon-Perez, LM, Larkin, R, Liao, Y-S, Balazic, E, Cowart, JR, Arocha, R, Redler, T, Febo, M, Vickroy, T, Martyniuk, CJ, Reznikov, LR and Zubcevic, J (2019) Impaired butyrate absorption in the proximal colon, low serum butyrate and diminished central effects of butyrate on blood pressure in spontaneously hypertensive rats. Acta Physiologica 226(2), e13256. https://doi.org/10.1111/apha.13256CrossRefGoogle ScholarPubMed
Yang, T, Santisteban, MM, Rodriguez, V, Li, E, Ahmari, N, Carvajal, JM, Zadeh, M, Gong, M, Qi, Y, Zubcevic, J, Sahay, B, Pepine, CJ, Raizada, MK and Mohamadzadeh, M (2015) Gut dysbiosis is linked to hypertension. Hypertension 65(6), 13311340. https://doi.org/10.1161/HYPERTENSIONAHA.115.05315CrossRefGoogle Scholar
Zheng, P, Zeng, B, Zhou, C, Liu, M, Fang, Z, Xu, X, Zeng, L, Chen, J, Fan, S, Du, X, Zhang, X, Yang, D, Yang, Y, Meng, H, Li, W, Melgiri, ND, Licinio, J, Wei, H and Xie, P (2016) Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Molecular Psychiatry 21, 786796. https://doi.org/10.1038/mp.2016.44CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Overview of different types of dietary fibres with their concomitant origin, structure and degree of solubility, viscosity and fermentability.

Figure 1

Table 2. Human studies assessing the effect of dietary fibre interventions on mood and cognition.

Figure 2

Figure 1 Overview of potential biological mechanisms underlying dietary fibres’ effects on mood and cognition according to physicochemical properties. The degree of viscosity, fermentability, and solubility of dietary fibres influences gut microbiota composition and function, the immune system, as well as cholesterol levels, through which mood and cognition can be modulated. Via the direct immune route, viscous, fermentable, and soluble fibres can reduce pro-inflammatory cytokines, lower numbers and activation of splenic macrophages and DCs, and increase neutrophils in the blood. Cholesterol levels can be lowered by viscous, fermentable, and soluble fibres and impact the brain, but can also indirectly affect it by reducing hypertension. Fermentability and solubility of dietary fibre modulate the gut microbiota, by which it can influence affective and cognitive processes via SCFA-dependent mechanisms or BDNF levels. SCFAs can lower hypertension as well as improve immune function and intestinal barrier integrity. Via the indirect immune route, SCFAs suppress pro-inflammatory reactions by reducing pro-inflammatory cytokines, increasing anti-inflammatory cytokines and Treg cells, as well as restoring microglial cell morphology and reversing microglial immaturity. All these mechanisms have been associated with modulation of mood and cognition in health and disease. The likelihood that the different biological mechanisms underlie dietary fibres’ effect on mood and cognition are displayed in proportion, with larger cubicle areas reflecting increased potential. The green plus sign indicates beneficial effects. BDNF, brain-derived neurotrophic factor; DCs, dendritic cells; Treg cells, regulatory T-cells; SCFAs, short chain fatty acids.