1,874
Views
0
CrossRef citations to date
0
Altmetric
Review

Diet-microbiota associations in gastrointestinal research: a systematic review

ORCID Icon, , , , &
Article: 2350785 | Received 04 Dec 2023, Accepted 29 Apr 2024, Published online: 09 May 2024

ABSTRACT

Interactions between diet and gastrointestinal microbiota influence health status and outcomes. Evaluating these relationships requires accurate quantification of dietary variables relevant to microbial metabolism, however current dietary assessment methods focus on dietary components relevant to human digestion only. The aim of this study was to synthesize research on foods and nutrients that influence human gut microbiota and thereby identify knowledge gaps to inform dietary assessment advancements toward better understanding of diet–microbiota interactions. Thirty-eight systematic reviews and 106 primary studies reported on human diet-microbiota associations. Dietary factors altering colonic microbiota included dietary patterns, macronutrients, micronutrients, bioactive compounds, and food additives. Reported diet-microbiota associations were dominated by routinely analyzed nutrients, which are absorbed from the small intestine but analyzed for correlation to stool microbiota. Dietary derived microbiota-relevant nutrients are more challenging to quantify and underrepresented in included studies. This evidence synthesis highlights advancements needed, including opportunities for expansion of food composition databases to include microbiota-relevant data, particularly for human intervention studies. These advances in dietary assessment methodology will facilitate translation of microbiota-specific nutrition therapy to practice.

Introduction

The gastrointestinal (GI) microbiota is increasingly implicated in maintaining good overall health and is important in both preventing and managing GI and extraintestinal diseases.Citation1,Citation2 Dietary intake affects the colonization, abundance, diversity, and metabolic influences of the GI microbiota.Citation3 Methods used to assess and analyze human dietary intake focus on nutrients that are specific to human digestion, absorption and metabolism,Citation4 not to human GI microbial metabolism, which means the extent and nature of these influences are currently not well characterized.Citation3,Citation5

Knowledge of GI microbes increased exponentially when gene sequencing superseded microbial culture. Assessment of the abundance and diversity of microbial taxa has become more detailed with use of next-generation sequencing technologies such as 16S rRNA amplicon gene sequencing and metagenomic shotgun sequencing (MGS). With the reduction of cost for MGS, this is fast becoming the preferred approach to microbiota profiling as it captures the taxonomic composition in a non-biased manner and reports the microbial metabolic functional properties.Citation6,Citation7 These technological advances have revealed that gut-derived metabolites are intermediaries between the gut microbiota and host metabolism and that differential responses to diet between individuals may relate to the interplay between microbes, the food metabolome,Citation8,Citation9 and human host factors, including their genetic profile and gene expression.Citation10,Citation11

Improvements in sample collection methods have further enhanced specificity and sensitivity of microbial analysis, allowing for identification of microbial niches along the GI tract. For example, duodenal mucosal samples can now be extracted using aseptic techniques, facilitating accurate analysis of mucosa-associated microbiota in this important part of the digestive tract.Citation12 Despite these advances, mechanistic microbiome studies are challenging to conduct and prone to error due to the complexity of the gut microbiota, and intra- and inter-individual variability.Citation5

The vastly expanded horizon of understanding regarding gut microbiota afforded by gene sequencing is likely to revolutionize research into, and understanding of digestion, absorption, and metabolism of nutrients in food. Nutrients and compounds in foods that have low bioavailability could ‘feed’ the colonic microbiota. Additives that are generally recognized as safe for human health have not been adequately assessed in relation to microbiota interactions.Citation13

Consolidation of diet-microbiota data for meta-analysis or meaningful synthesis is limited by the variability in microbiological samples, sampling techniques, dietary intake data, study designs and outcome measures in studies conducted to date. Systematic reviews consistently report limitations to dietary assessment methods for analyzing microbiota-relevant diet,Citation14 foods and nutrients.Citation15–18 While some data reported in these reviews are from human studies, research in the microbiome field continues to be highly reliant on animal data,Citation19,Citation20 which may not be well distinguished from human data in some reviews.

To progress toward the goal of diet-focused microbiota manipulation for health status optimization, the specificity and sensitivity of dietary assessment and analysis methods used in diet-microbiome research needs to improve. Such progress is reliant on better differentiation and delineation of human from animal study findings, increased ability to discern correlation or association from causationCitation21 and increased scrutiny of what nutrients are accessible to the microbiota in respective parts of the GI tract. As the purported effects of microbiota on human metabolism relate to the metabolites produced by those microorganisms, there also needs to be mechanisms for dietary, microbiota, and metabolomic data to be triangulated in analysis.

Therefore, the aim of this systematic review was to determine which foods, food components and whole foods consumed by humans, influence gut microbiota composition, and to identify gaps and opportunities for more relevant dietary assessment in diet-microbiome research.

Results

Of 241 papers retrieved, 38 systematic reviews,Citation13–18,Citation22–52 and 106 primary studiesCitation53–159 reported on diet-microbiome associations in humans or in-vitro laboratory studies with human fecal samples or human models, and were included in this review. The dietary aspects studies were categorized and reported as dietary patterns, dietary carbohydrate, protein, fat, micronutrients, bioactive food compounds, food additives, and individual foods.

Dietary patterns and the microbiota

Six systematic reviewsCitation13,Citation14,Citation22–24,Citation43 and 10 primary studiesCitation53–61,Citation137 reported on associations between dietary patterns and microbiota (Supplementary Table S1). Of these, four systematic reviewsCitation13,Citation22–24 and six primary studiesCitation53–57,Citation137 reported on associations between vegan or vegetarian, ‘healthy’, ‘plant-based’, or ‘Mediterranean’ dietary patterns and microbiota, with up to six microbial species associations reported in one study.Citation54 One systematic reviewCitation14 and four primary studiesCitation54,Citation58–60 investigated the effect of ‘western’, less healthy’, or ‘animal-based’ dietary patterns on microbiota, and reported associations with up to seven associations across taxonomic levels. Two systematic reviewsCitation14,Citation43 and two primary studiesCitation60,Citation61 reported on the effects of alterations in total energy intake on microbiota, with up to eight bacterial associations reported.Citation60 Studies that reported on associations between dietary patterns and intestinal microbiota differentiated the total diet dichotomously as either ‘plant’ or ‘animal’ based, vegan/vegetarian or omnivorous or as a ‘healthy’ or ‘unhealthy’ diet pattern based on total energy and macronutrient distribution.

Criteria for delineating specific dietary patterns were heterogenous in both review papersCitation14,Citation22–24 and primary studies.Citation53–55,Citation58–60 In studies that assessed or reviewed vegan or vegetarian diets compared to an omnivorous diet or ‘animal-based’ diet, inclusion criteria often included a predetermined period of adherence to the diet under study, which was either self-reported by participants (retrospective and usually longer duration of up to one year) or prospectively prescribed or provided as part of the intervention (usually 1 to 3 months).Citation22 For comparison of Mediterranean style or ‘healthy’ with a ‘Western’ or ‘less healthy’ dietary pattern, a classification tool was used to rank or score the diet, before, associations with microbiota or microbiome were analyzed statistically. Some studies used validated dietary assessment tools, while others involved more arbitrary classification systems or post-hoc delineation to categorize dietary intakes as ‘healthy’ or ‘unhealthy’ based on study-specific criteria.Citation57

In reviews and primary studies included, dietary patterns were compared with species-level microbial abundance,Citation22,Citation23,Citation54,Citation58–60 microbial diversityCitation54 and microbial enterotype.Citation14,Citation53 ‘Healthy’, ‘plant-based’ and ‘Mediterranean’ dietary patterns were generally associated with higher or increased Bacteroidetes (phylum),Citation13,Citation22 Firmicutes (phylum),Citation13,Citation54 Prevotella,Citation13,Citation22 Roseburia,Citation54,Citation57 Faecalibacterium prausnitzii,Citation54 Bifidobacteria, and Haemophilus.Citation55 Xylanibacter, Prevotella, Butyrivibrio and Treponemaare genera were found to be exclusive to people on a plant-based diet in one study.Citation56 One review reported an association between the Mediterranean diet and microbial metabolite ((short chain fatty acid (SCFA)) productionCitation13 and other studies reported the functional attributes of specific microbes that were relevant to the dietary pattern.Citation54,Citation58–60 A ‘Western’, ‘animal-based’ or ‘less healthy’ dietary pattern generally reported lower abundance of the same microbiota,Citation58–60 often because of the dichotomous, relative nature of analysis. These dietary patterns were specifically associated with increased genera Bacteroides,Citation14,Citation58 Clostridium,Citation54 AlistipesCitation58 and Bilophila wadsworthia.Citation58

Studies that reported on associations between total energy (Calorie or kilojoule) intake and GI microbiota varied in the primary and secondary outcomes assessed. One systematic review focused on interventions that achieved either increased or decreased energy intake while maintaining macronutrient ratios.Citation14 One review investigated the impact of a very low energy, ultra-processed dietCitation43 and another compared microbiota outcomes before and after energy restriction with modified macronutrient ratios.Citation60 Despite these differences, lower (or reducing) energy intake compared to higher (or increasing) total energy intake generally resulted in increased abundance of Bacteroidetes,Citation14,Citation43 decreased FirmicutesCitation43,Citation60 and increased GI microbial diversity.Citation14

Dietary carbohydrates, fibres and the microbiota

Twelve systematic reviewsCitation14–17,Citation25–30,Citation42,Citation45 and 32 primary studiesCitation44,Citation58–61,Citation65,Citation74–95,Citation135,Citation136,Citation140,Citation150 reported associations between microbiota and either total digestible carbohydrates, total fibers, specific digestible carbohydrates or specific fiber types, with up to 52 associations reported across six studies reported in one reviewCitation25 (Supplementary Table S2).

Carbohydrate constituents of food reported in papers included in this review included digestible carbohydrate intake (total, high/low diet, saccharide type), fiber amount (total fiber or as higher/lower fiber), human enzyme digestibility (level of processing, resistant starch), availability to microbiota (reported as prebiotic, total fermentable carbohydrate, total fructans, fructooligosaccharides, galactooligosaccharides, oligofructose, raffinose, arabinoxylose, trehalose, polydextrose, sialic acid, resistant starch), fermentable oligosaccharide, disaccharide, monosaccharide and polyol (FODMAP) content (total, low FODMAP diet, fructose, lactose, polyols), by cereal grain amount and type (barley, rye, oats, wheat), grain fiber type (wheat, rye and lupine fiber, insoluble fiber) or grain processing (total wholegrains, intact cereal grains, grain size).

Carbohydrate or fiber-microbiota associations reported were generally as expected, with higher dietary fiber associated with higher or increased beneficial microbesCitation14–17,Citation25–27,Citation30,Citation42,Citation45,Citation60,Citation61,Citation65,Citation74,Citation76–84,Citation88–91,Citation136,Citation140 and the influence of carbohydrate varying depending on the food source and carbohydrate type.Citation17,Citation28,Citation29,Citation58–60,Citation75,Citation82,Citation85–87,Citation92–95,Citation135,Citation150

A systematic review that included 34 (intervention and cross-sectional) studies reported that high dietary fiber intake was associated with greater gut bacteria diversity and increased Firmicutes, Proteobacteria (phylum), Prevotella, Bifidobacterium and Lactobacillus genera, and increased and decreased Actinobacteria (phylum).Citation14 Reported associations between fiber and Bacteroidetes varied between studies, depending on whether phylum or species level data was measured, and the study design or granularity of dietary intake analysis.

One systematic reviewCitation14 reported that resistant starch intake (from supplementation or specific formulated food sources) increased Eubacteria and Ruminococcus. One review reported that higher (or addition of) resistant starch increased Bifidobacteria.Citation17 Interestingly, a lack of Ruminococcus bromii at baseline was associated with lower resistant starch digestion of 20–30% compared to 100% in participants with Ruminococcus bromii in their gut microbiotaCitation14 indicating the importance of baseline microbiota assessment and consideration of intra-individual responses. The type of resistant starch being investigated influenced the microbial response, although few studies have compared microbial responses to all resistant starch types in the same study sampleCitation78 and study populations also differ between studies.Citation60,Citation61

By definition, prebiotic fibers promote proliferation of beneficial bacteria or a beneficial microbial profile. Prebiotic fibers can be intrinsic to foods or consumed as supplements and were reported in studies in various ways. Total prebiotic fiber (intrinsic and supplemental) intake was reported together in some studies and separately in others, while other studies reported associations between individual prebiotic fibers and bacteria separately. Total prebiotic fiber, specific prebiotic fiber-rich foods, or supplements and total or specific whole grain foods or supplements were reported to have similar effects on microbiota, despite diverse study designs. Commonly reported associations included increased microbial diversity and abundance, increased Firmicutes, Ruminococcaceae (family), Bacteroides, Bifidobacterium, Lactobacillus, Anaerostipes, F. prausnitzii, Prevotella, Clostridium spp. and fecal butyrate,Citation14,Citation15,Citation17,Citation26–29,Citation44,Citation85,Citation86,Citation90,Citation91 lower relative abundance of Bacteroidetes, Escherichia coliCitation26,Citation28,Citation88 and inconsistent results reported for Clostridia class.Citation15,Citation26,Citation28,Citation91

Dietary approaches to the management of functional GI disorders involve manipulation of fermentable carbohydrate intake to alleviate GI pain and bowel symptoms. For example, the low FODMAP diet is an evidence-based dietary management approach for IBS, and its impact on bacterial fermentation has been investigated in both primary studies and systematic reviews.Citation16,Citation79 One systematic reviewCitation16 and a primary studyCitation79 that involved healthy individuals and people with IBS reported that a high FODMAP (compared to low FODMAP) diet resulted in microbiota changes that were comparable with those reported for supplementation with other prebiotic fibers. Associations with higher FODMAP intake included higher microbial abundance, fecal volume, gene richness, Ruminococcaceae (family), Lachnospiraceae (family), Lactobacillus (genus), Clostridium cluster XIVa (spp.), Akkermansia muciniphila (sp.), F. prausnitzii (sp.), Bilophila wadsworthia (sp.) and lower Clostridia (class) and Enterococcus (genus).Citation16,Citation79 A low FODMAP diet has also been associated with increased Clostridiales family XIII, Porphyromonas IVCitation16 (genus), Bacteroides,Citation45 BacteroidetesCitation65 and lower Propionibacteriaceae (family), Bifidobacterium, Clostridium cluster IV (spp.) and Ruminococcus torques.Citation16,Citation30,Citation79 A recent systematic review and meta-analysis of low FODMAP diets and microbiota outcomes in IBS reported that lower relative abundance of Actinobacteria (4 studies) was the only consistently reported association, with variable outcomes at phylum and genus level for all other reported microbes and SCFA.Citation45

Dietary protein, amino acids and the microbiota

Five reviewsCitation14,Citation31,Citation32,Citation40,Citation41 and 20 primary studiesCitation7,Citation46,Citation62–73,Citation138,Citation139,Citation154–157 reported on associations between microbiota and total dietary proteins, specific peptides or amino acid, with up to 14 associations across taxonomic levels reportedCitation62 (Supplementary Table S3).

Reported associations between dietary protein and microbiota range from very general bacterial enterotype and total dietary protein to highly specific bacterial species and individual amino acids. Studies in this review indicated that increased or higher total dietary protein intake is usually associated with less beneficial bacterial taxonomic and metabolite profiles,Citation14 although it is not possible to differentiate the effect of protein separately from the difference or change in total energy or the proportion of dietary fat or carbohydrate (and associated fiber).

Across the two reviews and four primary studies that reported on microbiota and dietary gluten intake, associations with 28 different bacterial species were reported. There were few consistent findings between studies, most likely due to the wide range of study designs. Inclusion criteria ranged from gluten-free to ‘low’ versus ‘high’ gluten intake, and study populations ranged from children to adults with or without celiac disease, IBS or non-celiac gluten sensitivity (NCGS), and analysis of samples from saliva, duodenal tissue or stool.Citation32,Citation62–66

Dietary intake of individual amino acids is not routinely assessed as part of nutrient analysis of total diet intake. One study reported reduced Firmicutes:Bacteroidetes following L-glutamine supplementation.Citation70 One study each reported on microbial metabolism of proline,Citation71 sulfur amino acids,Citation72 biogenic aminesCitation73 and L-carnitine.Citation14

Dietary fat and the microbiota

Two systematic reviewsCitation14,Citation18 and 11 primary studiesCitation20,Citation74,Citation96–103,Citation148 reported associations between the microbiota and total, saturated or unsaturated fats, with up to four bacterial associations in primary studiesCitation74,Citation101–103 (Supplementary Table S4).

Assessment of the absolute effect of total fat, dietary fat profile and individual fatty acids on microbiota in humans is complex. Altering total fat intake influences total energy intake or macronutrient proportions, as well as bile production, which all independently influence microbiota.Citation43,Citation60,Citation160 Overall, data in this review indicate that a high or increased intake of total fat and saturated fatty acids have a consistently negative effect on microbiota richness and diversity.Citation14,Citation18

Studies in this review reported high total fat intakes as being associated with higher or increased Bacteroidetes, Actinobacteria, Alistipes, and lower or decreased relative abundance of Firmicutes, Proteobacteria, Bifidobacteria, butyrate and total SCFAs,Citation14 with saturated fats specifically linked to an unfavorable gut microbiota profile,Citation18 increased Bacteroides, Bilophila (genus) and F. prausnitzii.Citation3,Citation97 One whole food-based dietary intervention reported 57 genus-level taxon differences between high saturated fat and a high MUFA diet.Citation99 The same study suggested that high animal fat intake for 1 month was associated with a Bacteroides enterotype.Citation99

Studies investigating the influence of dietary fats on microbiota did not consistently report similar or opposite effects of unsaturated fats on microbiota compared to saturated fats. Diets high in unsaturated fat and manipulation of unsaturated fatty acids had variable but generally less detrimental influences on relative microbial abundance, microbial richness or diversityCitation14,Citation18 (Supplementary Table S4).

Micronutrients and the microbiota

Four reviewsCitation14,Citation33–35 and one primary studyCitation104 reported on a range of direct and indirect associations between micronutrients and gut microbes (Supplementary Table S5). Some microbes, such as Eubacterium rectale and Porphyromonas gingivalis (sp) are reported to be dependent on vitamin B6 for enzymatic activities.Citation14 Vitamin B12 intake and supplementation have been associated with gut microbiome diversity, relative abundance, functional capacity and SCFA production.Citation33 Altered gut microbiota have also been reported following vitamin D supplementation.Citation34,Citation35

In relation to dietary or supplemental mineral intake, Clostridium (spp.) and fecal SCFAs were increased following administration of phosphorous and calcium in one study.Citation104 Reduced Lactobacilli (g) has been reported in iron-deficiency anemia.Citation14

In addition to some gut microbes being micronutrient dependent, microbes may also be involved in micronutrient production. Gut microbiota are involved in synthesis of b-carotene, vitamin B6 and folate, with vitamin B12 specifically reported as being synthesized by Propionibacterium freudenreichii (sp), Listeria innocua (sp) and Lactobacillus reuteri (sp).Citation14 One review reported that Bifidobacterium (g) used resistant starch to produce folate.Citation14

Bioactive food compounds and the microbiota

Eight reviewsCitation14,Citation27,Citation30,Citation36,Citation37 reported phytochemical-microbiota associations (Supplementary Table S6). The phytochemicals that were most commonly reported to be associated with gut microbiota in this review were dietary fibers (see carbohydrate section), vitamin and minerals (see micronutrients section) and polyphenols, a heterogeneous collection of plant-derived compounds, with the structural unit of hydroxylated aromatic or phenol rings in common.Citation161 Polyphenols were reported to influence gut microbiota to a similar extent to oligosaccharide prebiotics,Citation36 with evidence from systematic reviews of human trials, intervention studies and some preclinical studies supporting this strong relationship.

Polyphenol consumption positively influenced gut microbiota, with bifidogenic effects reported in three systematic reviewsCitation14,Citation36,Citation37 and higher or increased Lactobacillus in two systematic reviews.Citation14,Citation36 Other bacteria reported to be higher or increase in relation to polyphenol intakes included Ruminococcaceae, Lachnospiraceae (family), Lachnospira (genus), Anaerostipes, Bacteroides, Roseburia (genus), Prevotella, Enterococcus (genus) and Eggerthella (genus) Acetitomaculum (genus), Faecalicoccus (genus), Kopriimonas (genus), Verrucomicrobia (phylum), Flavonifractor (genus), Christensenellaceae (family), Mogibacteriacea (family), Akkermansia muciniphila, Bacteroidetes, Sutterella (genus), Butyricicoccus (genus), Lactobacillus, Bacteroides, and AlistipesCitation36

Polyphenol intake was associated with beneficial bacteria including Ruminococcus bromii assisting in resistant starch degradation in the human gut.Citation36 F. prausnitzii, Roseburia and Anaerostipes produce butyrate, which stimulates production of mucins that improve gut integrity.Citation36 Both Eggerthella lenta and Bacteroides uniformis can break down resveratrol into dihydroresveratrol, Proteobacteria including Gammaproteobacteria (class) metabolizes uric acid, and Akkermansia has been consistently associated with reduced fat mass.Citation36 Some polyphenols can reduce the detrimental effects of potentially harmful microbial by-products. For example, the glycoside Allicin inhibits gut microbial production of pro-inflammatory TMAOCitation5

In parallel to these prebiotic and health-promoting characteristics, Lactobacillus, Bifidobacteria and other commensals also halt the growth of, or outcompete pathogenic microbes.Citation36 Clostridia (class) and some Clostridium spp. were reported as being substantially inhibited by polyphenols in one systematic reviewCitation14 and in human and preclinical trials (Supplementary Table S6). This association was attributed to procyanidins and the catechins in foods like green and black tea.Citation36 Other pathogens reported to be inhibited by phytochemicals included Salmonella (genus) and Bacteroides.Citation14 The different types of polyphenols exert their respective effects by preferentially binding to specific bacterial cell membrane types, altering functional aspects of the membrane thereby preventing bacterial growth. Other mechanisms of action include changing the pH or releasing signal molecules that are not conducive to the growth of pathogenic bacteria.Citation161 Some included studies reported no statistically significant associations between phytochemicals and microbiota but this could generally be explained by the studies being either under-powered, short-term studies without genus-level analysis or in ‘healthy’ populations, without existing dysbiosis.Citation36

Naturally occurring food chemicals implicated in food intolerances include salicylates, amines, and glutamates. Although interaction with chemoreceptors is the most commonly proposed mechanism of action for natural food chemicals in gastrointestinal disorder management,Citation162 salicylates, biogenic amines and glutamates (see also protein section) are each reported to have associations with gut microbiotaCitation132 and microbial metabolites.Citation30,Citation133 Further research is warranted to determine whether natural food chemicals act directly on chemoreceptors or whether microbiota or bioactive microbial metabolites play any role in etiology or symptom induction in these disorders.

Food additives and the microbiota

Two reviewsCitation38,Citation39 and 43 individual studiesCitation105–134,Citation152,Citation153 reported an association between microbiota and an emulsifier, thickener, stabilizer, preservative, flavoring, sweetener or food coloring (Supplementary Table S7). A high proportion of studies investigating the effects of food additives on the gut microbiota have been animalCitation38,Citation118 or in vitro studies,Citation106,Citation130 with human studies conducted to date being disease-specific.Citation163 The emulsifiers carboxymethylcellulose and polysorbate-80 induced dysbiosis in one review that incorporated studies using animal and human models,Citation38 with inconsistent findings reported in primary studies with human subjects or samples.Citation105,Citation106

The only other association consistently reported at the systematic review level was for maltodextrin, which was associated with altered microbiota Firmicutes, Bacteroidetes, Lactobacillus or Bifidobacterium in half of the 42 studies in one review.Citation39 Maltodextrin is a food thickener and stabilizer that is often used as placebo for the control group in microbiome interventions, so it is notable that research indicates that this additive is not inert and independently influences the microbiota.Citation112,Citation116 It was notable that a daily dose of 2 g of the flavor enhancer monosodium glutamate resulted in no significant microbiota effects in one study.Citation152

Overall, beneficial Clostridium leptum and Blautia coccoides seemed particularly susceptible to food additives, with reduction in response to emulsifiers, colorings, detergent and preservatives.Citation106 The addition of dishwashing detergent to human fecal samples negatively affected many bacterial types, resulting in increased pathogenic E. coli, Shigella and Klebsiella, increased Bacteroides/Prevotella ratio,Citation106 decreased relative abundance and decreased beneficial butyrate-producing Bifidobacterium, Firmicutes, B. coccoides and C. leptum.Citation106 Carrageenan emulsifier and cinnamaldehyde flavoring exerted similarly detrimental effects on microbiota as detergent, although fewer microbial species were reported to be affected.Citation106 Thickening agents gum ArabicCitation107,Citation108 and xanthan gumCitation109 were reported to be cleaved by specific primary degrading bacteria, producing metabolites for secondary degradation by other bacteria. These gums therefore exert differential effects on microbiota depending on the relative abundance of primary and secondary degraders. In two studies that tested the effects of preservatives on human fecal samples, pathogenic bacteria were generally increased, with detrimental effects on commensals.Citation106,Citation110 The natural preservative nisin exerted a beneficial effect by inhibiting Clostridium difficile and altering gram-positive bacteriaCitation111 (Supplementary Table S7).

Sweeteners that have been assessed for association with gut microbiota include aspartame (amino acid derived), trehalose (non-reducing disaccharide), stevioside (steviol glycosides), erythritol, xylitol, maltitol, lactitol (sugar alcohols), saccharin (chemical compound) sucralose (chlorinated sucrose) and polydextrose (glucose polymer, contains sorbitol). Study design varied considerably in terms of the dosage of sweeteners, method of analysis for associations and testing of singleCitation106,Citation116,Citation117,Citation128 or combinations of sweeteners.Citation106,Citation118 Of the sweeteners identified as being associated with gut microbiota in this review, those reported to have a detrimental effect on microbiota included trehalose which increased C. difficile,Citation116 and sucralose,Citation119 saccharineCitation119 and a combination of saccharine, sucralose and aspartame that had a dysbiotic effect on microbiota.Citation106 Stevioside, sugar alcohols, aspartame and acesulfame-KCitation112 were reported to influence gut bacteriaCitation112 and bacterial metabolites.Citation106 The polyol sweetener mannitol was also reported to alter the metaproteome across microbiomes, whereas erythritol did not significantly alter metaproteomes in one study.Citation153

Evidence relating to the influence of food coloring intake on gut microbiota is restricted to animal studies and in vitro studies using human fecal samples. Comparisons between studies are limited by the diverse study designs with high variability between colors investigated, dosage and sample preparation. The influence of colorants on microbiota included altered growth of Bacteroides, Bifidobacterium, Clostridium, Enterococcus, Escherichia, Lactobacillus and RuminococcusCitation131 and SaccharomycesCitation129 species, decreased microbial metabolism of isobutyrate, isovalerate and methanol,Citation130 decreased production of acetate, butyrate and propionateCitation130 and increased bacterial degradation liberating potentially carcinogenic metabolites.Citation164

Individual foods and the microbiota

Foods and beverages assessed for associations with microbiota separately from their nutrient components were predominantly investigated for a prebiotic or probiotic effect. Items included nuts, specific nut varieties, alcoholic drinks, coffee, and fermented foods or drinks, and were reported across three reviewsCitation14,Citation47,Citation48 and fourteen primary studiesCitation54,Citation141–144,Citation146,Citation147,Citation149–151,Citation158,Citation159,Citation165,Citation166 (Supplementary Table S8)

The combined effect of the fiber, phytochemical and fatty acid content on microbiota was assessed for individual nut varieties in some studiesCitation48,Citation141,Citation143,Citation144,Citation146,Citation147,Citation149,Citation165 and collectively in two reviews.Citation47,Citation48 The influence of these foods on microbiota was generally prebiotic and consistent with reported separate effects of constituent fibers, phytochemicals, and fatty acids. One review reported that significant increases in Roseburia and Clostridium were negated if walnut studies were removed from analyses.Citation47

One reviewCitation14 and one primary studyCitation151 reported on the influence of alcoholic drinks on microbiota, with the findings generally consistent with those reported for phytochemicals (see Section 6). In one study, red wine consumption increased Proteobacteria, Firmicutes and Bacteroidetes regardless of whether it contained alcohol or not, but dealcoholized red wine also increased Fusobacteria (phylum).Citation151

Analysis of stool samples from 6,811 individuals from the American Gut Project revealed statistically significant differences in beta diversity and differential taxa between fermented food consumers and non-consumers, with the metabolome of fermented food consumers enriched with putatively health-promoting conjugated linoleic acid.Citation158 Two similar fermented dairy products were reported to influence microbiota.Citation54,Citation142 Buttermilk increased bacterial diversityCitation142 and yogurt increased Lactobacillus, Bifidobacterium and Streptococcus thermophilus.Citation54 A diet rich in fermented foods steadily increased microbiota diversity and decreased inflammatory markers in another intervention studyCitation159

Dietary factor classification by dietary assessment and microbial degradation

Dietary factors in this review identified as being associated with microbiota were categorized as macronutrients (carbohydrate, fat, protein), micronutrients, other bioactive food compounds, food additives and specific foods. The foods or nutrients that were assessed for associations with microbiota in the current review could therefore be classified into four broad groups based on human digestibility, microbiota degradability and whether they can be assessed in the human diet using existing methods:

  1. Routinely analyzed using standard food composition database and relevant to microbiota because they are not absorbed for human metabolism during digestion. These were classified as ‘degradable, assessable’ and included carbohydrate, fat and protein;

  2. able to be analyzed using standard methods but predominantly absorbed in the small intestine so may be of less or indirect relevance to large bowel microbiota. These were classified as ‘digestible, assessable’ and included carbohydrate, protein, fat and micronutrients;

  3. foods or nutrients regularly investigated in relation to gastrointestinal microbiota but that can only be assessed and analyzed in isolation, not as part of the whole food matrix or whole-of-diet analysis, because they are not listed in food composition databases. These were classified as ‘degradable, individually assessable’ and included proteins and protein components (peptides and amino acids), or:

  4. foods, food components or nutrients that are important or potentially relevant to gastrointestinal microbiota research but were rarely assessed or only reported in animal studies. These were classified as ‘degradable, not assessed or not assessable’ and included food additives, bioactive compounds and specific foods.

Discussion

This systematic review synthesizes existing research findings regarding relationships between dietary intake and the microbiota, and progresses understanding of which food components may be most relevant to diet-microbiota research. It is evident from this review that some food components that are not quantified in current food composition databases do impact microbiota composition, function and potential health influences. Studies investigating such microbiota-relevant food components therefore currently need to analyze food components of interest individually, which is expensive and inefficient and has the potential for error. This review highlights the need for more comprehensive methods to assess and analyze intake of specific food components relevant to microbial metabolism. This level of detail in dietary assessment for diet-microbiota research will be necessary before it is even possible to consider future goals of personally tailored microbiota-specific nutrition therapy for maintaining or improving health status.

Degradable, assessable nutrients (carbohydrate, protein, fat)

Some food components routinely analyzed using standard dietary assessment methods are highly relevant to microbiota because they are not absorbed during human digestion and metabolism. The most obvious example is the expected and evident positive influence of a high or increased dietary fiber intake on bacterial profile, microbiota diversity and metabolite production.Citation14,Citation25,Citation74–77 This prebiotic effect of fiber is consistent with existing literature and consensus in the field.Citation3,Citation50 The strong alignment of microbiota-relevant dietary factors that are comprehensively captured in existing dietary assessment and analysis methods was evidenced by the high proportion of studies that reported on these dietary factors and high number of reported fiber-microbiota associations.

As dietary fibers are commonly found in high carbohydrate foods, it can be difficult to conclude whether observed effects are related to total dietary fiber intake or the whole food containing the carbohydrate or other nutrients associated with those foods. Total dietary fiber was the most commonly reported fiber measure in studies included in this review, but various types of dietary fiber may differentially impact the microbiome, as does the form in which they are consumed (i.e., supplemented or within a whole-food matrix). This highlights the need for further characterization of the physicochemical characteristics and combinations of fibers, doses and duration of intake to achieve clinically meaningful GI health benefits.Citation167

The overlap between effects of prebiotic fibers and high FODMAP diets and opposing findings with low FODMAP diets on microbiota are expected, given the prebiotic capabilities of oligosaccharide FODMAPs.Citation168 Many bacteria can break down short-chain oligofructose but only specific Bifidobacterium spp. digest long-chain fructans.Citation168 The individual effects of other constituent FODMAPs on human microbiota have not been as thoroughly investigated. The availability of fructose for bacterial degradation is highly determined by dietary load and subsequent availability of the fructose supply to bacteria, as well as the gut microenvironment, with availability of fructose being much higher in the duodenum compared to the large intestine.Citation169 Lactose degradation is determined by relative availability of lactase enzyme within the villi in the duodenum. Although lactose, fructose and polyols are grouped together as osmotic rather than gas producing FODMAPs, they each influence gut microbes differently, and individual polyols may also have different effects.

Other dietary associations with microbiota that were expected and readily analyzed included very high intakes of dietary fatCitation96,Citation97 or proteinCitation14 that could not feasibly be completely absorbed in the small intestine. Amino acids are consumed in food both as ‘free’ form or as part of peptide chains that are enzymatically digested to amino acids. Some microbes also cleave peptides to amino acids or ferment amino acids, producing microbial fermentation metabolites, including ammonia, amines, phenols, indoles, and hydrogen sulfides.Citation50 The wide range of reported protein-microbiome associations is partially attributable to the diversity of protein and amino acid structures, and the relative ability of different microbes to cleave bonds between amino acids or to utilize their nitrogen or sulfur. Mechanistic, in vitro and some experimental studies have investigated microbial metabolism of amino acidsCitation46,Citation54,Citation68,Citation69 and identify microbiota-mediated degradation pathwaysCitation51 and dominant amino acids metabolisers in each part of the digestive tract.Citation46 Very few studies have reported on associations between dietary amino acid intake and microbiota because the necessary analysis methods are not available.

The implications of increased availability of fats and proteins to microbiota in the large bowel as a result of dietary manipulation, nutrient-binding medications or cooking methods are interesting to consider and plausible to analyze. Although measuring the exact amount of specific nutrients available for microbial degradation is not yet possible, the effect of very high intake of particular macronutrients can be assessed in interventions or in research with population subsets that follow specific dietary regimes that involve extreme macronutrient alternations, such as ketogenic diets.Citation170 The effects of nutrient-binding medications are harder to assess because of the potential confounding effect of concurrently prescribed diet and lifestyle changes, and the possibility that medications may independently influence gut microbiota.Citation171

Digestible, assessable nutrients (carbohydrate, protein, fat, micronutrients)

Some nutrients or food components that can be assessed and analyzed using standard methods are relevant to upper but less relevant to lower GI microbiota because they are readily absorbed in the small intestine. Nutrients in this group from this review included digestible carbohydrate (total sugars, starches), total protein, some amino acids (e.g., tryptophan), dietary fat and most micronutrients. Far fewer associations were reported for total dietary proteins,Citation31,Citation61 carbohydratesCitation3 and fatsCitation61 than for their constituents or foods containing these macronutrients. The relatively low number of diet-microbiota associations reported in reviews and primary studies for these nutrients is indicative of misalignment in timing between study design regarding microbiota sampling and dietary assessment or analysis methods.

As humans efficiently digest and absorb protein, the vast majority of dietary protein in an average diet is enzymatically digested and absorbed before the end of the small intestine, leaving little available for colonic microbial fermentationCitation172 (). However, dietary peptides can be a microbial fermentation substrate across the intestine from the oral cavity to the colon. Bacteroides, Clostridium, Propionibacterium, Fusobacterium, Streptococcus, Bacillus, Lactobacillus and Proteobacteria are all contain proteolytic bacteria involved in catabolism and recycling nitrogen compounds, including amino acids.Citation46,Citation154 Bacteria with proteolytic capability that can survive or proliferate in upper parts of the intestinal tract have a competitive advantage in terms of sourcing nutrients. For example, some oral microbes (Rothia, Actinomyces odontolyticus, Streptococcus) have gluten-cleaving capability,Citation155 suggesting that bacterial cleavage of gluten may be initiated in the oral cavity before enzymatic digestion commences. Intestinal proteolytic digestion of gluten produces proline- and glutamine-rich polypeptides that can be cleaved by specific GI bacteriaCitation32,Citation156 and Pseudomonas spp. including P. aeruginosa have gluten-degrading capabilities.Citation64,Citation157

Figure 1. Accessibility of nutrients and other food components to intestinal microbiota at different sites within the human digestive tract, and associated dietary assessment methods.

Created in BioRender
Figure 1. Accessibility of nutrients and other food components to intestinal microbiota at different sites within the human digestive tract, and associated dietary assessment methods.

Micronutrients contribute to human and gut microbial health. Assessing associations between micronutrient intake and microbiota is complex because of the bi-directional nature of influence between bacteria and some vitamins or minerals as some microbiota utilize micronutrients remaining from human digestion and some contribute to production of essential vitamins.Citation14 For example, the function of certain gut microbes is dependent on vitamin B availabilityCitation14,Citation173 and the relative abundance of some microbes is associated with vitamin E intake.Citation174 Alterations to gut microbiota have been reported following supplementation with vitamins B2 and B3,Citation174 vitamin B12Citation33 vitamin D,Citation34,Citation35 iron,Citation175 calcium and phosphorous.Citation174 Vitamins reported to be produced by gut microbes included vitamin A,Citation14,Citation176 vitamins B1 and B2,Citation177 folateCitation14 and vitamin K.Citation14 Microbe composition and activity may also influence mineral absorption. Bioavailability of iron,Citation175 calcium and phosphorousCitation174 have been reported to improve as a result of SCFAs reducing the pH in colon. Beneficial Lactobacillus spp. increase bioavailability of magnesium, which in turn improves the composition of gut microbiota.Citation174 Despite awareness of these bidirectional relationships, micronutrient bioavailability to microbiota and human hosts are not as well characterized.Citation173

Although intake of micronutrients from food and supplements can be measured using standard dietary assessment methods, this review highlighted the considerable potential for improved characterization of the relationships between micronutrient bioavailability and microbiota in human hosts.Citation173 For example, body stores and pools of some micronutrients are higher than indicated by dietary intake, suggesting they are produced in the colon by microbiota then absorbed.Citation173 Genetic abnormalities, such as vitamin D receptor deficiency, have also been used to investigate gut dysbiosis. Bioavailability and modulation of micronutrient status via microbiota are emerging and important areas of research. The implications of improved understanding of these relationships are broad, extending across dietary intake, nutritional supplements, pharmaceuticals and disease management. The lower proportion of studies that reported no significant microbiota alterations for the specified nutrients is consistent with conclusions of Jardon et al. (2022), who indicate a need for improved understanding of the intersect between human digestion and microbial fermentation in nutrient metabolism.Citation10 They propose that assessment and analysis of GI site-specific fermentation will advance our ability to influence interplay between diet, gut microbiota and metabolic processes.Citation10 It is likely that the lack of specificity when measuring total macronutrients, rather than component saccharides, amino acids and fatty acids, results in lack of sensitivity to changes in gut microbiota. Such changes result from delivery of specific nutrients to symbionts that possess specific systems for their metabolism.Citation178

Degradable, individually assessed nutrients (protein, including peptides and amino acids)

The third group of food components identified in our review were those predicted to influence the GI microbiota but were usually assessed and analyzed in isolation, rather than as part of the whole food matrix. This limits attribution of causation to the individual food or nutrient.

Gluten is an example of a food component that is highly relevant to GI health and the microbiota, but unable to be analyzed using current national food composition databases. Analyzing the presence or absence of gluten using ELISACitation179 is not useful for determining total dietary gluten intake, but is the only widely available method used in the food industry to date. Investigating the influence of gluten on microbiota is further complicated because much of the research is related to avoidance of gluten by people with celiac disease, or for other health-related reasons. This could mean that metabolic factors influence intestinal microbiota responses to gluten, in addition to the changes induced by altered dietary gluten intake.Citation62

Amino acid-based natural food chemicals (glutamates, biogenic amines) and microbial fermentation metabolites (short- or branched chain fatty acids, phenols, amines, sulfides, ammonia and trimethylamine N-oxide) need to be considered in diet-microbiome analysis because of their potential implications for the gut-microbiome-brain axis and related metabolic and health outcomes.Citation14,Citation40,Citation73 Histamine is of particular relevance in relation to GI conditions, being implicated in inflammatory mast cell activationCitation180 and chemosensor activation.Citation181 The influences of potentially immunogenic peptides resulting from microbial cleavage of dietary proteins may also have a role in immune-related GI manifestations.Citation182 Differentiating the relative influence of dietary histamine in the context of diet-microbiota associations is complicated by the fact that mast cells and many bacteria, as well as some yeasts and molds have the capacity for histamine formation.Citation41

Other foods, food components and nutrients with potential to influence microbiota (food additives, bioactive compounds, specific foods)

A fourth group of food components important to GI microbiota research were identified, which were rarely quantified or only reported in animal studies. These food components included food additives and microbiota-relevant bioactive food compounds such as phytochemicals, food additives, salicylates, and glutamates.

Incorporating all chemicals in plants, phytochemicals have varying, but potentially profound influences on gut microbes that are exerted by inhibiting growth of pathogenic microbes, producing bioactive microbial metabolites and promoting growth of commensal microbes.Citation36 Phytochemicals can be grouped by their structure, function or by their colorCitation36 and their health-promoting efficacy depends on bioavailability, structural complexity, and positioning within the food matrix. To optimally assess the influence of phytochemicals in the context of the food matrix, high phytochemical foods need to be consumed as part of a whole diet, then coded and analyzed within the same food composition database as other nutrients being reported.

The salicylate and glutamate contents of foods have been semi-quantified on low food chemical dietary guidance, but not in the context of diet-microbiota assessment. To determine whether associations between natural food chemicals and microbiota exist, these bioactive compounds need to be assessable in the context of the whole diet.

Data sources used to quantify dietary intake of food additives vary depending on the additive or specific health condition being assessed, and range from population-level data linked to branded food databases to individual-level assessment using food frequency questionnaires, 24-h recall or food diaries.Citation183 Food additive analysis methods need to become more sensitive and specific for accurate assessment of the effects of food additives on the gut microbiota.

It was evident from this review that some dietary factors influence the microbiota via less direct mechanisms. For example, associations between fat and microbiota are likely to be attributable to increased bile acids in response to dietary fat ingestion, particularly saturated fat. Although nearly all fatty acids are absorbed in the small intestine, bile released in response to dietary fat intake can impact on microbiota throughout the digestive tract. Medications that bind fat can also alter microbial fermentation potential because they are not absorbed in the small intestine and become a fermentation substrate in the colon. It remains a substantial challenge in this field to differentiate the direct effects of macronutrient intake on microbiota from the indirect effects of metabolic factors, disease states or predisposition and medications.Citation184

The relationships between fermented foods and microbiota are complex, with the findings that diet steadily increased microbiota diversity consistent with a narrative review that reported high fermented food intake to influence more than 50 bacterial species across 20 studies.Citation185

Microbiota sampling

Although microbiota sampling and microbiome analysis methodologies were not the primary focus of this review, some findings did highlight potential future directions that were particularly relevant to assessment of diet-microbiota associations. For example, reported diet-microbiota associations needed to be considered in relation to the location within the digestive tract being investigated. Intestinal microbes that differentially populate the various niche environments of the digestive tract have different fermentation capabilities and substrate preferences, which has implications for analysis and interpretation of diet-microbiota associations.Citation178 Few studies in this review reported on upper GI microbiota data, which would be more specific and sensitive for analyzing microbiota associations with some nutrients.

Niche environments also exist in between the mucosal surface and lumen in the different compartments along the GI tract. In the current review, the vast majority of microbiota and microbiota data originated from stool samples and are therefore representative of luminal colonic microbes, which may be considerably different to the diversity and abundance of microbial species in the colonic mucosa or the lumen or mucosa of other parts of the digestive tract.

Another consideration is matching the timing of stool sampling to dietary intake. Microbial species respond at different rates to dietary changes, so alignment of sample collection and dietary assessment is important. In their longitudinal study with 34 individuals who provided a stool sample daily for 17 days, Johnson et al. (2019) reported that microbiota composition aligned best with multiple days of dietary data history, aligned more strongly with food than nutrients and that microbCreated in BioRenderial responses to diet were highly individual.Citation148 Timing of stool sample collection is not often outlined in study methods, which may influence accuracy of reported outcomes.

Improving dietary assessment and analysis in microbiome research

There is considerable scope to improve dietary assessment methods in diet-microbiome research to differentially measure associations with mucosal or luminal microbiota from samples collected throughout the digestive tract from mouth to anus. Accurate, longitudinal dietary intake assessment and analysis, combined with collection of multiple microbiota samples from relevant compartments in the digestive tract will contribute to more reliable, rigorous diet-microbiome data. Baseline gut microbial characteristics and habitual dietary intake may predict an individual’s response to dietary interventions. It is therefore crucial that both dietary data and microbiota samples are collected at this timepoint, as well as during and post-study periods, in order to better understand factors influencing intervention responses.Citation186 Analysis methods may also substantially influence reported associations, depending on whether individual or group-level analysis has been used. For example, Tap et al. (2015) reported no significant inter-individual differences but significant intra-individual genus-level microbiota differences in a study investigating differences in microbiota with low or high fiber intake.Citation77 Understanding the mechanisms of the differential responses to diet is essential to move forward in the field of precision nutrition.Citation10

Some interesting considerations evidenced from this review include the need for more research in which ‘target’ foods are consumed and analyzed within the whole diet context, the exciting potential for progress in this field and the substantial opportunity for secondary analysis of existing dietary data collected in diet-microbiome research.

Two substantial shifts in dietary assessment methodology are needed for diet-microbiome research to progress. The main shift from a dietary assessment perspective is to consider what is not digested by human digestive enzymes, and what might be degraded in different parts of the digestive tract to where enzymatic digestion occurs. For example, gluten degrading microbes are present in the oral cavity, so it is feasible that gliadin peptides from gluten degradation are present in the esophagus.Citation187 The change in focus from a dietary analysis viewpoint is to consider what data from whole-diet intake assessment can and cannot be quantified using available dietary analysis methods, and how data from the wide range of different sources described in this review could potentially be consolidated. These major considerations will require combined dietary assessment, microbiome and metabolomic expertise and considerable investment, but could stand to contribute to quantum diet-microbiome knowledge gains, applications and opportunities.

Recommendations

  1. To optimize analysis of diet-microbiome associations: Highly specific microbiota data needs to be compared with equally robust and specific dietary intake data. Where dichotomous dietary categorization is appropriate to the research question or necessary due to budget constraints, it’s use in microbiome research could be enhanced by use of standardization of macronutrient proportions and energy intake categorization and by use of validated, accessible tools for delineating dietary patterns. Improved use of multiomics modeling of metabolic pathways specific to individual microbiome signatures could facilitate precision nutrition to modulate metabolites associated with human health.Citation171

  2. Optimizing diet assessment and analysis in study design: In future studies that involve shotgun metagenomics, it is recommended that an expert in dietary assessment is involved in planning the dietary assessment and analysis to ensure the research questions can be answered using the methods employed and that use of the dietary data is optimized in the context of study variables. Additionally, future studies should ensure that dietary assessment methods report details of the food composition database used to analyze intake. Consensus methods and/or a core outcome set for diet–microbiome studies would facilitate the achievement of this recommendation.

  3. Opportunities for secondary analysis: A high proportion of included studies reported dietary data collection that far exceeded what was used in diet-microbiota analysis and reported as study findings. This suggested there is a considerable dietary data across studies already conducted that could be used for secondary analyses. We recommend that researchers consider involving dietary assessment specialists to optimize dietary data use, and potentially consider retrospective, secondary analyses. For example, it is possible for data from food frequency questionnaires to be disaggregated, reanalyzed using additional microbiota-relevant dietary data and then for associations with microbiota or microbiome data to be reevaluated.

  4. Matching dietary assessment methods to microbiome sampling methods: Accurate dietary intake quantification will reduce systematic and random errors. The choice of dietary assessment method needs to be appropriately aligned with research question in relation to microbiome sampling and analysis. For example, food frequency questionnaires are appropriate for assessing associations between dietary patterns and the microbiome, whereas repeat 24-h recalls are more appropriate to assess short-term changes in microbiome in relation to daily variability in dietary intake.

  5. To improve dietary assessment of fat-microbiota associations: Consensus around grouping of fatty acids for analysis is needed. Approximation of usual dietary intake requires consumption of fat within the food matrix, rather than analysis of supplementary fats only. Particular consideration should be given to the production and influence of bile and dietary fibers is recommended.

  6. Improved assessment and analysis of carbohydrate-microbiota associations: This will depend on increased consideration of bacterial metabolism or foods in addition to usual consideration of enzymatic digestion, and consolidation of data from the range of industry and research sources from which it is currently drawn. Considerable progress in understanding of carbohydrate-microbiota associations could be made by conducting secondary analysis of already collected data, but using a consolidated analysis database that includes whole grain, resistant starches, fiber and FODMAP data.

  7. To improve dietary assessment of protein-microbiota associations: More accurate tools for assessing dietary intake of amino acids and microbiota-relevant peptides such as gluten and gliadin are needed.

  8. Combining comparator data to increase accuracy: Use of dietary intake in combinations with phenotypic and genotypic profiles, and with microbial metabolites and metabolomic data will increase accuracy for assessment of associations with microbiota data.

  9. Differentiating animal and human studies: Animal and human studies are not clearly defined or differentiated in the findings of all trials or reviews, adding further complexity to analysis of associations. It was also evident that animal trials are used in cases where the safety of a new food additive or supplement has not been established. For example, one animal study reported that high doses of inulin induced microbiota-related hepatic damage, and cautioned high-dose supplementation in humans.Citation84 These findings are inconsistent with the findings from human studies on inulin supplementation that used physiologically justifiable doses of inulin, but nevertheless warrant further investigation.

  10. Differentiating digestive health, disorder, or disease state of participants: Although the review focused on healthy individuals, some studies included or compared healthy individuals and those with gut disorders. It was interesting that two reviews reported similar effects of prebiotic fibers on bacterial profiles between healthy individuals and those with intestinal diseases.Citation16,Citation168

Strengths and limitations

This systematic review comprehensively assesses associations between diet and microbiota in human studies, describing relevant dietary assessment methods where possible. As the aim of this review was predominantly focused on capturing all reported diet-microbiota associations, systematic reviews and primary studies were included, so additional screening was conducted to mitigate overlap between reporting of review and primary study findings. It was not possible to consistently report whether 16S or MGS was used for microbial sequencing, as it was not always reported. While this is a limitation in terms of comparability between studies, the intention of this review was to describe the range of food constituents that have been assessed and those that should be considered in the future for more sensitive and specific analysis of diet-microbiota associations.

Conclusion

This systematic review highlights considerable opportunities to improve diet-microbiome research methodology. Dietary assessment methodology needs to expand to include microbiota-relevant data and integrate with nutritional metabolomic data, which requires dietary assessment expertise. Study designs need to align dietary assessment methods with standardized microbiota sample collection and analysis, using deeply phenotyped participant cohorts. Refinement and consolidation of food composition databases should include microbiome-relevant data, particularly from human intervention studies. A substantial shift in dietary and nutritional metabolomic quantification methodology has the potential to make microbiome-specific nutrition therapy a reality.

Materials and methods

This review involved a systematic literature search and data extraction process, with narrative synthesis of extracted data.

Step 1: systematic search to identify studies for inclusion

PUBMED search: ((((((((((((((((fiber) OR (fiber)) OR (carb*)) OR (FODMAP)) OR (protein)) OR (fat)) OR (food additive)) OR (emulsifier)) OR (food chemical)) OR (diet[MeSH Terms])) OR (nutritive value[MeSH Terms])) OR (additive, food[MeSH Terms])) AND (microbiota)) OR (microbial)) OR (microbiome)) AND (gut)) OR (gastrointestinal microbiome[MeSH Terms]).

Systematic reviews that addressed at least one dietary factor in relation to the human gut microbiome were included. While focused on healthy individuals, reviews were included if they included both human and animal data, or they compared healthy individuals and those with gastrointestinal conditions, if these data could be differentiated and extracted. The search state date of January 1, 2010, was based on the rationale that this would result in a higher proportion of studies that reported MGS and less 16s only microbiome data, and therefore be more comparable. Reviews were excluded if they focused on very specific population groups (e.g. people who had undergone bariatric surgery, cancer treatment, dialysis).

Two reviewers (JH, KD) independently screened titles and abstracts for relevant papers, with conflicts discussed to achieve consensus on inclusion and exclusion of retrieved titles.

Step 2: identification of primary studies for inclusion

  1. Data from primary studies identified in the search were extracted to data extraction tables

  2. Studies identified in systematic reviews retrieved above, where only one study was identified as reporting on a diet-microbiome association of interest, were extracted as primary studies.

  3. The reference lists of narrative reviews identified during literature searches, including those identified via Step 1, were screened by one reviewer (JH) for reporting of other diet-microbiome factors. Any new studies identified were extracted as primary studies.

  4. Dietary factors with potential microbiome associations (identified in preclinical studies) were identified, with individual searches for relevant human studies conducted in PubMed using a similar search strategy as described above, without AND (systematic review).

Step 3: data extraction and synthesis

Data were extracted by one reviewer (JH) to an excel spreadsheet with one main spreadsheet, then discussed and grouped (JH, KD) before individual sheets (KD) were created for each dietary factor identified (dietary profile, carbohydrates and fibers, proteins, fats, bioactive food compounds, food additives and individual foods). Data retrieved from systematic reviews were differentiated from primary studies, with the number of studies from the review that reported on each specific dietary factor reported as identified as (SR, x or x/y studies) depending on whether or not the number of studies that reported non-significant findings were specified. Study design and characteristics relating to dietary assessment and analysis methods were also extracted, including retrieval of information from protocols and supplementary materials, if indicated in the article (Supplementary Tables S1–S8). Extracted data were checked by a second researcher to ensure that findings from systematic reviews and primary studies that may have contributed to systematic reviews were differentiated as far as possible, to avoid replication of reported findings. Data from reviews and primary studies were then consolidated, with the number of studies reporting on each dietary factor and number of reported associations with bacterial numbers, diversity or abundance tallied and presented as tables and narrative reporting in the manuscript.

Consolidated data describing diet-microbiome associations and dietary assessment methods were then described narratively. Supplementary tables that include diet-microbiota associations were prepared and are available on request by e-mail to the corresponding author.

Supplemental material

Supplemental Material

Download Zip (115.7 KB)

Acknowledgments

The authors gratefully acknowledge JH for contributions to the literature search and data extraction for this review.

Disclosure statement

KD, GW, ECH, CEC, SK report no disclosures in relation to this work. NJT reports non-financial support from: Norgine (2021)(IBS interest group), personal fees from Allakos (gastroduodenal eosinophilic disease) (2021), Bayer (IBS) (2020), Planet Innovation (Gas capsule IBS) (2020), twoXAR Viscera Labs, (USA 2021) (IBS-diarrhoea), Dr Falk Pharma (2020) (EoE), Sanofi-aventis, Glutagen (2020) (celiac disease), IsoThrive (2021) (esophageal microbiome), BluMaiden (microbiome advisory board) (2021), Rose Pharma (IBS) (2021), Intrinsic Medicine (2022) (human milk oligosaccharide), Comvita Mānuka Honey (2021) (digestive health), Astra Zeneca (2022), outside the submitted work. In addition, Dr. Talley has a patent Nepean Dyspepsia Index (NDI) 1998, Biomarkers of IBS licensed, a patent Licensing Questionnaires Talley Bowel Disease Questionnaire licensed to Mayo/Talley, a patent Nestec European Patent licensed, and a patent Singapore Provisional Patent “Microbiota Modulation of BDNF Tissue Repair Pathway” issued, “Diagnostic marker for functional gastrointestinal disorders” Australian Provisional Patent Application 2021901692. Committees: OzSage; NHMRC Principal Committee (Research Committee) Asia Pacific Association of Medical Journal Editors, Rome V Working Team Member (Gastroduodenal Committee), International Plausibility Project Co-Chair (Rome Foundation funded), COVID-19 vaccine forum member (by invitation only). Community group: Advisory Board, IFFGD (International Foundation for Functional GI Disorders), AusEE. Editorial: Medical Journal of Australia (Editor in Chief), Mayo Clinic Proceedings (Assoc Ed), Up to Date (Section Editor), Precision and Future Medicine, Sungkyunkwan University School of Medicine, South Korea, Med (Journal of Cell Press). Dr. Talley is supported by funding from the National Health and Medical Research Council (NHMRC) to the Centre for Research Excellence in Digestive Health and he holds an NHMRC Investigator grant.

Data availability statement

The data that support the findings will be available in Open Science Foundation at https://osf.io/mw2ek/following a 12-month embargo from the date of publication to allow for commercialization of research findings.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19490976.2024.2350785

Additional information

Funding

This work was supported by NHMRC Centre of Research Excellence in Digestive Health pilot grant funding under University of Newcastle grant GS210011.NJT reports non-financial support from: Norgine (2021)(IBS interest group), personal fees from Allakos (gastroduodenal eosinophilic disease) (2021), Bayer (IBS) (2020), Planet Innovation (Gas capsule IBS) (2020), twoXAR Viscera Labs (USA 2021) (IBS-diarrhea), Dr Falk Pharma (2020) (EoE), Sanofi-aventis, Glutagen (2020) (celiac disease), IsoThrive (2021) (esophageal microbiome), BluMaiden (microbiome advisory board) (2021), Rose Pharma (IBS) (2021), Intrinsic Medicine (2022) (human milk oligosaccharide), Comvita Mānuka Honey (2021) (digestive health), Astra Zeneca (2022), outside the submitted work. In addition, Dr Talley has a patent Nepean Dyspepsia Index (NDI) 1998, Biomarkers of IBS licensed, a patent Licensing Questionnaires Talley Bowel Disease Questionnaire licensed to Mayo/Talley, a patent Nestec European Patent licensed, and a patent Singapore Provisional Patent “Microbiota Modulation of BDNF Tissue Repair Pathway” issued, “Diagnostic marker for functional gastrointestinal disorders” Australian Provisional Patent Application 2,021,901,692. Committees: OzSage; NHMRC Principal Committee (Research Committee) Asia Pacific Association of Medical Journal Editors, Rome V Working Team Member (Gastroduodenal Committee), International Plausibility Project Co-Chair (Rome Foundation funded), COVID-19 vaccine forum member (by invitation only). Community group: Advisory Board, IFFGD (International Foundation for Functional GI Disorders), AusEE. Editorial: Medical Journal of Australia (Editor in Chief), Mayo Clinic Proceedings (Assoc Ed), Up to Date (Section Editor), Precision and Future Medicine, Sungkyunkwan University School of Medicine, South Korea, Med (Journal of Cell Press). Dr Talley is supported by funding from the National Health and Medical Research Council (NHMRC) to the Centre for Research Excellence in Digestive Health and he holds an NHMRC Investigator grant.

References

  • Eetemadi A, Rai N, Pereira BMP, Kim M, Schmitz H, Tagkopoulos I. The computational diet: a review of computational methods across diet, microbiome, and health. Front Microbiol. 2020;11:393. doi:10.3389/fmicb.2020.00393.
  • Shreiner AB, Kao JY, Young VB. The gut microbiome in health and in disease. Current opinion in gastroenterology. Curr Opin Gastroenterol. 2015;31(1):69. doi:10.1097/MOG.0000000000000139.
  • Singh RK, Chang H-W, Yan D, Lee KM, Ucmak D, Wong K, Abrouk M, Farahnik B, Nakamura M, Zhu TH. et al. Influence of diet on the gut microbiome and implications for human health. J Transl Med. 2017;15(1):1–26. doi:10.1186/s12967-017-1175-y.
  • Rollo ME, Williams RL, Burrows T, Kirkpatrick SI, Bucher T, Collins CE. What are they really eating? A review on new approaches to dietary intake assessment and validation. Curr Nutr Rep. 2016;5(4):307–314. doi:10.1007/s13668-016-0182-6.
  • Leeming ER, Louca P, Gibson R, Menni C, Spector TD, Le Roy CI. The complexities of the diet-microbiome relationship: advances and perspectives. Genome Med. 2021;13(1):1–14. doi:10.1186/s13073-020-00813-7.
  • Shanahan ER, McMaster JJ, Staudacher HM. Conducting research on diet–microbiome interactions: a review of current challenges, essential methodological principles, and recommendations for best practice in study design. J Hum Nutr Diet. 2021;34(4):631–644. doi:10.1111/jhn.12868.
  • Kim CH. Immune regulation by microbiome metabolites. Immunology. 2018;154(2):220–229. doi:10.1111/imm.12930.
  • Scalbert A, Brennan L, Manach C, Andres-Lacueva C, Dragsted LO, Draper J, Rappaport SM, van der Hooft JJ, Wishart DS. The food metabolome: a window over dietary exposure. Am J Clin Nutr. 2014;99(6):1286–1308. doi:10.3945/ajcn.113.076133.
  • LeVatte M, Keshteli AH, Zarei P, Wishart DS. Applications of metabolomics to precision nutrition. Lifestyle Genomics. 2022;15(1):1–9. doi:10.1159/000518489.
  • Jardon KM, Canfora EE, Goossens GH, Blaak EE. Dietary macronutrients and the gut microbiome: a precision nutrition approach to improve cardiometabolic health. Gut. 2022;71(6):1214–1226. doi:10.1136/gutjnl-2020-323715.
  • Lewis CM, Vassos E. Polygenic risk scores: from research tools to clinical instruments. Genome Med. 2020;12(1):1–11. doi:10.1186/s13073-020-00742-5.
  • Shah A, Talley NJ, Koloski N, Macdonald GA, Kendall BJ, Shanahan ER, Walker MM, Keely S, Jones MP, Morrison M. et al. Duodenal bacterial load as determined by quantitative polymerase chain reaction in asymptomatic controls, functional gastrointestinal disorders and inflammatory bowel disease. Aliment Pharmacol Ther. 2020;52(1):155–167. doi:10.1111/apt.15786.
  • Gibiino G, De Siena M, Sbrancia M, Binda C, Sambri V, Gasbarrini A, Fabbri C. Dietary habits and gut microbiota in healthy adults: focusing on the right diet. A systematic review. Int J Mol Sci. 2021;22(13):6728. doi:10.3390/ijms22136728.
  • Frame LA, Costa E, Jackson SA. Current explorations of nutrition and the gut microbiome: a comprehensive evaluation of the review literature. Nutr Rev. 2020;78(10):798–812. doi:10.1093/nutrit/nuz106.
  • Jefferson A, Adolphus K. The effects of intact cereal grain fibers, including wheat bran on the gut microbiota composition of healthy adults: a systematic review. Front Nutr. 2019;6:33. doi:10.3389/fnut.2019.00033.
  • Vandeputte D, Joossens M. Effects of low and high FODMAP diets on human gastrointestinal microbiota composition in adults with intestinal diseases: a systematic review. Microorganisms. 2020;8(11):1638. doi:10.3390/microorganisms8111638.
  • Williams GM, Tapsell LC, O’Brien CL, Tosh SM, Barrett EM, Beck EJ. Gut microbiome responses to dietary intake of grain-based fibers with the potential to modulate markers of metabolic disease: a systematic literature review. Nutr Rev. 2021;79(11):1274–1292. doi:10.1093/nutrit/nuaa128.
  • Wolters M, Ahrens J, Romaní-Pérez M, Watkins C, Sanz Y, Benítez-Páez A, Stanton C, Günther K. Dietary fat, the gut microbiota, and metabolic health–A systematic review conducted within the MyNewGut project. Clin Nutr. 2019;38(6):2504–2520. doi:10.1016/j.clnu.2018.12.024.
  • Healey GR, Murphy R, Brough L, Butts CA, Coad J. Interindividual variability in gut microbiota and host response to dietary interventions. Nutrition Reviews. 2017;75(12):1059–1080. doi:10.1093/nutrit/nux062.
  • Johnson AJ, Zheng JJ, Kang JW, Saboe A, Knights D, Zivkovic AM. A guide to diet-microbiome study design. Front Nutr. 2020;7:79. doi:10.3389/fnut.2020.00079.
  • Shang Q. From correlation to causation: the missing point in the study of functional foods and gut microbiota. J Funct Foods. 2019;61:103466. doi:10.1016/j.jff.2019.103466.
  • Losno EA, Sieferle K, Perez-Cueto FJA, Ritz C. Vegan diet and the gut microbiota composition in healthy adults. Nutrients. 2021;13(7):2402. doi:10.3390/nu13072402.
  • Trefflich I, Jabakhanji A, Menzel J, Blaut M, Michalsen A, Lampen A, Abraham K, Weikert C. Is a vegan or a vegetarian diet associated with the microbiota composition in the gut? Results of a new cross-sectional study and systematic review. Crit Rev Food Sci Nutr. 2020;60(17):2990–3004. doi:10.1080/10408398.2019.1676697.
  • Kimble R, Gouinguenet P, Ashor A, Stewart C, Deighton K, Matu J, Griffiths A, Malcomson FC, Joel A, Houghton D. et al. Effects of a Mediterranean diet on the gut microbiota and microbial metabolites: a systematic review of randomized controlled trials and observational studies. Crit Rev Food Sci Nutr. 2022;63:1–22.
  • So D, Whelan K, Rossi M, Morrison M, Holtmann G, Kelly JT, Shanahan ER, Staudacher HM, Campbell KL. Dietary fiber intervention on gut microbiota composition in healthy adults: a systematic review and meta-analysis. Am J Clin Nutr. 2018;107(6):965–983. doi:10.1093/ajcn/nqy041.
  • Le Bastard Q, Chapelet G, Javaudin F, Lepelletier D, Batard E, Montassier E. The effects of inulin on gut microbial composition: a systematic review of evidence from human studies. Eur J Clin Microbiol Infect Dis. 2020;39(3):403–413. doi:10.1007/s10096-019-03721-w.
  • Verhoog S, Taneri PE, Roa Díaz ZM, Marques-Vidal P, Troup JP, Bally L, Franco OH, Glisic M, Muka T. Dietary factors and modulation of bacteria strains of akkermansia muciniphila and faecalibacterium prausnitzii: a systematic review. Nutrients. 2019;11(7):1565. doi:10.3390/nu11071565.
  • Tangestani H, Emamat H, Ghalandari H, Shab-Bidar S. Whole grains, dietary fibers and the human gut microbiota: a systematic review of existing literature. Recent Pat Food, Nutr & Agric. 2020;11(3):235–248. doi:10.2174/2212798411666200316152252.
  • Valido E, Stoyanov J, Bertolo A, Hertig-Godeschalk A, Zeh RM, Flueck JL, Minder B, Stojic S, Metzger B, Bussler W. et al. Systematic review of the effects of oat intake on gastrointestinal health. J Nutr. 2021;151(10):3075–3090. doi:10.1093/jn/nxab245.
  • Su H, Li Y-T, Heitkemper MM, Zia J. Effects of low-FODMAPS diet on irritable bowel syndrome symptoms and gut microbiome. Gastroenterol Nurs. 2019;42(2):150–158. doi:10.1097/SGA.0000000000000428.
  • Albracht-Schulte K, Islam T, Johnson P, Moustaid-Moussa N. Systematic review of beef protein effects on gut microbiota: implications for health. Adv Nutr. 2021;12(1):102–114. doi:10.1093/advances/nmaa085.
  • Chander AM, Yadav H, Jain S, Bhadada SK, Dhawan DK. Cross-talk between gluten, intestinal microbiota and intestinal mucosa in celiac disease: recent advances and basis of autoimmunity. Front Microbiol. 2018;9:2597. doi:10.3389/fmicb.2018.02597.
  • Guetterman HM, Huey SL, Knight R, Fox AM, Mehta S, Finkelstein JL. Vitamin B-12 and the gastrointestinal microbiome: a systematic review. Adv Nutr. 2022;13(2):530–558. doi:10.1093/advances/nmab123.
  • Waterhouse M, Hope B, Krause L, Morrison M, Protani MM, Zakrzewski M, Neale RE. Vitamin D and the gut microbiome: a systematic review of in vivo studies. Eur J Nutr. 2019;58(7):2895–2910. doi:10.1007/s00394-018-1842-7.
  • Bellerba F, Muzio V, Gnagnarella P, Facciotti F, Chiocca S, Bossi P, Cortinovis D, Chiaradonna F, Serrano D, Raimondi S. et al. The association between Vitamin D and gut microbiota: a systematic review of human studies. Nutrients. 2021;13(10):3378. doi:10.3390/nu13103378.
  • Moorthy M, Chaiyakunapruk N, Jacob SA, Palanisamy UD. Prebiotic potential of polyphenols, its effect on gut microbiota and anthropometric/clinical markers: a systematic review of randomised controlled trials. Trends Food Sci & Technol. 2020;99:634–649. doi:10.1016/j.tifs.2020.03.036.
  • Khairudin MAS, Mhd Jalil AM, Hussin N. Effects of polyphenols in tea (Camellia sinensis sp.) on the modulation of gut microbiota in human trials and animal studies. Gastroenterol Insights. 2021;12(2):202–216. doi:10.3390/gastroent12020018.
  • Abiega-Franyutti P, Freyre-Fonseca V. Chronic consumption of food-additives lead to changes via microbiota gut-brain axis. Toxicology. 2021;464:153001. doi:10.1016/j.tox.2021.153001.
  • Almutairi R, Basson AR, Wearsh P, Cominelli F, Rodriguez-Palacios A. Validity of food additive maltodextrin as placebo and effects on human gut physiology: systematic review of placebo-controlled clinical trials. Eur J Nutr. 2022;61(6):1–19. doi:10.1007/s00394-022-02802-5.
  • Comas-Basté O, Sánchez-Pérez S, Veciana-Nogués MT, Latorre-Moratalla M, Vidal-Carou MDC. Histamine intolerance: the current state of the art. Biomolecules. 2020;10(8):1181. doi:10.3390/biom10081181.
  • Hrubisko M, Danis R, Huorka M, Wawruch M. Histamine intolerance—the more we know the less we know. A review. Nutrients. 2021;13(7):2228. doi:10.3390/nu13072228.
  • Mora-Flores LP, Moreno-Terrazas Casildo RMT, Fuentes-Cabrera J, Pérez-Vicente HA, de Anda-Jáuregui G, Neri-Torres EE. The role of carbohydrate intake on the gut microbiome: a weight of evidence systematic review. Microorganisms. 2023;11(7):1728. doi:10.3390/microorganisms11071728.
  • Lane M, Howland G, West M, Hockey M, Marx W, Loughman A, O’Hely M, Jacka F, Rocks T. The effect of ultra-processed very low-energy diets on gut microbiota and metabolic outcomes in individuals with obesity: a systematic literature review. Obesity Res & Clin Pract. 2020;14(3):197–204. doi:10.1016/j.orcp.2020.04.006.
  • Myhrstad MC, Tunsjø H, Charnock C, Telle-Hansen VH. Dietary fiber, gut microbiota, and metabolic regulation—Current status in human randomized trials. Nutrients. 2020;12(3):859. doi:10.3390/nu12030859.
  • So D, Loughman A, Staudacher H. Effects of a low FODMAP diet on the colonic microbiome in irritable bowel syndrome: a systematic review with meta-analysis. Am J Clin Nutr. 2022;116(4):943–952. Early online. doi: 10.1093/ajcn/nqac176.
  • Dai Z-L, Wu G, Zhu W-Y. Amino acid metabolism in intestinal bacteria: links between gut ecology and host health. Front Biosci. 2011;16(1):1768–1786. doi:10.2741/3820.
  • Creedon AC, Hung ES, Berry SE, Whelan K. Nuts and their effect on gut microbiota, gut function and symptoms in adults: a systematic review and meta-analysis of randomised controlled trials. Nutrients. 2020;12(8):2347. doi:10.3390/nu12082347.
  • Fitzgerald E, Lambert K, Stanford J, Neale EP. The effect of nut consumption (tree nuts and peanuts) on the gut microbiota of humans: a systematic review. Br J Nutr. 2021;125(5):508–520. doi:10.1017/S0007114520002925.
  • Costa MADC, Vilela DL, Fraiz GM, Lopes IL, Coelho AI, Castro LC, Martin JG. Effect of kombucha intake on the gut microbiota and obesity-related comorbidities: a systematic review. Crit Rev Food Sci Nutr. 2021;63:1–16.
  • Peled S, Livney YD. The role of dietary proteins and carbohydrates in gut microbiome composition and activity: a review. Food Hydrocoll. 2021;120:106911. doi:10.1016/j.foodhyd.2021.106911.
  • Ma N, Ma X. Dietary amino acids and the gut‐microbiome‐immune axis: physiological metabolism and therapeutic prospects. Compr Rev Food Sci Food Saf. 2019;18(1):221–242. doi:10.1111/1541-4337.12401.
  • Arias N, Arboleya S, Allison J, Kaliszewska A, Higarza SG, Gueimonde M, Arias JL. The relationship between choline bioavailability from diet, intestinal microbiota composition, and its modulation of human diseases. Nutrients. 2020;12(8):2340. doi:10.3390/nu12082340.
  • De Angelis M, Ferrocino I, Calabrese FM, De Filippis F, Cavallo N, Siragusa S, Rampelli S, Di Cagno R, Rantsiou K, Vannini L. et al. Diet influences the functions of the human intestinal microbiome. Sci Rep. 2020;10(1):4247. doi:10.1038/s41598-020-61192-y.
  • Asnicar F, Berry SE, Valdes AM, Nguyen LH, Piccinno G, Drew DA, Leeming E, Gibson R, Le Roy C, Khatib HA. et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med. 2021;27(2):321–332. doi:10.1038/s41591-020-01183-8.
  • Stanford J, Charlton K, Stefoska-Needham A, Zheng H, Bird L, Borst A, Fuller A, Lambert K. Associations among plant-based diet quality, uremic toxins, and gut microbiota profile in adults undergoing hemodialysis therapy. J Renal Nutr. 2021;31(2):177–188. doi:10.1053/j.jrn.2020.07.008.
  • De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA. 2010;107(33):14691–14696. doi:10.1073/pnas.1005963107.
  • Haro C, Montes-Borrego M, Rangel-Zúñiga OA, Alcalá-Díaz JF, Gómez-Delgado F, Pérez-Martínez P, Delgado-Lista J, Quintana-Navarro GM, Tinahones FJ, Landa BB. et al. Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population. J Clin Endocr Metab. 2016;101(1):233–242. doi:10.1210/jc.2015-3351.
  • David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA. et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563. doi:10.1038/nature12820.
  • Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Appl Environ Microbiol. 2007;73(4):1073–1078. doi:10.1128/AEM.02340-06.
  • Salonen A, Lahti L, Salojärvi J, Holtrop G, Korpela K, Duncan SH, Date P, Farquharson F, Johnstone AM, Lobley GE. et al. Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men. ISME J. 2014;8(11):2218–2230. doi:10.1038/ismej.2014.63.
  • Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P, Bergerat A. et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5(2):220–230. doi:10.1038/ismej.2010.118.
  • Hansen L, Roager HM, Søndertoft NB, Gøbel RJ, Kristensen M, Vallès-Colomer M, Vieira-Silva S, Ibrügger S, Lind MV, Mærkedahl RB. et al. A low-gluten diet induces changes in the intestinal microbiome of healthy Danish adults. Nat Commun. 2018;9(1):1–13. doi:10.1038/s41467-018-07019-x.
  • Ercolini D, Francavilla R, Vannini L, De Filippis F, Capriati T, Di Cagno R, Iacono G, De Angelis M, Gobbetti M. From an imbalance to a new imbalance: Italian-style gluten-free diet alters the salivary microbiota and metabolome of African celiac children. Sci Rep. 2015;5(1):1–11. doi:10.1038/srep18571.
  • Garcia-Mazcorro JF, Rivera-Gutierrez X, Cobos-Quevedo O, Grube-Pagola P, Meixueiro-Daza A, Hernandez-Flores K, Cabrera-Jorge F, Vivanco-Cid H, Dowd S, Remes-Troche J. et al. First insights into the gut microbiota of Mexican patients with celiac disease and non-celiac gluten sensitivity. Nutrients. 2018;10(11):1641. doi:10.3390/nu10111641.
  • Naseri K, Dabiri H, Rostami-Nejad M, Yadegar A, Houri H, Olfatifar M, Sadeghi A, Saadati S, Ciacci C, Iovino P. et al. Influence of low FODMAP-gluten free diet on gut microbiota alterations and symptom severity in Iranian patients with irritable bowel syndrome. BMC Gastroenterol. 2021;21(1):1–14. doi:10.1186/s12876-021-01868-5.
  • Costa A, Constante M, Stefanolo JP, Temprano P, Sugai E, Sanchez MIP, Moreno ML, Vázquez H, Mauriño E, Gonzalez AF. et al. Tu1459 Symptomatic and asymptomatic celiac disease patients on long-term gluten-free diet exhibit differences in fecal microbial taxa. Gastroenterology. 2020;158(6):S–1116. doi:10.1016/S0016-5085(20)33459-4.
  • Bonder MJ, Tigchelaar EF, Cai X, Trynka G, Cenit MC, Hrdlickova B, Zhong H, Vatanen T, Gevers D, Wijmenga C. et al. The influence of a short-term gluten-free diet on the human gut microbiome. Genome Med. 2016;8(1):1–11. doi:10.1186/s13073-016-0295-y.
  • Sánchez‐Jiménez F, Ruiz‐Pérez MV, Urdiales JL, Medina MA. Pharmacological potential of biogenic amine–polyamine interactions beyond neurotransmission. British J Pharmacology. 2013;170(1):4–16. doi:10.1111/bph.12109.
  • Baj A, Moro E, Bistoletti M, Orlandi V, Crema F, Giaroni C. Glutamatergic signaling along the microbiota-gut-brain axis. IJMS. 2019;20(6):1482. doi:10.3390/ijms20061482.
  • de Souza AZZ, Zambom AZ, Abboud KY, Reis SK, Tannihão F, Guadagnini D, Saad MJA, Prada PO. Oral supplementation with L-glutamine alters gut microbiota of obese and overweight adults: a pilot study. Nutrition. 2015;31(6):884–889. doi:10.1016/j.nut.2015.01.004.
  • Mayneris-Perxachs J, Castells-Nobau A, Arnoriaga-Rodríguez M, Martin M, de la Vega-Correa L, Zapata C, Burokas A, Blasco G, Coll C, Escrichs A. et al. Microbiota alterations in proline metabolism impact depression. Cell Metab. 2022;34(5):681–701. e10. doi:10.1016/j.cmet.2022.04.001.
  • Yao CK, Rotbart A, Ou JZ, Kalantar-Zadeh K, Muir JG, Gibson PR. Modulation of colonic hydrogen sulfide production by diet and mesalazine utilizing a novel gas-profiling technology. Gut Microbes. 2018;9(6):510–522. doi:10.1080/19490976.2018.1451280.
  • Spano G, Russo P, Lonvaud-Funel A, Lucas P, Alexandre H, Grandvalet C, Coton E, Coton M, Barnavon L, Bach B. et al. Biogenic amines in fermented foods. Eur J Clin Nutr. 2010;64(3):S95–S100. doi:10.1038/ejcn.2010.218.
  • Mayorga Reyes L, González Vázquez R, Cruz Arroyo SM, Melendez Avalos A, Reyes Castillo PA, Chavaro Pérez DA, Ramos Terrones I, Ramos Ibáñez N, Rodríguez Magallanes MM, Langella P. et al. Correlation between diet and gut bacteria in a population of young adults. Int J Food Sci Nutr. 2016;67(4):470–478. doi:10.3109/09637486.2016.1162770.
  • De Filippo C, Di Paola M, Ramazzotti M, Albanese D, Pieraccini G, Banci E, Miglietta F, Cavalieri D, Lionetti P. Diet, environments, and gut microbiota. A preliminary investigation in children living in rural and urban Burkina Faso and Italy. Front Microbiol. 2017;8:1979. doi:10.3389/fmicb.2017.01979.
  • David LA, Materna AC, Friedman J, Campos-Baptista MI, Blackburn MC, Perrotta A, Erdman SE, Alm EJ. Host lifestyle affects human microbiota on daily timescales. Genome Biol. 2014;15(7):1–15. doi:10.1186/gb-2014-15-7-r89.
  • Tap J, Furet J-P, Bensaada M, Philippe C, Roth H, Rabot S, Lakhdari O, Lombard V, Henrissat B, Corthier G. et al. Gut microbiota richness promotes its stability upon increased dietary fibre intake in healthy adults. Environ Microbiol. 2015;17(12):4954–4964. doi:10.1111/1462-2920.13006.
  • Martínez I, Kim J, Duffy PR, Schlegel VL, Walter J. Resistant starches types 2 and 4 have differential effects on the composition of the fecal microbiota in human subjects. PLOS One. 2010;5(11):e15046. doi:10.1371/journal.pone.0015046.
  • Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Gibson PR, Muir JG. Diets that differ in their FODMAP content alter the colonic luminal microenvironment. Gut. 2015;64(1):93–100. doi:10.1136/gutjnl-2014-307264.
  • Drabińska N, Jarocka-Cyrta E, Markiewicz L, Krupa-Kozak U. The effect of oligofructose-enriched inulin on faecal bacterial counts and microbiota-associated characteristics in celiac disease children following a gluten-free diet: results of a randomized, placebo-controlled trial. Nutrients. 2018;10(2):201. doi:10.3390/nu10020201.
  • Davis LM, Martínez I, Walter J, Goin C, Hutkins RW. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLOS ONE. 2011;6(9):e25200. doi:10.1371/journal.pone.0025200.
  • Chu N, He J, Ma R, Kong A, Chan J, Chow E. 556-P: the relationship between short-chain fermentable carbohydrates (FODMAPs) , gut microbiome, and glucose intolerance: an exploratory analysis. Diabetes. 2022;71(Supplement_1). doi:10.2337/db22-556-P.
  • Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N. et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585–588. doi:10.1038/nature12480.
  • Singh V, Yeoh BS, Chassaing B, Xiao X, Saha P, Aguilera Olvera R, Lapek JD, Zhang L, Wang W-B, Hao S. et al. Dysregulated microbial fermentation of soluble fiber induces cholestatic liver cancer. Cell. 2018;175(3):679–694. doi:10.1016/j.cell.2018.09.004.
  • Carvalho-Wells AL, Helmolz K, Nodet C, Molzer C, Leonard C, McKevith B, Thielecke F, Jackson KG, Tuohy KM. Determination of the in vivo prebiotic potential of a maize-based whole grain breakfast cereal: a human feeding study. Br J Nutr. 2010;104(9):1353–1356. doi:10.1017/S0007114510002084.
  • van Trijp MPH, Schutte S, Esser D, Wopereis S, Hoevenaars FPM, Hooiveld GJEJ, Afman LA. Minor changes in the composition and function of the gut microbiota during a 12-week whole grain wheat or refined wheat intervention correlate with liver fat in overweight and obese adults. J Nutr. 2021;151(3):491–502. doi:10.1093/jn/nxaa312.
  • Connolly ML, Lovegrove JA, Tuohy KM. In vitro evaluation of the microbiota modulation abilities of different sized whole oat grain flakes. Anaerobe. 2010;16(5):483–488. doi:10.1016/j.anaerobe.2010.07.001.
  • Martínez I, Lattimer JM, Hubach KL, Case JA, Yang J, Weber CG, Louk JA, Rose DJ, Kyureghian G, Peterson DA. et al. Gut microbiome composition is linked to whole grain-induced immunological improvements. ISME J. 2013;7(2):269–280. doi:10.1038/ismej.2012.104.
  • Lappi J, Salojärvi J, Kolehmainen M, Mykkänen H, Poutanen K, de Vos WM, Salonen A. Intake of whole-grain and fiber-rich rye bread versus refined wheat bread does not differentiate intestinal microbiota composition in Finnish adults with metabolic syndrome. J Nutr. 2013;143(5):648–655. doi:10.3945/jn.112.172668.
  • Aoe S, Nakamura F, Fujiwara S. Effect of wheat bran on fecal butyrate-producing bacteria and wheat bran combined with barley on bacteroides abundance in Japanese healthy adults. Nutrients. 2018;10(12):1980. doi:10.3390/nu10121980.
  • Smith SC, Choy R, Johnson SK, Hall RS, Wildeboer-Veloo ACM, Welling GW. Lupin kernel fiber consumption modifies fecal microbiota in healthy men as determined by rRNA gene fluorescent in situ hybridization. Eur J Nutr. 2006;45(6):335–341. doi:10.1007/s00394-006-0603-1.
  • Eid N, Enani S, Walton G, Corona G, Costabile A, Gibson G, Rowland I, Spencer JPE. The impact of date palm fruits and their component polyphenols, on gut microbial ecology, bacterial metabolites and colon cancer cell proliferation. J Nutr Sci. 2014;3:3. doi:10.1017/jns.2014.16.
  • Zhao S, Jang C, Liu J, Uehara K, Gilbert M, Izzo L, Zeng X, Trefely S, Fernandez S, Carrer A. et al. Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate. Nature. 2020;579(7800):586–591. doi:10.1038/s41586-020-2101-7.
  • Jones RB, Alderete TL, Kim JS, Millstein J, Gilliland FD, Goran MI. High intake of dietary fructose in overweight/obese teenagers associated with depletion of eubacterium and streptococcus in gut microbiome. Gut Microbes. 2019;10(6):712–719. doi:10.1080/19490976.2019.1592420.
  • Francavilla R, Calasso M, Calace L, Siragusa S, Ndagijimana M, Vernocchi P, Brunetti L, Mancino G, Tedeschi G, Guerzoni E. et al. Effect of lactose on gut microbiota and metabolome of infants with cow’s milk allergy. Pediatr Allergy Immunol. 2012;23(5):420–427. doi:10.1111/j.1399-3038.2012.01286.x.
  • Wu GD, Chen J, Hoffmann C, Bittinger K, Chen Y-Y, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108. doi:10.1126/science.1208344.
  • Fava F, Gitau R, Griffin BA, Gibson GR, Tuohy KM, Lovegrove JA. The type and quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty acid excretion in a metabolic syndrome ‘at-risk’population. Int J Obes. 2013;37(2):216–223. doi:10.1038/ijo.2012.33.
  • Wan Y, Wang F, Yuan J, Li J, Jiang D, Zhang J, Li H, Wang R, Tang J, Huang T. et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: a 6-month randomised controlled-feeding trial. Gut. 2019;68(8):1417–1429. doi:10.1136/gutjnl-2018-317609.
  • Lang JM, Pan C, Cantor RM, Tang WHW, Garcia-Garcia JC, Kurtz I, Hazen SL, Bergeron N, Krauss RM, Lusis AJ. et al. Impact of individual traits, saturated fat, and protein source on the gut microbiome. MBio. 2018;9(6):e01604–18. doi:10.1128/mBio.01604-18.
  • Lim RRX, Park MA, Wong LH, Haldar S, Lim KJ, Nagarajan N, Henry CJ, Jiang YR, Moskvin OV. Gut microbiome responses to dietary intervention with hypocholesterolemic vegetable oils. Npj Biofilms Microbiomes. 2022;8(1):1–12. doi:10.1038/s41522-022-00287-y.
  • Simoes CD, Maukonen J, Kaprio J, Rissanen A, Pietiläinen KH, Saarela M. Habitual dietary intake is associated with stool microbiota composition in monozygotic twins. J Nutr. 2013;143(4):417–423. doi:10.3945/jn.112.166322.
  • Pu S, Khazanehei HR, Krause DO, West SG, Kris‐Etherton PM, Jenkins DJ, Lamarche B, Jones PJ, Khafipour E. Effects of unsaturated fatty acids (USFA) on human gut microbiome profile in a subset of canola oil multicenter intervention trial (COMIT). FASEB; 2013;27:1056.7. doi:10.1096/fasebj.27.1_supplement.1056.7.
  • Fernandes J, Su W, Rahat-Rozenbloom S, Wolever TMS, Comelli EM. Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr & Diabetes. 2014;4(6):e121–e121. doi:10.1038/nutd.2014.23.
  • Trautvetter U, Camarinha-Silva A, Jahreis G, Lorkowski S, Glei M. High phosphorus intake and gut-related parameters–results of a randomized placebo-controlled human intervention study. Nutr J. 2018;17(1):1–11. doi:10.1186/s12937-018-0331-4.
  • Chassaing B, Compher C, Bonhomme B, Liu Q, Tian Y, Walters W, Nessel L, Delaroque C, Hao F, Gershuni V. et al. Randomized controlled-feeding study of dietary emulsifier carboxymethylcellulose reveals detrimental impacts on the gut microbiota and metabolome. Gastroenterology. 2022;162(3):743–756. doi:10.1053/j.gastro.2021.11.006.
  • Gerasimidis K, Bryden K, Chen X, Papachristou E, Verney A, Roig M, Hansen R, Nichols B, Papadopoulou R, Parrett A. et al. The impact of food additives, artificial sweeteners and domestic hygiene products on the human gut microbiome and its fibre fermentation capacity. Eur J Nutr. 2020;59(7):3213–3230. doi:10.1007/s00394-019-02161-8.
  • Pellock SJ, Redinbo MR. Glucuronides in the gut: sugar-driven symbioses between microbe and host. J Biol Chem. 2017;292(21):8569–8576. doi:10.1074/jbc.R116.767434.
  • Calame W, Weseler AR, Viebke C, Flynn C, Siemensma AD. Gum Arabic establishes prebiotic functionality in healthy human volunteers in a dose-dependent manner. Br J Nutr. 2008;100(6):1269–1275. doi:10.1017/S0007114508981447.
  • Ostrowski MP, La Rosa SL, Kunath BJ, Robertson A, Pereira G, Hagen LH, Varghese NJ, Qiu L, Yao T, Flint G. et al. Mechanistic insights into consumption of the food additive xanthan gum by the human gut microbiota. Nat Microbiol. 2022;7(4):556–569. doi:10.1038/s41564-022-01093-0.
  • Hrncirova L, Hudcovic T, Sukova E, Machova V, Trckova E, Krejsek J, Hrncir T. Human gut microbes are susceptible to antimicrobial food additives in vitro. Folia Microbiol (Praha). 2019;64(4):497–508. doi:10.1007/s12223-018-00674-z.
  • Le Lay C, Fernandez B, Hammami R, Ouellette M, Fliss I. On Lactococcus lactis UL719 competitivity and nisin (Nisaplin®) capacity to inhibit Clostridium difficile in a model of human colon. Front Microbiol. 2015;6:1020. doi:10.3389/fmicb.2015.01020.
  • Frankenfeld CL, Sikaroodi M, Lamb E, Shoemaker S, Gillevet PM. High-intensity sweetener consumption and gut microbiome content and predicted gene function in a cross-sectional study of adults in the United States. Annals of Epidemiology. 2015;25(10):736–742. e4. doi:10.1016/j.annepidem.2015.06.083.
  • Horwitz DL, McLane M, Kobe P. Response to single dose of aspartame or saccharin by NIDDM patients. Diabetes Care. 1988;11(3):230–234. doi:10.2337/diacare.11.3.230.
  • Tordoff MG, Alleva AM. Effect of drinking soda sweetened with aspartame or high-fructose corn syrup on food intake and body weight. Am J Clin Nutr. 1990;51(6):963–969. doi:10.1093/ajcn/51.6.963.
  • Ahmad SY, Friel J, Mackay D. The effects of non-nutritive artificial sweeteners, aspartame and sucralose, on the gut microbiome in healthy adults: secondary outcomes of a randomized double-blinded crossover clinical trial. Nutrients. 2020;12(11):3408. doi:10.3390/nu12113408.
  • Collins J, Robinson C, Danhof H, Knetsch CW, van Leeuwen HC, Lawley TD, Auchtung JM, Britton RA. Dietary trehalose enhances virulence of epidemic Clostridium difficile. Nature. 2018;553(7688):291–294. doi:10.1038/nature25178.
  • Gatea F, Sârbu I, Vamanu E. In vitro modulatory effect of stevioside, as a partial sugar replacer in sweeteners, on human child microbiota. Microorganisms. 2021;9(3):590. doi:10.3390/microorganisms9030590.
  • Suez J, Korem T, Zeevi D, Zilberman-Schapira G, Thaiss CA, Maza O, Israeli D, Zmora N, Gilad S, Weinberger A. et al. Artificial sweeteners induce glucose intolerance by altering the gut microbiota. Nature. 2014;514(7521):181–186. doi:10.1038/nature13793.
  • Suez J, Cohen Y, Valdés-Mas R, Mor U, Dori-Bachash M, Federici S, Zmora N, Leshem A, Heinemann M, Linevsky R. et al. Personalized microbiome-driven effects of non-nutritive sweeteners on human glucose tolerance. Cell. 2022;185(18):3307–3328.e19. doi:10.1016/j.cell.2022.07.016.
  • Serrano J, Smith KR, Crouch AL, Sharma V, Yi F, Vargova V, LaMoia TE, Dupont LM, Serna V, Tang F. et al. High-dose saccharin supplementation does not induce gut microbiota changes or glucose intolerance in healthy humans and mice. Microbiome. 2021;9(1):1–18. doi:10.1186/s40168-020-00976-w.
  • Arrigoni E, Brouns F, Amado R. Human gut microbiota does not ferment erythritol. Br J Nutr. 2005;94(5):643–646. doi:10.1079/BJN20051546.
  • Beards E, Tuohy K, Gibson G. A human volunteer study to assess the impact of confectionery sweeteners on the gut microbiota composition. Br J Nutr. 2010;104(5):701–708. doi:10.1017/S0007114510001078.
  • Gostner A, Blaut M, Schäffer V, Kozianowski G, Theis S, Klingeberg M, Dombrowski Y, Martin D, Ehrhardt S, Taras D. et al. Effect of isomalt consumption on faecal microflora and colonic metabolism in healthy volunteers. Br J Nutr. 2006;95(1):40–50. doi:10.1079/BJN20051589.
  • Finney M, Smullen J, Foster HA, Brokx S, Storey DM. Effects of low doses of lactitol on faecal microflora, pH, short chain fatty acids and gastrointestinal symptomology. Eur J Nutr. 2007;46(6):307–314. doi:10.1007/s00394-007-0666-7.
  • Ballongue J, Schumann C, Quignon P. Effects of lactulose and lactitol on colonic microflora and enzymatic activity. Scand J Gastroenterol. 1997;32(sup222):41–44. doi:10.1080/00365521.1997.11720716.
  • Ouwehand AC, Tiihonen K, Saarinen M, Putaala H, Rautonen N. Influence of a combination of Lactobacillus acidophilus NCFM and lactitol on healthy elderly: intestinal and immune parameters. Br J Nutr. 2008;101(3):367–375. doi:10.1017/S0007114508003097.
  • Salminen S, Salminen E, Koivistoinen P, Bridges J, Marks V. Gut microflora interactions with xylitol in the mouse, rat and man. Food Chem Toxicol. 1985;23(11):985–990. doi:10.1016/0278-6915(85)90248-0.
  • Costabile A, Fava F, Röytiö H, Forssten SD, Olli K, Klievink J, Rowland IR, Ouwehand AC, Rastall RA, Gibson GR. et al. Impact of polydextrose on the faecal microbiota: a double-blind, crossover, placebo-controlled feeding study in healthy human subjects. Br J Nutr. 2012;108(3):471–481. doi:10.1017/S0007114511005782.
  • Jabeen HS, Ur Rahman S, Mahmood S, Anwer S. Genotoxicity assessment of amaranth and Allura red using Saccharomyces cerevisiae. Bull Environ Contam Toxicol. 2013;90(1):22–26. doi:10.1007/s00128-012-0870-x.
  • Ivusic Polic I. Evaluation of the impact of azo dyes on the metabolism of stabilized fecal communities and in vitro cell culture. Ontario, Canada: University of Guelph; 2018. https://atrium.lib.uoguelph.ca/server/api/core/bitstreams/ce048094-a395-447f-bc83-8e35e54084e6/content.
  • Pan H, Feng J, He G-X, Cerniglia CE, Chen H. Evaluation of impact of exposure of Sudan azo dyes and their metabolites on human intestinal bacteria. Anaerobe. 2012;18(4):445–453. doi:10.1016/j.anaerobe.2012.05.002.
  • O’Connor K, Morrissette M, Strandwitz P, Ghiglieri M, Caboni M, Liu H, Khoo C, D’Onofrio A, Lewis K. Cranberry extracts promote growth of bacteroidaceae and decrease abundance of Enterobacteriaceae in a human gut simulator model. PLoS One. 2019;14(11):e0224836. doi:10.1371/journal.pone.0224836.
  • Hatch M. Gut microbiota and oxalate homeostasis. Ann Transl Med. 2017;5(2). doi:10.21037/atm.2016.12.70.
  • Lerner A, Benzvi C. Microbial transglutaminase is a very frequently used food additive and is a potential inducer of autoimmune/neurodegenerative diseases. Toxics. 2021;9(10):233. doi:10.3390/toxics9100233.
  • Mano F, Ikeda K, Joo E, Fujita Y, Yamane S, Harada N, Inagaki N. The effect of white rice and white bread as staple foods on gut microbiota and host metabolism. Nutrients. 2018;10(9):1323. doi:10.3390/nu10091323.
  • Healey G, Murphy R, Butts C, Brough L, Whelan K, Coad J. Habitual dietary fibre intake influences gut microbiota response to an inulin-type fructan prebiotic: a randomised, double-blind, placebo-controlled, cross-over, human intervention study. Br J Nutr. 2018;119(2):176–189. doi:10.1017/S0007114517003440.
  • Rejeski JJ, Wilson FM, Nagpal R, Yadav H, Weinberg RB. The impact of a Mediterranean diet on the gut microbiome in healthy human subjects: a pilot study. Digestion. 2022;103(2):133–140. doi:10.1159/000519445.
  • Kung H-F, Lee Y-C, Huang Y-L, Huang Y-R, Su Y-C, Tsai Y-H. Degradation of histamine by lactobacillus plantarum isolated from miso products. J Food Prot. 2017;80(10):1682–1688. doi:10.4315/0362-028X.JFP-17-135.
  • Schink M, Konturek PC, Tietz E, Dieterich W, Pinzer TC, Wirtz S, Neurath MF, Zopf Y. Microbial patterns in patients with histamine intolerance. J Physiol Pharmacol. 2018;69(4):579–593.
  • Holscher HD, Caporaso JG, Hooda S, Brulc JM, Fahey GC, Swanson KS. Fiber supplementation influences phylogenetic structure and functional capacity of the human intestinal microbiome: follow-up of a randomized controlled trial. Am J Clin Nutr. 2015;101(1):55–64. doi:10.3945/ajcn.114.092064.
  • Ukhanova M, Wang X, Baer DJ, Novotny JA, Fredborg M, Mai V. Effects of almond and pistachio consumption on gut microbiota composition in a randomised cross-over human feeding study. Br J Nutr. 2014;111(12):2146–2152. doi:10.1017/S0007114514000385.
  • Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S. et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352(6285):565–569. doi:10.1126/science.aad3369.
  • Holscher HD, Guetterman HM, Swanson KS, An R, Matthan NR, Lichtenstein AH, Novotny JA, Baer DJ. Walnut consumption alters the gastrointestinal microbiota, microbially derived secondary bile acids, and health markers in healthy adults: a randomized controlled trial. J Nutr. 2018;148(6):861–867. doi:10.1093/jn/nxy004.
  • Holscher HD, Taylor A, Swanson K, Novotny J, Baer D. Almond consumption and processing affects the composition of the gastrointestinal microbiota of healthy adult men and women: a randomized controlled trial. Nutrients. 2018;10(2):126. doi:10.3390/nu10020126.
  • Dieterich W, Schuppan D, Schink M, Schwappacher R, Wirtz S, Agaimy A, Neurath MF, Zopf Y. Influence of low FODMAP and gluten-free diets on disease activity and intestinal microbiota in patients with non-celiac gluten sensitivity. Clin Nutr. 2019;38(2):697–707. doi:10.1016/j.clnu.2018.03.017.
  • García-Mantrana I, Calatayud M, Romo-Vaquero M, Espín JC, Selma MV, Collado MC. Urolithin metabotypes can determine the modulation of gut microbiota in healthy individuals by tracking walnuts consumption over three days. Nutrients. 2019;11(10):2483. doi:10.3390/nu11102483.
  • Gargari G, Deon V, Taverniti V, Gardana C, Denina M, Riso P, Guardamagna O, Guglielmetti S. Evidence of dysbiosis in the intestinal microbial ecosystem of children and adolescents with primary hyperlipidemia and the potential role of regular hazelnut intake. FEMS Microbiol Ecol. 2018;94(5):fiy045. doi:10.1093/femsec/fiy045.
  • Johnson AJ, Vangay P, Al-Ghalith GA, Hillmann BM, Ward TL, Shields-Cutler RR, Kim AD, Shmagel AK, Syed AN, Walter J. Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host & Microbe. 2019;25(6):789–802.e5. doi:10.1016/j.chom.2019.05.005.
  • Dhillon J, Li Z, Ortiz RM. Almond snacking for 8 wk increases alpha-diversity of the gastrointestinal microbiome and decreases bacteroides fragilis abundance compared with an isocaloric snack in college freshmen. Curr Dev Nutr. 2019;3(8):nzz079. doi:10.1093/cdn/nzz079.
  • Farhangi MA, Dehghan P, Namazi N. Prebiotic supplementation modulates advanced glycation end-products (AGEs), soluble receptor for AGEs (sRAGE), and cardiometabolic risk factors through improving metabolic endotoxemia: a randomized-controlled clinical trial. Eur J Nutr. 2020;59(7):3009–3021. doi:10.1007/s00394-019-02140-z.
  • Queipo-Ortuño MI, Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM, Clemente-Postigo M, Estruch R, Cardona Diaz F, Andrés-Lacueva C, Tinahones FJ. Influence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical biomarkers. Am J Clin Nutr. 2012;95(6):1323–1334. doi:10.3945/ajcn.111.027847.
  • Peng Q, Huo D, Ma C, Jiang S, Wang L, Zhang J. Monosodium glutamate induces limited modulation in gut microbiota. J Funct Foods. 2018;49:493–500. doi:10.1016/j.jff.2018.09.015.
  • Sun Z, Wang W, Li L, Zhang X, Ning Z, Mayne J, Walker K, Stintzi A, Figeys D. Comprehensive assessment of functional effects of commonly used sweeteners on ex vivo human gut microbiome. Microbiol. Spectr. 2022:10(4):e00412–e00422.
  • Macfarlane G, Allison C, Gibson SAW, Cummings JH. Contribution of the microflora to proteolysis in the human large intestine. J Appl Bacteriol. 1988;64(1):37–46. doi:10.1111/j.1365-2672.1988.tb02427.x.
  • Fernandez-Feo M, Wei G, Blumenkranz G, Dewhirst FE, Schuppan D, Oppenheim FG, Helmerhorst EJ. The cultivable human oral gluten-degrading microbiome and its potential implications in coeliac disease and gluten sensitivity. Clin Microbiol Infect. 2013;19(9):E386–E394. doi:10.1111/1469-0691.12249.
  • Herrán AR, Pérez-Andrés J, Caminero A, Nistal E, Vivas S, Ruiz de Morales JM, Casqueiro J. Gluten-degrading bacteria are present in the human small intestine of healthy volunteers and celiac patients. Res Microbiol. 2017;168(7):673–684. doi:10.1016/j.resmic.2017.04.008.
  • Caminero A, Herrán AR, Nistal E, Pérez-Andrés J, Vaquero L, Vivas S, Ruiz de Morales JMG, Albillos SM, Casqueiro J. Diversity of the cultivable human gut microbiome involved in gluten metabolism: isolation of microorganisms with potential interest for coeliac disease. FEMS Microbiol Ecol. 2014;88(2):309–319. doi:10.1111/1574-6941.12295.
  • Taylor BC, Lejzerowicz F, Poirel M, Shaffer JP, Jiang L, Aksenov A, Litwin N, Humphrey G, Martino C, Miller-Montgomery S. et al. Consumption of fermented foods is associated with systematic differences in the gut microbiome and metabolome. mSystems. 2020;5(2):e00901–19. doi:10.1128/mSystems.00901-19.
  • Wastyk HC, Fragiadakis GK, Perelman D, Dahan D, Merrill BD, Yu FB, Topf M, Gonzalez CG, Van Treuren W, Han S. et al. Gut-microbiota-targeted diets modulate human immune status. Cell. 2021;184(16):4137–4153.e14. doi:10.1016/j.cell.2021.06.019.
  • Sánchez B. Bile acid–microbiota crosstalk in gastrointestinal inflammation and carcinogenesis: a role for bifidobacteria and lactobacilli? Nat Rev Gastro Hepat. 2018;15(4):205–205. doi:10.1038/nrgastro.2018.23.
  • Kumar Singh A, Cabral C, Kumar R, Ganguly R, Kumar Rana H, Gupta A, Rosaria Lauro M, Carbone C, Reis F, Pandey AK. et al. Beneficial effects of dietary polyphenols on gut microbiota and strategies to improve delivery efficiency. Nutrients. 2019;11(9):2216. doi:10.3390/nu11092216.
  • Tuck CJ, Malakar S, Barrett JS, Muir JG, Gibson PR. Naturally‐occurring dietary salicylates in the genesis of functional gastrointestinal symptoms in patients with irritable bowel syndrome: Pilot study. JGH Open. 2021;5(8):871–878. doi:10.1002/jgh3.12578.
  • Bancil AS. et al. Food additive emulsifiers and their impact on gut microbiome, permeability, and inflammation: mechanistic insights in inflammatory bowel disease. J Crohn’s Colitis. 2021;15(6):1068–1079. doi:10.1093/ecco-jcc/jjaa254.
  • Chen H, Xu H, Heinze TM, Cerniglia CE. Decolorization of water and oil-soluble azo dyes by Lactobacillus acidophilus and lactobacillus fermentum. J Ind Microbiol Biotechnol. 2009;36(12):1459. doi:10.1007/s10295-009-0633-9.
  • Bamberger C, Rossmeier A, Lechner K, Wu L, Waldmann E, Fischer S, Stark R, Altenhofer J, Henze K, Parhofer K. et al. A walnut-enriched diet affects gut microbiome in healthy caucasian subjects: a randomized, controlled trial. Nutrients. 2018;10(2):244. doi:10.3390/nu10020244.
  • Mills CE, Tzounis X, Oruna-Concha M-J, Mottram DS, Gibson GR, Spencer JPE. In vitro colonic metabolism of coffee and chlorogenic acid results in selective changes in human faecal microbiota growth. Br J Nutr. 2015;113(8):1220–1227. doi:10.1017/S0007114514003948.
  • Gill SK, Rossi M, Bajka B, Whelan K. Dietary fibre in gastrointestinal health and disease. Nat Rev Gastroenterol Hepatol. 2021;18(2):101–116. doi:10.1038/s41575-020-00375-4.
  • Holscher HD. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes. 2017;8(2):172–184. doi:10.1080/19490976.2017.1290756.
  • Febbraio MA, Karin M. “Sweet death”: fructose as a metabolic toxin that targets the gut-liver axis. Cell Metab. 2021;33(12):2316–2328. doi:10.1016/j.cmet.2021.09.004.
  • Ang QY, Alexander M, Newman JC, Tian Y, Cai J, Upadhyay V, Turnbaugh JA, Verdin E, Hall KD, Leibel RL. et al. Ketogenic diets alter the gut microbiome resulting in decreased intestinal Th17 cells. Cell. 2020;181(6):1263–1275.e16. doi:10.1016/j.cell.2020.04.027.
  • Nogal B, Blumberg JB, Blander G, Jorge M. Gut microbiota–Informed precision nutrition in the generally healthy individual: are we there yet? Curr Dev Nutr. 2021;5(9):nzab107. doi:10.1093/cdn/nzab107.
  • Diether NE, Willing BP. Microbial fermentation of dietary protein: an important factor in diet–microbe–host interaction. Microorganisms. 2019;7(1):19. doi:10.3390/microorganisms7010019.
  • Celis AI, Relman DA. Competitors versus collaborators: micronutrient processing by pathogenic and commensal human-associated gut bacteria. Mol Cell. 2020;78(4):570–576. doi:10.1016/j.molcel.2020.03.032.
  • Barone M, D’Amico F, Brigidi P, Turroni S. Gut microbiome–micronutrient interaction: the key to controlling the bioavailability of minerals and vitamins? BioFactors. 2022;48(2):307–314. doi:10.1002/biof.1835.
  • Ramakrishna BS. Role of the gut microbiota in human nutrition and metabolism. J Gastroen Hepatol. 2013;28(S4):9–17. doi:10.1111/jgh.12294.
  • Naliyadhara N, Kumar A, Kumar Gangwar S, Nair Devanarayanan T, Hegde M, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara A. Interplay of dietary antioxidants and gut microbiome in human health: what has been learnt thus far? J Funct Foods. 2023;100:105365. doi:10.1016/j.jff.2022.105365.
  • Biesalski HK. Nutrition meets the microbiome: micronutrients and the microbiota. Ann NY Acad Sci. 2016;1372(1):53–64. doi:10.1111/nyas.13145.
  • Glowacki RW, Martens EC. If you eat it or secrete it, they will grow: the expanding list of nutrients utilized by human gut bacteria. J Bacteriol. 2020;203(9):e00481–20. doi:10.1128/JB.00481-20.
  • Diaz-Amigo C, Popping B. Accuracy of ELISA detection methods for gluten and reference materials: a realistic assessment. J Agric Food Chem. 2013;61(24):5681–5688. doi:10.1021/jf3046736.
  • Lackner S, Malcher V, Enko D, Mangge H, Holasek SJ, Schnedl WJ. Histamine-reduced diet and increase of serum diamine oxidase correlating to diet compliance in histamine intolerance. Eur J Clin Nutr. 2019;73(1):102–104. doi:10.1038/s41430-018-0260-5.
  • Barrett JS, Gibson PR. Fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs) and nonallergic food intolerance: FODMAPs or food chemicals? Therap Adv Gastroenterol. 2012;5(4):261–268. doi:10.1177/1756283X11436241.
  • Burns G, Pryor J, Holtmann G, Walker MM, Talley NJ, Keely S. Immune activation in functional gastrointestinal disorders. Gastroenterol Hepatol (N Y). 2019;15:539.
  • Lee D, Swan CK, Suskind D, Wahbeh G, Vanamala J, Baldassano RN, Leonard MB, Lampe JW. Children with Crohn’s disease frequently consume select food additives. Dig Dis Sci. 2018;63(10):2722–2728. doi:10.1007/s10620-018-5145-x.
  • Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19(1):55–71. doi:10.1038/s41579-020-0433-9.
  • Leeuwendaal NK, Stanton C, O’Toole PW, Beresford TP. Fermented foods, health and the gut microbiome. Nutrients. 2022;14(7):1527. doi:10.3390/nu14071527.
  • Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, Ben-Yacov O, Lador D, Avnit-Sagi T, Lotan-Pompan M. et al. Personalized nutrition by prediction of glycemic responses. Cell. 2015;163(5):1079–1094. doi:10.1016/j.cell.2015.11.001.
  • Marietta EV, Geno DM, Smyrk TC, Becker A, Alexander JA, Camilleri M, Murray JA, Katzka DA. Presence of intraepithelial food antigen in patients with active eosinophilic oesophagitis. Aliment Pharmacol Ther. 2017;45(3):427–433. doi:10.1111/apt.13877.