Hostname: page-component-586b7cd67f-gb8f7 Total loading time: 0 Render date: 2024-11-27T11:28:18.649Z Has data issue: false hasContentIssue false

Fibre & fermented foods: differential effects on the microbiota-gut-brain axis

Published online by Cambridge University Press:  25 October 2024

Elizabeth Schneider*
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland
Ramya Balasubramanian
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Cork, Ireland
Aimone Ferri
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
Paul D. Cotter
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Cork, Ireland
Gerard Clarke
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
John F. Cryan
Affiliation:
APC Microbiome Ireland, University College Cork, Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
*
*Corresponding author: Elizabeth Schneider, email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

The ability to manipulate brain function through the communication between the microorganisms in the gastrointestinal tract and the brain along the gut-brain axis has emerged as a potential option to improve cognitive and emotional health. Dietary composition and patterns have demonstrated a robust capacity to modulate the microbiota-gut-brain axis. With their potential to possess pre-, pro-, post-, and synbiotic properties, dietary fibre and fermented foods stand out as potent shapers of the gut microbiota and subsequent signalling to the brain. Despite this potential, few studies have directly examined the mechanisms that might explain the beneficial action of dietary fibre and fermented foods on the microbiota-gut-brain axis, thus limiting insight and treatments for brain dysfunction. Herein, we evaluate the differential effects of dietary fibre and fermented foods from whole food sources on cognitive and emotional functioning. Potential mediating effects of dietary fibre and fermented foods on brain health via the microbiota-gut-brain axis are described. Although more multimodal research that combines psychological assessments and biological sampling to compare each food type is needed, the evidence accumulated to date suggests that dietary fibre, fermented foods, and/or their combination within a psychobiotic diet can be a cost-effective and convenient approach to improve cognitive and emotional functioning across the lifespan.

Type
Conference on ‘Diet and lifestyle strategies for prevention and management of multimorbidity’
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - SA
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike licence (https://creativecommons.org/licenses/by-nc-sa/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the same Creative Commons licence is used to distribute the re-used or adapted article and the original article is properly cited. The written permission of Cambridge University Press must be obtained prior to any commercial use.
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Introduction

The interplay between diet and brain health is receiving ever more attention(Reference Marx, Lane and Hockey1Reference Medawar, Huhn and Villringer3). In tandem, over the past two decades, the importance of the bidirectional communication between the gut microbiota (bacteria, viruses, fungi, protozoa, archaea) and the brain has also surfaced, positioning this community of microorganisms as an accessible target to alter brain function and behaviour. It is now well-established that diet is a potent manipulator of the gut microbiota, with the capacity to alter both microbial abundance and functionality(Reference Rothschild, Weissbrod and Barkan4,Reference Johnson, Vangay and Al-Ghalith5) , and therefore its potential to improve brain health, is emerging as a plausible therapeutic intervention strategy(Reference Long-Smith, O’Riordan and Clarke6). While several dietary foods and patterns have been shown to modify the gut microbiota, dietary fibre and fermented foods with their capacity to act as substrates for microbial digestion and to supply live microorganisms as well as the associated enzymatically converted food components, respectively, have been identified as key modulators of the gut microbiota and subsequent signalling to the brain(Reference Berding, Vlckova and Marx7Reference Stiemsma, Nakamura and Nguyen9).

Increasing attention is now given to the concept of a diet-microbiota-gut-brain axis(Reference Berding, Vlckova and Marx7) with an appreciation that it is not just what microbes are present in the gut but what they are doing and how they interact with diet to affect host health(Reference Sonnenburg and Bäckhed10). Undigested fibre is a major energy source for the gut microbiota and short-chain fatty acids (SCFAs) that are produced upon gut microbial fermentation of fibre influence cognitive and emotional responses of the host(Reference O’Riordan, Collins and Moloney11,Reference Dalile, Van Oudenhove and Vervliet12) . Similarly, fermented foods produced through enzymatic conversion of food components by live microorganisms also house a large community of microbes at the time of consumption (e.g. kefir, kimchi)(Reference Lang, Eisen and Zivkovic13) that can increase gut microbiota diversity(Reference Le Roy, Kurilshikov and Leeming14,Reference Wastyk, Fragiadakis and Perelman15) . These new or expanded members of the gut microbiota can subsequently produce neuroactive metabolites (e.g. SCFAs, polyphenolic, tryptophan, bile, gamma-aminobutyric acid (GABA)). Intriguingly, there is a growing appreciation that, behaviourally, both food types have been shown to benefit cognitive and emotional processing(Reference Aslam, Green and Jacka16Reference Balasubramanian, Schneider, Gunnigle and Cotter20), effects which are suggested to be mediated via the microbiota-gut-brain axis(Reference Berding, Carbia and Cryan19,Reference Marx, Scholey and Firth21) . It is therefore worth evaluating the relative efficacy of fibre and fermented foods to alter brain functioning and signalling along the microbiota-gut-brain axis to inform future interventional research and move toward personalised medicine approaches for brain health management. Currently, there are very few studies investigating the biological basis explaining the beneficial actions of fibre and fermented foods(Reference Wastyk, Fragiadakis and Perelman15,Reference Balasubramanian, Schneider, Gunnigle and Cotter20) . While the studies that have been conducted provide tentative evidence of the scope of influence that fibre and fermented foods have on human health, it is clear that observational and interventional studies directly/systematically evaluating each food component on the microbiota-gut-brain axis are, in general, lacking. In this narrative review, we evaluate the differential neuroactive potential of dietary fibre and fermented foods to modulate cognitive and emotional functioning in humans. We further discuss possible pathways through which fibre and fermented foods can act on the brain via the microbiota-gut-brain axis. Future directions for the field and the therapeutic potential of fibre and fermented foods to improve brain health are highlighted.

Fibre

The widely-adopted definition from Codex Alimentarius defines fibre, natural and synthetic, as ‘carbohydrate polymers with ten or more monomeric units, which are not hydrolysed by the endogenous enzymes’(Reference Alimentarius22). Although the terms are often used interchangeably, prebiotics only represent the subset of fibres that are ‘substrates that are selectively utilised by host microorganisms conferring a health benefit’(Reference Gibson, Hutkins and Sanders23). While most prebiotics are dietary fibres, not all fibres are prebiotics, nor are all prebiotics fibres. Indeed, certain polyphenols and omega-3 fatty acids also exhibit prebiotic-like effects on the gut microbiota(Reference Gibson, Hutkins and Sanders23,Reference Alves-Santos, Sugizaki and Lima24) , although not being classed as dietary fibres. Despite the large body of research examining the impact of fibre supplements on the microbiota-gut-brain axis, whole dietary approaches more closely resemble the variety of foods that humans consume each day, particularly in terms of the fibre delivery matrix. Therefore, only studies that examine fibre from dietary sources, including whole grains, legumes, fruits and vegetables, are included in this review.

Dietary fibre and the gut microbiota

Fructo- and galacto- oligosaccharides, inulin and pectins from plants, animal tissue, or food-borne microbes are important energy sources for the gut microbiota(Reference Sonnenburg and Sonnenburg25). Fermentation of dietary fibre increases the abundance of Bifidobacterium and lactobacilli(Reference So, Whelan, Rossi and Morrison26). It has been proposed that roughly 30 % of the dietary fibre found in grain products is accessible for microbial metabolism, while estimates suggest that 75–90 % of fibres from fruits and vegetables are metabolised by the gut microbiota(Reference Nyman, Asp and Cummings27,Reference Wisker, Daniel and Rave28) . However, insoluble fibres can increase faecal bulk to reduce gut transit time, which also shapes gut microbiota composition(Reference Procházková, Falony and Dragsted29).

The gut microbiota is highly responsive to both acute and chronic fibre intake. A dramatic restructuring of the gut microbiota, including a significant alteration of over 25 bacterial genera, was observed after just 24 hours with an intervention of reduced carbohydrate and fibre diet in participants with obesity and metabolic dysfunction-associated steatotic liver disease. Moreover, the carbohydrate reduction expectedly decreased the abundance of fibre-degrading bacteria (Lactococcus, Eggerrthella and Streptococcus), which resulted in decreased levels of SCFAs(Reference Mardinoglu, Wu and Bjornson30). On the other hand, observational data showing a prolonged loss of fibre intake linked to the Western Diet was associated with reduced fibre-fermenting Prevotella strains in Asians who emigrated to the United States. Further losses in the functional capacity to degrade complex fibres were observed, alongside reductions in microbial diversity that decreased with each generation(Reference Vangay, Johnson and Ward31). This data is consistent with observational comparisons of individuals from cultures who consistently consume higher quantities of fibre. For example, children from rural Burkina Faso whose staple diet includes larger quantities of cereals, legumes and vegetables displayed a greater abundance of bacteria from Prevotella and Xylanibacter genera and increased levels of SCFAs than European children(Reference De Filippo, Cavalieri and Di Paola32). An overall increased microbial diversity has also been detected in observational studies examining regular fibre consumers compared to non-consumers(Reference Schnorr, Candela and Rampelli33,Reference Martínez, Stegen and Maldonado-Gómez34) . Fibre intake has further been shown to manipulate gut microbiota functionality.

Fermented foods

Fermented foods are defined as ‘foods made through desirable microbial growth and enzymatic conversions of food components’(Reference Marco, Sanders and Ganzle35). Fermented foods have been culturally embedded as staple foods in nearly all societies since the beginning of human civilisation. Given this rich history, it is unsurprising that the process of fermentation is often relatively simple and requires few ingredients and minimal preparation/processing to achieve an end-product that (1) naturally prolongs shelf-life, (2) reduces toxicity of raw materials and increases digestibility, and (3) alters the flavour profile(Reference Balasubramanian, Schneider, Gunnigle and Cotter20,Reference Marco, Heeney and Binda36) . As such, fermented foods are an affordable and convenient food option that are already available and frequently consumed in most societies.

The primary microbes involved in fermented foods include yeast, acetic acid bacteria, lactic acid bacteria (such as Leuconostoc, members of the former genus Lactobacillus, and Streptococcus), Propionbacterium freudenreichii, Bacillus, and moulds(Reference Marco, Heeney and Binda36). There is an assumption that the bacteria in fermented foods are probiotic, but this is not necessarily so. Probiotics are defined as ‘live microorganisms, when administered in adequate amounts confer a health benefit on the host’(Reference Hill, Guarner and Reid37). Fermented foods house a rich repertoire of microorganisms (some of which have the potential to be probiotics), bioactive peptides, phytochemicals and peptides that can influence human health(Reference Balasubramanian, Schneider, Gunnigle and Cotter20). Predominantly by manipulating the composition and enhancing microbial diversity, fermented foods can also modulate the production of several metabolites with neuroactive potential including SCFAs, polyphenolic, tryptophan and bile metabolites(Reference Marco, Heeney and Binda36,Reference Mukherjee, Breselge and Dimidi38,Reference Valentino, Magliulo and Farsi39) .

Because the ingestion of live microbes may be an important aspect underlying their health effects, for the purpose of this review, we focus on fermented foods with viable microorganisms at the time of consumption (e.g. sauerkraut, kombucha, yogurt). Foods where the microorganisms have been deactivated through heat (tea, coffee, bread, cocoa) or through the addition of vinegar (pickles) are also interesting but outside of the scope of the current narrative. Due to the known direct behavioural properties resulting from acute consumption of alcoholic products (e.g. memory lapse, reduced alertness, changes in affect), alcohol is also not relevant to the current discussion.

Fermented foods and the gut microbiota

Observational reports demonstrate increased microbial diversity with fermented food consumption(Reference Noh, Jang and Kim40,Reference Taylor, Lejzerowicz and Poirel41) . For example, a higher alpha diversity and enrichment of Lactobacillus, Ruminococcus and Eubacterium was reported within an observational study comparing Koreans who consume large amounts of fermented legumes and high-fibre foods, including vegetables and nuts/seeds, showing potential synergistic effects from fermented food and fibre intake combinations(Reference Noh, Jang and Kim40). Differences in beta diversity were also observed in another observational study comparing regular fermented food consumers to occasional consumers, alongside increased abundances of Faecalibacterium prausnitzii, Prevotella spp, Pseudomonas spp, Clostridiales, Enterobacteriaceae, Lachnospiraceae and Bacteroides spp.(Reference Taylor, Lejzerowicz and Poirel41). Conversely, interventions with fermented foods largely fail to alter microbial diversity(Reference Berding, Bastiaanssen and Moloney8,Reference Le Roy, Kurilshikov and Leeming14,Reference Alvarez, Tap and Chambaud42,Reference Walsh, Walsh and Garcia-Perez43) . This discrepancy could be due to a number of factors that include differences in the food matrix of the fermented foods, minimal frequency/quantity of fermented foods consumed, differences in baseline microbial composition and provenance of raw materials, heterogeneity in analysis techniques with 16S rRNA sequencing being unable to capture changes at the strain level, and, importantly, diversity of the fermented foods selected for intervention. Moreover, the duration of the intervention may be insufficient to significantly shift the composition of gut microbiota, potentially masking effects detected in observational studies that reflect regular eating habits.

Cognition

Much of the evidence linking fibre intake to cognitive performance comes from observational data investigating whole dietary patterns such as fruit and vegetable intake, often in older adults. Greater fibre intake was associated with protection against age-related cognitive decline(Reference Ortega, Requejo and Andrés44Reference Fortune, Harville and Guralnik49). Similar beneficial effects of fibre on cognitive inhibition(Reference Khan, Raine and Drollette50) and reasoning(Reference Naveed, Venäläinen, Eloranta and Erkkilä51) have been observed in children aged 6–9 years. Observational studies linking fibre to cognitive performance in young-middle adulthood is lacking, but the fact that higher intake of fruits and vegetables at 18–30 years was associated with better executive functioning, attention and verbal memory in middle adulthood(Reference Mao, Chen and Pengcheng52) provides tentative support for a potential relationship.

Evidence from randomised-controlled trials (RCTs) with fibre intervention is mixed, both in terms of study quality and results obtained. A mixed-grain dietary intervention for 9 weeks to high school students improved sustained attention and inhibition alongside increased plasma brain-derived neurotrophic factor (BDNF) levels(Reference Chung, Park and Kwon53). In undergraduates, increased fibre intake from various sources improved memory performance and microbial richness(Reference Zhu, Ming and Wu54). In adults aged 50–70, a berry beverage containing 11 g of fibre and 795 mg of polyphenols improved working memory performance(Reference Nilsson, Salo and Plaza55). However, administration of a whole grain rye bread diet supplemented with resistant starch for 3 days to adults aged 52–70 improved mood but failed to improve cognitive performance despite increasing plasma SCFAs(Reference Sandberg, Björck and Nilsson56). The discrepant RCT data suggests that habitual intake/longer intervention may be needed to capture/facilitate the effects of fibre on cognition, and that elevated BDNF levels may mediate these effects. Supporting evidence can be seen in the fact that 3 ounces of almonds consumed daily for 6 months improved learning, planning, working memory, and visual memory in adults 50–75 years old, but no effects were seen after 3 months(Reference Mustra Rakic, Tanprasertsuk and Scott57). However, this delayed effect could be due to the accumulation of tocopherol, which is also related to cognitive functioning(Reference Ashley, Bradburn and Murgatroyd58,Reference Dorey, Gierhart and Fitch59) . The combination of fibre with other bioactives that are known to be associated with healthier eating patterns, including fatty acids and polyphenols(Reference Caruso, Torrisi and Mogavero60Reference Ross, Mayer and Horn62), and also to influence cognitive function is problematic and requires unravelling to parse specific mechanisms of action of fibre per se.

The majority of RCTs with fermented food measure age-related cognitive decline in older adults to which fermented foods show a protective effect(Reference Hwang, Park and Paik63Reference Ohsawa, Nakamura and Uchida68), which is also reported in the observational literature(Reference Hogervorst, Sadjimim and Yesufu69,Reference Tessier, Presse and Rahme70) . The RCTs conducted in older adults largely lack biological sampling with the exception of a small number of studies. In older adults, elevated serum BDNF levels from consumption of fermented soybean for 12 weeks positively correlated with improved attentional and memory performance(Reference Hwang, Park and Paik63), but camembert consumption for 12 weeks failed to improve memory despite an increase in serum BDNF levels(Reference Suzuki, Kojima and Osuka71), suggesting divergent results from different food substrates. Such heterogenous results, in general, are unsurprising given the broad definition of fibre and fermented foods.

While studies that examine the effects of fermented foods on cognitive processes in younger/middle-aged adults are sparse, the fact that a fermented milk drink altered brain activity in the frontal cortex of healthy women with a mean age of 30(Reference Tillisch, Labus and Kilpatrick72) suggests potential effects on executive functioning in addition to the observed hippocampal effects. Moreover, in younger adults aged 25–45, 4-week kefir consumption improved performance on hippocampal-dependent relational memory-associated tasks, but memory improvements were not correlated with the increased Lactobacillus levels(Reference Cannavale, Mysonhimer and Bailey73), suggesting indirect effects of microbial signalling to the brain that were not measured in that study.

Taken together, fibre intake appears to alter attention, memory, and executive functioning when consumed habitually for longer periods across the lifespan, although more research in younger/middle-aged adults is still warranted. For older adults, in which much of the data is derived, fermented foods exhibit a protective effect against global age-related cognitive decline, both over time and following briefer interventions. Nevertheless, additional effects of fermented foods on executive functioning in younger/middle-aged adults cannot be ruled out given the lack of studies that directly assess these cognitive domains in younger populations. The effects of fermented foods on cognitive processes in children and adolescents also remain largely unknown. Moreover, the lack of functional imaging data to determine a psychophysiological interaction for each food type precludes neurological insights. Increased BDNF could be a mediating signalling pathway, but formal exploration within a statistical mediation model is needed wherein BDNF is entered as a predictor to determine if it partially or fully explains the relationship between fibre/fermented foods on cognition. By determining the mechanism or pathway through which an independent variable influences a dependent variable, mediation models are useful analyses for statistically disentangling causal factors(Reference Pearl74). Moreover, the relationship between peripheral and central BDNF is disputed(Reference Klein, Williamson and Santini75), thus warranting some caution in interpreting effects on the brain. More attention is required to parse the mechanisms from the gut to the brain that are mediating such effects(Reference Ribeiro, Ferri and Clarke76).

Emotion

Fibre consumption alone and as part of a healthy dietary pattern has been consistently associated with better mood in clinical and non-clinical populations(Reference Kim, Byeon and Shin77Reference Jaatinen, Korpela and Poussa81), but a systematic review and meta-analysis concluded that the extant RCT data is inconsistent with observational findings(Reference Aslam, Lotfaliany and So18). Recently, it was shown that an 8-week diet with a high potential prebiotic content improved anxiety, stress, and sleep in adults compared to a probiotic alone, a combination of the pre- and probiotic (synbiotic), or placebo and diet as usual. The prebiotic diet comprised a minimum of 5 g/d of asparagus, garlic, onion, oats, whole wheat, chickpeas, or watermelon. The probiotic contained Bifidobacterium bifidum (Bb-06), Bifidobacterium animalis subsp. lactis (HNO19), Bifidobacterium longum (R0175), Lactobacillus acidophilus (La-14), Lacticaseibacillus helveticus (R0052), Lacticaseibacillus casei (Lc-11), Lactiplantibacillus plantarum (Lp-115), Lacticaseibacillus rhamnosus (HN001)(Reference Freijy, Cribb and Oliver82). The beneficial effects observed in this study may be attributed to the high dosage of prebiotic-specific foods, meriting further attention. On the other hand, very few studies have explored the link between fermented foods and emotion, indicating a gap in understanding their chronic effects on mood. In young adults, higher frequency of fermented food consumption from sources that included live and inactive fermented foods in addition to non-fermented foods (i.e. yogurt, kefir, soy beverages and foods, miso soup, sauerkraut, dark chocolate, juices that contain microalgae, pickles, tempeh, kimchi) over the previous 30 days was linked to reduced social anxiety symptoms(Reference Hilimire, DeVylder and Forestell83). Conversely, Karbownik et al. discovered that higher total fermented food consumption from a variety of sources (cheese, yogurt, kefir, soured milk, kvass, unpasteurised beer, fermented and pickled vegetables with brine) over the previous 7 days was associated with increased depressive and anxiety symptoms in healthy medical students, but the inverse was found for medical students who reported an ongoing psychiatric disorder(Reference Karbownik, Mokros and Kowalczyk84). The difference in the timeframe of the assessment of fermented foods intake and the discrepancy in criteria for fermented foods might explain the inconsistent results.

RCTs with fermented food intervention show inconsistencies in effects, which could be due to differences in participant demographics. For example, fermented dairy products for 2 weeks(Reference Cannavale, Mysonhimer and Bailey73), 8 weeks(Reference Ohsawa, Nakamura and Uchida68), or 12 weeks(Reference Chung, Jin and Cui66) failed to improve mood in healthy adults. However, yogurt and the fermented dairy product, quark, improved mood only in participants with poor health status (i.e. immune depressed or chronic disease) and not for healthy participants(Reference Baars, Berge and Garssen85). Similarly, fermented kefir improved mood in participants with overweight(Reference Pražnikar, Kenig and Vardjan86) and fermented bonita fish broth improved fatigue and mood disturbances in adults with chronic fatigue(Reference Kuroda, Ishizaki and Maruyama87). Despite reports of improved mood in healthy women after consuming bonita broth for 2 weeks(Reference Nozawa, Ishizaki and Kuroda88), the results collectively suggest that fermented foods have larger effects on mood for individuals with compromised health status.

Similar to the cognitive data, prolonged fibre intake is likely to improve mood more than briefer periods of intake. Moreover, research that examines the impact of fibre and fermented foods on emotion in adolescence or middle adulthood is currently lacking. Considering this is a period with greater mood disturbances(Reference Pedrelli, Shapero and Archibald89,Reference Black and Rofey90) , more research is urgently needed in this population. The effects of fermented foods on mood in clinical and non-clinical populations, especially over longer time periods, is still largely unknown(Reference Ribera, Sánchez-Ortí and Clarke91). However, it appears that fermented foods may have preferential effects on individuals with comorbid health conditions.

Signalling pathways

Few studies incorporate the assessment of potential biological mechanisms to support behavioural findings, which precludes insight into mediating pathways. Given the demonstrated capacity for fibre and fermented foods to concurrently modulate gut microbiota and the brain, each food likely alters signalling pathways from the gut to the brain. It is now accepted that the gut microbiota communicates with the brain along several pathways including immune, hypothalamic-pituitary-adrenal (HPA) axis, serotonin/tryptophan/kynurenine, vagal, neuroendocrine, and metabolome signalling(Reference Cryan, O’Riordan and Cowan92) (see Fig. 1). In the next sections we outline the effects of fibre and fermented foods on each communication pathway, prioritising evidence from humans where possible and available, to determine potential mechanisms of action underlying cognitive and emotional effects.

Fig. 1. Fermented foods are rich in microorganisms, bioactive peptides, phytochemicals, and peptides that can modulate brain function through enhancing microbial diversity leading to an enrichment of diverse microbial metabolites. Dietary fibre increases microbial richness, and neuroactive SCFAs are produced as a result of microbial fermentation of dietary fibre. Both fermented foods and fibre support intestinal and BBB integrity to prevent peripheral and central inflammation for optimal cognitive and emotional functioning. BBB: blood-brain barrier. GLP-1: glucagon-like peptide 1. IL: interleukin. PP: polyphenol. PYY: peptide YY. SCFA: short-chain fatty acid. Vit K, B: Vitamin K and B. Created with BioRender.com.

Immune system

The immune system is one of the most direct signalling pathways along the microbiota-gut-brain axis and has been shown to be fundamental in modifying behaviour across the lifespan(Reference Ratsika, Cruz Pereira and Lynch93Reference Macpherson, Pachnis and Prinz95). The highest concentration of immune cells is located within the gastrointestinal tract, and these cells are in constant communication with the gut microbiota for the identification of potentially harmful pathogens. In mice, short-term exposure to a Western Diet deprived of fibre was sufficient to impair mucosal and systemic immunity, which created a window for opportunistic pathogens to invade intestinal tissue(Reference Siracusa, Schaltenberg and Kumar96). Re-introduction of dietary fibre re-programmed T cell metabolism and restored mucosal and systemic immunity. In this same study, healthy adults received a high-fibre diet comprised of a variety of food sources for five days before receiving a low-fibre diet for an additional five days. Not only did fibre deprivation reduce the abundance of fibre-fermenting bacteria (Eubacterium and bacteria from Lachnospiraceae family), but the main butyrate producer in the human microbiota (Agathobaculum butyriciproducens and Faecalibacterium prausnitzii) was decreased. These results were mirrored by a significant peripheral reduction of systemic TH17 cells co-expressing IL-17A and TNF-α and TH1 cells(Reference Siracusa, Schaltenberg and Kumar96). However, the anti-inflammatory effects of fibre have been shown to be moderated by baseline conditions of the individual. For example, Wastyk et al. found differential effects of fibre on inflammation that was dependent on baseline inflammation levels(Reference Wastyk, Fragiadakis and Perelman15). Similarly, the efficacy of soluble fibre intake on inflammation markers was reduced in individuals with lower microbiota richness(Reference Cotillard, Kennedy and Kong97).

In a recent RCT that compared administration of fermented yogurt against heat-treated yogurt with inactivated bacteria and two unfermented controls of whole milk and chemically acidified milk for 16 weeks to men with obesity, there were no differences between fermented yogurt and controls on c-reactive protein (CRP), IL-6, and TNF alpha levels(Reference Sandby, Magkos and Chabanova98). Despite these null effects, it is worth noting the sophisticated design of this study which incorporated several different fermented controls in line with our previous recommendations to enhance the scientific quality of future research(Reference Balasubramanian, Schneider, Gunnigle and Cotter20). These findings are also accordant with the results of a meta-analysis on the effects of fermented food intervention on inflammation in heterogeneous cohorts, which revealed that there was no dose-dependent association between fermented food consumption and inflammatory cytokine profile (IL-6, TNF-α, CRP)(Reference SaeidiFard, Djafarian and Shab-Bidar99). However, similar to dietary fibre, these effects interact with other factors such as age and food substrate. Differential results were shown when the analysis was split between participants below or above 50 years of age, such that fermented foods enhanced CRP and IL-6 levels in participants under 50 but significantly reduced IL-6 levels in participants over 50. Fermented foods decreased TNF-α levels in both age groups. The authors suggested the moderating effects of age are likely due to increased inflammation associated with aging. Furthermore, subgroup analyses of fermented dairy products demonstrated a reduction in CRP and elevation in IFN-γ, and no effect on other inflammatory markers (IL-10, IL-6 and Il-12)(Reference SaeidiFard, Djafarian and Shab-Bidar99). This may explain the anti-inflammatory effects observed following a varied, but predominantly yogurt, 10-week dietary intervention in healthy adults(Reference Wastyk, Fragiadakis and Perelman15). Pooling fermented foods for a meta-analysis likely obscures physiological effects of each food individually given their diverse substrate nature, bioactive profile, and microbial consortia that they host.

Hypothalamic-pituitary-adrenal (HPA) axis and stress

The HPA axis and its hormone, cortisol, is activated in response to acute and chronic stress and has been shown to be crucially regulated by the microbiota-gut-brain axis(Reference Leigh, Uhlig and Wilmes100Reference Nagpal and Cryan102). Germ-free rodents, deprived of exposure to microorganisms at birth and reared in sterile environments, display exaggerated HPA axis responsiveness to stressors compared to colonised controls(Reference Sudo, Chida and Aiba103Reference Lyte, Gheorghe and Goodson106), and Bifidobacterium infantis administration reverses this response in a time-window dependent manner(Reference Sudo, Chida and Aiba103). Additionally, administration of strains of Lactobacillus and Bifidobacterium has been shown to attenuate anxiety and depressive-like behaviour brought about by early-life stress(Reference Eutamene, Lamine, Chabo and Theodorou107,Reference Desbonnet, Garrett and Clarke108) , indicating a key signalling pathway from the gut to the brain(Reference Cryan, O’Riordan and Cowan92).

In humans, responses to stressors can be evaluated from naturally occurring acute and chronic stressors to provide greater generalisability. Conversely, experimental induction of acute stress in a laboratory setting has the advantage of reducing extraneous factors, but insights into chronic stress cannot be gained(Reference Allen, Kennedy and Cryan109). Reductions in salivary cortisol levels from fermented milk with Lacticaseibacillus casei strain Shirota(Reference Takada, Nishida and Kataoka-Kato110,Reference Kato-Kataoka, Nishida and Takada111) , but not from Lactobacillus casei DN-114001 fermented milk(Reference Marcos, Wärnberg and Nova112), have been observed in students undergoing academic exams and in school-aged students consuming a West African fermented milk product containing sugar and millet(Reference Brett, Koko and Doumbia113). Despite this encouraging evidence, more studies are needed that assess the impact of non-dairy fermented foods on acute and chronic stress in heterogeneous populations including older aged adults and patients with clinical stress disorders.

Much of the evidence for a link between dietary fibre and cortisol comes from observational or RCT studies that examine dietary patterns as opposed to fibre alone. For example, acute intake of low-fibre/high-sugar meal to ethnic minority adolescents with obesity was related to elevated cortisol levels compared to a low-sugar/high-fibre meal(Reference Wen, Chou and Belcher114). Chronic adherence to a healthy diet rich in fibrous foods and polyunsaturated fats decreased cortisol levels in response to an acute laboratory stressor in women with overweight/obesity(Reference Soltani, Keim and Laugero115). Intriguingly, sweet, fatty and snack food consumption frequency was positively linked to cortisol levels in a large sample of children aged 5–10, but fruit and vegetable intake had no effect(Reference Michels, Sioen, Braet and Huybrechts116). Combined, these results suggest that the negative effects of high-fat/sugar foods may have a greater impact on cortisol levels than from fibre alone(Reference Pearlmutter, DeRose and Samson117). While dietary patterns more closely resemble actual eating patterns, specificity in determining the mechanism of action of the efficacious compound is needed to determine if the effects observed are due to fibre or polyunsaturated fat intake.

Vagus nerve

The vagus nerve is the most direct pathway linking the gut to the brain(Reference Fülling, Dinan and Cryan118). Interestingly, specific probiotic bacteria, such as Limosilactobacillus reuteri and Lacticaseibacillus rhamnosus, that are present in some fermented foods have demonstrated dependency on vagal signalling to exert effects on the brain(Reference Sgritta, Dooling and Buffington119Reference Bercik, Park and Sinclair121). Similarly, SCFAs have been shown to stimulate vagal signalling(Reference Bruning, Chapp and Kaurala122,Reference Goswami, Iwasaki and Yada123) , suggesting another pathway through which fibre influences cognition. In rodents, a high-fat, high-sugar carbohydrate diet impaired vagus nerve signalling of satiety(Reference Loper, Leinen and Bassoff124), and administration of potato-derived resistant starch inhibits remodelling of vagal satiety signalling from a high-fat diet(Reference Klingbeil, Cawthon and Kirkland125).

Short-chain fatty acids

Microbial fermentation of undigested dietary fibre stimulates the formation of organic acids (lactic, succinic acid) and SCFAs (acetate, propionate, and butyrate). There are several pathways through which SCFAs influence brain activity, indicating the prominence of this pathway between fibre intake and the brain. First, SCFAs can modulate concentrations of neurotransmitters (e.g. GABA, serotonin, glutamate) and neurotropic factors(Reference Silva, Bernardi and Frozza126) that are directly involved in cognitive and emotional processes. Through supporting the intestinal and blood-brain barrier (BBB), SCFAs protect against neuroinflammation(Reference O’Riordan, Collins and Moloney11,Reference Khoshbin and Camilleri127) . Finally, a combination of SCFAs (acetate, propionate, and butyrate), but not butyrate alone(Reference Dalile, Fuchs and La Torre128), alters the HPA axis to attenuate stress-induced cortisol increases(Reference Dalile, Vervliet and Bergonzelli129), suggesting synergistic effects of different SCFAs on stress. Fermented foods have also been shown to alter SCFAs. For example, Spanish residents who consumed greater amounts of cheese had higher levels of all SCFAs(Reference González, Fernández-Navarro and Arboleya130). However, given that SCFAs are produced as a result of fibre fermentation, fibre is likely to have stronger effect on concentrations than fermented foods.

Other signalling pathways

Enteroendocrine

Enteroendocrine L cells within the intestinal epithelial cells secrete glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) in the postprandial state. Beyond their effects on satiety, gut-derived peptides GLP-1 and PYY have be linked to cognitive and emotional processing(Reference McIntyre, Powell and Kaidanovich-Beilin131,Reference Stadlbauer, Woods and Langhans132) . Short- and long-term dietary fibre intake concurrently increases self-report feelings of satiety and the release of gut peptides GLP-1 and PYY(Reference Wanders, van den Borne and de Graaf133Reference Al-Mana and Robertson135). Moreover, SCFAs can alter the production of GLP-1 and PYY, indicating indirect pathways(Reference Zhao, Zheng and Ding136). GLP-1 can also be stimulated by certain Lactobacillus strains present in fermented foods(Reference Simon, Strassburger and Nowotny137), but further research is needed that assesses GLP-1 and PYY release from whole foods in human participants.

Bile acids

Primary bile acids that are formed in the liver from cholesterol breakdown are further transformed into secondary bile acids by gut microbiota(Reference Rowland, Gibson and Heinken138). Bile acids can cross the blood-brain barrier to influence brain function. Indeed, altered bile acid profiles have been shown in mild cognitive impairment and Alzheimer’s Disease(Reference Mahmoudian Dehkordi, Arnold and Nho139,Reference Varma, Wang and An140) , but bile acids have also shown neuroprotective effects on the brain in preclinical models(Reference Theofilopoulos, Wang and Kitambi141). The binding of dietary fibre to conjugated bile acids prior to metabolism by the gut microbiota hints at the connection between dietary intake and microbial metabolites(Reference Singh, Metrani and Shivanagoudra142). Indeed, increased secondary bile acid, taurolithocholic acid, was observed following a four-week diet high in whole grains, legumes, fruits, and vegetables in healthy adults(Reference Ginos, Navarro and Schwarz143). Although the relationship between fermented foods and bile acids is less established, fermented milk kefir increased secondary bile acids that were decreased from high-fat diet in rats(Reference Gao, Mao and Wang144).

Other metabolites

Polyphenols and polyamines

Fibrous and fermented foods are rich sources of polyphenols, and fermentation of plant and vegetables through lactic acid bacteria enhances the conversion of phenolic compounds to biologically active metabolites leading to an increase the production of phenolic metabolites(Reference Johnson and de Mejia145,Reference Filannino, Bai and Di Cagno146) . Moreover, polyamines (spermine, spermidine and putrescine), which are also produced by the gut microbiota and affect the brain(Reference Makletsova, Rikhireva and Kirichenko147), are found in plant-derived foods linked to phenolic compounds(Reference Di Martino, Campilongo and Casalino148,Reference Muñoz-Esparza, Latorre-Moratalla and Comas-Basté149) . Gut microbiota utilisation of polyphenols results in phenolic compounds, which has shown associations with cognitive resilience in rats(Reference Frolinger, Sims and Smith150). As such, it is still largely unknown if polyphenols in fibrous and fermented foods are driving their beneficial impact on health.

Neurotransmitters

The gut microbiota indirectly and directly stimulates the production of neurotransmitters to influence central nervous system activity. There is a growing appreciation that tryptophan and its metabolic pathways to serotonin or kynurenine can be key signalling pathways within the microbiota-gut-brain axis(Reference Gheorghe, Martin and Manriquez151Reference Rothhammer, Mascanfroni and Bunse153). Although the gut produces the majority of serotonin(Reference Appleton154), it is unable to cross the BBB directly. However, dietary-derived tryptophan is absorbed by the gut and then transported, along with its metabolite kynurenine, to the brain to influence behaviour(Reference O’Mahony, Clarke and Borre155). A strain of Bifidobacterium infantis, a bacterium present in fermented foods, has been shown to elevate plasma tryptophan levels to increase supply to the central nervous system for serotonin production(Reference Desbonnet, Garrett and Clarke108). Evidence for neuromodulatory capacity of dietary fibre and fermented foods in humans is sparse. One study showed that Lacticaseibacillus casei fermented milk product consumption for 8 weeks increased faecal serotonin and decreased gastrointestinal distress without altering serum tryptophan or kynurenine levels(Reference Kato-Kataoka, Nishida and Takada111). High dietary fibre intake positively correlates with indolepropionic acid, a microbial metabolite of tryptophan(Reference Tuomainen, Lindström and Lehtonen156), and administration of whole grain rye lowered plasma serotonin concentrations in healthy adults(Reference Keski-Rahkonen, Kolehmainen and Lappi157).

Like serotonin, dopamine produced within the gut(Reference Asano, Hiramoto and Nishino158) cannot cross the BBB, but its precursor L-3,4-dihydroxyphenylalanine (L-DOPA) can via large neutral amino acid transporters(Reference Kageyama, Nakamura and Yamasaki159). Given the known relationship between dopamine and cognitive and emotional processing(Reference Alexander, Aragon and Bookwala160), and the fact that hyper-palatable foods deprived of fibre consumed chronically blunts central dopaminergic activity(Reference Volkow, Wang and Baler161), it is necessary to understand how the gut microbiota and brain communicate through food-induced/microbiota mediated alteration of peripheral and central dopamine.

The primary inhibitory neurotransmitter, GABA, plays a key role in affective and cognitive processing(Reference Koh, Kwak and Cheong162,Reference Luscher, Maguire and Rudolph163) . GABA is synthesised by bacterial strains found in fermented foods(Reference Richard and Foster164,Reference Siragusa, De Angelis and Di Cagno165) , and fermented foods that contain high levels of lactobacilli frequently also contain millimolar levels of GABA(Reference Li, Gao, Cao and Xu166). Intriguingly, fermentation by Lactiplantibacillus plantarum VTT E-133328 of faba bean flour enhanced GABA levels of the faba bean flour(Reference Coda, Melama and Rizzello167), indicating additive effects from fermenting high-fibre foods. Notably, the ability of GABA to cross the BBB in appreciable quantities is disputed and further research in warranted to understand the contribution of peripheral v. central mechanisms(Reference Boonstra, de Kleijn and Colzato168).

The cholinergic neurotransmitter, acetylcholine, is found in both the central and peripheral nervous system. Acetylcholine is highly implicated in shaping emotional and cognitive processes(Reference Picciotto, Higley and Mineur169). Several bacteria commonly found in fermented foods produce acetylcholine(Reference Horiuchi, Kimura and Kato170), demonstrating the potential for fermented food interventions to modulate peripheral acetylcholine and possibly its signalling to the brain. It was recently shown that a cafeteria diet deficient in dietary fibre impaired hippocampal-dependent memory and hippocampal acetylcholine signalling in rats(Reference Hayes, Tierno Lauer and Kao171). It will be interesting to test whether fibrous or fermented foods can rescue deficits in acetylcholinergic signalling induced by the Western Diet.

Vitamins and minerals

The effects of vitamin and mineral deficiencies on cognitive and emotional health are well-defined(Reference Muscaritoli172). Vitamin K and several B vitamins are produced by gut microbiota and host intake of these vitamins increases microbial diversity and richness, alongside an increase in the production of SCFAs(Reference Pham, Dold and Rehman173). Dietary fibre and substrates used for fermentation are rich in vitamins, and fermentation further increases the abundance of vitamins(Reference Septembre-Malaterre, Remize and Poucheret174). Similarly, minerals, such as magnesium and zinc found in both fibrous and fermented foods, have limited bioavailability when obtained from vegetable sources due to the presence of phytates and oxalates(Reference Gibson, Raboy and King175), which form complexes with minerals and limit their absorption. Fermentation through different microorganisms improves their bioavailability and absorption by breaking down phytate and oxalate complexes with minerals(Reference Ahmed, Xu and Sulieman176,Reference Wadamori, Vanhanen and Savage177) . Like polyphenols, more granularity is needed to understand mechanisms of action specific to the vitamin and mineral content present in fibrous and fermented foods.

Future directions

In addition to evaluating the unique effects of fermented foods or fibre on the microbiota-gut-brain axis, we have highlighted several gaps in the literature that are summarised in Fig. 2 and described herein.

Fig. 2. Potential approaches to address challenges in nutrition studies for the microbiota-gut-brain axis. Created with BioRender.com.

A review of the cognitive and emotional literature has emphasised the need for multimodal outcomes that assess behavioural changes on a full battery of cognitive tasks and self-report measures of affect in addition to biological samples within the same study design. To the best of our knowledge, only two studies directly assess the impact of fermented foods on brain functioning, and no studies used imaging techniques to study the neurological effects of fibre in humans. Combined methodology will shed light on the biopsychological mechanisms necessary to inform the development and selection of potential psychobiotic treatments.

We continue to know very little about how genetic backgrounds moderate the effects of each food type on gut-brain signalling. For example, some individuals exhibit differences in the metabolism of fermentation byproducts including biogenic amines such as histamine, which can indirectly affect cognitive and emotional processing through physical side effects and alterations of arousal(Reference Maintz and Novak178). Alternatively, fermented foods can also indirectly improve brain activity through enhancing digestibility in individuals with certain food intolerances by, for example, converting lactose into lactic acid. Future studies should investigate how individuals from different populations and genetic backgrounds respond to interventions with fibre or fermented foods. Baseline gut microbiota composition and functionality is also highly individual, and baseline differences in the fermentation and/or colonic absorption of fibre can impact butyrate production(Reference So, Whelan, Rossi and Morrison26,Reference Holmes, Villa and Durand179) . Employment of a crossover design, where each participant serves as their own control, is a possible solution to this confound. It is imperative, however, to counterbalance experimental arms in a crossover design and include a washout period to ensure the absence of prolonged effects on the gut microbiota that can spillover into the next experimental arm. The duration of the intervention should also be prolonged, because effects of fibre on cognition were observed at six months, but not three months, which may explain the discrepancies between observational and interventional data(Reference Mustra Rakic, Tanprasertsuk and Scott57). It is likely that longer interventions are needed in healthy populations for which changes may be small in effect size. Longer interventions are also useful to allow for tolerance to a high-fibre and/or fermented food diet that may initially cause gastric distress and therefore affect cognitive/emotional measures. Testing in healthy populations may further necessitate selection of behavioural measures that can capture small changes alongside utilisation of next generation sequencing of the microbiota and metabolome (e.g. shotgun metagenomics) to detect resolution at the strain level alongside functional changes(Reference Petrone, Aqeel and Jiang180). Moreover, biomarkers are needed to identify food consumption patterns and their potential downstream metabolites that are produced as a consequence of intake and can affect the overall health of the consumer(Reference Li, Burton-Pimentel and Brouwer-Brolsma181). Throughout the intervention, longitudinal captures of the gut microbiota via repeat faecal sampling can provide valuable information about the time course of effects.

A granular understanding of specific fibre subtypes on health outcomes is needed for personalised/precision approaches. However, the fact that fibrous foods can simultaneously comprise fermentable soluble and insoluble fibre provides a challenge. Collaboration with bioinformaticians to establish an open-source database that catalogues the constituents of each fibre is needed to advance mechanistic insight. More specificity is similarly required for fermented foods. There are several studies that evaluate the effects of singular fermented foods, but heterogeneity in the participants, length of the intervention, and outcomes limits insights gained from aggregating data(Reference Marx, Scholey and Firth21). In an RCT where participants consumed a variety of fermented foods, a stronger correlation to alpha diversity from yogurt and vegetable brine drink consumption was observed, but this could be due to the higher consumptions rates of these items relative to the other fermented food types (kefir, cottage cheese, kombucha, and fermented vegetables)(Reference Wastyk, Fragiadakis and Perelman15). Exploitation of large datasets from individuals who consume a variety of fermented foods is needed to avoid floor effects and to achieve adequate statistical power required to determine which fermented foods are superior in modulating brain activity. It is worth noting that metagenomics revealed that, of those studied, water kefirs, sauerkrauts, and kvasses contained the greatest concentration of potential health-associated gene clusters(Reference Leech, Cabrera-Rubio and Walsh182). Combining in silico methods with functional behavioural readouts is a promising approach to identify top-performing fermented foods to improve neurobehaviour. Another limitation in the isolation of specific fermented foods is the lack of validated, culturally sensitive and/or feasible tools to measure fermented food intake(Reference Li, Brouwer-Brolsma and Burton183,Reference Campbell, Hauptmann and Campbell184) . Specifically, fermented foods are currently assessed via unvalidated self-report measures of frequency or through self- or dietitian-led food logging or food recall, which is onerous for the participant and/or the researcher.

Towards new psychobiotic therapies

To date, only one study has directly compared the differential effects of fibre and fermented foods in humans within the same experimental design(Reference Wastyk, Fragiadakis and Perelman15). In this study, healthy adults were randomly allocated to consume a high quantity of a variety of fibrous foods (n=18) or fermented foods (n=18) for 10 weeks with no control group. Distinct effects on the gut microbiome were found such that fibre altered microbial functionality by decreasing the production of branched-chain fatty acids (isobutyric, isovaleric, valeric acid), while fermented foods increased alpha diversity. Fermented foods further reduced inflammatory markers, while fibre inflammatory effects were person-dependent. Unexpectedly, no differences were observed on self-reported perceived stress, wellbeing, fatigue, or cognition(Reference Wastyk, Fragiadakis and Perelman15), but this could be due to a reliance on self-report measures of cognitive performance and the small sample size that obscured effects.

The results from Wastyk et al. suggested potential additive effects from combining fermented and fibrous foods on the microbiota-gut-brain axis(Reference Wastyk, Fragiadakis and Perelman15). When combined with fermented foods, fibre can enhance the colonisation, survival and function of the microbes within fermented foods and from host microbes(Reference Preidis and Versalovic185). Recently, we have shown in our own lab that a combined diet rich in fibre and fermented foods from a variety of food sources for four weeks improved perceived stress in a dose-dependent manner in healthy adults compared to a healthy diet in line with Irish dietary guidelines(Reference Berding, Bastiaanssen and Moloney8). However, the combined diet had only subtle effects on gut microbiota composition and local gastrointestinal functional outputs, but this could be due the short duration of the intervention. Similarly, an inulin-enriched fermented yogurt decreased self-report scores of anxiety and depression in menopausal women compared to yogurt alone(Reference Shafie, Homayouni Rad and Mirghafourvand186). Future work should investigate whether consuming fermented and fibrous foods within the same meal elicits additive benefits, both acutely and chronically. The collective data provides support for the use of psychobiotics(Reference Sarkar, Lehto and Harty187), as a combination of pre-, pro-, post-, and/or synbiotics, to treat disorders related to dysfunction in cognitive and/or emotional processes. Psychobiotics have already demonstrated efficacy in attenuating depressive symptoms(Reference Zhang, Chen and Zhang188) and improving cognition in healthy populations(Reference Nieto-Ruiz, Garcia-Santos and Verdjo-Roman189) and populations with cognitive impairments(Reference Louzada and Ribeiro190), but more research with whole-food interventions in healthy and clinical populations is needed.

Conclusion

Taken together, both fibre and fermented foods exert protective effects on cognitive and emotional processes. Although only one study has investigated the differential mechanisms of fibre and fermented foods on the microbiota-gut-brain axis(Reference Wastyk, Fragiadakis and Perelman15), there is a growing interest in understanding the role of the gut microbiota underlying dietary patterns that have been shown to alter central nervous system processes(Reference Link, Subramanian and Cheung191). Such advancements are essential for development of future therapeutics to treat brain dysfunction.

In considering future therapeutics, the challenge in consuming high-fibre foods is reflected by reports of inadequate consumption of fibre in nearly all Western societies(Reference Burley, Champ and Cloran192,Reference Clemens, Kranz and Mobley193) . For some, fermented foods could be an alternative solution for individuals who struggle to consume dietary fibre due to issues with taste and/or tolerance, for example. It is worth highlighting the encouraging work currently underway by Jeff Gordon and colleagues. In this pioneering work, Gordon et al. aims to treat cognitive dysfunction in children brought upon by malnutrition to the gut microbiota in early life by fibrous food formulations that are culturally appropriate, environmentally sustainable, and affordable to produce(Reference Goyal, Venkatesh and Milbrandt194Reference Gehrig, Venkatesh and Chang196). This work suggests that the inexpensive and ubiquitous nature of fibre and fermented foods is a viable means of targeting brain health, at least partially via the gut microbiota, to ensure optimal functioning across all stages of life.

Acknowledgements

None.

Financial support

This work was supported by a Science Foundation Ireland (SFI) grant to APC Microbiome Ireland through the Irish Government’s National Development Plan [grant SFI/12/RC/2273_P2]. ES is funded by an Irish Research Council Postdoctoral Fellowship [GOIPD/2024/273]. ES and AF are supported by a private philanthropy donation awarded to JFC. The funding sources had no involvement in the decision to submit this manuscript for publication.

Author contributions

Authors ES and JFC were responsible for the design, and ES drafted the article. Authors ES, RB, AF, PDC, GC and JFC assisted in the interpretation and revision of the content.

Competing interests

Author ES has received honorarium from Janssen Sciences Ireland UC. PC is a co-founder and is the CTO of SeqBiome Ltd. and also serves as the Field Chief Editor for Frontiers in Microbiology. He has also been occasionally paid, or received hospitality, to deliver talks on his research. Research in the Cotter laboratory has been funded by Danone, PepsiCo, Friesland Campina and the PrecisionBiotics Group. GC has received honoraria from Janssen, Probi and Apsen as an invited speaker, is in receipt of research funding from Pharmavite, Reckitt, Tate and Lyle, Nestle and Fonterra, and has received payments as a consultant from Yakult, Zentiva and Heel Pharmaceuticals. JFC has received research funding from 4D Pharma, Cremo, Dupont, Mead Johnson, Nutricia and Pharmavite; has been an invited speaker at meetings organised by Alimentary Health, Alkermes, Ordesa, and Yakult; and has served as a consultant for Alkermes and Nestle.

References

Marx, W, Lane, MM, Hockey, M et al. (2021) Diet and depression: exploring the biological mechanisms of action. Mol Psychiatry 26, 134150.CrossRefGoogle ScholarPubMed
Muth, A-K & Park, SQ (2021) The impact of dietary macronutrient intake on cognitive function and the brain. Clin Nutr 40, 39994010.CrossRefGoogle ScholarPubMed
Medawar, E, Huhn, S, Villringer, A et al. (2019) The effects of plant-based diets on the body and the brain: a systematic review. Transl Psychiatry 9, 226.CrossRefGoogle ScholarPubMed
Rothschild, D, Weissbrod, O, Barkan, E et al. (2018) Environment dominates over host genetics in shaping human gut microbiota. Nat 555, 210215.CrossRefGoogle ScholarPubMed
Johnson, AJ, Vangay, P, Al-Ghalith, GA et al. (2019) Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host Microbe 25, 789802.e5.CrossRefGoogle ScholarPubMed
Long-Smith, C, O’Riordan, KJ, Clarke, G et al. (2020) Microbiota-gut-brain axis: new therapeutic opportunities. Annu Rev Pharmacol Toxicol 60, 477502.CrossRefGoogle ScholarPubMed
Berding, K, Vlckova, K, Marx, W et al. (2021) Diet and the microbiota–gut–brain axis: sowing the seeds of good mental health. Adv Nutr 12, 12391285.CrossRefGoogle ScholarPubMed
Berding, K, Bastiaanssen, TFS, Moloney, GM et al. (2022) Feed your microbes to deal with stress: a psychobiotic diet impacts microbial stability and perceived stress in a healthy adult population. Mol Psychiatry 28, 601610.CrossRefGoogle Scholar
Stiemsma, LT, Nakamura, RE, Nguyen, JG et al. (2020) Does consumption of fermented foods modify the human gut microbiota? J Nutr 150, 16801692.CrossRefGoogle ScholarPubMed
Sonnenburg, JL & Bäckhed, F (2016) Diet–microbiota interactions as moderators of human metabolism. Nat 535, 5664.CrossRefGoogle ScholarPubMed
O’Riordan, KJ, Collins, MK, Moloney, GM et al. (2022) Short chain fatty acids: microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol 546, 111572.CrossRefGoogle ScholarPubMed
Dalile, B, Van Oudenhove, L, Vervliet, B et al. (2019) The role of short-chain fatty acids in microbiota–gut–brain communication. Nat Rev Gastroenterol Hepatol 16, 461478.CrossRefGoogle ScholarPubMed
Lang, JM, Eisen, JA & Zivkovic, AM (2014) The microbes we eat: abundance and taxonomy of microbes consumed in a day’s worth of meals for three diet types. PeerJ 2, e659.CrossRefGoogle Scholar
Le Roy, CI, Kurilshikov, A, Leeming, E et al. (2022) Yoghurt consumption is associated with changes in the composition of the human gut microbiome and metabolome. BMC Microbiol 22, 39.CrossRefGoogle ScholarPubMed
Wastyk, HC, Fragiadakis, GK, Perelman, D et al. (2021) Gut-microbiota-targeted diets modulate human immune status. Cell 184, 41374153.e14.CrossRefGoogle ScholarPubMed
Aslam, H, Green, J, Jacka, F et al. (2020) Fermented foods, the gut and mental health: a mechanistic overview with implications for depression and anxiety. Nutr Neurosci 23, 659671.CrossRefGoogle ScholarPubMed
Diez-Ozaeta, I & Astiazaran, OJ (2022) Fermented foods: an update on evidence-based health benefits and future perspectives. Food Res Int 156, 111133.CrossRefGoogle ScholarPubMed
Aslam, H, Lotfaliany, M, So, D et al. (2023) Fiber intake and fiber intervention in depression and anxiety: a systematic review and meta-analysis of observational studies and randomized controlled trials. Nutr Rev 0, nuad143.CrossRefGoogle Scholar
Berding, K, Carbia, C & Cryan, JF (2021) Going with the grain: fiber, cognition, and the microbiota-gut-brain-axis. Exp Biol Med (Maywood) 246, 796811.CrossRefGoogle ScholarPubMed
Balasubramanian, R, Schneider, E, Gunnigle, E, Cotter, PD et al. (2024) Fermented foods: harnessing their potential to modulate the microbiota-gut-brain axis for mental health. Neurosci Biobehav Rev 158, 105562.CrossRefGoogle ScholarPubMed
Marx, W, Scholey, A, Firth, J et al. (2020) Prebiotics, probiotics, fermented foods and cognitive outcomes: a meta-analysis of randomized controlled trials. Neurosci Biobehav Rev 118, 472484.CrossRefGoogle ScholarPubMed
Alimentarius, C (2010) Guidelines on Nutrition Labelling CAC/GL 2–1985 as Last Amended 2010. Rome: FAO.Google Scholar
Gibson, GR, Hutkins, R, Sanders, ME et al. (2017) Expert consensus document: the international scientific association for probiotics and prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat Rev Gastroenterol Hepatol 14, 491502.CrossRefGoogle ScholarPubMed
Alves-Santos, AM, Sugizaki, CSA, Lima, GC, et al. (2020) Prebiotic effect of dietary polyphenols: a systematic review. J Funct Foods 74, 104169.CrossRefGoogle Scholar
Sonnenburg, ED & Sonnenburg, JL (2014) Starving our microbial self: the deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab 20, 779786.CrossRefGoogle ScholarPubMed
So, D, Whelan, K, Rossi, M, Morrison, M et al. (2018) Dietary fiber intervention on gut microbiota composition in healthy adults: a systematic review and meta-analysis. Am J Clin Nutr 107, 965983.CrossRefGoogle ScholarPubMed
Nyman, M, Asp, NG, Cummings, J et al. (1986) Fermentation of dietary fibre in the intestinal tract: comparison between man and rat. Br J Nutr 55, 487496.CrossRefGoogle Scholar
Wisker, E, Daniel, M, Rave, G et al. (1998) Fermentation of non-starch polysaccharides in mixed diets and single fibre sources: comparative studies in human subjects and in vitro. Br J Nutr 80, 253261.CrossRefGoogle ScholarPubMed
Procházková, N, Falony, G, Dragsted, LO et al. (2023) Advancing human gut microbiota research by considering gut transit time. Gut 72, 180191.CrossRefGoogle ScholarPubMed
Mardinoglu, A, Wu, H, Bjornson, E et al. (2018) An integrated understanding of the rapid metabolic benefits of a carbohydrate-restricted diet on hepatic steatosis in humans. Cell Metab 27, 559571.e5.CrossRefGoogle ScholarPubMed
Vangay, P, Johnson, AJ, Ward, TL et al. (2018) US immigration westernizes the human gut microbiome. Cell 175, 962972.e10.CrossRefGoogle ScholarPubMed
De Filippo, C, Cavalieri, D, Di Paola, M et al. (2010) Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci 107, 1469114696.CrossRefGoogle ScholarPubMed
Schnorr, SL, Candela, M, Rampelli, S et al. (2014) Gut microbiome of the Hadza hunter-gatherers. Nat Commun 5, 3654.CrossRefGoogle ScholarPubMed
Martínez, I, Stegen, JC, Maldonado-Gómez, MX, et al. (2015) The gut microbiota of rural papua new guineans: composition, diversity patterns, and ecological processes. Cell Rep 11, 527538.CrossRefGoogle ScholarPubMed
Marco, ML, Sanders, ME, Ganzle, M et al. (2021) The international scientific association for probiotics and prebiotics (ISAPP) consensus statement on fermented foods. Nat Rev Gastroenterol Hepatol 18, 196208.CrossRefGoogle ScholarPubMed
Marco, ML, Heeney, D, Binda, S et al. (2017) Health benefits of fermented foods: microbiota and beyond. Curr Opin Biotechnol 44, 94102.CrossRefGoogle ScholarPubMed
Hill, C, Guarner, F, Reid, G et al. (2014) The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 11, 506514.CrossRefGoogle ScholarPubMed
Mukherjee, A, Breselge, S, Dimidi, E et al. (2023) Fermented foods and gastrointestinal health: underlying mechanisms. Nat Rev Gastroenterol Hepatol 21, 248266.CrossRefGoogle ScholarPubMed
Valentino, V, Magliulo, R, Farsi, D et al. (2024) Fermented foods, their microbiome and its potential in boosting human health. Microb Biotechnol 17, e14428.CrossRefGoogle ScholarPubMed
Noh, H, Jang, H-H, Kim, G et al. (2021) Taxonomic composition and diversity of the gut microbiota in relation to habitual dietary intake in Korean adults. Nutrients 13, 366.CrossRefGoogle ScholarPubMed
Taylor, BC, Lejzerowicz, F, Poirel, M et al. (2020) Consumption of fermented foods is associated with systematic differences in the gut microbiome and metabolome. mSystems 5, e0090119.Google ScholarPubMed
Alvarez, A-S, Tap, J, Chambaud, I et al. (2020) Safety and functional enrichment of gut microbiome in healthy subjects consuming a multi-strain fermented milk product: a randomised controlled trial. Sci Rep 10, 15974.CrossRefGoogle ScholarPubMed
Walsh, LH, Walsh, A, Garcia-Perez, I et al. (2023) Comparison of the relative impacts of acute consumption of an inulin-enriched diet, milk kefir or a commercial probiotic product on the human gut microbiome and metabolome. NPJ Sci Food 7, 41.CrossRefGoogle ScholarPubMed
Ortega, RM, Requejo, AM, Andrés, P et al. (1997) Dietary intake and cognitive function in a group of elderly people. Am J Clin Nutr 66, 803809.CrossRefGoogle Scholar
Vercambre, M-N, Boutron-Ruault, M-C, Ritchie, K et al. (2009) Long-term association of food and nutrient intakes with cognitive and functional decline: a 13-year follow-up study of elderly French women. Br J Nutr 102, 419427.CrossRefGoogle ScholarPubMed
Aparicio Vizuete, A, Robles, F, Rodríguez-Rodríguez, E et al. (2010) Association between food and nutrient intakes and cognitive capacity in a group of institutionalized elderly people. Eur J Nutr 49, 293300.CrossRefGoogle Scholar
Milte, CM, Ball, K, Crawford, D et al. (2019) Diet quality and cognitive function in mid-aged and older men and women. BMC Geriatr 19, 361.CrossRefGoogle ScholarPubMed
Prokopidis, K, Giannos, P, Ispoglou, T et al. (2022) Dietary fiber intake is associated with cognitive function in older adults: data from the national health and nutrition examination survey. Am J Med 135, e257e262.CrossRefGoogle ScholarPubMed
Fortune, NC, Harville, EW, Guralnik, JM et al. (2019) Dietary intake and cognitive function: evidence from the Bogalusa heart study. Am J Clin Nutr 109, 16561663.CrossRefGoogle ScholarPubMed
Khan, NA, Raine, LB, Drollette, ES et al. (2015) Dietary fiber is positively associated with cognitive control among prepubertal children. J Nutr 145, 143149.CrossRefGoogle ScholarPubMed
Naveed, S, Venäläinen, T, Eloranta, A-M, Erkkilä, AT et al. (2020) Associations of dietary carbohydrate and fatty acid intakes with cognition among children. Public Health Nutr 23, 16571663.CrossRefGoogle ScholarPubMed
Mao, X, Chen, C, Pengcheng, X et al. (2019) Intake of vegetables and fruits through young adulthood is associated with better cognitive function in midlife in the US general population. J Nutr 149, 14241433.CrossRefGoogle ScholarPubMed
Chung, Y-C, Park, C-H, Kwon, H-K et al. (2012) Improved cognitive performance following supplementation with a mixed-grain diet in high school students: a randomized controlled trial. Nutr 28, 165172.CrossRefGoogle ScholarPubMed
Zhu, L, Ming, Y, Wu, M et al. (2023) Effect of fiber-rich diet and rope skipping on memory, executive function, and gut microbiota in young adults: a randomized controlled trial. Mol Nutr Food Res 68, e2300673.CrossRefGoogle ScholarPubMed
Nilsson, A, Salo, I, Plaza, M et al. (2017) Effects of a mixed berry beverage on cognitive functions and cardiometabolic risk markers; a randomized cross-over study in healthy older adults. PLoS One 12, e0188173.CrossRefGoogle ScholarPubMed
Sandberg, JC, Björck, IME & 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. Nutr J 17, 102.CrossRefGoogle ScholarPubMed
Mustra Rakic, J, Tanprasertsuk, J, Scott, TM et al. (2022) Effects of daily almond consumption for six months on cognitive measures in healthy middle-aged to older adults: a randomized control trial. Nutr Neurosci 25, 14661476.CrossRefGoogle ScholarPubMed
Ashley, S, Bradburn, S & Murgatroyd, C (2021) A meta-analysis of peripheral tocopherol levels in age-related cognitive decline and Alzheimer’s disease. Nutr Neurosci 24, 795809.CrossRefGoogle ScholarPubMed
Dorey, CK, Gierhart, D, Fitch, KA et al. (2023) Low xanthophylls, retinol, lycopene, and tocopherols in grey and white matter of brains with Alzheimer’s disease. J Alzheimers Dis 94, 117.CrossRefGoogle ScholarPubMed
Caruso, G, Torrisi, SA, Mogavero, MP et al. (2022) Polyphenols and neuroprotection: therapeutic implications for cognitive decline. Pharmacol Ther 232, 108013.CrossRefGoogle ScholarPubMed
Alex, A, Abbott, KA, McEvoy, M et al. (2020) Long-chain omega-3 polyunsaturated fatty acids and cognitive decline in non-demented adults: a systematic review and meta-analysis. Nutr Rev 78, 563578.CrossRefGoogle ScholarPubMed
Ross, FC, Mayer, DE, Horn, J et al. (2024) Potential of dietary polyphenols for protection from age-related decline and neurodegeneration: a role for gut microbiota? Nutr Neurosci 27, 10581076.CrossRefGoogle ScholarPubMed
Hwang, Y-H, Park, S, Paik, J-W et al. (2019) Efficacy and safety of lactobacillus plantarum C29-fermented soybean (DW2009) in individuals with mild cognitive impairment: a 12-week, multi-center, randomized, double-blind, placebo-controlled clinical trial. Nutrients 11, 305.CrossRefGoogle ScholarPubMed
Reid, SNS, Ryu, J-K, Kim, Y et al. (2018) The effects of fermented laminaria Japonica on short-term working memory and physical fitness in the elderly. Evid Based Complement Alternat Med 2018, 8109621.CrossRefGoogle ScholarPubMed
Handajani, YS, Turana, Y, Yogiara, Y et al. (2020) Tempeh consumption and cognitive improvement in mild cognitive impairment. Dement Geriatr Cogn Disord 49, 497502.CrossRefGoogle ScholarPubMed
Chung, Y-C, Jin, H-M, Cui, Y et al. (2014) Fermented milk of Lactobacillus helveticus IDCC3801 improves cognitive functioning during cognitive fatigue tests in healthy older adults. J Funct Foods 10, 465474.CrossRefGoogle Scholar
Beauchet, O, Launay, CP, Galery, K et al. (2019) Effects of vitamin D and calcium fortified yogurts on gait, cognitive performances, and serum 25-hydroxyvitamin D concentrations in older community-dwelling females: results from the GAit, MEmory, dietary and vitamin D (GAME-D2) randomized controlled tria. Nutrients 11, 2880.CrossRefGoogle Scholar
Ohsawa, K, Nakamura, F, Uchida, N et al. (2018) Lactobacillus helveticus-fermented milk containing lactononadecapeptide (NIPPLTQTPVVVPPFLQPE) improves cognitive function in healthy middle-aged adults: a randomised, double-blind, placebo-controlled trial. Int J Food Sci Nutr 69, 369376.CrossRefGoogle ScholarPubMed
Hogervorst, E, Sadjimim, T, Yesufu, A et al. (2008) High tofu intake is associated with worse memory in elderly indonesian men and women. Dement Geriatr Cogn Disord 26, 5057.CrossRefGoogle ScholarPubMed
Tessier, AJ, Presse, N, Rahme, E et al. (2021) Milk, yogurt, and cheese intake is positively associated with cognitive executive functions in older adults of the Canadian longitudinal study on aging. J Gerontol A Biol Sci Med Sci 76, 22232231.CrossRefGoogle ScholarPubMed
Suzuki, T, Kojima, N, Osuka, Y et al. (2019) The effects of mold-fermented cheese on brain-derived neurotrophic factor in community-dwelling older Japanese women with mild cognitive impairment: a randomized, controlled, crossover trial. J Am Med Dir Assoc 20, 15091514.e2.CrossRefGoogle ScholarPubMed
Tillisch, K, Labus, J, Kilpatrick, L et al. (2013) Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 144, 13941401.e4.CrossRefGoogle ScholarPubMed
Cannavale, CN, Mysonhimer, AR, Bailey, MA et al. (2023) Consumption of a fermented dairy beverage improves hippocampal-dependent relational memory in a randomized, controlled cross-over trial. Nutr Neurosci 26, 265274.CrossRefGoogle Scholar
Pearl, J (2014) Interpretation and identification of causal mediation. Psychol Methods 19, 459481.CrossRefGoogle ScholarPubMed
Klein, AB, Williamson, R, Santini, MA et al. (2011) Blood BDNF concentrations reflect brain-tissue BDNF levels across species. Int J Neuropsychopharmacol 14, 347353.CrossRefGoogle ScholarPubMed
Ribeiro, G, Ferri, A, Clarke, G et al. (2022) Diet and the microbiota - gut - brain-axis: a primer for clinical nutrition. Curr Opin Clin Nutr Metab Care 25, 443450.CrossRefGoogle ScholarPubMed
Kim, C-S, Byeon, S & Shin, D-M (2020) Sources of dietary fiber are differently associated with prevalence of depression. Nutrients 12, 2813.CrossRefGoogle ScholarPubMed
Li, D, Tong, Y & Li, Y (2020) Dietary fiber is inversely associated with depressive symptoms in premenopausal women. Front Neurosci 14, 373.CrossRefGoogle ScholarPubMed
Saghafian, F, Sharif, N, Saneei, P et al. (2021) Consumption of dietary fiber in relation to psychological disorders in adults. Front Psychiatry 12, 587468.CrossRefGoogle ScholarPubMed
Burlutksy, G, Flood, VM, Gopinath, B et al. (2016) Association between carbohydrate nutrition and prevalence of depressive symptoms in older adults. Br J Nutr 116, 21092114.Google Scholar
Jaatinen, N, Korpela, R, Poussa, T et al. (2014) Effects of daily intake of yoghurt enriched with bioactive components on chronic stress responses: a double-blinded randomized controlled trial. Int J Food Sci Nutr 65, 507514.CrossRefGoogle ScholarPubMed
Freijy, TM, Cribb, L, Oliver, G et al. (2022) Effects of a high-prebiotic diet versus probiotic supplements versus synbiotics on adult mental health: the ‘gut feelings’ randomised controlled trial. Front Neurosci 16, 1097278.CrossRefGoogle ScholarPubMed
Hilimire, MR, DeVylder, JE & Forestell, CA (2015) Fermented foods, neuroticism, and social anxiety: an interaction model. Psychiatry Res 228, 203208.CrossRefGoogle ScholarPubMed
Karbownik, MS, Mokros, Ł & Kowalczyk, E (2022) Who benefits from fermented food consumption? A comparative analysis between psychiatrically ill and psychiatrically healthy medical students. Int J Environ Res Public Health 19, 3861.CrossRefGoogle ScholarPubMed
Baars, T, Berge, AC, Garssen, J et al. (2019) Effect of raw milk consumption on perceived health, mood and immune functioning among US adults with a poor and normal health: a retrospective questionnaire based study. Complement Ther Med 47, 102196.CrossRefGoogle Scholar
Pražnikar, ZJ, Kenig, S, Vardjan, T et al. (2020) Effects of kefir or milk supplementation on zonulin in overweight subjects. J Dairy Sci 103, 39613970.CrossRefGoogle ScholarPubMed
Kuroda, M, Ishizaki, T, Maruyama, T et al. (2007) Effect of dried-bonito broth on mental fatigue and mental task performance in subjects with a high fatigue score. Physiol Behav 92, 957962.CrossRefGoogle ScholarPubMed
Nozawa, Y, Ishizaki, T, Kuroda, M et al. (2008) Effect of dried-bonito broth intake on peripheral blood flow, mood, and oxidative stress marker in humans. Physiol Behav 93, 267273.CrossRefGoogle ScholarPubMed
Pedrelli, P, Shapero, B, Archibald, A, et al. (2016) Alcohol use and depression during adolescence and young adulthood: a summary and interpretation of mixed findings. Curr Addict Rep 3, 9197.CrossRefGoogle Scholar
Black, JJ & Rofey, DL (2018) An overview of common psychiatric problems among adolescent and young adult females: focus on mood and anxiety. Best Pract Res Clin Obstet Gynaecol 48, 165173.CrossRefGoogle Scholar
Ribera, C, Sánchez-Ortí, JV, Clarke, G et al. (2024) Probiotic, prebiotic, synbiotic and fermented food supplementation in psychiatric disorders: a systematic review of clinical trials. Neurosci Biobehav Rev 158, 105561.CrossRefGoogle ScholarPubMed
Cryan, JF, O’Riordan, KJ, Cowan, CSM et al. (2019) The microbiota-gut-brain axis. Physiol Rev 99, 18772013.CrossRefGoogle ScholarPubMed
Ratsika, A, Cruz Pereira, JS, Lynch, CMK et al. (2023) Microbiota-immune-brain interactions: a lifespan perspective. Curr Opin Neurobiol 78, 102652.CrossRefGoogle ScholarPubMed
Bostick, JW, Schonhoff, AM & Mazmanian, SK (2022) Gut microbiome-mediated regulation of neuroinflammation. Curr Opin Immunol 76, 102177.CrossRefGoogle ScholarPubMed
Macpherson, AJ, Pachnis, V & Prinz, M (2023) Boundaries and integration between microbiota, the nervous system, and immunity. Immun 56, 17121726.CrossRefGoogle ScholarPubMed
Siracusa, F, Schaltenberg, N, Kumar, Y et al. (2023) Short-term dietary changes can result in mucosal and systemic immune depression. Nat Immunol 24, 14731486.CrossRefGoogle ScholarPubMed
Cotillard, A, Kennedy, SP, Kong, LC et al. (2013) Dietary intervention impact on gut microbial gene richness. Nat 500, 585588.CrossRefGoogle ScholarPubMed
Sandby, K, Magkos, F, Chabanova, E et al. (2024) The effect of dairy products on liver fat and metabolic risk markers in males with abdominal obesity – a four-arm randomized controlled trial. Clin Nutr 43, 534542.CrossRefGoogle ScholarPubMed
SaeidiFard, N, Djafarian, K & Shab-Bidar, S (2020) Fermented foods and inflammation: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN 35, 3039.CrossRefGoogle ScholarPubMed
Leigh, S-J, Uhlig, F, Wilmes, L et al. (2023) The impact of acute and chronic stress on gastrointestinal physiology and function: a microbiota–gut–brain axis perspective. J Physiol 601, 44914538.CrossRefGoogle ScholarPubMed
Foster, JA, Rinaman, L & Cryan, JF (2017) Stress & the gut-brain axis: regulation by the microbiome. Neurobiol Stress 7, 124136.CrossRefGoogle ScholarPubMed
Nagpal, J & Cryan, JF (2021) Microbiota-brain interactions: moving toward mechanisms in model organisms. Neuron 109, 39303953.CrossRefGoogle ScholarPubMed
Sudo, N, Chida, Y, Aiba, Y et al. (2004) Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol 558, 263275.CrossRefGoogle ScholarPubMed
Crumeyrolle-Arias, M, Jaglin, M, Bruneau, A et al. (2014) Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 42, 207217.CrossRefGoogle ScholarPubMed
Clarke, G, Grenham, S, Scully, P et al. (2013) The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 18, 666673.CrossRefGoogle Scholar
Lyte, JM, Gheorghe, CE, Goodson, MS et al. (2020) Gut-brain axis serotonergic responses to acute stress exposure are microbiome-dependent. Neurogastroenterology Motil 32, e13881.CrossRefGoogle ScholarPubMed
Eutamene, H, Lamine, F, Chabo, C, Theodorou, V et al. (2007) Synergy between lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr 137, 19011907.CrossRefGoogle ScholarPubMed
Desbonnet, L, Garrett, L, Clarke, G et al. (2010) Effects of the probiotic bifidobacterium infantis in the maternal separation model of depression. Neurosci 170, 11791188.CrossRefGoogle ScholarPubMed
Allen, AP, Kennedy, PJ, Cryan, JF et al. (2014) Biological and psychological markers of stress in humans: focus on the trier social stress test. Neurosci Biobehav Rev 38, 94124.CrossRefGoogle ScholarPubMed
Takada, M, Nishida, K, Kataoka-Kato, A et al. (2016) Probiotic lactobacillus casei strain Shirota relieves stress-associated symptoms by modulating the gut-brain interaction in human and animal models. Neurogastroenterology Motil 28, 10271036.CrossRefGoogle ScholarPubMed
Kato-Kataoka, A, Nishida, K, Takada, M et al. (2016) Fermented milk containing lactobacillus casei strain Shirota prevents the onset of physical symptoms in medical students under academic examination stress. Benef Microbes 7, 153156.CrossRefGoogle ScholarPubMed
Marcos, A, Wärnberg, J, Nova, E et al. (2004) The effect of milk fermented by yogurt cultures plus lactobacillus casei DN-114001 on the immune response of subjects under academic examination stress. Eur J Nutr 43, 381389.CrossRefGoogle ScholarPubMed
Brett, BE, Koko, BK, Doumbia, HOY et al. (2021) Salivary biomarkers of stress and inflammation in first graders in Côte d’Ivoire: effects of a probiotic food intervention. Psychoneuroendocrinology 129, 105255.CrossRefGoogle ScholarPubMed
Wen, CKF, Chou, C-P, Belcher, BR et al. (2021) The acute relationship between affective states and stress biomarkers in ethnic minority youths. Int J Environ Res Public Health 18, 12670.CrossRefGoogle ScholarPubMed
Soltani, H, Keim, NL & Laugero, KD (2019) Increasing dietary carbohydrate as part of a healthy whole food diet intervention dampens eight week changes in salivary cortisol and cortisol responsiveness. Nutrients 11, 2563.CrossRefGoogle ScholarPubMed
Michels, N, Sioen, I, Braet, C, Huybrechts, I et al. (2013) Relation between salivary cortisol as stress biomarker and dietary pattern in children. Psychoneuroendocrinology 38, 15121520.CrossRefGoogle ScholarPubMed
Pearlmutter, P, DeRose, G, Samson, C et al. (2020) Sweat and saliva cortisol response to stress and nutrition factors. Sci Rep 10, 19050.CrossRefGoogle ScholarPubMed
Fülling, C, Dinan, TG & Cryan, JF (2019) Gut microbe to brain signaling: what happens in Vagus…. Neuron 101, 9981002.CrossRefGoogle ScholarPubMed
Sgritta, M, Dooling, SW, Buffington, SA et al. (2019) Mechanisms underlying microbial-mediated changes in social behavior in mouse models of autism spectrum disorder. Neuron 101, 246259.e6.CrossRefGoogle ScholarPubMed
Bravo, JA, Forsythe, P, Chew, M V et al. (2011) Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci 108, 1605016055.CrossRefGoogle Scholar
Bercik, P, Park, AJ, Sinclair, D et al. (2011) The anxiolytic effect of bifidobacterium longum NCC3001 involves vagal pathways for gut–brain communication. Neurogastroenterology Motil 23, 11321139.CrossRefGoogle ScholarPubMed
Bruning, J, Chapp, A, Kaurala, GA et al. (2020) Gut microbiota and short chain fatty acids: influence on the autonomic nervous system. Neurosci Bull 36, 9195.CrossRefGoogle ScholarPubMed
Goswami, C, Iwasaki, Y & Yada, T (2018) Short-chain fatty acids suppress food intake by activating vagal afferent neurons. J Nutr Biochem 57, 130135.CrossRefGoogle ScholarPubMed
Loper, H, Leinen, M, Bassoff, L et al. (2021) Both high fat and high carbohydrate diets impair vagus nerve signaling of satiety. Sci Rep 11, 10394.CrossRefGoogle ScholarPubMed
Klingbeil, EA, Cawthon, C, Kirkland, R et al. (2019) Potato-resistant starch supplementation improves microbiota dysbiosis, inflammation, and gut–brain signaling in high fat-fed rats. Nutrients 11, 2710.CrossRefGoogle ScholarPubMed
Silva, YP, Bernardi, A & Frozza, RL (2020) The role of short-chain fatty acids from gut microbiota in gut-brain communication. Front Endocrinol (Lausanne) 11, 25.CrossRefGoogle ScholarPubMed
Khoshbin, K & Camilleri, M (2020) Effects of dietary components on intestinal permeability in health and disease. Am J Physiol Gastrointest Liver Physiol 319, G589G608.CrossRefGoogle ScholarPubMed
Dalile, B, Fuchs, A, La Torre, D et al. (2024) Colonic butyrate administration modulates fear memory but not the acute stress response in men: a randomized, triple-blind, placebo-controlled trial. Prog Neuropsychopharmacol Biol Psychiatry 131, 110939.CrossRefGoogle ScholarPubMed
Dalile, B, Vervliet, B, Bergonzelli, G et al. (2020) Colon-delivered short-chain fatty acids attenuate the cortisol response to psychosocial stress in healthy men: a randomized, placebo-controlled trial. Neuropsychopharmacol 45, 22572266.CrossRefGoogle Scholar
González, S, Fernández-Navarro, T, Arboleya, S et al. (2019) Fermented dairy foods: impact on intestinal microbiota and health-linked biomarkers. Front Microbiol 10, 1046.CrossRefGoogle ScholarPubMed
McIntyre, RS, Powell, AM, Kaidanovich-Beilin, O et al. (2013) The neuroprotective effects of GLP-1: possible treatments for cognitive deficits in individuals with mood disorders. Behav Brain Res 237, 164171.CrossRefGoogle ScholarPubMed
Stadlbauer, U, Woods, SC, Langhans, W et al. (2015) PYY3-36: beyond food intake. Front Neuroendocrinol 38, 111.CrossRefGoogle ScholarPubMed
Wanders, AJ, van den Borne, JJGC, de Graaf, C et al. (2011) Effects of dietary fibre on subjective appetite, energy intake and body weight: a systematic review of randomized controlled trials. Obesity Rev 12, 724739.CrossRefGoogle ScholarPubMed
Henry, CJ, Leow, MKS, Lim, WX et al. (2016) Combination of soya protein and polydextrose reduces energy intake and glycaemic response via modulation of gastric emptying rate, ghrelin and glucagon-like peptide-1 in Chinese. Br J Nutr 115, 21302137.Google Scholar
Al-Mana, NM & Robertson, MD (2018) Acute effect of resistant starch on food intake, appetite and satiety in overweight/obese males. Nutrients 10, 1993.CrossRefGoogle ScholarPubMed
Zhao, L, Zheng, F, Ding, X et al. (2018) Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Sci (1979) 359, 11511156.Google ScholarPubMed
Simon, M-C, Strassburger, K, Nowotny, B et al. (2015) Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: a proof of concept. Diabetes Care 38, 18271834.CrossRefGoogle ScholarPubMed
Rowland, I, Gibson, G, Heinken, A et al. (2018) Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 57, 124.CrossRefGoogle ScholarPubMed
Mahmoudian Dehkordi, S, Arnold, M, Nho, K et al. (2019) Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease-an emerging role for gut microbiome. Alzheimers Dement 15, 7692.CrossRefGoogle ScholarPubMed
Varma, VR, Wang, Y, An, Y et al. (2021) Bile acid synthesis, modulation, and dementia: a metabolomic, transcriptomic, and pharmacoepidemiologic study. PLoS Med 18, e1003615.CrossRefGoogle ScholarPubMed
Theofilopoulos, S, Wang, Y, Kitambi, SS et al. (2013) Brain endogenous liver X receptor ligands selectively promote midbrain neurogenesis. Nat Chem Biol 9, 126133.CrossRefGoogle ScholarPubMed
Singh, J, Metrani, R, Shivanagoudra, SR et al. (2019) Review on bile acids: effects of the gut microbiome, interactions with dietary fiber, and alterations in the bioaccessibility of bioactive compounds. J Agric Food Chem 67, 91249138.CrossRefGoogle ScholarPubMed
Ginos, BNR, Navarro, SL, Schwarz, Y et al. (2018) Circulating bile acids in healthy adults respond differently to a dietary pattern characterized by whole grains, legumes and fruits and vegetables compared to a diet high in refined grains and added sugars: a randomized, controlled, crossover feeding stud. Metab 83, 197204.CrossRefGoogle Scholar
Gao, J, Mao, K, Wang, X et al. (2021) Tibet kefir milk regulated metabolic changes induced by high-fat diet via amino acids, bile acids, and equol metabolism in human-microbiota-associated rats. J Agric Food Chem 69, 67206732.CrossRefGoogle ScholarPubMed
Johnson, MH & de Mejia, EG (2016) Phenolic compounds from fermented berry beverages modulated gene and protein expression to increase insulin secretion from pancreatic β-cells in vitro. J Agric Food Chem 64, 25692581.CrossRefGoogle ScholarPubMed
Filannino, P, Bai, Y, Di Cagno, R et al. (2015) Metabolism of phenolic compounds by lactobacillus spp. during fermentation of cherry juice and broccoli puree. Food Microbiol 46, 272279.CrossRefGoogle ScholarPubMed
Makletsova, MG, Rikhireva, GT, Kirichenko, EY et al. (2022) The role of polyamines in the mechanisms of cognitive impairment. J Neurochem 16, 283294.CrossRefGoogle Scholar
Di Martino, ML, Campilongo, R, Casalino, M et al. (2013) Polyamines: emerging players in bacteria-host interactions. Int J Med Microbiol 303, 484491.CrossRefGoogle ScholarPubMed
Muñoz-Esparza, NC, Latorre-Moratalla, ML, Comas-Basté, O et al. (2019) Polyamines in food. Front Nutr 6, 108.CrossRefGoogle ScholarPubMed
Frolinger, T, Sims, S, Smith, C et al. (2019) The gut microbiota composition affects dietary polyphenols-mediated cognitive resilience in mice by modulating the bioavailability of phenolic acids. Sci Rep. 9, 3546.CrossRefGoogle ScholarPubMed
Gheorghe, CE, Martin, JA, Manriquez, FV et al. (2019) Focus on the essentials: tryptophan metabolism and the microbiome-gut-brain axis. Curr Opin Pharmacol 48, 137145.CrossRefGoogle ScholarPubMed
Purton, T, Staskova, L, Lane, MM et al. (2021) Prebiotic and probiotic supplementation and the tryptophan-kynurenine pathway: a systematic review and meta analysis. Neurosci Biobehav Rev 123, 113.CrossRefGoogle ScholarPubMed
Rothhammer, V, Mascanfroni, ID, Bunse, L et al. (2016) Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med 22, 586597.CrossRefGoogle ScholarPubMed
Appleton, J. (2018) The gut-brain axis: influence of microbiota on mood and mental health. Integr Med (Encinitas) 17, 2832.Google ScholarPubMed
O’Mahony, SM, Clarke, G, Borre, YE et al. (2015) Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res 277, 3248.CrossRefGoogle ScholarPubMed
Tuomainen, M, Lindström, J, Lehtonen, M et al. (2018) Associations of serum indolepropionic acid, a gut microbiota metabolite, with type 2 diabetes and low-grade inflammation in high-risk individuals. Nutr Diabetes 8, 35.CrossRefGoogle ScholarPubMed
Keski-Rahkonen, P, Kolehmainen, M, Lappi, J et al. (2019) Decreased plasma serotonin and other metabolite changes in healthy adults after consumption of wholegrain rye: an untargeted metabolomics study. Am J Clin Nutr 109, 16301639.CrossRefGoogle ScholarPubMed
Asano, Y, Hiramoto, T, Nishino, R et al. (2012) Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol 303, G128895.CrossRefGoogle ScholarPubMed
Kageyama, T, Nakamura, M, Yamasaki, Y et al. (2000) The 4F2hc/LAT1 complex transports l-DOPA across the blood–brain barrier. Brain Res 879, 115121.CrossRefGoogle ScholarPubMed
Alexander, R, Aragon, O, Bookwala, J et al. (2021) The neuroscience of positive emotions and affect: implications for cultivating happiness and wellbeing. Neurosci Biobehav Rev 121, 220249.CrossRefGoogle ScholarPubMed
Volkow, ND, Wang, GJ, Baler, RD. (2011) Reward, dopamine and the control of food intake: implications for obesity. Trends Cogn Sci 15, 3746.CrossRefGoogle ScholarPubMed
Koh, W, Kwak, H, Cheong, E et al. (2023) GABA tone regulation and its cognitive functions in the brain. Nat Rev Neurosci 24, 523539.CrossRefGoogle ScholarPubMed
Luscher, B, Maguire, JL, Rudolph, U et al. (2023) GABA(A) receptors as targets for treating affective and cognitive symptoms of depression. Trends Pharmacol Sci 44, 586600.CrossRefGoogle ScholarPubMed
Richard, HT & Foster, JW (2003) Acid resistance in escherichia coli. Adv Appl Microbiol 52, 167186.CrossRefGoogle ScholarPubMed
Siragusa, S, De Angelis, M, Di Cagno, R (2007) Synthesis of γ-aminobutyric acid by lactic acid bacteria isolated from a variety of Italian cheeses. Appl Environ Microbiol 73, 72837290.CrossRefGoogle ScholarPubMed
Li, H, Gao, D, Cao, Y, Xu, H. (2008) A high γ-aminobutyric acid-producingLactobacillus brevis isolated from Chinese traditionalpaocai. Ann Microbiol 58, 649653.CrossRefGoogle Scholar
Coda, R, Melama, L, Rizzello, CG et al. (2015) Effect of air classification and fermentation by lactobacillus plantarum VTT E-133328 on faba bean (Vicia faba L.) flour nutritional properties. Int J Food Microbiol 193, 3442.CrossRefGoogle ScholarPubMed
Boonstra, E, de Kleijn, R, Colzato, LS et al. (2015) Neurotransmitters as food supplements: the effects of GABA on brain and behavior. Front Psychol 6, 1520.CrossRefGoogle ScholarPubMed
Picciotto, MR, Higley, MJ & Mineur, YS (2012) Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron 76, 116129.CrossRefGoogle ScholarPubMed
Horiuchi, Y, Kimura, R, Kato, N et al. (2003) Evolutional study on acetylcholine expression. Life Sci 72, 17451756.CrossRefGoogle Scholar
Hayes, AMR, Tierno Lauer, L, Kao, AE et al. (2024) Western diet consumption impairs memory function via dysregulated hippocampus acetylcholine signaling. Brain Behav Immun 118, 408422.CrossRefGoogle ScholarPubMed
Muscaritoli, M (2021) The impact of nutrients on mental health and well-being: insights from the literature. Front Nutr 8, 656290.CrossRefGoogle ScholarPubMed
Pham, VT, Dold, S, Rehman, A et al. (2021) Vitamins, the gut microbiome and gastrointestinal health in humans. Nutr Res 95, 3553.CrossRefGoogle ScholarPubMed
Septembre-Malaterre, A, Remize, F & Poucheret, P (2018) Fruits and vegetables, as a source of nutritional compounds and phytochemicals: changes in bioactive compounds during lactic fermentation. Food Res Int 104, 8699.CrossRefGoogle ScholarPubMed
Gibson, RS, Raboy, V & King, JC (2018) Implications of phytate in plant-based foods for iron and zinc bioavailability, setting dietary requirements, and formulating programs and policies. Nutr Rev 76, 793804.CrossRefGoogle Scholar
Ahmed, MI, Xu, X, Sulieman, AA et al. (2020) The effect of fermentation time on in vitro bioavailability of iron, zinc, and calcium of kisra bread produced from koreeb (dactyloctenium aegyptium) seeds flour. Microchem J 154, 104644.CrossRefGoogle Scholar
Wadamori, Y, Vanhanen, L & Savage, GP (2014) Effect of kimchi fermentation on oxalate levels in silver beet (beta vulgaris var. cicla). Foods 3, 269278.CrossRefGoogle Scholar
Maintz, L & Novak, N (2007) Histamine and histamine intolerance Am J Clin Nutr 85, 11851196.CrossRefGoogle ScholarPubMed
Holmes, ZC, Villa, MM, Durand, HK et al. (2022) Microbiota responses to different prebiotics are conserved within individuals and associated with habitual fiber intake. Microbiome 10, 114.CrossRefGoogle ScholarPubMed
Petrone, BL, Aqeel, A, Jiang, S et al. (2023) Diversity of plant DNA in stool is linked to dietary quality, age, and household income. Proc Natl Acad Sci 120, e2304441120.CrossRefGoogle ScholarPubMed
Li, KJ, Burton-Pimentel, KJ, Brouwer-Brolsma, EM et al. (2023) Identifying plasma and urinary biomarkers of fermented food intake and their associations with cardiometabolic health in a Dutch observational cohort. J Agric Food Chem 71, 44264439.CrossRefGoogle Scholar
Leech, J, Cabrera-Rubio, R, Walsh, A et al. (2020) Fermented-food metagenomics reveals substrate-associated differences in taxonomy and health-associated and antibiotic resistance determinants. mSystems 5, e0052220.CrossRefGoogle ScholarPubMed
Li, KJ, Brouwer-Brolsma, EM, Burton, KJ et al. (2020) Prevalence of fermented foods in the Dutch adult diet and validation of a food frequency questionnaire for estimating their intake in the NQplus cohort. BMC Nutr 6, 114.CrossRefGoogle ScholarPubMed
Campbell, R, Hauptmann, A, Campbell, K et al. (2022) Better understanding of food and human microbiomes through collaborative research on inuit fermented foods. Microbiome Res Rep 1, 5.Google ScholarPubMed
Preidis, GA & Versalovic, J (2009) Targeting the human microbiome with antibiotics, probiotics, and prebiotics: gastroenterology enters the metagenomics era. Gastroenterology 136, 20152031.CrossRefGoogle ScholarPubMed
Shafie, M, Homayouni Rad, A & Mirghafourvand, M (2022) Effects of prebiotic-rich yogurt on menopausal symptoms and metabolic indices in menopausal women: a triple-blind randomised controlled trial. Int J Food Sci Nutr 73, 693704.CrossRefGoogle ScholarPubMed
Sarkar, A, Lehto, SM, Harty, S et al. (2016) Psychobiotics and the manipulation of bacteria-gut-brain signals. Trends Neurosci 39, 763781.CrossRefGoogle ScholarPubMed
Zhang, Q, Chen, B, Zhang, J et al. (2023) Effect of prebiotics, probiotics, synbiotics on depression: results from a meta-analysis. BMC Psychiatry 23, 477.CrossRefGoogle ScholarPubMed
Nieto-Ruiz, A, Garcia-Santos, JA, Verdjo-Roman, J et al. (2022) Infant formula supplemented with milk fat globule membrane, long-chain polyunsaturated fatty acids, and synbiotics is associated with neurocognitive function and brain structure of healthy children aged 6 years: the COGNIS study. Front Nutr 9, 820224.CrossRefGoogle ScholarPubMed
Louzada, ER & Ribeiro, SML (2020) Synbiotic supplementation, systemic inflammation, and symptoms of brain disorders in elders: a secondary study from a randomized clinical trial. Nutr Neurosci 23, 93100.CrossRefGoogle Scholar
Link, VM, Subramanian, P, Cheung, F et al. (2024) Differential peripheral immune signatures elicited by vegan versus ketogenic diets in humans. Nat Med 30, 560572.CrossRefGoogle ScholarPubMed
Burley, VJ, Champ, MM-J, Cloran, SJ et al. (2017) Dietary fibre in Europe: current state of knowledge on definitions, sources, recommendations, intakes and relationships to health. Nutr Res Rev 30, 149190.Google Scholar
Clemens, R, Kranz, S, Mobley, AR et al. (2012) Filling America’s fiber intake gap: summary of a roundtable to probe realistic solutions with a focus on grain-based foods. J Nutr 142, 1390S1401S.CrossRefGoogle ScholarPubMed
Goyal, MS, Venkatesh, S, Milbrandt, J et al. (2015) Feeding the brain and nurturing the mind: linking nutrition and the gut microbiota to brain development. Proc Natl Acad Sci 112, 1410514112.CrossRefGoogle ScholarPubMed
Chen, RY, Mostafa, I, Hibberd, M et al. (2021) A microbiota-directed food intervention for undernourished children. N Engl J Med 384, 15171528.CrossRefGoogle ScholarPubMed
Gehrig, JL, Venkatesh, S, Chang, H-W et al. (2019) Effects of microbiota-directed foods in gnotobiotic animals and undernourished children. Sci (1979) 365, eaau4732.Google ScholarPubMed
Figure 0

Fig. 1. Fermented foods are rich in microorganisms, bioactive peptides, phytochemicals, and peptides that can modulate brain function through enhancing microbial diversity leading to an enrichment of diverse microbial metabolites. Dietary fibre increases microbial richness, and neuroactive SCFAs are produced as a result of microbial fermentation of dietary fibre. Both fermented foods and fibre support intestinal and BBB integrity to prevent peripheral and central inflammation for optimal cognitive and emotional functioning. BBB: blood-brain barrier. GLP-1: glucagon-like peptide 1. IL: interleukin. PP: polyphenol. PYY: peptide YY. SCFA: short-chain fatty acid. Vit K, B: Vitamin K and B. Created with BioRender.com.

Figure 1

Fig. 2. Potential approaches to address challenges in nutrition studies for the microbiota-gut-brain axis. Created with BioRender.com.