Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Nov 14.
Published in final edited form as: Br J Nutr. 2020 Jun 1;124(9):931–942. doi: 10.1017/S0007114520001853

Dietary inflammatory potential in relation to the gut microbiome: results from a cross-sectional study

Jiali Zheng 1, Kristi L Hoffman 1,2, Jiun-Sheng Chen 1,3, Nitin Shivappa 4, Akhil Sood 1,5, Gladys J Browman 1, Danika D Dirba 5, Samir Hanash 6, Peng Wei 3,7, James R Hebert 4, Joseph F Petrosino 2, Susan M Schembre 5,8, Carrie R Daniel 1,3
PMCID: PMC7554089  NIHMSID: NIHMS1598966  PMID: 32475373

Abstract

Diet has direct and indirect effects on health through inflammation and the gut microbiome. We investigated total dietary inflammatory potential via the literature-derived index (DII®) with gut microbiota diversity, composition, and function. In cancer-free patient volunteers initially approached at colonoscopy and healthy volunteers recruited from the medical center community, we assessed 16S rDNA in all subjects who provided dietary assessments and stool samples (n=101) and the gut metagenome in a subset of patients with residual fasting blood samples (n=34). Associations of energy-adjusted DII scores with microbial diversity and composition were examined using linear regression, permutational multivariate analysis of variance, and linear discriminant analysis. Spearman correlation was used to evaluate associations of species and pathways with DII and circulating inflammatory markers. Alpha- and beta-diversity did not significantly differ across DII levels; however, Ruminococcus torques, Eubacterium nodatum, Acidaminococcus intestini, and Clostridium leptum were more abundant in the most pro-inflammatory diet group, while Akkermansia muciniphila was enriched in the most anti-inflammatory diet group. With adjustment for age and BMI, R. torques, E. nodatum, A. intestini remained significantly associated with a more pro-inflammatory diet. In the metagenomic and fasting blood subset, A. intestini was correlated with circulating PAI-1, a pro-inflammatory marker (rho=0.40), but no associations remained significant upon correction for multiple testing. An index reflecting overall inflammatory potential of the diet was associated with specific microbes, but not overall diversity of the gut microbiome in our study. Findings from this preliminary study warrant further research in larger samples and prospective cohorts.

Keywords: diet, inflammation, gut microbiota, cross-sectional study, circulating markers

INTRODUCTION

Diet is one of the most influential and accessible modulators of the gut microbiome, the human intestine’s vast and diverse microbial ecosystem increasingly recognized as a key player in the development of obesity, type 2 diabetes, cardiovascular disease and cancer 16. The composition and collective function of the gut community affects how and what the human host is able to extract from the diet by providing the machinery that converts dietary content into biological signals with profound systemic effects on host health 7

Not unlike the complex and interactive nature of the microbiome, dietary habits are multidimensional with many interrelated components. Recently, Bowyer et al. and Maskarinec et al. demonstrated the utility of a priori dietary patterns or indices for capturing and controlling for variation in the gut microbiome due to the effects of participants’ diets 812. The Dietary Inflammatory Index (DII®) assesses the balance of pro- and anti-inflammatory dietary factors based on literature-derived associations between various dietary components and inflammatory biomarkers. This index, with its specific focus on inflammation, inherently differs from other index-based scores assessed in previous studies measuring adherence to established dietary guidelines or healthy eating patterns [e.g., the Healthy Eating Index (HEI) or Mediterranean Diet Score (MDS)] 13.

Diet-microbiome interactions may be one of the most promising targets to reduce chronic inflammation, a key pathophysiological mechanism underlying diet’s influence on multiple chronic diseases through the common mechanism of the NF-κB pathway and a complex interaction of cellular, molecular and metabolic factors 1422. Given that diet can modulate inflammation through both pro-inflammatory and anti-inflammatory mechanisms, we hypothesized that the inflammatory potential of usual dietary habits would be associated with the overall composition and functional capacity of the gut microbiome. In this study, we investigated total dietary inflammatory potential, as assessed by the literature-derived DII, in relation to gut microbiota diversity and composition. In a subset of individuals with fasting blood samples and metagenomic data, we further explored microbes and microbial gene pathways associated with DII score and circulating inflammatory markers.

METHODS

Study population

The study population is composed of 101 cancer-free individuals, including patient volunteers initially approached in the colonoscopy clinic of MD Anderson Cancer Center (N=36) and community volunteers recruited from the medical center community (N=65). Detailed study recruitment and eligibility criteria are provided in Supplemental Figure 1. Initially, from 2013 to 2016, a total of 132 patient volunteers with no history of cancer and 449 community volunteers expressed an interest in participating. Both subgroups were screened and interviewed by a clinical provider and/or trained study coordinator with regard to their medical history and medication use and asked to provide stool samples using the same collection protocol. Study ineligibility criteria included current smoker, antibiotic use within the past month, incident or prevalent cancer other than non-melanoma skin cancer, one or more chronic conditions that restricts dietary intake (e.g., Celiac disease), major intestinal surgery (e.g., gastric bypass), currently pregnant or lactating. BMI was calculated from measured weight (kg)/height (m)2 and categorized based on WHO criteria 23. Study subjects were included in the current analysis if they completed the dietary questionnaires, provided a stool sample, and passed microbiome data quality filtering criteria. All procedures were reviewed and approved by The University of Texas MD Anderson Institutional Review Board. Informed consent was obtained from all research participants; and all methods were performed in accordance with relevant guidelines and regulations.

Dietary assessment

Participants completed dietary histories via one of two versions of the National Cancer Institute (NCI)-developed food frequency questionnaire (FFQ). For consistency with historic recruitment of patients in prior and ongoing MD Anderson studies, patient volunteers completed a modified version of the NCI Health Habits and History Questionnaire (NCI-HHHQ) 24,25, which queries the frequency of intake and portion size of 165 food and beverage items, including ethnic foods commonly consumed in the Texas region. Daily nutrient consumption was estimated using the US Department of Agriculture Food and Nutrient Database for Dietary Studies 26. Community volunteers completed the NCI Dietary History Questionnaire II (DHQ II), a more recent web-based adaptation of the NCI-HHHQ 26, which queries 134 food items. DHQ II responses were processed using Diet*Calc software 27.

E-DII score calculation

Food and nutrient intake derived from responses to FFQs were linked to the corresponding inflammatory effect scores designated in the DII to calculate the energy adjusted (E-DII) score for each participant 13. The DII is a literature-derived, population-based dietary index designed to quantify the overall inflammatory potential of an individual’s entire diet. The details of the development and scoring algorithm are described elsewhere 13. In short, approximately 2,000 primary research articles published through 2010 which investigated the effects of 45 different food parameters (mostly macronutrients, micronutrients, some bioactive components and individual food items such as garlic and tea) on six inflammatory markers [i.e., IL-1β, IL-4, IL-6, IL-10, TNF-α and C-reactive protein (CRP)] were identified and scored to derive the component-specific inflammatory effect scores 13. Thirty-one DII components (plus garlic in the NCI-HHHQ) were used to calculate the instrument-specific, energy-adjusted E-DII score for analysis. Both FFQs lacked information on some less commonly consumed spices (ginger, saffron, turmeric, pepper, oregano, rosemary) and phenols/flavonoids (eugenol, flavan-3-ol, flavones, flavonols, flavonones, anthocyanidins, isoflavones). Food and nutrient consumption was first energy-adjusted per 1000 kcal; and subsequently standardized for each component using mean and standard deviation data derived from a composite dietary database representing energy-adjusted intake from 11 populations around the world 13. The energy-adjusted and standardized dietary intakes were then converted to centered proportions to account for skewness, multiplied by the inflammatory effect score for each available DII parameter, and summed across all DII components to obtain the overall E-DII score 13. Higher (i.e., more positive) E-DII scores represent more pro-inflammatory diets while lower (i.e., more negative) E-DII scores indicate more anti-inflammatory diets. The E-DII score has been construct-validated and consistently associated with elevated inflammatory biomarker levels such as IL-6 28, TNF-α 28 and CRP 29.

Stool and blood sample collection

All participants were provided the same stool sample collection kit with detailed instructions. Following defecation into a plastic “toilet hat,” gloved participants used a sterile BBL culture swab collection and transport system (Becton, Dickinson France) to collect a small portion of their sample. Samples were either Express (overnight or same day) shipped or brought to their next scheduled visit. All fecal samples were received within less than 48 hours of collection, stored at −80°C, and processed within one year of collection 30. Residual fasting blood samples drawn at the colonoscopy clinic visit were available in a subset of 34 patient volunteers with FFQ and stool samples.

Microbiome characterization

Total genomic DNA was extracted from fecal samples using the MoBio PowerSoil DNA Isolation Kit (Carlsbad, CA) following manufacturer’s instructions. The 16S rRNA gene was amplified using V4-targed primers (GGACTACHVGGGTWTCTAAT and GTGCCAGCMGCCGCGGTAA) 31, and amplicons were sequenced using the MiSeq platform (Illumina; CA, US). Raw FASTQ sequences were processed as previously described 32. Following quality filtering and chimera removal, sequence reads were clustered into operational taxonomic units (OTUs) using UPARSE 33, with OTUs subsequently mapped to a V4-optimized version of the SILVA database (v.123) at 97% similarity level. The remaining samples were rarefied to 2,742 reads/sample and relative abundances were calculated (Supplemental Table 1). Basic Local Alignment Search Tool (BLAST) was used to identify the likely species represented by each OTU centroid sequence 34.

Whole genome shotgun (WGS) sequencing was performed to comprehensively assess microbial genomic DNA of fecal samples in a subset of 34 patient volunteers who also had FFQ and residual fasting blood samples. WGS data provides species-level taxonomy of the gut microbiome, as well as the metabolic or functional gene content pathways represented within the microbial community. Individual libraries constructed from extracted total gDNA for each sample were pooled and sequenced via HiSeq 2000 (Illumina) using the 1×100 bp paired-end read protocol. Pooling resulted in a sequencing depth >3 Gb/sample. Quality filtering, trimming, demultiplexing, and read mapping were carried out by an in-house pipeline described previously 35.To determine metabolic pathway content of the entire metagenome, reads aligning to known orthologues in the Kyoto Encyclopedia of Genes and Genomes (KEGG) database were tabulated and pathways constructed by calculating the minimum set through MinPath45 36.

Circulating inflammatory markers

Plasma leptin, TNF-α, IL-6, lipocalin 2 (LCN2), plasminogen activator inhibitor-1 (PAI-1), C-peptide, monocyte chemoattractant protein-1 (MCP-1) were assessed via multiplex assay (Millipore). All samples were run in duplicate with internal standards (pooled cancer case plasma), healthy control samples (normal plasma), and kit quality controls to assure plate to plate consistency.

Statistical analyses

The E-DII score was categorized according to instrument-specific tertiles. Characteristics of study participants were described with medians and standard errors of mean for continuous variables, and frequencies and percentages for categorical variables. We examined the difference of categorical variables across E-DII tertiles by Chi-square test and the difference of continuous variables by Kruskal-Wallis non-parametric test. We also examined the associations of these covariates with bacterial alpha and beta diversity to assess potential confounders for inclusion in the adjusted models of E-DII and microbiota associations.

Alpha-diversity was assessed by observed OTU, Shannon diversity index, Chao1 index, and Simpson’s diversity index. Differences in alpha-diversity across E-DII tertiles among all subjects and by study subgroup were assessed via the Kruskal-Wallis non-parametric test. We also examined associations of the continuous E-DII score with alpha-diversity in each subgroup using linear regression models. None of the potential confounders examined, including age (continuous), sex (males and females), BMI (continuous), medication use (yes/no), and study subgroup (patient vs. community volunteers) were associated with alpha-diversity or appreciably changed the model estimates; and thus, were not included in the final adjusted-model.

Similarly, Bray-Curtis dissimilarity, Weighted Unifrac, Unweighted Unifrac and Jaccard (beta-diversity metrics) between the highest versus the lowest E-DII tertile was assessed via permutational multivariate analysis of variance (PERMANOVA) among all the subjects and by study subgroup 37. While none of the measured covariates were clearly associated with beta-diversity, we further examined whether BMI-status modified associations of beta diversity with E-DII in stratified analysis.

Differentially abundant bacterial taxa by E-DII tertiles were assessed using Linear Discriminant Analysis (LDA) Effect Size (LEfSe) under Galaxy environment 38, applying the one-against-all strategy with a logarithmic LDA score threshold of 3 and α of 0.1 for factorial Kruskal-Wallis test among classes. Analysis was restricted to bacteria present in ≥20% of the study population. We further assessed the associations between differentially abundant candidate taxa identified from LEfSe and potential confounders as mentioned above. Given some taxa were associated with BMI, we subsequently evaluated associations of LEfSe-identified taxa in association with E-DII tertiles via a negative binomial regression model adjusted for age and BMI.

Due to the large number of low abundance species in metagenomic data, we employed the least absolute shrinkage and selection operator (LASSO) method to identify bacterial species and microbial gene content pathways associated with E-DII among 34 patient volunteers with residual fasting blood samples using the glmnet package in R 39. Spearman’s rank correlation method was subsequently used to estimate the correlations of selected species and functional pathways based on LASSO and 16S LEfSe analyses with 7 circulating inflammatory biomarkers including leptin, TNF-α, IL-6, LCN2, PAI-1, C-peptide, and MCP-1. The Benjamini-Hochberg (B-H) method was used to adjust P values in the multiple correlation analyses while controlling for the expected false discovery rate at 0.05.

Sensitivity analysis was performed by rerunning all the analyses described above after removing one subject identified as an outlier based on his/her Shannon index value, defined as 1.5 interquartile range below the 25th or above the 75th percentile of the population’s value. All analyses were performed in SAS, R, Galaxy 38, Agile Toolkit for Incisive Microbial Analyses (ATIMA) 40. All tests were two-sided, with P values <0.05 considered statistically significant unless otherwise noted.

RESULTS

Characteristics of participants

The median E-DII score was 1.79 (range was from −5.15 to −3.08) for patient volunteers and −0.85 (range was from −4.04 to 2.83) for community volunteers. Subjects with higher E-DII scores (i.e., more pro-inflammatory diets) tended to have higher BMI (Table 1). However, there was no statistically significant difference across E-DII levels in the distribution of age, sex, history of precancerous colorectal polyps and medication use. When we examined these factors in relation to overall microbial diversity, none of the factors were strongly associated with alpha-diversity (all P>=0.15) or beta-diversity (all rho<=0.05; data not shown).

Table 1.

Characteristics of participants by E-DII score (n=101)

Most anti-inflammatory diet Most pro-inflammatory diet P-valueb
Tertile 1a Tertile 2a Tertile 3a
N 33 34 34
Median (SE) Median (SE) Median (SE)
Age, years 36.0 (2.49) 45.0 (2.44) 41.5 (2.47) 0.78
BMI, kg/m2 23.10 (1.04) 28.6 (1.52) 30.2 (1.09) 0.001
Microbial alpha-diversity measures
 Shannon Index 2.45 (0.12) 2.61 (0.10) 2.63 (0.11) 0.63
 Observed OTUs 70.58 (4.18) 76.29 (4.24) 80.12 (4.83) 0.41
 Chao1 Index 88.94 (5.58) 100.11 (6.60) 104.15 (6.61) 0.22
 Simpson Index 0.83 (0.03) 0.87 (0.02) 0.85 (0.02) 0.65
N (%) N (%) N (%)
Sex 0.33
 Male 9 (27.27) 11 (32.35) 15 (44.12)
 Females 24 (72.73) 23 (67.65) 19 (55.88)
Study subgroup 0.91
 Community volunteers 22 (66.67) 21 (61.67) 22 (64.71)
 Patient volunteers 11 (33.33) 13 (38.24) 12 (35.29)
History of precancerous colorectal polyps 0.37
 Yes 8 (24.24) 11 (32.35) 6 (17.65)
 No 25 (75.76) 23 (67.65) 28 (82.35)
Medication use for the following conditions
 Hyperlipidemia 0.53
  Yes 3 (9.09) 6 (17.65) 6 (17.65)
  No 30 (90.91) 28 (82.35) 28 (82.35)
 High blood sugar 0.60
  Yes 1 (3.03) 0 (0) 1 (2.94)
  No 32 (96.97) 34 (100) 33 (97.06)
Gastroesophageal reflux 0.67
 Yes 2 (6.06) 4 (11.76) 4 (11.76)
 No 31 (93.94) 30 (88.24) 30 (88.24)
Medication use for one or more of above conditions 0.56
 Yes 6 (18.18) 8 (23.53) 10 (29.41)
 No 27 (81.82) 26 (76.47) 24 (70.59)
a.

E-DII values across the three levels were calculated from FFQ-specific E-DII scores and categorized in to tertiles based on the distributions in each study subgroup.

b.

The difference of categorical variable across E-DII tertiles was tested by Chi-square test and difference of continuous variables was tested using Kruskal-Wallis non-parametric test as they are not normally distributed.

E-DII, energy-adjusted dietary inflammatory index

Associations of E-DII score with microbial diversity

Alpha diversity (within sample diversity), a measure of microbial richness and/or evenness, did not differ across E-DII levels among all participants in the crude analysis [Shannon index: P=0.63 (Figure 1a); other metrics (Table 1)]. Null results were similarly consistent within study subgroup (Figure 1b for Shannon Index; data not shown for other metrics). Null results also were observed when examining continuous associations of E-DII and alpha-diversity by subgroup (data not shown).

Figure 1.

Figure 1.

Microbial alpha-diversity, as assessed by Shannon index, across E-DII tertiles among (a) all study subjects (n=101) and (b) by study subgroup (n=65 community volunteers, n=36 patient volunteers)

We examined several beta diversity (between sample diversity) metrics, representing different measures of dissimilarity or distance between groups by highest versus lowest E-DII level. We observed modest visual differences in overall community composition (Figure 2 and Supplemental Figure 2). Although the results of Unweighted Unifrac reached statistical significance, E-DII clusters, comparing the highest (pro-inflammatory) vs. lowest (anti-inflammatory) levels were not overly distinct (P=0.03, rho=0.02; Supplemental Figure 2). Similar results were observed when we examined E-DII and beta-diversity associations stratified by BMI status.

Figure 2.

Figure 2.

Microbial community differences, as assessed by Bray-Curtis dissimilarity, between individuals with the most anti-inflammatory vs. pro-inflammatory diet among (a) all study subjects and (b) by study subgroup

Differentially abundant taxa by E-DII score

We identified several differentially abundant taxa in the LEfSe analysis across the three levels of E-DII score (Figure 3). Ruminococcus torques, Eubacterium nodatum, Acidaminococcus intestini, Clostridium leptum were more abundant in subjects consuming the most pro-inflammatory diets, while Akkermansia muciniphila was enriched in subjects with the most anti-inflammatory diets. With adjustment for age and BMI in the negative binomial model, E-DII associations with these 5 taxa did not appreciably change compared to crude model (Table 2). R. torques, E. nodatum, A. intestini remained significantly associated with a more pro-inflammatory diet.

Figure 3.

Figure 3.

Differentially abundant taxa across E-DII tertiles using LEfSe approach among 101 subjects.

Table 2.

Crude and age- and BMI-adjusted associations of E-DII with 5 candidate taxaa (n=101)

Most anti-inflammatory diet
Tertile 1
N=33
Tertile 2
N=34
Most pro-inflammatory diet
Tertile 3
N=34
P-valuea
eβ (95% CI)b eβ (95% CI)b eβ (95% CI)b
R. torques
 Crude model 1.00 (ref) 1.17 (0.62–2.21) 3.16 (1.68–5.93) 0.001
 Age- and BMI-adjusted model 1.00 (ref) 1.12 (0.55–2.28) 3.03 (1.48–6.20) 0.002
A. intestine
 Crude model 1.00 (ref) 18.93 (2.23160.93) 257.21 (30.90–2140.72) <.0001
 Age- and BMI adjusted model 1.00 (ref) 7.59 (0.83–69.58) 127.96 (9.85–1661.54) 0.0005
E. nodatum
 Crude model 1.00 (ref) 0.57 (0.19–1.70) 2.10 (0.86–5.16) 0.03
 Age- and BMI-adjusted model 1.00 (ref) 0.43 (0.13–1.36) 1.74 (0.65–4.67) 0.018
C. Leptum
 Crude model 1.00 (ref) 0.59 (0.17–2.04) 1.26 (0.38–4.20) 0.48
 Age- and BMI-adjusted model 1.00 (ref) 0.33 (0.08–1.34) 1.24 (0.36–4.26) 0.13
A. muciniphila
 Crude model 1.00 (ref) 0.74 (0.18–3.09) 0.23 (0.06–0.97) 0.16
 Age- and BMI-adjusted model 1.00 (ref) 2.30 (0.45–11.65) 0.59 (0.10–3.72) 0.17
a.

Computed for the association between E-DII and 5 LEfSe selected taxa; the significant P-value indicates E-DII is significantly associated with abundance of the taxa

b.

Negative binomial model with log link was used to estimate eβ and 95% CI. Magnitude indicates how much more abundant the taxa (treated as count), e.g. 3-times more abundant in the highest, as compared to the lowest E-DII tertile.

Associations of E-DII with microbial species, pathways, and circulating inflammatory markers

Although the E-DII has been construct validated in several large studies4143, no significant associations were observed with inflammatory markers among the subset of subjects with fasting blood samples available, but all were in the expected direction (n=34; Supplemental Table 2). In the correlation analysis of microbial species and functional pathways with inflammatory biomarkers (n=34), Luteimonas mephitis, a low abundance species identified via LASSO based on its non-zero negative correlation with E-DII, was found to be inversely related to PAI-1 (rho= −0.40, P=0.02; B-H adjusted P=0.24). A. intestini, the taxa associated with the most pro-inflammatory diet in the overall (16S) analysis, was positively related to PAI-1 (rho=0.40, P=0.02; B-H adjusted P=0.24) in the WGS subset. Another LASSO-selected Lachnospiraceae bacterium strain associated with a more pro-inflammatory E-DII was also positively correlated with C-peptide (rho=0.41, P=0.02; B-H adjusted P=0.24). No other taxa selected from LEfSe in the larger 16S analysis, or LASSO in the WGS subset analysis were associated with circulating markers (all P values>0.05; Figure 4). Polyketide sugar unit biosynthesis was significantly positively correlated with E-DII (rho=0.32, P=0.03; B-H adjusted P=0.50). Secondary bile acid biosynthesis was positively correlated with IL-6 (rho=0.35, P=0.04; B-H adjusted P=0.50), but appeared to be modestly inversely correlated with E-DII scores favoring a more anti-inflammatory diet. Mammalian AMP-activated protein kinase (AMPK) signaling and carbon metabolism pathways, which were each modestly correlated with E-DII scores favoring a more anti-inflammatory diet, were inversely correlated with C-peptide (rho=−0.36 for AMPK and rho=−0.40 for carbon metabolism, respectively; both P values<0.05; B-H adjusted P>=0.50; Figure 5). Of note, none of these associations were statistically significant following correction for multiple testing.

Figure 4.

Figure 4.

Correlation heatmap of E-DII associated species and circulating markers among 34 subjects with residual fasting blood samples and WGS sequencing of the gut microbiome. A. intestini, A. muciniphila, R. torques, H. biformis were selected as differentially abundant OTUs in the 16S analyses while other species were selected using the LASSO method based on non-zero estimates of correlation with E-DII. **Marks the statistically significant Spearman correlations with P<0.05 before correction for multiple testing (none of the correlations were significant after B-H adjustment).

Figure 5.

Figure 5.

Correlation heatmap of E-DII associated pathways and circulating markers among 34 subjects with residual fasting blood samples and WGS sequencing of the gut microbiome. A total of 7 WGS characterized pathways were selected using the LASSO method based on non-zero estimates of correlation with E-DII. **Marks the statistically significant Spearman correlations with P<0.05 before correction for multiple testing (none of the correlations were significant after B-H adjustment).

DISCUSSION

Dietary inflammatory potential as measured by the E-DII score was associated with differential composition of specific microbes, but not overall diversity of the gut microbiome in this cross-sectional sample of 101 cancer-free individuals. In the overall analysis, R. torques, E. nodatum, A. intestini, C. leptum were more abundant in subjects consuming the most pro-inflammatory diets, while A. muciniphila was enriched in subjects with the most anti-inflammatory diets. In analysis adjusted for both age and BMI, R. torques, E. nodatum, A. intestini remained significantly associated with a more pro-inflammatory diet. In an exploratory subset of individuals with fasting blood samples and metagenomic characterization of the gut microbiome, pathways reflecting the functional capacity of the gut microbiome to support AMPK signaling, carbon metabolism, polyketide and secondary bile acid biosynthesis were associated with the inflammatory potential of the diet and/or systemic inflammation in the host.

We found several differentially abundant microbes by E-DII level. Four bacteria: R. torques, E. nodatum, A. intestini, C. leptum were associated with the most pro-inflammatory diet; and A. muciniphila was enriched in participants with the most anti-inflammatory diet. A. mucinphila and R. torques have previously been highlighted in a randomized cross-over intervention trial of the fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet among 27 irritable bowel syndrome patients and 6 healthy subjects 44. Results of this trial showed that low compared with moderate FODMAP intake was associated with a 5-fold reduction in A. mucinphila and a 1.5-fold increase in R. torques 44. FODMAPs substrates are fermented by gut bacteria to short chain fatty acids (SCFA) with anti-carcinogenic and anti-inflammatory actions 17. A. muciniphila is a producer of SCFA, primarily acetate and propionate, through mucin degradation. Mouse studies have demonstrated that a high-fat diet (i.e., pro-inflammatory diet) decreases the abundance of this species and support a causative role for this species in lowering adipose tissue inflammation and improving insulin sensitivity and lipid metabolism 45. However, the role of A. muciniphila in modulating inflammation in humans remains unclear. Two studies reported no association of A. muciniphila with systemic inflammatory markers, including high sensitivity-CRP, IL-6 and lipopolysaccharides 46,47. In a recently published randomized, double-blind, placebo-controlled pilot study of 32 overweight/obese insulin-resistant volunteers, a 3-month administration of pasteurized A. muciniphila was found to significantly decrease two enzyme activities that are thought to be involved in modulating inflammation, i.e, dipeptidyl peptidase 4 (DPP4) and γ-glutamyltransferase (GGT) 48. Previous human studies that investigated other a priori dietary patterns or indices (e.g., HEI, MDS and Healthy Food Diversity (HFD)-index) also showed that greater adherence to healthy diets was associated with increased abundance of other SCFA-producing microbes, such as Faecalibacterium prausnitzii 8,10.

Less is known with regard to other potential inflammation-related taxa observed in our study and their interactions with the inflammatory potential of diet. E. nodatum is a periodontal pathogen49 that has been linked to active rheumatoid arthritis and psoriatic arthritis characterized by chronic inflammation affecting joints and connective tissues50. C. leptum is a carbohydrate-fermenting bacteria associated with Treg responses known to modulate intestinal and systemic inflammation in experimental models; and found to be lower in patients with inflammatory bowel disease5153. Although in our study higher C. leptum was observed in healthy individuals with a more pro-inflammatory diet, this cross-sectional association was attenuated with adjustment for age and BMI. A. intestini, previously recovered from various human clinical samples of hospitalized patients54, appears to be responsive to supplementation with quercetin and partially hydrolyzed guar gum in human fecal samples55,56; and is positively associated with LPS-stimulated TNFα production57. This is generally consistent with our study finding of higher A. intestini in the most pro-inflammatory diet group; and its positive correlation with circulating PAI-1, a pro-inflammatory marker closely associated with obesity, type-2 diabetes, and CVD risk 58. Conversely, L. mephitis, a species of Proteobacteria that reduces nitrite to nitrous oxide without production of nitrogen59, was inversely correlated with E-DII and PAI-1 in our study.

Metagenomic pathways are expected to more closely reflect the functional nature of the gut microbiome as an interacting community of multiple microorganisms that supports (or hinders) the host. We found that the AMPK signaling pathway, a central regulator of cellular energy homeostasis and glucose and lipid metabolism 60 was inversely associated with E-DII and C-peptide. In both human and animal studies, activation of AMPK in adipose tissue has been linked to several anti-inflammatory dietary factors, such as n-3 polyunsaturated fatty acids, polyphenolic compounds, and fiber 61,62. However, there is little evidence to support our findings for microbial AMPK and carbon metabolism pathways with C-peptide, an insulin resistance marker 63. We also identified a positive association of secondary bile acids biosynthesis with IL-6. Primary bile acids are converted to secondary bile acids through microbial modifications in the gut; and modulate signaling via the nuclear bile acid receptors, i.e, farnesoid X receptor (FXR) and G-protein-coupled bile acid receptor (TGR5). FXR and TGR5 signaling influence many different metabolic processes in the host, including energy homeostasis, glucose homeostasis, obesity, and inflammatory responses, involving IL-6 and TNF-α 6466. The positive association between E-DII and polyketide biosynthesis is also interesting, as bacterially-derived polyketides exhibit a number of bioactive properties that modulate antibacterial, antitumor, and antiviral activities 6769. The direction of this relationship and some others (e.g., the suggestive inverse association of DII with secondary bile acids biosynthesis) are somewhat unexpected and may be chance findings due to very limited sample size (n=34). While these pathways reflect the presence of genes needed to perform a particular metabolic function, they do not necessarily reflect the gut-derived metabolites that may or may not be produced from interactions between microbes and host diet or other exposures (e.g., medications). Additionally, some pathological pathways may not be activated given the study exclusion criteria and focus on healthy individuals. Larger prospective studies are warranted to confirm or refute our findings, as this analysis was exploratory in nature and none of our findings were significant following adjustment for multiple testing.

The biologic processes underlying diet-microbiome interactions that modulate inflammation are not fully known. With a diet enriched with more anti-inflammatory components such as fiber- and polyphenol-rich plant foods, saccharolytic fermentation of carbohydrates by gut microbes can produce SCFA (i.e., butyrate, acetate and propionate). SCFAs promote anti-inflammatory responses in the host through a series of mechanisms, including intestinal homeostasis, genetic/epigenetic regulation, and immunomodulatory signaling 17. Conversely, a pro-inflammatory diet (e.g., high in fat, simple carbohydrates and meat but low in fiber-rich plant foods) creates a pro-inflammatory milieu of protein catabolites and deconjugated bile acid residues, leading to increased inflammation-related phenotypes in the host 17. These include impairment of the mucosal barrier and altered gut permeability and immune responses 19. A local inflammatory environment further alters gut microbiota to affect systemic inflammation via adherence to the gut epithelium, passage through the gut barrier to enter systemic circulation, activation of an immune response through toll-like receptor binding and/or activation of regulatory T cells, and through the synthesis and secretion of cytotoxic biomolecules or metabolites17,19,70. Additional human research is needed to elucidate the microbial pathways and metabolites that modulate diet-induced inflammation.

Interestingly, despite our findings for specific microbes and functional pathways, we did not observe significant or striking associations between gut microbial (alpha and beta) diversity and E-DII score in either the crude or adjusted analyses. While this could be attributed to insufficient variation, sample size, or residual confounding due to unmeasured factors, several studies comparing fecal microbiota diversity across participants following distinct diets (e.g., vegetarian, vegan and omnivore) also reported no, or only modest, differences in microbial diversity between diet groups 7175. Notably, the DII focuses on a balance of dietary components similar to other diet indices, but it may not be directly comparable to other studies due to its specific focus on inflammation. The HEI and MDS were significantly associated with various alpha diversity metrics in an assessment of three different dietary indices based on FFQ data from 2,070 members of the TwinsUK cohort 8. One 9 of three additional observational studies focused on increased adherence to the MDS or other diet quality indices reported a concurrent increase or difference in microbial alpha diversity 76,77. Similarly, a dietary intervention among overweight and obese subjects, as well as a companion observational study focused on healthy vs. unhealthy dietary patterns, reported increased fecal microbial gene richness (total gene counts) among participants with healthier diets 78,79.

Our study is novel in its specific focus on diet-related inflammation and its interaction with microbiome, a hypothesis rooted in biologically plausible relationships that are highly relevant to host health 14,80. Using a construct-validated tool which converts major inflammation-related dietary factors commonly consumed by Americans to an overall interpretable diet score, the E-DII provides a comprehensive summary of the dietary inflammatory potential of participants’ entire diets 4143. Although not all of the DII components were queried in our study, majority of the missing dietary components, like spices, are typically consumed in very low amounts in the US. As previously reported, the range of DII scores may rely more on the amount of foods actually consumed rather than on the number of available DII components.81 Furthermore, misclassification in dietary inflammatory potential due to the missing dietary factors would likely be non-differential, thus attenuating our results toward the null. Other limitations of nutritional epidemiologic studies also apply here, as the FFQ is prone to response bias and measurement error. Importantly, other important factors or exposures that may affect the microbiota composition (e.g., medications) were carefully collected in personal interviews and medical charts. However, we are still learning the complexity and breadth of factors that affect the microbiome or mask observed diet-microbiome associations, and our sample size limited our ability to explore multiple factors simultaneously. At the time of recruitment, we excluded individuals who reported antibiotic use in the past month, however, this may not have been sufficient for some individual’s microbial communities to “return to normal”. Recent studies demonstrate that the recovery time may vary by the type, dose, and duration of antibiotic use 82,83. Our inclusion of cancer-free patient and community volunteers was designed to maximize the variation needed to identify associations of diet with the microbiome. However, homogeneity due to exclusion criteria coupled with limitations in sample size could explain the low variation observed in alpha-diversity, beta-diversity, and other associations. Also, it may have been easier to detect differences had the range in DII scores gone towards their theoretical extremes (i.e., −9 to + 8). We were particularly limited in our subset analysis of patients with residual fasting blood samples, for which we also conducted WGS sequencing of the gut microbiome. To address this, we used LASSO, which is an efficient method to select species and pathways given a small number of subjects and the large number of zero-inflated species revealed in WGS sequencing. Taken overall, this study is of a preliminary and hypothesis-generating nature given the cross-sectional design with evaluation of diet, microbiome and blood markers at a single time point, as well as the lack of significant associations following adjustment for multiple comparisons, all of which prohibit causal and mechanistic inferences.

Dietary inflammatory potential was associated with differential composition of specific microbes but not overall gut microbiota diversity in this well-defined sample of 101 individuals. Our analysis highlighted several biologically plausible microbes potentially related to diet-induced inflammation. R. torques, E. nodatum, A. intestini, C leptum were more abundant in subjects consuming the most pro-inflammatory diets, while A. muciniphila was enriched in subjects with the most anti-inflammatory diets. Correlations between E-DII, microbes, functional gene content pathways, and inflammatory biomarkers in an exploratory subset further support the role of diet-microbiota interactions in modulating systemic inflammation in the host. Future prospective studies of dietary inflammatory potential and its interactions with the microbiome are warranted. It is increasingly important to understand how diet as a whole shapes the composition and function of the gut microbiome to modulate host inflammation, an important mechanism in the development of cancer and other major chronic diseases.

Supplementary Material

1
2
3

ACKNOWLEGEMENTS

The authors would like to acknowledge the UT MD Anderson Center for Energy Balance in Cancer Prevention and Survivorship, the Center for Translational and Public Health Genomics, as well as Dr. G. S. Raju, gastroenterologist at MD Anderson, and Jonathan Busquets, summer intern from Rice University.

FINANCIAL SUPPORT

This work was supported by a grant from The University of Texas MD Anderson Cancer Center Duncan Family Institute for Cancer Prevention and Risk Assessment (to CRD). The authors were supported by grants from the American Cancer Society RSG-17-049-01-NEC (CRD), the Cancer Prevention and Research Institute of Texas RP170259 Postdoctoral Training Fellowship (JZ), Chandler Cox Foundation (SMS), the National Cancer Institute Cancer Prevention Research Training Program R25 CA056452 (AS) and R25 CA057730 (KLH), and the National Cancer Institute Cancer Center Support Grant to MD Anderson CCSG5P30 CA016672-37 Risk, Detection and Outcomes Program (CRD, SH). The funders had no role in the design, analysis or writing of this article.

Footnotes

CONFLICT OF INTEREST

Dr. James R. Hébert owns a controlling interest in Connecting Health Innovations LLC (CHI), a company has licensed the right to his invention of the dietary inflammatory index (DII®) from the University of South Carolina in order to develop computer and smartphone applications for patient counseling and dietary intervention in clinical settings. Dr. Nitin Shivappa is an employee of CHI. The other authors have no conflict of interest to disclose.

REFERENCES

  • 1.Cotillard A, Kennedy SP, Kong LC, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585–588. [DOI] [PubMed] [Google Scholar]
  • 2.Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Flint HJ. The impact of nutrition on the human microbiome. Nutrition reviews. 2012;70 Suppl 1:S10–13. [DOI] [PubMed] [Google Scholar]
  • 4.Ley RE, Lozupone CA, Hamady M, Knight R, Gordon JI. Worlds within worlds: evolution of the vertebrate gut microbiota . Nature reviews Microbiology. 2008;6(10):776–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kinross JM, Darzi AW, Nicholson JK. Gut microbiome-host interactions in health and disease. Genome medicine. 2011;3(3):14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Daniel CR, McQuade JL. Nutrition and Cancer in the Microbiome Era. Trends in Cancer. 2019;5(9):521–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Flint HJ, Scott KP, Louis P, Duncan SH. The role of the gut microbiota in nutrition and health. Nature reviews Gastroenterology & hepatology. 2012;9(10):577–589. [DOI] [PubMed] [Google Scholar]
  • 8.Bowyer RC, Jackson MA, Pallister T, et al. Use of dietary indices to control for diet in human gut microbiota studies. Microbiome. 2018;6(1):77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Maskarinec G, Hullar MAJ, Monroe KR, et al. Fecal Microbial Diversity and Structure Are Associated with Diet Quality in the Multiethnic Cohort Adiposity Phenotype Study. J Nutr. 2019;149(9):1575–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tindall AM, Petersen KS, Kris-Etherton PM. Dietary Patterns Affect the Gut Microbiome-The Link to Risk of Cardiometabolic Diseases. The Journal of nutrition. 2018;148(9):1402–1407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC. Shifts on gut microbiota associated to mediterranean diet adherence and specific dietary intakes on general adult population. Frontiers in microbiology. 2018;9:890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016;65(11):1812–1821. [DOI] [PubMed] [Google Scholar]
  • 13.Shivappa N, Steck SE, Hurley TG, Hussey JR, Hebert JR. Designing and developing a literature-derived, population-based dietary inflammatory index. Public Health Nutr. 2014;17(8):1689–1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.O’Keefe SJD, Li JV, Lahti L, et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nature communications. 2015;6:6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mehta RS, Nishihara R, Cao Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacterium nucleatum in tumor tissue. JAMA oncology. 2017;3(7):921–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Daniel CR, McQuade JL. Nutrition and Cancer in the Microbiome Era. Trends Cancer. 2019;5(9):521–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.O’keefe SJ. Diet, microorganisms and their metabolites, and colon cancer. Nature reviews Gastroenterology & hepatology. 2016;13(12):691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hardman WE. Diet components can suppress inflammation and reduce cancer risk. Nutrition research and practice. 2014;8(3):233–240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu L, Tabung FK, Zhang X, et al. Diets That Promote Colon Inflammation Associate With Risk of Colorectal Carcinomas That Contain Fusobacterium nucleatum. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2018;16(10):1622–1631.e1623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yang J, Yu J. The association of diet, gut microbiota and colorectal cancer: what we eat may imply what we get. Protein & cell. 2018;9(5):474–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nature Reviews Microbiology. 2014;12:661. [DOI] [PubMed] [Google Scholar]
  • 22.Zitvogel L, Pietrocola F, Kroemer G. Nutrition, inflammation and cancer. Nature Immunology. 2017;18:843. [DOI] [PubMed] [Google Scholar]
  • 23.Seidell JC, Flegal KM. Assessing obesity: classification and epidemiology. Br Med Bull. 1997;53(2):238–252. [DOI] [PubMed] [Google Scholar]
  • 24.Melkonian SC, Daniel CR, Hildebrandt MAT, et al. Joint Association of Genome-Wide Association Study–Identified Susceptibility Loci and Dietary Patterns in Risk of Renal Cell Carcinoma Among Non-Hispanic Whites. American Journal of Epidemiology. 2014;180(5):499–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Block G, Coyle LM, Hartman AM, Scoppa SM. Revision of dietary analysis software for the Health Habits and History Questionnaire. American journal of epidemiology. 1994;139(12):1190–1196. [DOI] [PubMed] [Google Scholar]
  • 26.Venook AP, Papandreou C, Furuse J, de Guevara LL. The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective. Oncologist. 2010;15 Suppl 4:5–13. [DOI] [PubMed] [Google Scholar]
  • 27.Subar AF, Midthune D, Kulldorff M, et al. Evaluation of alternative approaches to assign nutrient values to food groups in food frequency questionnaires. American journal of epidemiology. 2000;152(3):279–286. [DOI] [PubMed] [Google Scholar]
  • 28.Phillips C, Shivappa N, Hébert J, Perry I. Dietary Inflammatory Index and Biomarkers of Lipoprotein Metabolism, Inflammation and Glucose Homeostasis in Adults. Nutrients. 2018;10(8):1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Shivappa N, Wirth MD, Murphy EA, Hurley TG, Hebert JR. Association between the Dietary Inflammatory Index (DII) and urinary enterolignans and C-reactive protein from the National Health and Nutrition Examination Survey-2003–2008. Eur J Nutr. 2018. [DOI] [PubMed] [Google Scholar]
  • 30.Sinha R, Chen J, Amir A, et al. Collecting Fecal Samples for Microbiome Analyses in Epidemiology Studies. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 2016;25(2):407–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Caporaso JG, Lauber CL, Walters WA, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proceedings of the National Academy of Sciences of the United States of America. 2011;108 Suppl 1:4516–4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hoffman KL, Hutchinson DS, Fowler J, et al. Oral microbiota reveals signs of acculturation in Mexican American women. PloS one. 2018;13(4):e0194100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. 2013;10(10):996–998. [DOI] [PubMed] [Google Scholar]
  • 34.Zhang Z, Schwartz S, Wagner L, Miller W. A greedy algorithm for aligning DNA sequences. J Comput Biol. 2000;7(1–2):203–214. [DOI] [PubMed] [Google Scholar]
  • 35.Stewart CJ, Ajami NJ, O’Brien JL, et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018;562(7728):583–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kanehisa M, Goto S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Research. 2000;28(1):27–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Morgan XC, Huttenhower C. Human microbiome analysis. PLoS computational biology. 2012;8(12):e1002808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.IARC MONOGRAPHS ON THE IDENTIFICATION OF CARCINOGENIC HAZARDS TO HUMANS. Agents Classified by the IARC Monographs, Volumes 1–125 WHO; 2019. https://monographs.iarc.fr/agents-classified-by-the-iarc/ Accessed 1/6/2020. [Google Scholar]
  • 39.Friedman J, Hastie T, Tibshirani R. Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw. 2010;33(1):1–22. [PMC free article] [PubMed] [Google Scholar]
  • 40.Paradis E, Claude J, Strimmer K. APE: Analyses of Phylogenetics and Evolution in R language. Bioinformatics (Oxford, England). 2004;20(2):289–290. [DOI] [PubMed] [Google Scholar]
  • 41.Tabung FK, Steck SE, Zhang J, et al. Construct validation of the dietary inflammatory index among postmenopausal women. Annals of epidemiology. 2015;25(6):398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wirth MD, Shivappa N, Davis L, et al. Construct Validation of the Dietary Inflammatory Index among African Americans. The journal of nutrition, health & aging. 2017;21(5):487–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shivappa N, Steck SE, Hurley TG, et al. A population-based dietary inflammatory index predicts levels of C-reactive protein in the Seasonal Variation of Blood Cholesterol Study (SEASONS). Public health nutrition. 2014;17(8):1825–1833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Gibson PR, Muir JG. Diets that differ in their FODMAP content alter the colonic luminal microenvironment. Gut. 2014. [DOI] [PubMed] [Google Scholar]
  • 45.Schneeberger M, Everard A, Gómez-Valadés AG, et al. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep. 2015;5:16643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Dao MC, Everard A, Aron-Wisnewsky J, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut. 2016;65(3):426–436. [DOI] [PubMed] [Google Scholar]
  • 47.de Moraes AC, Fernandes GR, da Silva IT, et al. Enterotype May Drive the Dietary-Associated Cardiometabolic Risk Factors. Frontiers in cellular and infection microbiology. 2017;7:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Depommier C, Everard A, Druart C, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nature Medicine. 2019;25(7):1096–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Haffajee A, Teles R, Socransky S. Association of Eubacterium nodatum and Treponema denticola with human periodontitis lesions. Oral microbiology and immunology. 2006;21(5):269–282. [DOI] [PubMed] [Google Scholar]
  • 50.Moen K, Brun J, Valen M, et al. Synovial inflammation in active rheumatoid arthritis and psoriatic arthritis facilitates trapping of a variety of oral bacterial DNAs. Clin Exp Rheumatol. 2006;24(6):656–663. [PubMed] [Google Scholar]
  • 51.Kabeerdoss J, Sankaran V, Pugazhendhi S, Ramakrishna BS. Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case–control study in India. BMC Gastroenterology. 2013;13(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Takaishi H, Matsuki T, Nakazawa A, et al. Imbalance in intestinal microflora constitution could be involved in the pathogenesis of inflammatory bowel disease. International Journal of Medical Microbiology. 2008;298(5–6):463–472. [DOI] [PubMed] [Google Scholar]
  • 53.Prosberg M, Bendtsen F, Vind I, Petersen AM, Gluud LL. The association between the gut microbiota and the inflammatory bowel disease activity: a systematic review and meta-analysis. Scandinavian Journal of Gastroenterology. 2016;51(12):1407–1415. [DOI] [PubMed] [Google Scholar]
  • 54.Jumas-Bilak E, Carlier JP, Jean-Pierre H, et al. Acidaminococcus intestini sp. nov., isolated from human clinical samples. Int J Syst Evol Microbiol. 2007;57(Pt 10):2314–2319. [DOI] [PubMed] [Google Scholar]
  • 55.Ghimire S, Wongkuna S, Sankaranarayanan R, Ryan EP, Bhat GJ, Scaria J. Rice Bran and Quercetin Produce a Positive Synergistic Effect on Human Gut Microbiota, Elevate the Level of Propionate, and Reduce the Population of Enterobacteriaceae family when Determined using a Bioreactor Model. bioRxiv. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Inoue R, Sakaue Y, Kawada Y, et al. Dietary supplementation with partially hydrolyzed guar gum helps improve constipation and gut dysbiosis symptoms and behavioral irritability in children with autism spectrum disorder. Journal of clinical biochemistry and nutrition. 2019;64(3):217–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell. 2016;167(4):1125–1136.e1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Yarmolinsky J, Bordin Barbieri N, Weinmann T, Ziegelmann PK, Duncan BB, Inês Schmidt M. Plasminogen activator inhibitor-1 and type 2 diabetes: a systematic review and meta-analysis of observational studies. Scientific reports. 2016;6:17714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Finkmann W, Altendorf K, Stackebrandt E, Lipski A. Characterization of N2O-producing Xanthomonas-like isolates from biofilters as Stenotrophomonas nitritireducens sp. nov., Luteimonas mephitis gen. nov., sp. nov. and Pseudoxanthomonas broegbernensis gen. nov., sp. nov. International journal of systematic and evolutionary microbiology. 2000;50(1):273–282. [DOI] [PubMed] [Google Scholar]
  • 60.Hardie DG, Ross FA, Hawley SA. AMPK - a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol. 2012;13(4):251–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wang ZQ, Yu Y, Zhang XH, et al. Comparing the effects of nano-sized sugarcane fiber with cellulose and psyllium on hepatic cellular signaling in mice. Int J Nanomedicine. 2012;7:2999–3012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Gauthier M-S, O’Brien EL, Bigornia S, et al. Decreased AMP-activated protein kinase activity is associated with increased inflammation in visceral adipose tissue and with whole-body insulin resistance in morbidly obese humans. Biochemical and biophysical research communications. 2011;404(1):382–387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Haidet J, Cifarelli V, Trucco M, Luppi P. Anti-inflammatory properties of C-Peptide. The review of diabetic studies : RDS. 2009;6(3):168–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wahlström A, Sayin SI, Marschall H-U, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell metabolism. 2016;24(1):41–50. [DOI] [PubMed] [Google Scholar]
  • 65.Cao H, Xu M, Dong W, et al. Secondary bile acid‐induced dysbiosis promotes intestinal carcinogenesis. International journal of cancer. 2017;140(11):2545–2556. [DOI] [PubMed] [Google Scholar]
  • 66.Kakiyama G, Hylemon PB, Zhou H, et al. Colonic inflammation and secondary bile acids in alcoholic cirrhosis. American journal of physiology Gastrointestinal and liver physiology. 2014;306(11):G929–937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Lombó F, Menéndez N, Salas JA, Méndez C. The aureolic acid family of antitumor compounds: structure, mode of action, biosynthesis, and novel derivatives. Applied Microbiology and Biotechnology. 2006;73(1):1–14. [DOI] [PubMed] [Google Scholar]
  • 68.Zhan J Biosynthesis of bacterial aromatic polyketides. Current topics in medicinal chemistry. 2009;9(17):1958–1610. [DOI] [PubMed] [Google Scholar]
  • 69.Arthur JC, Perez-Chanona E, Mühlbauer M, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. science. 2012;338(6103):120–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Candela M, Turroni S, Biagi E, et al. Inflammation and colorectal cancer, when microbiota-host mutualism breaks. World journal of gastroenterology: WJG. 2014;20(4):908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liszt K, Zwielehner J, Handschur M, Hippe B, Thaler R, Haslberger AG. Characterization of bacteria, clostridia and Bacteroides in faeces of vegetarians using qPCR and PCR-DGGE fingerprinting. Ann Nutr Metab. 2009;54(4):253–257. [DOI] [PubMed] [Google Scholar]
  • 72.Ferrocino I, Di Cagno R, De Angelis M, et al. Fecal Microbiota in Healthy Subjects Following Omnivore, Vegetarian and Vegan Diets: Culturable Populations and rRNA DGGE Profiling. PloS one. 2015;10(6):e0128669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zimmer J, Lange B, Frick J-S, et al. A vegan or vegetarian diet substantially alters the human colonic faecal microbiota. European journal of clinical nutrition. 2012;66(1):53. [DOI] [PubMed] [Google Scholar]
  • 74.Kabeerdoss J, Devi RS, Mary RR, Ramakrishna BS. Faecal microbiota composition in vegetarians: comparison with omnivores in a cohort of young women in southern India. British Journal of Nutrition. 2012;108(6):953–957. [DOI] [PubMed] [Google Scholar]
  • 75.Wu GD, Compher C, Chen EZ, et al. Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production. Gut. 2016;65(1):63–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Garcia-Mantrana I, Selma-Royo M, Alcantara C, Collado MC. Shifts on gut microbiota associated to Mediterranean diet adherence and specific dietary intakes on general adult population. Frontiers in Microbiology. 2018;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2015:gutjnl-2015–309957. [DOI] [PubMed] [Google Scholar]
  • 78.Cotillard A, Kennedy SP, Kong LC, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500:585. [DOI] [PubMed] [Google Scholar]
  • 79.Kong LC, Holmes BA, Cotillard A, et al. Dietary patterns differently associate with inflammation and gut microbiota in overweight and obese subjects. PloS one. 2014;9(10):e109434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Liu Y, Ajami NJ, El-Serag HB, et al. Dietary quality and the colonic mucosa–associated gut microbiome in humans. The American journal of clinical nutrition. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tabung FK, Steck SE, Liese AD, et al. Association between dietary inflammatory potential and breast cancer incidence and death: results from the Women’s Health Initiative. British journal of cancer. 2016;114(11):1277–1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Suez J, Zmora N, Zilberman-Schapira G, et al. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell. 2018;174(6):1406–1423 e1416. [DOI] [PubMed] [Google Scholar]
  • 83.Modi SR, Collins JJ, Relman DA. Antibiotics and the gut microbiota. The Journal of clinical investigation. 2014;124(10):4212–4218. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3

RESOURCES