Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Imbalanced dietary intake alters the colonic microbial profile in growing rats

  • Tae-Hwan Jung,

    Roles Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation Convergence Research Center, Sahmyook University, Seoul, Korea

  • Kyoung-Sik Han

    Roles Conceptualization, Formal analysis, Methodology, Supervision, Writing – review & editing

    kshan@syu.ac.kr

    Affiliations Convergence Research Center, Sahmyook University, Seoul, Korea, Department of Food and Nutrition, Sahmyook University, Seoul, Korea

Abstract

An imbalanced dietary intake is associated with alteration of intestinal ecosystem. We investigated the impact of imbalanced diets on colonic microbiota, concentrations of short chain fatty acid in colonic digesta and serum immunoglobulins (Igs) of growing rats. Compared to the control diet, consuming diets high in fat, sucrose, or processed meat, or low in iron, increased the abundance of the pathogenic bacteria such as Clostridium, Escherichia coli, and Salmonella species, and decreased the beneficial bacteria, like Bifidobacteria, Lactobacillus, Akkermansia, Phascolarctobacterium, Alistipes, and butyrate producing species of bacteria in the colon of growing rats. The heatmap of metagenomics indicated that each group was separated into distinct clusters, and the ID group in particular, showed significantly (P < 0.01) reduced alpha diversity of colonic microbiota in comparison to the control group. All experimental groups showed significantly (P < 0.05 or P < 0.01) decreased concentration of acetate and butyrate in the colonic digesta and lower levels of serum IgG or IgA, compared to the control. These results indicated that the imbalanced dietary intake negatively altered intestinal ecosystem and immunity.

Introduction

The genetic information of gut microbiota is regarded as the second genome of humans. A variety of studies have been conducted on the gut microbiota because its abnormal alteration is closely related with various health disorders such as asthma, obesity, and diabetes [13]. Composition of the gut microbiota is influenced by different factors like race, region, and diet [4]. Particularly, dietary intakes may have an exceedingly important effect on the gut microbiota [5], the balance of which may be associated with serum immunoglobulins (Igs) and the fecal short-chain fatty acids (SCFAs) produced by the metabolism of dietary ingredients [6, 7].

The gut is connected to the brain via vagal sensory neurons [8]. The gut microbiota influences the enteric nervous system (ENS), which interacts with the central nervous system (CNS) of brain [9]. The balanced gut microbiota composition contributes to health promotion, whereas its abnormal state can result in the mental disorder by adversely affecting the ENS and CNS [10]. Therefore, the desirable modulation of gut microbiota may prevent, and improve such mental disorders [11].

Recently, it was reported that the gut microbiota composition could affect the mental disorders such as attention deficit hyperactivity disorder (ADHD) and autism [12, 13]. ADHD, a neurodevelopmental disorder, makes it difficult for a person to control impulsive behaviors. It is one of the most common medical conditions in childhood, that tends to persist in adulthood [14]. ADHD is associated with neurotransmitters involved in dopamine function, and also with immune system, which is greatly influenced by alteration in the gut microbiota [15, 16].

Dietary intake has an important role in the modulation of the gut microbiota composition and imbalanced diet leads to dysbiosis of the gut microbiota [5]. Recent studies have reported that aggravation of ADHD symptoms is associated with imbalanced dietary intakes such as high fat (HF), high sucrose (HS), iron deficiency (ID) or processed meat (PM) diet [17, 18]. Prior to study on correlation between dietary patterns and ADHD in human, we have undertaken the present study, designed to investigate the effect of dietary imbalance on the colonic microbiota, production of SCFAs in the colon and serum Igs in growing rats.

Materials and methods

Animal study

This work was approved by the Sahmyook University Animal Ethics Committee (SYUIACUC2017-002). The animal procedures were conducted in strict accordance with the National Research Council and Institutional Animal Care and Use Committee (Seoul, Korea).

This study was carried out in the animal facility of Sahmyook University (Seoul, Korea) and all efforts were made to minimize suffering of animals. Experimental animals and feed were purchased from Duyeol Biotech (Seoul, Korea). Sixty Sprague-Dawley male growing rats (140–160 g body weight) were housed singly in stainless steel cages in a room maintained at 22 ± 2°C with a 12 h light-dark cycle. The rats were given one week to acclimatize, during which time they consumed the basal diet and water ad libitum. After adaptation, the rats were randomly allocated to one of the 5 diets (n = 12) during the four weeks of the study (Table 1). The standard diet AIN-93G (Envigo, Indianapolis, IN, USA) was used as a control diet. The HF diet was prepared with adding lard to the control diet. The corn starch was eliminated to increase composition of sucrose in the HS diet. The ID diet was similar to that of control diet, but ferric citrate was excluded from mineral mix. The composition of PM diet was same as that of control diet, but additionally spam (10 g/kg, Austin, MN, USA) was supplied daily. On the last day of the experiment, the rats were quickly anesthetized with carbon dioxide to alleviate pain and sacrified by dislocating the cervical vertebrae. The serum and the colonic digesta of each rat were carefully collected to analyze the composition of gut microbiota, SCFAs, and Igs.

thumbnail
Table 1. Composition of the control and experimental diets.

https://doi.org/10.1371/journal.pone.0253959.t001

DNA extraction and quantitative real-time PCR

Bacterial DNA was extracted from the colonic digesta using a QIAamp DNA stool mini kit (Qiagen, Hilden, Germany) according to the manufacturer’s guidelines. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed using the DT-prime 5 (DNA-Technology, Moscow, Russia) with the power SYBR Green PCR master mix (Applied Biosystems, Foster City, CA, USA) and specific primers to measure the number of total bacteria (forward, GCA GGC CTA ACA CAT GCA AGT C; reverse, CTG CCT CCC GTA GGA GT), Bacteroides (forward, GAA GGT CCC CCA CAT TG; reverse, CAA TCG GAG TTC GTG), Lactobacillus (forward, CGA TGA GTG CTA GGT GTT GGA; reverse, CAA GAT GTC AAG ACC TGG TAA G), Bifidobacteria (forward, CTC CTG GAA ACG GGT GG; reverse, GGT GTT CTT CCC GAT ATC TAC A), Clostridium (forward, ATG CAA GTC GAG CGA KG; reverse, TAT GCG GTA TTA ATC TYC CTT T), Enterobacteria (forward, ATG GCT GTC AGC TCG T; reverse, CTA CTT TTG CAA CCC ACT C), and Escherichia coli (forward, GTT AAT ACC TTT GCT CAT TGA; reverse, ACC AGG GTA TCT AAT CCT GTT) based on the methods of Han et al. [19]. The reaction mixture consisted of 10 μL SYBR green master mix, 1 μL each of specific primers (10 pM) and 2 μL DNA sample in a final volume of 20 μL. An initial DNA denaturation step at 95°C for 10 min was followed by 40 cycles of amplification (denaturation at 95°C for 15 s, primer annealing at 55–61°C for 25 s, and extension at 72°C for 30–40 s) and cooling at 4°C. The calibration curve was constituted by Ct values, depending on serial dilutions of bacterial DNA isolated from each variety of bacteria using the DNeasy Blood & Tissue Kit (Qiagen, Hilden, Germany).

PCR-DGGE

The PCR-DGGE was conducted using the method described by Jung et al. [20]. Extracted bacterial DNA was amplified by PCR using the Veriti 96 well thermal cycler (Applied Biosystems, Foster city, USA), TaKaRa Ex Taq polymerase (Takara, Kusatsu, Japan), and primers (forward, CGC CCG GGG CGC GCC CCG GGC GGG GCG GGG GCA CGG GGG GAA CGC GAA CCT TAC; reverse, CGG TGT GTA CAA GAC CC). The PCR amplicons were subjected to PCR-denaturing gradient gel electrophoresis (DGGE) using the Dcode system (Biorad, Hurcules, CA, USA). A polyacrylamide gel (8%) containing the urea-formamide gradient of 35–50% was used for the separation of the PCR amplicons, and the electrophoresis was conducted at 130 V for 5 h at 60°C. The intensity of each band was measured using the Image Lab 5.1 Software (Biorad) and sixteen bands of specific interest, indicating different intensities among the groups were resected. The DNA from each band was cloned separately into the pGEM-T Easy Vector (Promega, Madison, WI, USA), and sequenced at Bionics company (Seoul, Korea). Nucleotide sequence data was analyzed using the GenBank database with the BLAST search program.

Metagenomics analysis

Metagenomic analysis for gut microbiota was conducted at Chunlab company (Seoul, Korea). PCR amplification was performed using fusion primers targeting from V3 to V4 regions of the 16S rRNA gene in the extracted DNA. For bacterial amplification, fusion primers of 341F (5’-TGATACGGCGACCACCGAGATCTACAC-XXXXXXXXTCGTCGGCAGCG TCAGATGTGTATAAGAGACAG-CCTACGGGNGGCWGCAG-3’; underlining sequence indicates the target region primer) and 805R (5’- CAAGCAGAAGACGGCATACGAGAT-XXXXXXXX-GTCTCGTGGGCTCGG-AGATGTGTATAAGAGACAG-ACTACHVGGGTATCTAATCC-3’). The Fusion primers are constructed in the following order which is P5 graft binding, i5 index, nextera consensus, sequencing adaptor, and target region sequence. The amplifications were carried out under the following conditions: initial denaturation at 95°C for 3 min, followed by 25 cycles of denaturation at 95°C for 30 sec, primer annealing at 55°C for 30 s, and extension at 72°C for 30 s, with a final elongation at 72°C for 5 min. The PCR product was confirmed by using 1% agarose gel electrophoresis and visualized under a Gel Doc system (BioRad). The amplified products were purified with the CleanPCR (CleanNA, Waddinxveen, Netherlands). Equal concentrations of purified products were pooled together, from which short fragments (non-target products) were removed with CleanPCR (CleanNA). The quality and product size were assessed on a Bioanalyzer 2100 (Agilent, Palo Alto, CA, USA) using a DNA 7500 chip. Mixed amplicons were pooled and the sequencing was carried out at Chunlab, Inc. (Seoul, Korea), with Illumina MiSeq Sequencing system (Illumina, Sandiego, CA, USA) according to the manufacturer’s instructions. The data analysis was performed using CLcommunityTM ver 3.46 software (Chunlab) and EZBioCloud database.

Measurement of SCFA

The SCFAs (acetate, propionate, butyrate) were measured using gas chromatography. Ten milligrams of the dried colonic digesta were mixed with 1 mL of the methanol, and shaken at 200 rpm for 90 mins at 25°C. After the mixture was centrifuged at 10,000 rpm for 10 min at 25°C, the supernatants were filtered through a 0.22 μm syringe filter, and 2 μL of the filtrates were injected into YL 6100 GC column (30 m x 0.25 mm, Youngin Chromass, Gyeonggido, Korea) to analyze SCFAs from the colonic digesta. The calibration curves were plotted using the standard reagents (Sigma-Aldrich, Saint Louis, MO, USA).

Measurement of immunoglobulins

Concentration of immunoglobulins (IgG, IgM, and IgA) in the serum were measured using the Rat IgG total uncoated ELISA kit (Invitrogen, Carlsbad, CA, Austria), Rat IgM ELISA Kit (Bethyl, Montgomery, TX, USA), and Rat IgA ELISA kit (Bethyl) according to the manufacturer’s guidelines using coating and detection antibodies at room temperature. The absorbance values were measured at 450 nm using the microplate reader (Emax, Molecular Devices, San Jose, CA, USA).

Statistical analysis

The results were presented as mean ± SEM. Significance of the differences among the groups was determined using X2 test and the SAS/PROC GLM software (SAS version 9.1; SAS Institute Inc., Cary, NC, USA). The two-tailed p value less than 0.05 were considered significant.

Results

Measurement of colonic microbiota by qRT-PCR

The qRT-PCR was conducted to estimate the number of total bacteria, Bacteroides, Lactobacillus, Bifidobacteria, Clostridium, Enterobacteria, and E. coli strains. As shown in Table 2, there was no significant difference between control and experimental groups on the number of total bacteria, Bacteroides group, and Lactobacillus strains. However, each experimental group showed considerable difference in abundance of Bifidobacteria, Clostridium, Enterobacteria, and E. coli strains in comparison to the control group. The number of Clostridium, Enterobacteria, and E. coli strains significantly (P < 0.05 or P < 0.01) increased in rats fed HF, ID, or PM diet compared to the control group, whereas those fed HS diet showed a significantly (P < 0.01) lower number of Bifidobacteria in comparison to the control group. A significantly (P < 0.05 or P < 0.01) lower ratio of Lactobacillus to Enterobacteria was also observed in animals on the ID or PM diet compared to the control.

thumbnail
Table 2. The counts (log10 16S rDNA gene copies g-1 of colonic digesta) of different bacteria groups measured by quantitative real-time PCR.

https://doi.org/10.1371/journal.pone.0253959.t002

PCR-DGGE and DNA sequencing

The DGGE band pattern of pooled samples of each group (n = 12) is shown in Fig 1. The rats maintained on the experimental diets showed distinct band patterns compared to the control. The DNA sequencing of 16 selected DGGE bands revealed that the intensity of bands (No. 2 and 3) corresponding to beneficial bacteria such as Akkermansia muciniphila and butyrate-producing bacterial strains in the experimental groups decreased compared to the control; and that of bands (No. 4 and 12) representing Lactobacillus strains was reduced in the experimental groups except the rats fed the HS diet when compared to the control group, whereas that of bands (No. 5, 7, 8, 9, 15, and 16) representing the pathogenic bacteria such as E. coli, Shigella, Salmonella, and Clostridium increased in rats fed the experimental diets in comparison to those on control diet (Table 3). In particular, the ID group showed that the most pronounced increase in the intensity of bands corresponding to harmful bacteria such as E. coli, Salmonella, and Clostridium strains increased compared to the control group.

thumbnail
Fig 1. PCR-DGGE bands from pooled DNA samples (n = 12) of colonic digesta used in DNA sequencing for identification of bacteria.

Arrows indicate the bands subject to DNA sequencing. M, marker; A, control; B, high fat; C, high sucrose; D, iron deficiency; E, processed meat.

https://doi.org/10.1371/journal.pone.0253959.g001

thumbnail
Table 3. Bacterial species identified using DNA sequencing of the PCR-DGGE bands.

https://doi.org/10.1371/journal.pone.0253959.t003

Metagenomic analysis

Metagenomic analysis was conducted to investigate the composition and diversity of microbiota in the colonic digesta of rats from experimental and control groups. Processing raw reads started with quality check and filtering of low quality (<Q25) reads by Trimmomatic 0.32 [21]. The paired-end sequence data were merged using fastq_mergepairs command of VSEARCH 2.13.4 [22] with default parameters. Primer sequences were then trimmed with ChunLab’s in-house program at a similarity cut off of 0.8. Non-specific amplicons that did not encode 16S rRNA were detected by using HMMER program with hmm profiles [23]. Unique reads were extracted and redundant reads were clustered with the unique reads by derep_fulllength command of VSEARCH. The EzBioCloud database was used for taxonomic assignment using usearch_global command of VSEARCH followed by more precise pairwise alignment [24]. Chimeric reads were filtered on reads with <97% similarity by reference based chimeric detection using UCHIME [25] algorithm6 and the non-chimeric EzBioCloud database. The sequence data were then clustered using CD-HIT [26], UCLUST [27], and defined OTUs in this step. The heatmap was generated based on strains containing at least 1% at the genus level using CLcommunityTM software. The heatmap analysis of different genera of the colonic microbiota showed that the microbiota profile of the control and experimental groups mostly belonged to distinct clusters (Fig 2A). The colonic microbiota profiles at genus level from control and experimental groups revealed that the proportion of harmful bacteria such as Escherichia or Clostridium significantly (P < 0.05) increased in the ID and PM groups, whereas the ratio of Phascolarctobacterium or Alistipes strains, which induce SCFA production, markedly (P < 0.05) decreased in rats fed with experimental diets, as compared to those on control diet (Fig 2B). In addition, we analyzed the alpha diversity using the number of observed OTUs. There was no significant difference in alpha diversity of colonic microbiota among the test groups except that the rats fed ID diet showed significantly (P < 0.01) decreased alpha diversity in comparison to the control group (Fig 2C).

thumbnail
Fig 2. Comparison of the gut microbiota at genus level between control and experimental groups.

(A) heatmap, (B) relative abundance, (C) alpha diversity. CON, control; HF, high fat; HS, high sucrose; ID, iron deficiency; PM, processed meat. Values are expressed as mean ± SEM. Statistical significance compared to control was accepted at *P < 0.05, **P < 0.01.

https://doi.org/10.1371/journal.pone.0253959.g002

Measurement of SCFAs and immunoglobulins

The SCFAs (acetate, propionate, and butyrate) in the colonic digesta were measured using gas chromatography. As shown in Fig 3, there was significant (P < 0.05) decrease in the acetate or butyrate among the experimental groups, compared to the control. HF and ID groups showed significant (P < 0.05) decrease in the acetate concentration, while animals in all experimental diet groups showed significantly (P < 0.05) lower concentration of butyrate, than those of the control group.

thumbnail
Fig 3. Concentration of short chain fatty acids in the colonic digesta according to dietary treatments.

HF, high fat; HS, high sucrose; ID, iron deficiency; PM, processed meat. Values are expressed as mean ± SEM. Statistical significance compared to control was accepted at *P < 0.05.

https://doi.org/10.1371/journal.pone.0253959.g003

The concentration of the IgG, IgM, and IgA in the serum was estimated using the ELISA kits. As shown in Fig 4, there was no statistical difference in IgM concentration of the serum between control group and experimental groups. However, The IgG concentration in the serum significantly (P < 0.05) decreased in the rats fed HF, ID or PM diets compared to the control group. Moreover, IgA concentration in the serum of ID group significantly (P < 0.05) reduced in comparison to the control group.

thumbnail
Fig 4. Immunoglobulin levels in the serum of growing rats with different dietary treatments.

(A) IgG, (B) IgM, (C) IgA. HF, high fat; HS, high sucrose; ID, iron deficiency; PM, processed meat. Values are expressed as mean ± SEM. Statistical significance compared to control was accepted at *P < 0.05, **P < 0.01.

https://doi.org/10.1371/journal.pone.0253959.g004

Discussion

Gut microbiota plays an important role in maintaining health by regulating the metabolites such as SCFAs and molecules like immunoglobulins, and protect the host from external pathogens and potentially harmful bacteria residing in the intestine [28].

Despite limitation of PCR-DGGE method that different species with similar sequences might migrate to the same position on DGGE gel, the method has been used to study the taxonimic of bacterial community in the rats. In addition, the qRT-PCR method can be used to investigate alteration of the abundance of particular species. The results of the qRT-PCR and PCR-DGGE indicated that the rats fed HF, HS, ID, or PM diet had increased abundance of the pathogenic bacteria such as Clostridium, E. coli, and Salmonella strains, and reduced abundance of the beneficial bacteria such as Bifidobacteria, Lactobacillus, Akkermansia or butyrate producing bacterial strains when compared to the control. The decrease in abundance of Bifidobacteria strains in the rats fed HS diet seems to be due to the low in corn starch compared to control diet. In addition, it was observed that the ratio of Lactobacillus to Enterobacteria was markedly reduced in the rats fed ID or PM diet in comparison to the control. This ratio is known as an index of healthy intestine and the high index indicates greater intestinal protection against opportunistic pathogens [29].

The beneficial bacterial strains, such as Lactobacillus and Bifidobacteria modulate the balance of gut microbiota composition, interfering with the toxicity of pathogenic strains, and may have beneficial effects on psychological health [30]. Akkermansia strains improve health by increasing the thickness of mucus and intestinal barrier function [31, 32]. Abundance of these bacteria usually lead to a relative decrease in the pathogenic strains of bacteria, like Salmonella, Staphylococcus, E. coli and Clostridium [33]. Furthermore, a variety of studies announced that the higher abundance of Clostridium and E. coli was observed in the children with psychological problems compared to healthy children [34, 35].

The heatmap analysis indicated the existence of five distinct clusters, which meant that each group had distinct profiles of gut microbiota. The relative abundance of the pathogenic bacteria such as Escherichia and Clostridium strains significantly increased in rats fed ID or PM diets compared to the control, and these observations match with the results of qRT-PCR and PCR-DGGE. It was also observed that in all experimental groups, the relative abundance of the Phascolarctobacterium or Alistipes strains significantly decreased in comparison to that in the control group [36]. These bacteria are closely associated with the production of butyrate, which is important for maintaining intestinal health by promoting a favorable environment for the growth of beneficial bacteria [37]. Alpha diversity of colonic digesta significantly decreased in the rats fed ID diet in comparison to that of the control. Pereira et al. [38] reported that growing rats fed iron-deficient diet were shown to have markedly lower microbial diversity in the feces than rats fed iron-sufficient diet; and this decrease was partially restored with optimum supplementation of iron. Moreover, iron deficiency induces increased fecal calprotectin level and inflammation of the intestine, resulting in the increase in pathogens [39].

In this study, there was no significant difference in the propionate concentration in colonic digesta, but rats fed HF or ID diet showed considerably decreased fecal concentrations of acetate and butyrate, when compared to the control. The SCFAs, such as acetate and butyrate, can regulate the intestinal homeostasis and have direct effects on the composition of gut microbiota [6]. It is reported that SCFAs inhibit the growth of pathogenic bacterial strains, such as Salmonella, E. coli, and Clostridium [40]. In addition, the butyrate has an important role in modulating the neuronal excitability, and the activity of ENS [41]. Dalile et al. [42] reported that the SCFAs are known to play a key role in the microbiota-gut-brain axis of interaction, although the definite mechanism and the pathway are not yet identified.

The serum IgM did not statistically differ among the experimental groups but the serum IgG and IgA significantly decreased in rat fed HF, ID, or PM diet compared to that in the control. IgG promotes the growth of beneficial bacteria, inhibiting that of the pathogens in the intestine, and IgA is also important in modulating the composition of gut microbiota and immune function [43]. Al-Saiady [44] found that increased concentration of serum IgG was associated with relative abundance of the beneficial bacteria such as Lactobacillus strains.

Conclusions

In the present study, we investigated the influence of imbalanced diets on the gut microbiota, and levels of SCFAs in the colonic digesta and serum immunoglobulins through animal experiment. The results indicated that imbalanced dietary intake, especially ID diet, induced dysbiosis of the gut microbiota, and reduction of fecal SCFAs and serum Igs. Although the result of this study has a limitation which is not accompanied by human trial, the imbalanced dietary intake negatively altered intestinal ecosystem and immunity.

Supporting information

S1 Raw images. PCR-DGGE bands from pooled DNA samples (n = 12) of colonic digesta used in DNA sequencing for identification of bacteria.

M, marker; A, control; B, high fat; C, high sucrose; D, iron deficiency; E, processed meat.

https://doi.org/10.1371/journal.pone.0253959.s001

(PDF)

Acknowledgments

We appreciate Jae-hwan Choi, Yu-Na Jo, and Da-Eon Lee for assist in the animal testing for this study.

References

  1. 1. Fung TC. The microbiota-immune axis as a central mediator of gut-brain communication. Neurobiol Dis. 2019; 136: 104714. pmid:31846737
  2. 2. Das B, Nair GB. Homeostasis and dysbiosis of the gut microbiome in health and disease. J Biosci. 2019; 44: 117. pmid:31719226
  3. 3. Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, et al. Gut microbiota, obesity and diabetes. Postgrad Med J. 2016; 92: 286–300. pmid:26912499
  4. 4. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. 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: 14691–14696. pmid:20679230
  5. 5. Bibbo S, Ianiro G, Giorgio V, Scaldaferri F, Masucci L, Gasbarrini A, et al. The role of diet on gut microbiota composition. Eur Rev Med Pharmacol Sci. 2016; 20: 4742–4749. pmid:27906427
  6. 6. Feng W, Ao H, Peng C. Gut microbiota, short-chain fatty acids, and herbal medicines. Front Pharmacol. 2018; 9: 1354. pmid:30532706
  7. 7. Zeng MY, Cisalpino D, Varadarajan S, Hellman J, Warren HS, Cascalho M, et al. Gut microbiota-induced immunoglobulin G controls systemic infection by symbiotic bacteria and pathogens. Immunity. 2016; 44: 647–658. pmid:26944199
  8. 8. Yu CD, Xu QJ, Chang RB. Vagal sensory neurons and gut-brain signaling. Curr Opin Neurobiol. 2020; 62:133–140. pmid:32380360
  9. 9. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016; 7: 189–200. pmid:26963409
  10. 10. Liu X, Cao S, Zhang X. Modulation of gut microbiota-brain axis by probiotics, prebiotics, and diet. J Agric Food Chem. 2015; 63: 7885–7895. pmid:26306709
  11. 11. Ghoshal UC. Gut microbiota-brain axis modulation by a healthier microbiological microenvironment: facts and fictions. J Neurogastroenterol Motil. 2018; 24: 4–6. pmid:29291603
  12. 12. Cenit MC, Nuevo IC, Codoñer-Franch P, Dinan TG, Sanz Y. Gut microbiota and attention deficit hyperactivity disorder: new perspectives for a challenging condition. Eur Child Adolesc Psychiatry. 2017; 26: 1081–1092. pmid:28289903
  13. 13. Ersöz Alan B, Gülerman F. The role of gut microbiota in autism spectrum disorder. Turk Psikiyatri Derg. 2019; 30: 210–219. pmid:31613980
  14. 14. Thapar A, Cooper M. Attention deficit hyperactivity disorder. Lancet. 2016; 387: 1240–1250. pmid:26386541
  15. 15. Seyedi M, Gholami F, Samadi M, Djalali M, Effatpanah M, Yekaninejad MS, et al. The effect of vitamin D3 supplementation on serum BDNF, dopamine, and serotonin in children with attention-deficit/hyperactivity disorder. CNS Neurol Disord Drug Targets. 2019; 18: 496–501. pmid:31269890
  16. 16. Pickard JM, Zeng MY, Caruso R, Núñez G. Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev. 2017; 279: 70–89. pmid:28856738
  17. 17. Millichap JG, Yee MM. The diet factor in attention-deficit/hyperactivity disorder. Pediatrics, 2012; 129: 330–337. pmid:22232312
  18. 18. Wang LJ, Yu YH, Fu ML, Yeh WT, Hsu JL, Yang YH, et al. Dietary profiles, nutritional biochemistry status, and attention-deficit/hyperactivity disorder: path analysis for a case-control study. J Clin Med. 2019; 8: 709.
  19. 19. Han KS, Balan P, Molist Gasa F, Boland M. Green kiwifruit modulates the colonic microbiota in growing pigs. Lett Appl Microbiol. 2011;25: 379–385. pmid:21255058
  20. 20. Jung TH, Jeon WM, Han KS. In vitro effects of dietary inulin on human fecal microbiota and butyrate production. J Microbiol Biotechnol. 2015; 25: 1555–1558. pmid:26095388
  21. 21. Bolger AM, Lohse M. Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014; 30:2114–2120. pmid:24695404
  22. 22. Rognes T, Flouri T, Nichols B, Quince C, Mahe F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016; 4:e2584. pmid:27781170
  23. 23. Eddy SR. Accelerated profile HMM searches. PloS Comput Biol. 2011; 7:e1002195. pmid:22039361
  24. 24. Myers EW, Miller W. Optimal alignments in linear space. Bioinformatics. 1988; 4:11–17. pmid:3382986
  25. 25. Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves senditivity and speed of chimera detection. Bioinformatics. 2011; 27:2194–2200. pmid:21700674
  26. 26. Fu L, Niu B, Zhu Z, Wu S, Li W. CD-HIT: accelerated for clustering the next-generation sequencing data. Bioinforatics. 2012; 28:3150–3152. pmid:23060610
  27. 27. Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010; 26:2460–2461. pmid:20709691
  28. 28. Di Paola M, Bonechi E, Provensi G, Costa A, Clarke G, Ballerini C, et al. Oleoylethanolamide treatment affects gut microbiota composition and the expression of intestinal cytokines in Peyer’s patches of mice. Sci Rep. 2018; 8: 1488. pmid:29367613
  29. 29. Castillo M, Martín-Orúe SM, Nofrarías M, Manzanilla EG, Gasa J. Changes in caecal microbiota and mucosal morphology of weaned pigs, Vet Microbiol. 2007; 124: 239–247. pmid:17532151
  30. 30. Mohammadi AA, Jazayeri S, Khosravi-Darani K, Solati Z, Mohammadpour N, Asemi Z, et al. The effects of probiotics on mental health and hypothalamic-pituitary-adrenal axis: a randomized, double-blind, placebo-controlled trial in petrochemical workers. Nutr Neurosci. 2016; 19: 387–395. pmid:25879690
  31. 31. Servin AL. Antagonistic activities of lactobacilli and bifidobacterial against microbial pathogens. FEMS Microbiol Rev. 2004; 28: 405–440. pmid:15374659
  32. 32. Ottman N, Geerlings SY, Aalvink S, de Vos WM, Belzer C. Action and function of Akkermansia muciniphila in microbiome ecology, health and disease. Best Pract Res Clin Gastroenterol. 2017; 31: 637–642. pmid:29566906
  33. 33. Allen SJ, Wareham K, Wang D, Bradley C, Hutchings H, Harris W, et al. Lactobacilli and Bifidobacteria in the prevention of antibiotic-associated diarrhoea and clostridium difficile diarrhoea in older inpatients (PLACIDE): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet. 2013; 382: 1249–1257. pmid:23932219
  34. 34. Argou-Cardozo I, Zeidán-Chuliá F. Clostridium bacteria and autism spectrum conditions: a systematic review and hypothetical contribution of environmental glyphosate levels. Med Sci. 2018; 6: 29. pmid:29617356
  35. 35. Carissim C, Laudadio I, Palone F, Fulci V, Cesi V, Cardona F, et al. Functional analysis of gut microbiota and immunoinflammation in children with autism spectrum disorders. Dig Liver Dis. 2019; 51: 1366–1374. pmid:31320306
  36. 36. Wu F, Guo X, Zhang J, Zhang M, Ou Z, Peng Y. Phascolarctobacterium faecium abundant colonization in human gastrointestinal tract. Exp Ther Med. 2017; 14: 3122–3126. pmid:28912861
  37. 37. Baxter NT, Schmidt AW, Venkataraman A, Kim KS, Waldron C, Schmidt TM. Dynamics of human gut microbiota and short-chain fatty acids in response to dietary interventions with three fermentable fibers. mBio. 2019; 10: e02566–18. pmid:30696735
  38. 38. Pereira DI, Aslam MF, Frazer DM, Schmidt A, Walton GE, McCartney AL, et al. Dietary iron depletion at weaning imprints low microbiome diversity and this is not recovered with oral nano Fe(III). Microbiologyopen. 2015; 4: 12–27. pmid:25461615
  39. 39. Rusu IG, Suharoschi R, Vodnar DC, Pop CR, Socaci SA, Vulturar R, et al. Iron supplementation influence on the gut microbiota and probiotic intake effect in iron deficiency-a literature-based review. Nutrients. 2020; 12: 1993. pmid:32635533
  40. 40. Fachi JL, Felipe JS, Pral LP, da Silva BK, Corrêa RO, de Andrade MCP, et al. Butyrate protects mice from clostridium difficile-induced colitis through an HIF-1-dependent mechanism. Cell Rep. 2019; 27: 750–761. pmid:30995474
  41. 41. Soret R, Chevalier J, De Coppet P, Poupeau G, Derkinderen P, Segain JP, et al. Short-chain fatty acids regulate the enteric neurons and control gastrointestinal motility in rats. Gastroenterology. 2010; 138: 1772–1782. pmid:20152836
  42. 42. Dalile B, Van Oudenhove L, Vervliet B, Verbeke K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat Rev Gastroenterol Hepatol. 2019; 16: 461–478. pmid:31123355
  43. 43. Balan P, Han KS, Rutherfurd-Markwick K, Singh H, Moughan PJ. Ovine serum immunoglobulin has immunomodulatory effects in growing rats gavaged with Salmonella enteritidis. J Nutr. 2011; 141: 950–956. pmid:21411611
  44. 44. Al-Saiady MY. Effect of probiotic bacteria on immunoglobulin G concentration and other blood components of newborn calves. J Anim Vet Adv. 2010; 9: 604–609.