Next Article in Journal
Chemical Synthesis and Biological Evaluation of 3-Substituted Estrone/Estradiol Derivatives as 17β-Hydroxysteroid Dehydrogenase Type 1 Inhibitors Acting via a Reverse Orientation of the Natural Substrate Estrone
Previous Article in Journal
Intermolecular Interactions of Edaravone in Aqueous Solutions of Ethaline and Glyceline Inferred from Experiments and Quantum Chemistry Computations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Food Additives on the Abundance and Composition of Gut Microbiota

1
Food Laboratory of Zhongyuan, Beijing Technology and Business University, Beijing 100048, China
2
Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
*
Author to whom correspondence should be addressed.
Molecules 2023, 28(2), 631; https://doi.org/10.3390/molecules28020631
Submission received: 23 November 2022 / Revised: 1 January 2023 / Accepted: 4 January 2023 / Published: 7 January 2023

Abstract

:
The gut microbiota has been confirmed as an important part in human health, and is even take as an ‘organ’. The interaction between the gut microbiota and host intestinal environment plays a key role in digestion, metabolism, immunity, inflammation, and diseases. The dietary component is a major factor that affects the composition and function of gut microbiota. Food additives have been widely used to improve the color, taste, aroma, texture, and nutritional quality of processed food. The increasing variety and quantity of processed food in diets lead to increased frequency and dose of food additives exposure, especially artificial food additives, which has become a concern of consumers. There are studies focusing on the impact of food additives on the gut microbiota, as long-term exposure to food additives could induce changes in the microbes, and the gut microbiota is related to human health and disease. Therefore, the aim of this review is to summarize the interaction between the gut microbiota and food additives.

1. Introduction

Food additives are an important part of modern food industry, as they are used in food to maintain or promote its taste, safety, freshness, appearance, and nutritional quality. There are 22 categories and more than 2000 kinds of additives permitted in China [1], whereas this number in European Union and United States is more than 1500 and 4000, respectively [2,3]. The regulations and rules set by different government organizations are not totally the same. The regulations and rules of food additives used in food are authorized in the Code of Federal Regulations Title 21 set by United States Food and Drug Administration. And a new food additive application must include the basic physicochemical properties and provide the information about safety assessment in human health [2]. For the European Union, food additives are administrated by European Food Safety Authority (EFSA) and the Regulation (EC) No. 1333/2008 of the European Parliament and of the Council [3]. In Canada, the additives used in food are administrated by the List of Permitted Food Additives and Marketing Authorization, whereas in Australia and New Zealand, the Australia New Zealand Food Standard Code—Standard 1.3.1—Food Additives regulates the additives that permitted using in food [2]. All those regulations and rules have limited the food categories and dose which the additives can be used. The category of food additives mainly includes flavor enhancers, antioxidants, preserves, colorants, and sweeteners. The food additives are obtained from natural substances extraction or artificial synthesis. The artificial food additives are widely used in processed food due to their low cost, better stability, and uniformity [4]. In the French market, 53.8% (N = 126, 556) of food products contained at least 1 food additive and 11.3% at least 5 food additives [5]. The safety and toxic characteristics must be evaluated systematically before those additives are used in food, including through in vivo and in vitro trial [6,7,8]. There are studies focusing on the safety qualities of additives used in food, such as artificial sweeteners; the risk of cardiovascular diseases; and the metabolization of Allura red in vivo [9,10]. Azo dyes, including Sunset Yellow, tartrazine, Allura Red, and amaranth, have been reported with developmental toxicity potential in zebrafish embryos [11]. The EFSA Panel on Food Additives and Flavourings [12] has revaluated the safety of phosphoric acid-phosphates as food additive. In an in vitro study, butylated hydroxytoluene (BHT) induced non-apoptotic cell death in rat thymocytes [13] and can potential in enhancing ELT-3 rat leiomyoma cell proliferation [14]. However, most researches focus on the safety properties of artificial food additives, and there have been fewer studies on the effect on gut microbiota characteristics induced by food additives used in processed food. As the function of gut microbiota on host gut health, metabolism, immune, inflammation, and diseases has been studied deeply [15,16,17,18], researchers have raised concerns about the effect of food additives on the gut microbiota. Food additives can affect the gut microbiota such as Firmicutes, Bacteroidetes, Barnesiella, Prevotella, Ruminococcaceae, Bifidobacterium, etc. [19], and those microbiomes were reported to maintain gut barrier, participate in short chain fatty acid (SCFAs) metabolism as well as decomposition of fiber and sugar, and are associated with obesity and diabetes. The question is whether the artificial food additives affect the health and diseases of the host by regulating the gut microbiota.
This project is focused on artificial food additives, including antioxidants, preservatives, sweeteners, flavor enhancers, and colorants. The keyword additives (flavor enhancer, antioxidants, preservative, artificial colorants, and sweeteners), gut microbiota (or intestinal microbiota, or gut flora) were searched in the title, abstract, and keywords that were published in past ten years on the websites Web of Science, ScienceDirect, and PubMed. This review was aimed at the impact of artificial food additives on gut microbiota.

2. Gut Microbiota

2.1. The Function of Gut Microbiota

The gut microbiota is the sum of different kinds of microbes in the gastrointestinal tract, which influence the host’s health through the ability of digestion, metabolism, and excretion. The main phyla of gut microbiota are Bacteroidetes, firmicutes, fusobacteria, proteobacteria, cyanobacteria, verrucomicrobia, and actinobacteria [20]. The gut microbiota functioned in human health has been reported to be associated with the digestion and adsorption of nutrients through fermentation and decomposition of nutrients in the colony [21,22]. The glycoside hydrolase gene representation in gut microbiota is associated with the degradation of fiber and production of SCFAs [23]. The relative abundance of Ruminococcus 2, Faecalibacterium, and Akkermansia was associated with increased neutrophil rates and the total bacterial abundances of Faecalibacterium was associated with neutrophil dynamics, which revealed the function of gut microbiota in the immune system [24]. In the gut-brain axis, the modification of gut microbiota can promote endocrinal cells to release neuropeptides, peptides, and enteric neuroendocrinal factors [25]. Meanwhile, in the gut-liver axis, the proinflammatory markers, iNOS and MMP-2, that released by gut microbiota are enriched due to the alteration of the gut microbe’s environment [26]. The gut microbiota also acts as biomarkers in diseases, such as gout and lung cancer [27,28]. Furthermore, a cohort study in an obese population found that the relative abundance of Prevotella was correlated with Body Mass Index (BMI) [29]. The gut microbiota is also part of the intestinal barrier and contributes to mucosal immunity [30,31]. The microbiota and its metabolites largely impact the intestinal barrier integrity, host metabolism, and pathogen colonization [16]. Fermentation of carbohydrates is a core function of the gut microbiota [32]. In the colon and cecum, the dietary fiber and some resistant starches are fermented by gut microbiota, such as Bifidum, Bifidobacterium, Butyrivibrio, and generated SCFAs [33]. The SCFAs mainly include acetate, propionate, butyrate, and pentanoate, which serve as signaling molecules that activate the G-protein-coupled receptors (GPCRs) and contribute to the formation of an anti-inflammatory environment in the intestinal trial [34]. Vernocchi et al. [28] have found a strong correlation between the presence of SCFAs and the normal healthy gut microbiome. Moreover, the microbial metabolite, urolithin A, derived from microbial transformation, displays serveries activities, including anti-inflammatory, antioxidative, and anti-ageing [35]. The physiologic processes of lipid digestion and absorption are central functions to the jejunum [36]. The bile acids can facilitate the digestion and adsorption of lipids after secretion into the small intestine, and a small portion that enters the colon can return to the liver after microbial biotransformation [37]. In addition, the function of gut microbiota-derived vitamins plays an important role in psychiatric health and disease [38]. Instead, the gut microbiota was associated with diseases regulation, such as type 2 diabetes and obesity. The interaction between the gut microbiota and the regulatory system have been considered to affect human neurophysiology and mental health [39]. Therefore, the microbiota and its metabolites play an important role in host physiology and pathology.

2.2. Effect of Dietary Component on Gut Microbiota

Intestinal microbiomes use dietary components as substrates for fermentation, and the produced metabolites participate in various physiological processes. Normally, the gastrointestinal microbiota is stable and maintains balance in homeostasis with its host [40]. The gut microbiota changes rapidly after dietary ingestion and responds rapidly to the altered diet, those features potentially facilitating the diversity of human dietary lifestyle [41]. The type and component of the food, non-nutritional materials, or the toxic substance that are ingested into the gastrointestinal system can all induce structure and abundance alternation of the gut microbiota in the intestine [23,42]. Therefore, metabolites generated by intestinal microbiota using dietary nutrients as fermentation substrates will be changed [43]. Dietary protein, including protein source [22,44], processing factors [45], and protein oxidation, can affect the dietary protein digestibility on gut microbiota by influencing fermentation, adsorption, and functional properties of protein in the intestine, and finally, can impact the composition of the gut microbiota and thus affect human health [46]. A randomized controlled-feeding trial with overweight adults has found that a fried meat diet lowered the richness of the microbial community and decreased the abundance of Lachnospiraceae and Flavonifractor, accordingly, the lower concentration of butyric acid, valeric acid, and 3-indolepropionic acid, and higher concentration of methylglutaric acid were observed; those metabolites were associated with inflammation levels [45]. Lachnospiraceae can prevent obesity, and the abundance of Flavonifractor is negatively correlated with obesity [45]. The stewed pork protein diet changed the production of SCFAs by increasing Sphingomonadaceae, while decreasing Eggerthellaceae and Lactobacillaceae [22]. Rats fed with hen egg white induced an increase in the Akkermansia relative abundance, while those fed with duck egg white observed a higher relative abundance of Peptostreptococcaceae and Proteobacteria, and decreased in the relative abundance of Lachnospiraceae [47]. Considering the function of those microbes in the intestine, Akkermansia were shown to be responsible for increased intestinal permeability and facilitating pathogen entry [48]. Peptostreptococcaceae has the potential to maintain intestinal homeostasis [47]. Proteobacteria can synthesize both pantothenic acid and coenzyme A (CoA) [38]. While the complex plant material that cannot be digested in the host gut, it can be decomposed by Lachnospiraceae [46]. Xie et al. [22] found that soybean protein was beneficial for the microbial metabolism, but may reduce the intestinal peristalsis of mice when compared with intake casein and processed meat protein. Omnivore and vegan diets (containing fiber) can alter fecal amino acid levels by promoting the growth of Firmicutes for amino acid metabolism [23]. The high-fiber diet increased the Clostridiaceae abundance in the jejunum and ileum of mice, and decreased the abundance of Bacteroidaceae and Bifidobacteriaceae in all intestinal tract [18]. Clostridiaceae can increase the expression of enzyme Dgat2 that is critical for lipid TG synthesis and storage both in in vitro and in vivo studies [18]. The increased abundance of genus Lachnoclostridium and Lactobacillus induced by collagen-induced arthritis (CIA) was eliminated in mice fed a high-fiber diet rich with resistant starch [49]. Lachnoclostridium has a known function in butyrate production [43]. Bifidobacteriaceae and Lactobacillus are two important components of the beneficial bacterial community and in cross-talk with the host through adhering to epithelial cells, colonization in gastrointestinal mucosa, defensing pathogens, and regulating the immune system [50]. Moreover, minerals and vitamins also alter the composition and diversity of gut microbiota, such as selenium [51] and vitamin E [21]. And the express genetic machinery of majority gut microbes enables them to synthesize and metabolize various vitamins, especially B vitamins [38]. Therefore, diet allows to affect the component and abundance of gut microbiota, and the gut microbes also functioned in the digestion and metabolism of dietary ingredients.

2.3. Gut Microbiota and Diseases

The dysbiosis of gut microbiota causes diseases or promotes the development of disease. The gut microbiota has been found with typical composition and abundance in special diseases. Microbial metabolites play an important role in developing obesity, acting as a bridge linking diet and obesity [43]. Obesity was associated with elevated levels of circulating succinate; the relative abundance of veillonellaceae and prevotellaceae, which produce succinate, was higher in obese individuals, while the abundance of clostridaceae and odoribacteraceae, which consume succinate, was lower [52]. Faecalibacterium prausnitzii is a marker of lower visceral fat mass, while Clostridium CAG 58 and Flavonifractor plautii are the taxa that associated with obesity [53]. Gut dysbiosis was observed preceding clinic symptoms in rheumatoid arthritis (RA) and has been implicated in the initiation and persistence of RA [49]. Furthermore, the increased abundances of Lachnospiraceae_NK4A136_group and Bacteroidales_S24-7_group were observed in CIA mice [49]. Gut microbiota can be used as a sensitive biomarker for diagnosing gout, since Bacteroides xylanisolvens and Bacteroides caccae are enriched while Bifidobacterium pseudocatenulatum and Faecalibacterium prausnitzii are depleted in diseased individuals [54]. In addition, the gut microbiota promotes catabolic metabolism of purines and uric acid, and its metabolites increase uric acid excretion [55]. Compared to healthy controls, the relative abundances of Bacteroides, Prevotella, and Fusobacterium were increased in gout patients, whereas Enterobacteriaceae and butyrate-producing species were decreased [27]. The gut microbiota and its metabolites regulate the gene expression in liver, which can induce imbalance in the gut-liver axis and may cause disease, including carcinogenesis [15]. Changes in the microbiomes in the gut were found to be related with breast cancer [56]. Enterococcus faecalis levels were associated with variants at the MED13L locus that linked to colorectal cancer [57]. The gut microbiota in non-small cell lung cancer (NSCLC) patients was imbalanced, wherein the abundance of Akkermansia muciniphila, Bacteroides, Rikenellaceae, Mogibacteriaceae, Clostridiaceae, and Peptostreptococcaceae were found lower than controls [28]. In the event of the COVID-19 pandemic, recent research has found that gut microbiota-mediated amino acids, sugar metabolites, and neurotransmitters are involved in multiple cytokine dynamics in COVID-19 [58]. Those reveal the role of microbes in the gut-lung axis. The disordered gut microbiota will disrupt the gut immune system and induce diseases, such as inflammatory bowel diseases (IBDs), ulcerative colitis (UC), and Crohn’s disease (CD) [17]. The previous study observed that Proteobacteria and Bacteroidetes were significantly increased in CD patients when compared to healthy subjects; indeed, the diversity of Bacteroidetes was far less than Firmicutes in CD patients [59]. Zhen et al. [60] found increased relative abundance of the Bacteroidetes phylum and decreased Firmicutes phylum in the gut of ABX mice, thus leading to a disorder of SCFAs metabolism. Moreover, recognition and working memory can be influenced by Lactobacillus acidophilus, Lactobacillus fermentum, Bifidobacteria, Bifidobacterium bifidum, Lactobacillus casei, and C. difficile, through the gut-brain axis in Alzheimer’s disease [61]. Trimethylamine-N-Oxide produced by Proteobacteria metabolization in the gut is implicated in developing Acute Coronary Syndrome (ACS) [62]. Based on those results, the gut microbiota was proved to be associated with the host’s diseases.

3. Effects of Food Additives on Gut Microbiota

3.1. Antioxidants

The food additives in one’s diet do affect the composition and abundance of the gut microbiota in the intestine. The food additives and their association with gut microbiomes summarized in this project are presented in Table 1.
Antioxidants are a kind of food additive that can be used in foodstuff with regulated amounts to avoid oxidation of food products and improve the storage duration [84]. The antioxidants include natural antioxidants (e.g., tocopherols) and synthetic antioxidants (e.g., phenolic antioxidants); these antioxidants can prevent free radicals chain reactions of oxidation [85]. Antioxidants are commonly used in the food processing industry, especially in edible oil and fat; thus, oil and fat are widely used as materials in different kinds of processed food.
A survey about the synthetic phenolic antioxidants (SPAs) in foodstuffs from ten provinces in China found that more than 99% samples detected at least one of the SPAs, the first three common SPAs being BHT, BHT-Q, and butylated hydroxyanisole (BHA), which totally accounted for 83.2% of total SPAs contents in thirteen food categories (N = 289) [86]. Although the antioxidants were considered safe within moderate amounts, the consumers were worried about the health effect induced by antioxidants added in food [87]. An in vitro study has evaluated the susceptibility of human gut microbes to phenolic compounds. Natural phenolic compounds (such as eugenol, ferulic acid, and vanillin) decreased the growth of Agathobacter and Clostridium strains, and the Bacteroidetes and Actinobacteria strains were mostly not susceptible to phenolics [88]. However, the effect of synthetic antioxidants on the gut microbiota still needs to be studied.

3.2. Preservatives

Food preservatives are used to ensure safety and prevent quality loss derived from physical-chemical, microbial, or enzymatic reaction [89]. Some of the preservatives are also active as antioxidants, such as sulfur dioxide, sodium metabisulphite, sodium sulfite, and potassium sorbate [1]. In this project, synthetic preservatives were of concern, including sodium benzoate, benzoic acid, ethylparaben, sodium nitrite, nitrite, sodium sulphite, and potassium sorbate.
An in vivo study was done in pigs fed with a benzoic acid-supplemented nursery diet. The transition of the bacterial community was mainly driven by the decreased abundance of the genus of Prevotella and the phylum of Bacteroidetes [65]. The abundance of Fusicatenibacter, Ruminococcus, and Escherichia-Shigella in pigs fed with a diet containing 90% benzoic acid and 10% essential oil (include thymol, 2-meth-oxyphenol, and eugenol) were significantly (p < 0.05) increased compared to control (without additive), while Prevotella, and Coprococcus 1 were significantly decreased [66]. In another piglet trial, 49% benzoic acid supplementation diet was observed with higher abundance of Ruminococcus (False Discovery Rate, FDR < 0.01), Fibrobacteraceae (FDR < 0.05), and Prevotellaceae (FDR < 0.01), bacteria which were confirmed with certain fiber fermenting abilities [67]. However, there is also research that found no significant difference of benzoic acid supplementation on pig jejunum and cecum microbial populations [90]. Meanwhile, the gut microbiota of wild-type C57BL/6 mice (male) fed with sodium benzoate-supplemented diet for 8 weeks was studied, and a significant decrease was observed in the Coriobacteriaceae family, which can convert carbohydrates to acetic acid and lactic acid in mice [63]. Lastly, in human volunteers, sodium benzoate promoted the growth of Bifidobacterium [69].
Xu et al. [68] found that both low dose nitrite (0.15 g/L) and high dose nitrite (0.30 g/L) could significantly upregulate α-diversity in C57BL/6 mice on day 120. The result of α-diversity includes the increase of Chao 1 and Shannon index, which revealed that the total number of operational taxonomic units (OTUs) is increased and the diversity is higher. In addition, the markedly different genera were higher in day 120 than in day 70. The low dose nitrite–treated mice uniquely upregulated the abundances of Alloprevotella, Coprococcus, Acetatifactor, and Falsiporphyromonas, while downregulated the abundances of Elusimicrobium, and Akkermansia. Those results revealed that long-term exposure to nitrite significantly alters the abundance of gut microbiota in C57BL/6 mice [68]. Akkermansia was reported as a next-generation beneficial microbe, which is negatively associated with obesity, diabetes, cardiometabolic diseases, and low-grade inflammation [91,92]. In a dextran sodium sulfate (DSS)–induced mouse model, genus level of Prevotellaceae_UCG-001, Ruminococcaceae_UCG-014, and Lactobacillus were increased in NaNO3 treated (2 mM in drinking water, 5 days) mouse; moreover, the enriched metabolic pathways of p53 signaling and colorectal cancer was partially decreased [71].
In an in vitro study, the human gut microbes were found to be highly susceptible to sodium nitrite, sodium benzoate, and potassium sorbate, especially, Clostridium tyrobutyricum or Lactobacillus paracasei, which have known anti-inflammatory properties, were significantly more susceptible to those three preservatives than Enterococcus faecalis or Bacteroides thetaiotaomicron that have known pro-inflammatory or colitogenic properties [64]. Potassium sorbate can significantly decrease the Coriobacteriaceae family, which can convert carbohydrates to acetic acid and lactic acid in mice [63]. Compared to control (sulfite free media), substantial decrease of Rhamnosus, Lactobacillus species casei, Streptococcus thermophilus, and Plantarum were observed in media containing concentrations of sulfites between 250 and 500 mg/L after being exposed to in vitro bacterial culture for two hours [70]. In a human volunteer’s trial, the propionic acid was found to increase while acetic acid decreased with the presence of sodium sulfite; indeed, the result of Shannon α-diversity showed that the addition of sodium sulfite increased the abundance of Escherichia/Shigella. In addition, sodium sulfite had an inhibitory effect on the growth of Bifidobacterium [69]. In wild C7BL/6 mice, ethylparaben showed significantly (p = 0.0424) hyperglycemic, and the relative abundance of Proteobacteria was enriched by ethylparaben compared to the control group [63].

3.3. Flavor Enhancers

Flavor enhancers are multiple substances used in food to promote taste, especially umami. Amino acids and nucleotides are flavor enhancers in common use, among which monosodium glutamate (MSG) is most widely used in processed food and is presented in this section. In addition, novel umami agents, such as protein hydrolysate and umami peptides [93,94], attract increasing attention and have the potential to become new flavor enhancers. However, the effect of flavor enhancers on gut microbiota is mainly focused on MSG, and relevant experimental data for those novel umami agents are still lacking.
The most commonly used flavor enhancer is monosodium glutamate (MSG, C5H8NO4Na), whose chemical structure is sodium salt from glutamic acid. Xu et al. [72] have studied the intestinal structure and the intestinal microbiota with MSG oral gavage to mice. The ratios of Bacteroidetes and Firmicutes in the 30 mg/kg (L-MSG) group were lower than those in the 300 mg/kg (M-MSG) and 1500 mg/kg (H-MSG) groups. Additionally, compared with the control group, the proteobacteria decreased in H-MSG group, but increased in M-MSG group. On the other hand, Peng et al. [73] have observed that MSG did not significantly alter the community structure and functional features of gut microbiota in human volunteers during a four-week experiment with 2 g MSG per day. Although some bacteria including Megamonas, Faecalibacterium, Collinsella, and Blautia tended to change, there was no significant difference in the alteration of all genera. At the functional level, the microbial functions were rich, mainly distributed in membrane transport, amino acid metabolism, and carbohydrate metabolism, but there was no significant difference between samples obtained at different times.

3.4. Sweeteners

Sweeteners are closely related to food flavor and human health, as consumers are more and more considering the health problems both certainly and potentially related to sugars. A prospective NutriNet-Santé cohort (103,388 participants) suggested that artificial sweeteners might represent a modifiable risk factor for cardiovascular disease prevention [10]. The effect of artificial sweeteners, acesulfame-K, aspartame, saccharin, sucralose, cyclamate, and neotame, on gut microbiota has been reviewed by Cao et al. [95], whereby those sweeteners could cause gut dysbiosis, which could lead to impaired glucose metabolism in rodents. Similar results were also reviewed by Ruiz-Ojeda et al. [96]. Gultekin et al. [97] have summarized that acesulfame-K, aspartame, saccharin, and sucralose are likely to destroy glucose tolerance and support weight gain by negatively affecting microbiota. Sugar alcohols are a group of polyols which are produced from sugars and are less digestible since they are difficult to totally digest in small intestine; therefore, some of them can be fermented in the colon [97]. The polyols can be used in sugar free food, since they do not induce salivation and do not interfere with the glucose levels in blood [98]. In a previous review, sugar alcohol was known to increase the number of bifidobacteria in the microbiomes and can induce dose-dependent flatulence in the colon [96]. Studies on the effect of sugar alcohol on the gut microbiota have been conducted within the last ten years. In this section, xylitol, sorbitol, erythritol, and lactitol are evaluated.
There are some in vivo data about the effect of xylitol on the gut microbiota in the intestine. Due to it characteristic of being less digestible in the intestine, the specific experiments on high-fat diet with xylitol supplement were evaluated in mice. Compared to the high-fat diet mice, the relative abundances of Proteobacteria, Bacteroidetes, and Actinobacteria were decreased, while the relative abundances of Firmicutes and ratio of Firmicutes/Bacteroidetes were increased in C57BL/6 mice that fed with high-fat diet supplemented with 10 g/L xylitol [74]. In addition, Uebanso et al. [75] gave a high-fat diet with 194 ± 25 mg/kg b.w. supplement of xylitol to C57BL/6J mice and found that the Bacteroidetes phylum and genus Barnesiella abundance were reduced, while the abundance of Firmicutes phylum and genus Prevotella were increased. Altered gut microbiota composition was present in the rats fed with 10% xylitol for 15 days, wherein the genera Ruminococcaceae and Prevotella was significant decreased, while Bacteroides was notably increased [76]. The results above showed similar changes of gut microbiota after the xylitol intake from feed. It has been reported that xylitol consumption by mice showed positive effect on the metabolic activity of a number of gut microbial populations [99]. However, in an in vitro single-phase continuous fermentation model, the gut microbiota composition was found differentiated after xylitol supplementation (1.67 g/L) only for the first 3 days; additionally, xylitol significantly enhanced the relative amount of Clostridium and Phascolarctobacterium, which act as butyrate synthesizing bacteria [77]. Meanwhile, xylitol has increased the production of butyrate and propionic acid. The same result was reported by Yue et al. [100] that xylitol produced mainly butyrate, which may play a major role in improving gut barrier function. The population sizes of Escherichia were increased beyond expectation after xylitol supplementation [77]. On the contrary, Xiang et al. [101] observed no significant of xylitol on the composition of gut microbiota both in vivo and in vitro, but observed the increasing contents of all SCFAs. This may be induced by key enzymes (xylulokinase, xylitol dehydrogenase, and xylulose phosphate isomerase) in xylitol metabolism which present in Bacteroides and Lachnospiraceae metabolites [101].
For long-term intake of sorbitol, Li et al. [78] found that the relative abundances of Bifidobacterium, Lachnospiraceae NK4A136, Lachnospiraceae UCG 001, Candidatus Arthromitus, Eubacterium ventriosum, and Ruminococcus torques were significantly decreased, while the relative abundances of Tyzzerella, Helicobacter, Prevotella 9, and Alistipes were increased in mice. An in vitro growth assay using no carbon-defined media with sugar alcohols supplement showed that Clostridia and Erysipelotrichia were isolated only in sorbitol as a carbon source [102]. Furthermore, Hattori et al. [79] found that the gut microbiota showed a positive impact on sorbitol-induced diarrhea; treatment with sorbitol resulted in the greatest increase at genus level of the abundance of Klebsiella, Escherichia, Proteus, and Enterobacter in the family Enterobacteriaceae. Those results revealed that sugar alcohols are a major carbon source for the fermentation of gut microbiota.
Erythritol (E968) was proposed as a food additive by EFSA in 2015 [103]. Ninety percent of erythritol is absorbed in the small intestine, and ten percent enters the colon, and the in vitro trial found that no consistent disruption in the α-diversity was observed in human gut community [104]. In participants (diabetic and non-diabetic patients) with lactitol administration for two weeks, the abundance of Actinobacteria, Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, and Bifidobacterium were found with an increasing trend [80]. Moreover, an in vitro colonic fermentation study observed that fermentation of lactitol produced mainly acetate [100]. This may result in gut microbiota that metabolize SCFAs.

3.5. Colorants

The synthetic food colorants used by food manufacturers have been increasing due to their low cost, better stability, high color intensity, and uniformity [4]. The food safety management of government and non-government organizations have strictly defined the range and dosage of using colorants. The synthetic colorants, including tartrazine, Sunset Yellow FCF, ponceau 4R, Allura Red AC, quinoline yellow, and carmoisine, have been reported associated with hyperactivity in children [25]. Another colorant, titanium dioxide, is forbidden for use in food in the European Union [105]. However, those additives were permitted for use in specific food categories with limited doses. This section evaluates the information about artificial colorants that are used in processed food with their effect on the gut microbiota.
Tartrazine exposure induced gut microbiota dysbiosis in the juvenile crucian carp fish (Carassius carassius) [106]. In an in vitro trial, Escherichia coli, Enterococcus faecium, Aerococcus viridans, and Bacillus cereus can decolorize Sunset Yellow, and tartrazine after 30 min contact, which means those microbiomes have azoreductase activity [82]. In animal studies, ponceau 4R was found merely absorbed in the digestive tract, where it is anaerobically reduced by microflora, with small levels of the resulting metabolites systemically absorbed [107]. Allura Red AC has been reported to induce colitis in the context of dysregulated interleukin -23 [108]. An in vivo challenge of primed mice with Red 40 (Allura Red AC) promoted rapid activation of CD4+ T cells [109], while in CD4+ T cells, the gut microbiota-reactive interleukin -17-producing Th17 cells are central to the pathogenesis of certain types of IBD [110]. The results presented that Allura Red AC can induce inflammation of intestine by regulating the immune cell secretion. At phylum level, the proportion of Verrucomicrobia after oral administration of micro-TiO2 (10, 40, 160 mg/kg bw) was significantly lower than that in the control group (p < 0.05), and the proportion of Bacteroidetes at 10 mg/kg group decreased to 28.20%, while that of Firmicutes increased significantly to 70.23% (p < 0.05) [81].

3.6. Other Food Additives

There are several artificial food additives which are not included above, such as emulsifiers carboxymethylcellulose, polysorbate 80, resistant starch, sodium stearoyl lactylate, maltodextrin, and carboxymethyl cellulose. Those food additives are evaluated in this section.
Emulsifiers, carboxymethylcellulose, and polysorbate 80 (P80) develop dysbiosis with overgrowth of mucus-degrading bacteria, as well as further deficiency in interleukin-10 or toll-like receptor 5 [111]. However, the emulsifiers used to maintain food-specific properties may increase the translocation of pathogenic microbes in the intestinal epithelial barrier and cause the initiation of intestinal inflammation and consequently cause the increase in the incidence of inflammatory bowel disease [30]. Maltodextrin and carboxymethyl cellulose induced the decreasing of α-diversity, and both decrease in acetic acid levels, whereas the lower acetic acid levels were correlated with higher Akkermansia abundance and lower abundance of Bacteroides and Streptococcus [112]. The increased Lachnoclostridium and Lactobacillus genera abundance concomitant with CIA were eliminated by a resistant starch-high fat diet. Notably, resistant starch supplement also led to a predominance of Bacteroidetes, and increased the abundances of Bacteroidales_S24-7_group and Lachnospiraceae_NK4A136_group genera in CIA mice [49]. The effect of sodium stearoyl lactylate (SSL) on fecal microbiota was studied in vitro, wherein 0.025% (w/v) of SSL was found to reduce the relative abundance of the Clostridia class. The relative abundance of the families Lachnospiraceae, Ruminococcaceae, and Clostridiaceae was substantially reduced, whereas that of Bacteroidaceae and Enterobacteriaceae, Desulfovibrionaceae was increased. The genome reconstruction analysis found that SSL significantly reduced concentrations of butyrate and increased concentrations of propionate compared to control cultures [83].

4. Summary

As summarized from the literatures, food additives can induce the alteration of the gut microbiota in the host intestine. The in vivo animal model and in vitro bacteria culture were used to evaluate the effect of food additives on the gut microbiota. The association between artificial antioxidants and gut microbiota was minor; conversely, the natural antioxidants attract more attention. Preservatives highly showed association with the microbes with SCFAs generation. Monosodium glutamate presented a positive (or no difference) effect on the composition of gut microbiota. The sugar alcohol sweeteners can be used as fermentation of gut microbiota, and most polyols will alter the composition of gut microbiota. However, the definite impact of food additives on gut microbiota is not illustrated clearly, even though various studies have been reviewed. Different food additives lead to different effects on the same phylum or genus of gut microbiota. It is difficult to summarize what kind of changes will be induced by those food additives.
Even though there are several studies on the additives that affect the gut microbiota, the knowledge about the effects on gut microbiota induced by multiple artificial, especially synthetic, additives are not sufficient. The systematic studies about the effects and functions of artificial antioxidants and synthetic colorants on gut microbiota are few. Therefore, those food additives should be studied further.

Author Contributions

X.Z., resources, methodology, investigation, writing—original draft preparation, review and editing; K.Q., resources, review and editing; H.W., resources; Y.Z., funding acquisition, project administration, review. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (No. 32122069), and Beijing Outstanding Young Scientist Program (No. BJJWZYJH01201910011025).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. CFDA&NHC. National Food Safty Standard for Uses of Food Additives; National Health Commission of the People’s Repulic of China: Beijing, China, 2014.
  2. China National Center for Food Safety Risk Assessment. Comparison and Analysis of National and International Food Additive Regulations and Standards; Zhang, J.Y., Zhang, J.Y., Wang, H.L., Eds.; China Quality and Standards Publishing & Media Co., Ltd: Beijing, China, 2019; p. 139. (In Chinese) [Google Scholar]
  3. Commission Regulation (EU), Regulation (EC) No 1333/2008 of the European Parliament and of the Council of 16 December 2008 on Food Additives. Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A02008R1333-20221031&qid=1672995237893 (accessed on 30 October 2022).
  4. Dey, S.; Nagababu, B.H. Applications of food color and bio-preservatives in the food and its effect on the human health. Food Chem. Adv. 2022, 1, 100019. [Google Scholar] [CrossRef]
  5. Chazelas, E.; Deschasaux, M.; Srour, B.; Kesse-Guyot, E.; Julia, C.; Alles, B.; Druesne-Pecollo, N.; Galan, P.; Hercberg, S.; Latino-Martel, P.; et al. Food additives: Distribution and co-occurrence in 126,000 food products of the French market. Sci. Rep. 2020, 10, 3980. [Google Scholar] [CrossRef] [Green Version]
  6. Xu, B. Safety Evaluation and Management of Food Additives by FAO/WHO. Adv. Mater. Res. 2013, 726–731, 812–815. [Google Scholar] [CrossRef]
  7. Leontopoulou, E. Re-evaluation program of food additives in the EU: Looking towards 2020. NutraCos 2018, 17, 30–33. [Google Scholar]
  8. EFSA. Food additives and nutrient sources added to food: Developments since the creation of EFSA. EFSA J. 2012, 10, s1006. [Google Scholar]
  9. Li, K.; Xia, Y.; Ma, G.; Zhao, Y.; Pidatala, V.R. New LC-MS/MS Method for the Analysis of Allura Red Level in Takeaway Chinese Dishes and Urine of an Adult Chinese Population. J. Agric. Food Chem. 2017, 65, 2588–2593. [Google Scholar] [CrossRef] [PubMed]
  10. Debras, C.; Chazelas, E.; Sellem, L.; Porcher, R.; Druesne-Pecollo, N.; Esseddik, Y.; de Edelenyi, F.S.; Agaësse, C.; De Sa, A.; Lutchia, R.; et al. Artificial sweeteners and risk of cardiovascular diseases: Results from the prospective NutriNet-Santé cohort. BMJ 2022, 378, e071204. [Google Scholar] [CrossRef]
  11. Jiang, L.-L.; Li, K.; Yan, D.-L.; Yang, M.-F.; Ma, L.; Xie, L.-Z. Toxicity assessment of 4 Azo Dyes in Zebrafish Embryos. Int. J. Toxicol. 2020, 39, 115–123. [Google Scholar] [CrossRef] [PubMed]
  12. EFSA FAF Panel (EFSA Panel on Food Additives and Flavourings); Younes, M.; Aquilina, G.; Castle, L.; Engel, K.-H.; Fowler, P.; Frutos Fernandez, M.J.; Fürst, P.; Gürtler, R.; Husøy, T.; et al. Scientific Opinion on the re-evaluation of phosphoric acid–phosphates—di-, tri- and polyphosphates (E 338–341, E 343, E 450–452) as food additives and the safety of proposed extension of use. EFSA J. 2019, 17, 5674. [Google Scholar]
  13. Mizobuchi, M.; Ishidoh, K.; Kamemura, N. A comparison of cell death mechanisms of antioxidants, butylated hydroxyanisole and butylated hydroxytoluene. Drug Chem. Toxicol. 2022, 45, 1899–1906. [Google Scholar] [CrossRef]
  14. Chiang, Y.-F.; Chen, H.-Y.; Ali, M.; Shieh, T.-M.; Huang, Y.-J.; Wang, K.-L.; Chang, H.-Y.; Huang, T.-C.; Hong, Y.-H.; Hsia, S.-M. The Role of Cell Proliferation and Extracellular Matrix Accumulation Induced by Food Additive Butylated Hydroxytoluene in Uterine Leiomyoma. Nutrients 2021, 13, 3074. [Google Scholar] [CrossRef] [PubMed]
  15. Silveira, M.A.D.; Bilodeau, S.; Greten, T.F.; Wang, X.W.; Trinchieri, G. The gut–liver axis: Host microbiota interactions shape hepatocarcinogenesis. Trends Cancer 2022, 8, 583–597. [Google Scholar] [CrossRef] [PubMed]
  16. Ghosh, S.; Whitley, C.S.; Haribabu, B.; Jala, V.R. Regulation of intestinal barrier function by microbial metabolites. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 1463–1482. [Google Scholar] [CrossRef]
  17. Shi, N.; Li, N.; Duan, X.; Niu, H. Interaction between the gut microbiome and mucosal immune system. Mil. Med. Res. 2017, 4, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Martinez-Guryn, K.; Hubert, N.; Frazier, K.; Urlass, S.; Musch, M.W.; Ojeda, P.; Pierre, J.F.; Miyoshi, J.; Sontag, T.J.; Cham, C.M.; et al. Small intestine microbiota regulate host digestive and absorptive adaptive responses to dietary lipids. Cell Host Microbe 2018, 23, 458–469.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Liu, C.; Zhan, S.; Tian, Z.; Li, N.; Li, T.; Wu, D.; Zeng, Z.; Zhuang, X. Food additives associated with gut microbiota alterations in inflammatory bowel disease: Friends or enemies? Nutrients 2022, 14, 3049. [Google Scholar] [CrossRef]
  20. Inan-Eroglu, E.; Ayaz, A. Effects of food additives on gut microbiota: Friend or foe? Nutr. Food Sci. 2019, 49, 955–964. [Google Scholar] [CrossRef]
  21. Liu, K.Y.; Nakatsu, C.H.; Jones-Hall, Y.; Kozik, A.; Jiang, Q. Vitamin E alpha- and gamma-tocopherol mitigate colitis, protect intestinal barrier function and modulate the gut microbiota in mice. Free Radic. Biol. Med. 2021, 163, 180–189. [Google Scholar] [CrossRef]
  22. Xie, Y.; Wang, C.; Zhao, D.; Zhou, G.; Li, C. Processing method altered mouse intestinal morphology and microbial composition by affecting digestion of meat proteins. Front. Microbiol. 2020, 11, 511. [Google Scholar] [CrossRef]
  23. Tanes, C.; Bittinger, K.; Gao, Y.; Friedman, E.S.; Nessel, L.; Paladhi, U.R.; Chau, L.; Panfen, E.; Fischbach, M.A.; Braun, J.; et al. Role of dietary fiber in the recovery of the human gut microbiome and its metabolome. Cell Host Microbe 2021, 29, 394–407.e5. [Google Scholar] [CrossRef]
  24. Schluter, J.P.J.; Taylor, B.P.; Markey, K.A.; Smith, M.; Taur, Y.; Niehus, R.; Staffas, A.; Dai, A.; Fontana, E.; Amoretti, L.A.; et al. The gut microbiota is associated with immune cell dynamics in humans. Nature 2020, 588, 303–307. [Google Scholar] [CrossRef]
  25. Abiega-Franyutti, P.; Freyre-Fonseca, V. Chronic consumption of food-additives lead to changes via microbiota gut-brain axis. Toxicology 2021, 464, 153001. [Google Scholar] [CrossRef] [PubMed]
  26. Bian, X.; Chi, L.; Gao, B.; Tu, P.; Ru, H.; Lu, K. Gut microbiome response to sucralose and its potential role in inducing liver inflammation in mice. Front. Physiol. 2017, 8, 487. [Google Scholar] [CrossRef] [PubMed]
  27. Chu, Y.; Sun, S.; Huang, Y.; Gao, Q.; Xie, X.; Wang, P.; Li, J.; Liang, L.; He, X.; Jiang, Y.; et al. Metagenomic analysis revealed the potential role of gut microbiome in gout. NPJ Biofilms Microb. 2021, 7, 66. [Google Scholar] [CrossRef]
  28. Vernocchi, P.; Gili, T.; Conte, F.; Del Chierico, F.; Conta, G.; Miccheli, A.; Botticelli, A.; Paci, P.; Caldarelli, G.; Nuti, M.; et al. Network analysis of gut microbiome and metabolome to discover microbiota-Linked biomarkers in patients affected by non-small cell lung cancer. Int. J. Mol. Sci. 2020, 21, 8730. [Google Scholar] [CrossRef]
  29. Stanislawski, M.A.; Dabelea, D.; Lange, L.A.; Wagner, B.D.; Lozupone, C.A. Gut microbiota phenotypes of obesity. NPJ Biofilms Microbiomes 2019, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  30. Glade, M.J.; Meguid, M.M. Dietary emulsifiers, the human intestinal mucus and microbiome, and dietary fiber. Nutrition 2016, 32, 609–614. [Google Scholar] [CrossRef]
  31. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef] [PubMed]
  32. Kastl, A.J.; Terry, N.A.; Wu, G.D.; Albenberg, L.G. The structure and function of the human small intestinal microbiota: Current understanding and future directions. Cell. Mol. Gastroenterol. Hepatol. 2020, 9, 33–45. [Google Scholar] [CrossRef] [Green Version]
  33. de Clercq, N.C.; Frissen, M.N.; Groen, A.K.; Nieuwdorp, M. Gut microbiota and the gut-brain axis: New insights in the pathophysiology of metabolic syndrome. Psychosom. Med. 2017, 79, 874–879. [Google Scholar] [CrossRef]
  34. Nicolas, G.R.; Chang, P.V. Deciphering the chemical lexicon of host-gut microbiota interactions. Trends Pharmacol. Sci. 2019, 40, 430–445. [Google Scholar] [CrossRef] [Green Version]
  35. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Volk, N.; Lacy, B. Anatomy and physiology of the small bowel. Gastrointest. Endosc. Clin. N. Am. 2017, 27, 1–13. [Google Scholar] [CrossRef] [PubMed]
  37. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Rudzki, L.; Stone, T.W.; Maes, M.; Misiak, B.; Samochowiec, J.; Szulc, A. Gut microbiota-derived vitamins—Underrated powers of a multipotent ally in psychiatric health and disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 107, 110240. [Google Scholar] [CrossRef]
  39. Shoubridge, A.P.; Choo, J.M.; Martin, A.M.; Keating, D.J.; Wong, M.L.; Licinio, J.; Rogers, G.B. The gut microbiome and mental health: Advances in research and emerging priorities. Mol. Psychiatry 2022, 27, 1908–1919. [Google Scholar] [CrossRef]
  40. Krga, I.; Glibetic, M. 2.16—Gut Microbiota in Health and Diseases. In Comprehensive Gut Microbiota; Glibetic, M., Ed.; Elsevier: Oxford, UK, 2022; pp. 182–198. [Google Scholar]
  41. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
  42. Roca-Saavedra, P.; Mendez-Vilabrille, V.; Miranda, J.M.; Nebot, C.; Cardelle-Cobas, A.; Franco, C.M.; Cepeda, A. Food additives, contaminants and other minor components: Effects on human gut microbiota-a review. J. Physiol. Biochem. 2018, 74, 69–83. [Google Scholar] [CrossRef]
  43. Zhou, M.; Johnston, L.J.; Wu, C.; Ma, X. Gut microbiota and its metabolites: Bridge of dietary nutrients and obesity-related diseases. Crit. Rev. Food Sci. Nutr. 2021, 1–18. [Google Scholar] [CrossRef]
  44. Zhao, J.; Zhang, X.; Liu, H.; Brown, A.M.; Qiao, S. Dietary protein and gut microbiota composition and function. Curr. Protein Pept. Sci. 2019, 20, 145–154. [Google Scholar] [CrossRef]
  45. Gao, J.; Guo, X.Y.; Wei, W.; Li, R.; Hu, K.; Liu, X.; Jiang, W.B.; Liu, S.Y.; Wang, W.Q.; Sun, H.; et al. The association of fried meat consumption with the gut microbiota and fecal metabolites and its impact on glucose homoeostasis, intestinal endotoxin levels, and systemic inflammation: A randomized controlled-feeding trial. Diabetes Care 2021, 44, 1970–1979. [Google Scholar] [CrossRef]
  46. Wu, S.; Bhat, Z.F.; Gounder, R.S.; Mohamed Ahmed, I.A.; Al-Juhaimi, F.Y.; Ding, Y.; Bekhit, A.E.D.A. Effect of dietary protein and processing on gut microbiota—A systematic review. Nutrients 2022, 14, 453. [Google Scholar] [CrossRef]
  47. Yu, H.; Qiu, N.; Meng, Y.; Keast, R. A comparative study of the modulation of the gut microbiota in rats by dietary intervention with different sources of egg-white proteins. J. Sci. Food Agric. 2020, 100, 3622–3629. [Google Scholar] [CrossRef] [PubMed]
  48. Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  49. Bai, Y.; Li, Y.; Marion, T.; Tong, Y.; Zaiss, M.M.; Tang, Z.; Zhang, Q.; Liu, Y.; Luo, Y. Resistant starch intake alleviates collagen-induced arthritis in mice by modulating gut microbiota and promoting concomitant propionate production. J. Autoimmun. 2021, 116, 102564. [Google Scholar] [CrossRef] [PubMed]
  50. Fan, Z.; Yang, B.; Paul Ross, R.; Stanton, C.; Zhang, F.; Sun, J.; Zhao, J.; Zhang, H.; Chen, W. Propionate restores disturbed gut microbiota induced by methotrexate in Rheumatoid Arthritis: From clinic to experiments. J. King Saud Univ. Sci. 2021, 33, 101545. [Google Scholar] [CrossRef]
  51. Zhai, Q.; Cen, S.; Li, P.; Tian, F.; Zhao, J.; Zhang, H.; Chen, W. Effects of Dietary Selenium Supplementation on Intestinal Barrier and Immune Responses Associated with Its Modulation of Gut Microbiota. Environ. Sci. Technol. Lett. 2018, 5, 724–730. [Google Scholar] [CrossRef]
  52. Serena, C.; Ceperuelo-Mallafre, V.; Keiran, N.; Queipo-Ortuno, M.I.; Bernal, R.; Gomez-Huelgas, R.; Urpi-Sarda, M.; Sabater, M.; Perez-Brocal, V.; Andres-Lacueva, C.; et al. Elevated circulating levels of succinate in human obesity are linked to specific gut microbiota. ISME J. 2018, 12, 1642–1657. [Google Scholar] [CrossRef] [Green Version]
  53. Asnicar, F.; Berry, S.E.; Valdes, A.M.; Nguyen, L.H.; Piccinno, G.; Drew, D.A.; Leeming, E.; Gibson, R.; Le Roy, C.; Khatib, H.A.; et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat. Med. 2021, 27, 321–332. [Google Scholar] [CrossRef] [PubMed]
  54. Guo, Z.; Zhang, J.; Wang, Z.; Ang, K.Y.; Huang, S.; Hou, Q.; Su, X.; Qiao, J.; Zheng, Y.; Wang, L.; et al. Intestinal microbiota distinguish gout patients from healthy humans. Sci. Rep. 2016, 6, 20602. [Google Scholar] [CrossRef] [Green Version]
  55. Wang, J.; Chen, Y.; Zhong, H.; Chen, F.; Regenstein, J.; Hu, X.; Cai, L.; Feng, F. The gut microbiota as a target to control hyperuricemia pathogenesis: Potential mechanisms and therapeutic strategies. Crit. Rev. Food Sci. Nutr. 2021, 62, 3979–3989. [Google Scholar] [CrossRef]
  56. Plaza-Diaz, J.; Alvarez-Mercado, A.I.; Ruiz-Marin, C.M.; Reina-Perez, I.; Perez-Alonso, A.J.; Sanchez-Andujar, M.B.; Torne, P.; Gallart-Aragon, T.; Sanchez-Barron, M.T.; Reyes Lartategui, S.; et al. Association of breast and gut microbiota dysbiosis and the risk of breast cancer: A case-control clinical study. BMC Cancer 2019, 19, 495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Qin, Y.; Havulinna, A.S.; Liu, Y.; Jousilahti, P.; Ritchie, S.C.; Tokolyi, A.; Sanders, J.G.; Valsta, L.; Brozynska, M.; Zhu, Q.; et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat. Genet. 2022, 54, 134–142. [Google Scholar] [CrossRef]
  58. Nagata, N.; Takeuchi, T.; Masuoka, H.; Aoki, R.; Ishikane, M.; Iwamoto, N.; Sugiyama, M.; Suda, W.; Nakanishi, Y.; Terada-Hirashima, J.; et al. Human gut microbiota and its metabolites impact immune responses in COVID-19 and its complications. Gastroenterology 2022, in press. [Google Scholar] [CrossRef]
  59. Gophna, U.; Sommerfeld, K.; Gophna, S.; Doolittle, W.F.; Zanten, S.J. Differences between tissue-associated intestinal microfloras of patients with Crohn’s Disease and Ulcerative Colitis. J. Clin. Microbiol. 2006, 44, 4136–4141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Zhen, Y.; Chen, Y.; Ge, L.; Wei, W.; Wang, Y.; Hu, L.; Loor, J.J.; Wang, M.; Yin, J. The short-day cycle induces intestinal epithelial purine metabolism imbalance and hepatic disfunctions in antibiotic-mediated gut microbiota perturbation mice. Int. J. Mol. Sci. 2022, 23, 6008. [Google Scholar] [CrossRef]
  61. Frausto, D.M.; Forsyth, C.B.; Keshavarzian, A.; Voigt, R.M. Dietary regulation of gut-brain axis in Alzheimer’s disease: Importance of microbiota metabolites. Front. Neurosci. 2021, 15, 736814. [Google Scholar] [CrossRef] [PubMed]
  62. Alhmoud, T.; Kumar, A.; Lo, C.C.; Al-Sadi, R.; Clegg, S.; Alomari, I.; Zmeili, T.; Gleasne, C.D.; McMurry, K.; Dichosa, A.E.K.; et al. Investigating intestinal permeability and gut microbiota roles in acute coronary syndrome patients. Hum. Microb. J. 2019, 13, 100059. [Google Scholar] [CrossRef]
  63. Li, P.; Li, M.; Wu, T.; Song, Y.; Li, Y.; Huang, X.; Lu, H.; Xu, Z.Z. Systematic evaluation of antimicrobial food preservatives on glucose metabolism and gut microbiota in healthy mice. NPJ Sci. Food 2022, 6, 42. [Google Scholar] [CrossRef]
  64. 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. 2019, 64, 497–508. [Google Scholar] [CrossRef]
  65. Zhai, H.; Luo, Y.; Ren, W.; Schyns, G.; Guggenbuhl, P. The effects of benzoic acid and essential oils on growth performance, nutrient digestibility, and colonic microbiota in nursery pigs. Anim. Feed Sci. Technol. 2020, 262, 114426. [Google Scholar] [CrossRef]
  66. Resende, M.; Chaves, R.F.; Garcia, R.M.; Barbosa, J.A.; Marques, A.S.; Rezende, L.R.; Peconick, A.P.; Garbossa, C.A.P.; Mesa, D.; Silva, C.C.; et al. Benzoic acid and essential oils modify the cecum microbiota composition in weaned piglets and improve growth performance in finishing pigs. Livest. Sci. 2020, 242, 104311. [Google Scholar] [CrossRef]
  67. Correa, F.; Luise, D.; Castillo, M.; Peris, S.; Palomo-Yague, A.; Bosi, P.; Trevisi, P. Effect of dietary supplementation with a blend of protected aromatic compounds, including benzoic acid, on growth performance and faecal microbial profile of weaned piglets as an alternative to Zinc Oxide. Livest. Sci. 2021, 246, 104455. [Google Scholar] [CrossRef]
  68. Xu, J.; Wang, M.; Liu, Q.; Lin, X.; Pu, K.; He, Z. Gut microbiota mediated the toxicity of high concentration of dietary nitrite in C57BL/6 mice. Ecotoxicol. Environ. Saf. 2022, 231, 113224. [Google Scholar] [CrossRef] [PubMed]
  69. Gerasimidis, K.; Bryden, K.; Chen, X.; Papachristou, E.; Verney, A.; Roig, M.; Hansen, R.; Nichols, B.; Papadopoulou, R.; Parrett, A. 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, 3213–3230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Irwin, S.V.; Fisher, P.; Graham, E.; Malek, A.; Robidoux, A. Sulfites inhibit the growth of four species of beneficial gut bacteria at concentrations regarded as safe for food. PLoS ONE 2017, 12, e0186629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Hu, L.; Jin, L.; Xia, D.; Zhang, Q.; Ma, L.; Zheng, H.; Xu, T.; Chang, S.; Li, X.; Xun, Z.; et al. Nitrate ameliorates dextran sodium sulfate-induced colitis by regulating the homeostasis of the intestinal microbiota. Free Radic. Biol. Med. 2020, 152, 609–621. [Google Scholar] [CrossRef]
  72. Xu, J.; Tang, M.; Liu, Y.; Xu, J.; Xu, X. Safety assessment of monosodium glutamate based on intestinal function and flora in mice. Food Sci. Hum. Wellness 2022, 11, 155–164. [Google Scholar] [CrossRef]
  73. 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. [Google Scholar] [CrossRef]
  74. Kong, F.; Kang, S.; Zhang, J.; Zhao, H.; Peng, Y.; Yang, M.; Zheng, Y.; Shao, J.; Yue, X. Whey protein and xylitol complex alleviate type 2 diabetes in C57BL/6 mice by regulating the intestinal microbiota. Food Res. Int. 2022, 157, 111454. [Google Scholar] [CrossRef]
  75. Uebanso, T.; Kano, S.; Yoshimoto, A.; Naito, C.; Shimohata, T.; Mawatari, K.; Takahashi, A. Effects of consuming xylitol on gut microbiota and lipid metabolism in mice. Nutrients 2017, 9, 756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Zuo, Q.L.; Cai, X.; Zheng, X.Y.; Chen, D.S.; Li, M.; Liu, Z.Q.; Chen, K.Q.; Han, F.F.; Zhu, X. Influences of xylitol consumption at different dosages on intestinal tissues and gut microbiota in rats. J. Agric. Food Chem. 2021, 69, 12002–12011. [Google Scholar] [CrossRef]
  77. Xu, Y.; Chen, Y.; Xiang, S.; Ye, K.; Bao, X.; Zhu, X.; Ge, Y.; Shi, L.; Lin, M. Effect of xylitol on gut microbiota in an in vitro colonic simulation. Turk. J. Biochem. 2019, 44, 646–653. [Google Scholar] [CrossRef]
  78. Li, C.H.; Wang, C.T.; Lin, Y.J.; Kuo, H.Y.; Wu, J.S.; Hong, T.C.; Chang, C.J.; Wu, H.T. Long-term consumption of the sugar substitute sorbitol alters gut microbiome and induces glucose intolerance in mice. Life Sci. 2022, 305, 120770. [Google Scholar] [CrossRef] [PubMed]
  79. Hattori, K.; Akiyama, M.; Seki, N.; Yakabe, K.; Hase, K.; Kim, Y.G. Gut microbiota prevents sugar alcohol-induced diarrhea. Nutrients 2021, 13, 2029. [Google Scholar] [CrossRef] [PubMed]
  80. Li, X.Q.; Zhang, X.M.; Wu, X.; Lan, Y.; Xu, L.; Meng, X.C.; Li, J.N. Beneficial effects of lactitol on the composition of gut microbiota in constipated patients. J. Dig. Dis. 2020, 21, 445–453. [Google Scholar] [CrossRef]
  81. Yan, J.; Wang, D.; Li, K.; Chen, Q.; Lai, W.; Tian, L.; Lin, B.; Tan, Y.; Liu, X.; Xi, Z. Toxic effects of the food additives titanium dioxide and silica on the murine intestinal tract: Mechanisms related to intestinal barrier dysfunction involved by gut microbiota. Environ. Toxicol. Pharmacol. 2020, 80, 103485. [Google Scholar] [CrossRef]
  82. Zahran, S.A.; Ali-Tammam, M.; Hashem, A.M.; Aziz, R.K.; Ali, A.E. Azoreductase activity of dye-decolorizing bacteria isolated from the human gut microbiota. Sci. Rep. 2019, 9, 5508. [Google Scholar] [CrossRef] [Green Version]
  83. Elmen, L.; Zlamal, J.E.; Scott, D.A.; Lee, R.B.; Chen, D.J.; Colas, A.R.; Rodionov, D.A.; Peterson, S.N. Dietary Emulsifier Sodium Stearoyl Lactylate Alters Gut Microbiota in vitro and Inhibits Bacterial Butyrate Producers. Front Microbiol. 2020, 11, 892. [Google Scholar] [CrossRef]
  84. Silva, M.M.; Lidon, F.C. An overview on applications and side effects of antioxidant food additives. Emir. J. Food Agric. 2016, 28, 823–832. [Google Scholar] [CrossRef]
  85. Gulcin, İ. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Wang, W.; Wang, X.; Zhu, Q.; Zhou, Q.; Wang, Y.; Liao, C.; Jiang, G. Occurrence of synthetic phenolic antioxidants in foodstuffs from ten provinces in China and its implications for human dietary exposure. Food Chem. Toxicol. 2022, 165, 113134. [Google Scholar] [CrossRef] [PubMed]
  87. Franco, R.; Navarro, G.; Martínez-Pinilla, E. Antioxidants versus food antioxidant additives and food preservatives. Antioxidants 2019, 8, 542. [Google Scholar] [CrossRef] [PubMed]
  88. Ruiz-Rico, M.; Renwick, S.; Allen-Vercoe, E.; Barat, J.M. In vitro susceptibility of human gut microbes to potential food preservatives based on immobilized phenolic compounds. Food Chem. 2022, 378, 132–136. [Google Scholar] [CrossRef]
  89. García-García, R.; Searle, S.S. Preservatives: Food use. In Encyclopedia of Food and Health; Caballero, B., Finglas, P.M., Toldrá, F., Eds.; Academic Press: Oxford, UK, 2016; pp. 505–509. [Google Scholar]
  90. Giannenas, I.; Doukas, D.; Karamoutsios, A.; Tzora, A.; Bonos, E.; Skoufos, I.; Tsinas, A.; Christaki, E.; Tontis, D.; Florou-Paneri, P. Effects of Enterococcus faecium, mannan oligosaccharide, benzoic acid and their mixture on growth performance, intestinal microbiota, intestinal morphology and blood lymphocyte subpopulations of fattening pigs. Anim. Feed Sci. Technol. 2016, 220, 159–167. [Google Scholar] [CrossRef]
  91. Cani, P.D.; de Vos, W.M. Next-generation beneficial microbes: The case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Gómez-Gallego, C.; Pohl, S.; Salminen, S.; Vos, W.M.D.; Kneifel, W. Akkermansia muciniphila: A novel functional microbe with probiotic properties. Benef. Microbes 2016, 7, 571–584. [Google Scholar] [CrossRef] [PubMed]
  93. Liang, L.; Zhou, C.; Zhang, J.; Huang, Y.; Zhao, J.; Sun, B.; Zhang, Y. Characteristics of umami peptides identified from porcine bone soup and molecular docking to the taste receptor T1R1/T1R3. Food Chem. 2022, 387, 132870. [Google Scholar] [CrossRef]
  94. Liang, L.; Duan, W.; Zhang, J.; Huang, Y.; Zhang, Y.; Sun, B. Characterization and molecular docking study of taste peptides from chicken soup by sensory analysis combined with nano-LC-Q-TOF-MS/MS. Food Chem. 2022, 383, 132455. [Google Scholar] [CrossRef]
  95. Cao, Y.; Liu, H.; Qin, N.; Ren, X.; Zhu, B.; Xia, X. Impact of food additives on the composition and function of gut microbiota: A review. Trends Food Sci. Technol. 2020, 99, 295–310. [Google Scholar] [CrossRef]
  96. Ruiz-Ojeda, F.J.; Plaza-Diaz, J.; Saez-Lara, M.J.; Gil, A. Effects of Sweeteners on the Gut Microbiota: A Review of Experimental Studies and Clinical Trials. Adv. Nutr. 2019, 10 (Suppl. S1), S31–S48. [Google Scholar] [CrossRef] [Green Version]
  97. Gultekin, F.; Oner, M.E.; Savas, H.B.; Dogan, B. Food additives and microbiota. North. Clin. Istanb. 2020, 7, 192–200. [Google Scholar] [CrossRef] [PubMed]
  98. Plaza-Diaz, J.; Pastor-Villaescusa, B.; Rueda-Robles, A.; Abadia-Molina, F.; Ruiz-Ojeda, F.J. Plausible biological interactions of low- and non-calorie sweeteners with the intestinal microbiota: An update of recent studies. Nutrients 2020, 12, 1153. [Google Scholar] [CrossRef] [PubMed]
  99. Tamura, M.; Hoshi, C.; Hori, S. Xylitol affects the intestinal microbiota and metabolism of daidzein in adult male mice. Int. J. Mol. Sci. 2013, 14, 23993–24007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Yue, Y.; Nielsen, D.S.G.; Forssten, S.D.; Knudsen, K.E.B.; Saarinen, M.T.; Ouwehand, A.C.; Purup, S. Effects of colonic fermentation products of polydextrose, lactitol and xylitol on intestinal barrier repair in vitro. Appl. Sci. 2021, 11, 4174. [Google Scholar] [CrossRef]
  101. Xiang, S.; Ye, K.; Li, M.; Ying, J.; Wang, H.; Han, J.; Shi, L.; Xiao, J.; Shen, Y.; Feng, X.; et al. Xylitol enhances synthesis of propionate in the colon via cross-feeding of gut microbiota. Microbiome 2021, 9, 62. [Google Scholar] [CrossRef]
  102. Tiffany, C.R.; Lee, J.Y.; Rogers, A.W.L.; Olsan, E.E.; Morales, P.; Faber, F.; Baumler, A.J. The metabolic footprint of Clostridia and Erysipelotrichia reveals their role in depleting sugar alcohols in the cecum. Microbiome 2021, 9, 174. [Google Scholar] [CrossRef]
  103. EFSA. Scientific opinion on the safety of the proposed extension of use of erythritol (E 968) as a food additive. EFSA J. 2015, 2015, 4033. [Google Scholar]
  104. Mahalak, K.K.; Firrman, J.; Tomasula, P.M.; Nunez, A.; Lee, J.J.; Bittinger, K.; Rinaldi, W.; Liu, L.S. Impact of steviol glycosides and erythritol on the human and cebus apella gut microbiome. J. Agric. Food Chem. 2020, 68, 13093–13101. [Google Scholar] [CrossRef]
  105. Schmid, N.; Verbeek, U. Titanium dioxide. The food additive E 171 will be banned in 2022. Dtsch. Lebensm. Rundsch. 2021, 117, 493–494. [Google Scholar]
  106. Wu, L.; Lv, X.; Zhang, Y.; Xin, Q.; Zou, Y.; Li, X. Tartrazine exposure results in histological damage, oxidative stress, immune disorders and gut microbiota dysbiosis in juvenile crucian carp (Carassius carassius). Aquat. Toxicol. 2021, 241, 105998. [Google Scholar] [CrossRef] [PubMed]
  107. Abbey, J.; Fields, B.; O’Mullane, M.; Tomaska, L.D. Food Additives: Colorants. In Encyclopedia of Food Safety; Motarjemi, Y., Ed.; Academic Press: Waltham, MA, USA, 2014; pp. 459–465. [Google Scholar]
  108. Yang, W.; Cong, Y. The disruption of intestinal homeostasis when foods are colored red. Cell. Mol. Immunol. 2022, 19, 855–857. [Google Scholar] [CrossRef]
  109. Chen, L.; He, Z.; Iuga, A.C.; Martins Filho, S.N.; Faith, J.J.; Clemente, J.C.; Deshpande, M.; Jayaprakash, A.; Colombel, J.-F.; Lafaille, J.J.; et al. Diet modifies colonic microbiota and CD4+ T-Cell repertoire to induce flares of colitis in mice with myeloid-cell expression of interleukin 23. Gastroenterology 2018, 155, 1177–1191.e16. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, L.; He, Z.; Reis, B.S.; Gelles, J.D.; Chipuk, J.E.; Ting, A.T.; Spicer, J.A.; Trapani, J.A.; Furtado, G.C.; Lira, S.A. IFN-gamma(+) cytotoxic CD4(+) T lymphocytes are involved in the pathogenesis of colitis induced by IL-23 and the food colorant Red 40. Cell. Mol. Immunol. 2022, 19, 777–790. [Google Scholar] [CrossRef]
  111. Laudisi, F.; Stolfi, C.; Monteleone, G. Impact of food additives on gut homeostasis. Nutrients 2019, 11, 2334. [Google Scholar] [CrossRef] [PubMed]
  112. Zangara, M.T.; Ponti, A.K.; Miller, N.D.; Engelhart, M.J.; Ahern, P.P.; Sangwan, N.; McDonald, C. Maltodextrin Consumption Impairs the Intestinal Mucus Barrier and Accelerates Colitis Through Direct Actions on the Epithelium. Front. Immunol. 2022, 13, 841188. [Google Scholar] [CrossRef] [PubMed]
Table 1. The effect of food additives on gut microbiota and its function.
Table 1. The effect of food additives on gut microbiota and its function.
Food AdditivesExperiment TrialMicrobesFunctionReferences
Sodium benzoateC57BL/6 miceCoriobacteriaceaeConvert carbohydrates acetic acid and lactic acid[63]
Sodium benzoate,
Sodium nitrite,
Potassium sorbate
In vitroClostridium tyrobutyricum or Lactobacillus paracaseiAnti-inflammatory properties[64]
Sodium benzoate,
Sodium nitrite,
Potassium sorbate
In vitroBacteroides thetaiotaomicron or Enterococcus faecalisPro-inflammatory or colitogenic properties[64]
Benzoic acidPigPhylum of Bacteroidetes and the genus of Prevotella in the phylum of Bacteroidetes/[65]
Benzoic acid (90%) and essential oil (10%, include thymol, 2-meth-oxyphenol, and eugenol)PigFusicatenibacter, Escherichia-Shigella, and Ruminococcus
Prevotella, and Coprococcus 1
/[66]
Potassium sorbateMiceCoriobacteriaceaeConvert carbohydrates to acetic acid and lactic acid[63]
Benzoic acid (49%)PigletRuminococcus, Prevotellaceae, and FibrobacteraceaeFiber fermenting abilities[67]
NitriteC57BL/6 miceFalsiporphyromonas, Alloprevotella, Acetatifactor, and Coprococcus
Akkermansia, and Elusimicrobium
/[68]
Sodium benzoateHumanBifidobacterium/[69]
Sodium sulphiteHumanEscherichia/Shigella
Bifidobacterium
/[69]
SulfiteIn vitroPlantarum, rhamnosus, Lactobacillus species casei, and Streptococcus thermophilus/[70]
NaNO3MouseLactobacillus, Ruminococcaceae_UCG-014, and Prevotellaceae_UCG-001/[71]
Monosodium glutamate
(300 mg/kg)
MicePatescibacteria/[72]
Monosodium glutamate
(1500 mg/kg)
MicePatescibacteria/[72]
Monosodium glutamate
(2 g/day)
HumanNo significant difference/[73]
Xylitol
(10 g/L in high fat diet)
C57BL/6 miceBacteroidetes, Actinobacteria, and Proteobacteria
Firmicutes
/[74]
Xylitol
(194 ± 25 mg/kg b.w. in high fat diet)
C57BL/6J miceBacteroidetes, Barnesiella
Firmicutes, Prevotella
/[75]
Xylitol (10%)RatRuminococcaceae, Prevotella
Bacteroides
/[76]
XylitolIn vitroClostridium, PhascolarctobacteriumButyrate synthesizing[77]
Escherichia/
SorbitolMiceBifidobacterium, Lachnospiraceae UCG 001, Lachnospiraceae NK4A136, Eubacterium ventriosum, Candidatus Arthromitus, Ruminococcus torques
Helicobacter, Tyzzerella, Alistipes, and Prevotella 9
/[78]
SorbitolC57BL/6 miceEscherichia, Klebsiella, Enterobacter, and Proteus in Enterobacteriaceae family/[79]
LactitolHumanActinobacteria, Actinobacteria, Bifidobacteriales, Bifidobacteriaceae and Bifidobacterium/[80]
Titanium dioxideICR miceVerrucomicrobia, Bacteroidetes
Firmicutes
/[81]
Sunset yellowIn vitroEscherichia coli, Enterococcus faecium, Aerococcus viridans, and Bacillus cereusDecolorized sunset yellow[82]
Resistant starchMiceLactobacillus, Lachnoclostridium/[49]
Sodium stearoyl lactylateIn vitroClostridia, Clostridiaceae, Lachnospiraceae, Ruminococcaceae
Bacteroidaceae and Enterobacteriaceae and Desulfovibrionaceae
/[83]
/ means there is no function concerned in the cited literature. ↑ positive influence, ↓ negative influence.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhou, X.; Qiao, K.; Wu, H.; Zhang, Y. The Impact of Food Additives on the Abundance and Composition of Gut Microbiota. Molecules 2023, 28, 631. https://doi.org/10.3390/molecules28020631

AMA Style

Zhou X, Qiao K, Wu H, Zhang Y. The Impact of Food Additives on the Abundance and Composition of Gut Microbiota. Molecules. 2023; 28(2):631. https://doi.org/10.3390/molecules28020631

Chicago/Turabian Style

Zhou, Xuewei, Kaina Qiao, Huimin Wu, and Yuyu Zhang. 2023. "The Impact of Food Additives on the Abundance and Composition of Gut Microbiota" Molecules 28, no. 2: 631. https://doi.org/10.3390/molecules28020631

Article Metrics

Back to TopTop