Skip to main content
Current Developments in Nutrition logoLink to Current Developments in Nutrition
. 2017 Nov 28;2(2):nzx004. doi: 10.3945/cdn.117.001180

Does Modification of the Large Intestinal Microbiome Contribute to the Anti-Inflammatory Activity of Fermentable Fiber?

Shiu-Ming Kuo 1,
PMCID: PMC6201682  PMID: 30377676

Abstract

Fiber is an inadequately understood and insufficiently consumed nutrient. This review examines the possible causal relation between fiber-induced microbiome changes and the anti-inflammatory activity of fiber. To demonstrate the dominant role of fermentable plant fiber in shaping the intestinal microbiome, animal and human fiber-feeding studies are reviewed. Using culture-, PCR-, and sequencing-based microbial analyses, a higher prevalence of Bifidobacterium and Lactobacillus genera was observed from the feeding of different types of fermentable fiber. This finding was reported in studies performed on several host species including human. Health conditions and medications that are linked to intestinal microbial alterations likely also change the nutrient environment of the large intestine. The unique gene clusters of Bifidobacterium and Lactobacillus that enable the catabolism of plant glycans and the ability of Bifidobacterium and Lactobacillus to reduce the colonization of proteobacteria probably contribute to their prevalence in a fiber-rich intestinal environment. The fiber-induced microbiome changes could contribute to the anti-inflammatory activity of fiber. Although most studies did not measure fecal microbial density or total daily fecal microbial output (colon microbial load), limited evidence suggests that the increase in intestinal commensal microbial load plays an important role in the anti-inflammatory activity of fiber. Various probiotic supplements, including Bifidobacterium and Lactobacillus, showed anti-inflammatory activity only in the presence of fiber, which promoted microbial growth as indicated by increasing plasma short-chain fatty acids. Probiotics alone or pure fiber administered under sterile conditions showed no anti-inflammatory activity. The potential mechanisms that could mediate the anti-inflammatory effect of common microbial metabolites are reviewed, but more in vivo trials are needed. Future studies including simultaneous microbial composition and load measurements are also important.

Keywords: fiber; microbiome, Bifidobacterium; inflammation; intestinal commensal microbial load

Introduction

The definition and classification of fibers have been reviewed previously (1). Although these plant-based materials have diverse structural features, none of them can be digested in the small intestine by mammalian digestive enzymes. In the anaerobic large intestinal lumen, some of them are known to be degraded by microbial enzymes to promote commensal bacterial growth (2, 3). This unique property of fiber to undergo fermentative degradation in the large intestine gave rise to nomenclature such as “fermentable fiber” and “prebiotics” (2, 4, 5). Fermentable fibers are mostly soluble in water and thus the term “soluble fiber” is also used. Chemically, fermentable fiber can be glucose-based (polydextrose and resistant starch), fructose-based (fructo-oligosaccharide and inulin), galactose-based (galacto-oligosaccharide and agaro-oligosaccharide), or hexose derivatives such as d-galacturonic acid polymer (pectin) or galactose- and mannose-based polymer (guar gum). Some pentose carbohydrates, such as arabinose and xylose-based arabinoxylan, are also fermentable fiber.

Fiber is essential for optimal health based on its unique ability to increase fecal volume and decrease fecal transit time (6, 7). Total fiber is included in the US Dietary Reference Intake tables (8).

It is not certain, however, whether a special requirement for fermentable fiber is needed. One limitation in understanding fiber nutrition is the lack of a comprehensive database for fiber, especially soluble or fermentable fiber, in food items. Using purified fermentable fiber with different structural features, anti-inflammatory activity has been observed in human and animal model studies (9). Inflammation is the cause and consequence of many diseases (10–16), and therefore fiber can also have an impact on health outside the gastrointestinal system. Indeed, anti-inflammatory activity has been observed with fibers under different physiologic and pathologic conditions (9).

However, the mechanism leading to the anti-inflammatory activity of fiber is not clear. Is it possible that this activity of fiber relates to the promotion of certain microbes in the large intestine? If this is the case, we can then make several predictions. First, we expect similarities among various fermentable fibers in their ability to modify the intestinal microbiome. This would explain why fermentable fibers with different structural features can show similar anti-inflammatory activity (9). Second, the presence or absence of fermentable fiber should serve as an important modifier of the intestinal microbiome. This is consistent with the anti-inflammatory activity of fermentable fiber having been observed under many different physiologic and pathologic conditions (9). Third, microbiome changes by themselves, such as an increase in the target bacterial taxonomic units, should also exhibit anti-inflammatory activity independent of fiber supplementation. This would support the hypothesis that a unique microbiome mediates the anti-inflammatory effect of fermentable fiber. Lastly, specific microbial metabolites should also exhibit anti-inflammatory activity. This last suggestion would provide a possible mechanism for the hypothesized causal relation between a unique microbiome and the anti-inflammatory activity of fermentable fiber. The purpose of this review is to use the relevant literature to address the above 4 points.

Current status of knowledge

Intestinal microbiome and the method of analysis

The presence of an extensive, mostly anaerobic, microbial population in the large intestine of healthy humans and animals has long been recognized. This symbiotic relationship is important for host health as germ-free animals show poor intestinal development (17). The earliest method for the analysis of the intestinal microbiome depended on the ability to culture isolated microbes (18). The development of sequence-based analyses such as PCR and restriction fragment length polymorphism increased the capability to quantify microbes that are difficult to culture. Further development of 16S ribosomal RNA sequence-based operational taxonomic unit (OTU) analysis allowed a more complete understanding of the host-bacteria symbiotic relationship (19–22). The advancement in DNA sequencing technology and data analysis algorithms has led to the application of 16S-independent shotgun metagenomic sequencing for comprehensive OTU analysis (23).

OTU analysis has both strengths and limitations. Because of its sequence-driven comprehensive coverage at multiple taxonomic levels, it is better at predicting bacterial diversity than earlier techniques. However, because most intestinal bacterial species have not been cultured and sequenced, OTU analysis is still limited in resolving diversity below the genus level. Moreover, OTU analysis by itself gives information on the relative prevalence but not the absolute quantity of different genera. Although the older methods of analyses detect only a limited fraction of the intestinal microbiome, the data can be expressed as per gram physiologic sample and thus provide information on the absolute amount of a particular bacterial genus or species.

Shotgun metagenomic sequencing of 1135 fecal samples from a Dutch cohort revealed the presence of DNA from all 4 domains: bacteria, archaea, viruses, and nonhost eukaryotes (23). Among the sequences amplified from the fecal samples, 97.6% came from bacteria, consistent with the previous conclusion on bacterial dominance in the intestinal microbiome (24).

Multiple factors influence the composition of an individual's intestinal microbiome. For example, a maternal contribution to the initial microbiome has been observed. The first stool of newborn mice has been shown to have genetically similar microorganisms to those that were orally inoculated in their pregnant mothers (25). The exact route of maternal-fetal transmission is not clear. The birthing process can further modify the intestinal microbiome. Vaginally delivered newborns harbored bacteria found in their mother's vaginal environment, whereas skin microbiome contributed more to those born via cesarean delivery (26). However, the effect of delivery is not permanent (23). Living environment further modifies the intestinal microbiome. For example, pigs raised in an outdoor environment had a significantly higher abundance of Firmicutes, particularly Lactobacillus, than did those raised indoors (27). As expected, even subtle differences in host genetic background can influence the intestinal microbiome as shown in studies where different inbred strains of mice were raised on the same diet and in the same environment (28, 29). Nevertheless, these studies also reported a crucial effect of diet on the intestinal microbiome.

Intestinal microbiome changes upon fiber supplementation in animal studies

Results from animal studies on the effect of fermentable fiber feeding are summarized in Table 1. Observations from in vitro fiber fermentation were not included because of their limited relevance to the complex fermentation occurring in the human large intestinal lumen. Also excluded were studies using ruminants, aquatic, or avian species, studies with confounding factors including studies with disease models, studies without detailed dietary information, and studies with supplements that simultaneously introduced other nutrients.

TABLE 1.

Summary of changes in the intestinal microbiome after increasing the intake of fermentable fiber in animal studies1

Effect of fiber on microbiome
Host (age, sex, n/group) Basal diet Treatment Duration (d)2 Increase3 Decrease3 Diversity Density Ref4
Mouse
 BALB/c (3 wk, M, 8–11) No-fiber diet Supplement 3% FOS/d in tap water 14CC Bifidobacterium A (gum arabic or XOS had no effects) NA NA (33)C
 C57/BL/6J (9 wk, M, 8) Purified 35% fat, 26% CHO diet 10% arabinoxylan mixed with 90% basal diet 28C Bacteroides/Prevotella B Bifidobacterium A Roseburia F NA NA ↓ (↑total) (30)G
 ICR (4–6 wk, F, 10) AIN-93M 10% rice bran into basal diet 28F Lactobacillus F (from days 11–25) NA NA NA (44)C
 BALB/c (adult, F, 10) 2% guar gum diet 10% guar gum into basal diet 14F E. coli P NA (51)S
 C57/BL/6J (10 wk, M, 10) Cereal-based 4% fiber diet Supplement 0.3 g FOS/d in water (∼10% of diet) 56C Actinobacteria Proteobacteria Verrucomicrobia Firmicutes NA (37)S
 BALB/c (8 wk, F, 6) AIN-93G 7.5% resistant maltodextrin into basal diet 14C CoriobacteriaceaeA ClostridialesF NA NA (174)T
 C57/BL/6J (18 wk, obese M, 6) AIN-93G-based 45% fat diet 10% FOS or 10% inulin in basal diet 28F CoriobacteriaceaeALactobacillusF VerrucomicrobiaceaeV ClostridiaceaeF RuminococcaceaeF NA (45)S
 C57/BL/6N (5 wk, M, 8) 32% fat diet, plain water Basal diet and 3% wt:wt AGOS in drinking water 28F Clostridium XVIII F Lactobacillus F Prevotella B Clostridium XIVa F NA NA (46)T
 C57/BL/6J (4 wk, M, 15) Diet with 45% kcal from fat and supplemented with digestible corn starch Basal diet with 20% resistant corn starch instead 70C LactobacillaceaeF RuminococcaceaeF Clostridium F CoriobacterialesF LachnospiraceaeFRuminococcusF NA NA (43)S
Rat
 Wistar (4 wk, M, 12) AIN-93G 1% GOS in basal diet 14F Bacteroides B Bifidobacterium A Lactobacillus F NA NA (38)P
 Wistar (adult, M, 4–5) Rapeseed oil and lard-based high-fat diet 9% pectin or 8% guar gum in basal diet 21C Proteobacteria Bacteroidetes (pectin group only) Firmicutes NA (50)S
 Wistar (4 wk, F, 5) Purified diet with 5% cellulose 5% FOS replacing cellulose 28C No significant difference in BacteroidesB, BifidobacteriumA, LactobacillusF NA (52)P,G
 Sprague Dawley (22 wk, F, 14) High fat/sucrose diet Basal diet plus 10% wt:wt FOS 40F (pregnancy & lactation) Bacteroides B Bifidobacterium A Clostridium I F Clostridium IV F Clostridium XI F Roseburia F NA NA (40)P
 Wistar (4 wk, M, 6) 20% soy protein diet without fiber 2% soy fiber added in the basal diet 14C Prevotella B Clostridium F Roseburia F NA NA (175)G
Pig
 Yorkshire × Hampshire × Landrace (6 wk, MF, 4) Corn-, soy-based diet 4% different types of inulin replacing corn starch in basal diet 35CC Bifidobacterium A Lactobacillus F NA NA NA (34)T
 Duroc × Landrace × Yorkshire (28 d, MF, 5) Maize-, soy-based diet 10% wheat bran or pea fiber replacing 10% maize in basal diet 30IC Bifidobacterium A (colon) LactobacillusF (ileum) E. coli P NA (35)P
 Duroc × Landrace × Yorkshire (28 d, MF, 5) Maize-, soy-based diet 10–30% pea fiber replacing 10–30% maize in basal diet 132C Lactobacillus F NA NA (41)P
 Shade Oak × Duroc × Hypor (12 wk, MF, 3) Potato-, soy-based diet 33% resistant tapioca starch replacing potato starch in basal diet 84F ErysipelotrichaceaeF LachnospiraceaeF PrevotellaceaeB VeillonellaceaeF ClostridiaceaeF NA (49)S
 DanBred × Duroc (10 d, M, 8) Cereal-based diet 2% inulin in basal diet 40CC Bifidobacterium A (colon) NA NA NA (36)P
 Landrace × Yorkshire (1 d, MF, 10) Commercial milk replacer diet Milk replacer supplemented with 0.8% GOS 26F Bifidobacterium A Lactobacillus F NA NA (39)P
 Seghers hybrid × Piétrain (28 d, MF, 6) Purified fiber-free diet 5% arabinoxylan replacing corn starch in basal diet 30C Lactobacillus F NA NA (42)P
1

AGOS, agaro-oligosaccharide; CHO, carbohydrate; FOS, fructo-oligosaccharide; GOS, galacto-oligosaccharide; NA, information not available; XOS, xylo-oligosaccharide; —, no effect; ↑, increase; ↓, decrease.

2

The superscript abbreviation shown after the duration description indicates the sample type used: C, cecal content; CC, cecal and colon content; F, feces; IC, ileum and colon content

3

List of bacteria follows alphabetical order. The superscript abbreviation shown after each indicates the phylum it belongs to: A, Actinobacteria; B, Bacteroidetes; F, Firmicutes; P, Proteobacteria; V, Verrucomicrobiaceae.

4

The letter shown after the reference number indicates the method used for microbial analysis: C, CFU determined by plating; G, 16S-rRNA-based denaturing gradient gel electrophoresis; P, quantitative PCR; S, 16S-rRNA-based sequencing and operational taxonomic unit analysis; T, terminal restriction fragment length polymorphism.

Among the animal studies summarized in Table 1, cecal and fecal samples were primarily used to determine the intestinal microbiome, although some studies also analyzed intestinal contents. Analytical methods ranging from culture to OTU analysis were used. It is important to point out that only sequencing-based analyses have the ability to cover the entire microbiome and thus be useful for performing comprehensive diversity analyses. Other earlier methods can only quantify selective taxonomic groups where the analytic tools are available.

Three different types of microbiome-related observations could be found in the studies in Table 1: prevalence, diversity, and density. Prevalence describes the quantitative presence of a particular taxonomic group within a fixed amount of biological sample. Diversity can be used to describe the fiber feeding-induced microbial population pattern changes, if any. Density is defined as the total microbes within a fixed amount of cecal, colon, or fecal sample. A decrease in density does not necessarily reflect a reduction in total microbes at an anatomic site. For example, in the only study that measured total bacteria in the biological sample from each animal, an increase in the cecal total microbial load was reported despite a lower cecal microbial density after fiber feeding (30). Without the information on daily total fecal output in different groups, comparing fecal microbial density may not be useful.

Of the studies summarized in Table 1, only 6 studies used the sequencing-based analysis of prevalence and thus the results on prevalence are incomplete. Nevertheless, most studies found an increased presence in the phyla Actinobacteria (especially the genus Bifidobacterium), and/or Firmicutes such as Lactobacillus after fiber feeding. Some studies also observed a decrease in Clostridiaceae or Clostridium. Two studies reported a decrease in Escherichia coli, a member of Proteobacteria, although 2 others found an increase in Proteobacteria.

The review summarized in Table 1 further uncovered several important pieces of information. The studies included here were done using different basal diets and fermentable fiber supplementations given to male and female mice and rats of different strains as well as pigs of different breeds. The ages of the animals ranged from newly weaned to adult (Table 1, Host). The consistent outcome of increasing Bifidobacterium and Lactobacillus suggests that age, gender, and host genetic background do not alter the microbiome response to fermentable fibers. Although one study lasted for >100 d, a few studies found similar effects after 2 wk of fiber feeding (Table 1, Duration).

Because of the differences in substrate and oxygen availability along the gastrointestinal tract, it is expected that microbial composition varies among different parts of this tract (31, 32). Cecal samples and feces show different physical characteristics: the former freely dissociate from each other whereas the latter tend to aggregate (unpublished observation from the author's laboratory). However, based on the analyses done on cecal, colonic, and fecal samples in Table 1, it seems that the microbiome pattern change in feces represents the microbial response to fiber in the intestine reasonably well. Of the 21 studies in Table 1, an increase in Actinobacteria or Bifidobacterium in response to fermentable fiber was observed in 4 studies that examined the combined cecal and ileal and colonic contents (33–36); 2 studies that examined cecal content (30, 37); and 3 studies that examined feces (38–40). Also, of the 21 studies in Table 1, an increase in Lactobacillaceae or Lactobacillus in response to fermentable fiber was observed in 2 studies that examined the cecal and ileal and colonic contents (34, 35); 3 studies that examined cecal content (41–43); and 5 studies that examined feces (38, 39, 44–46). In a preliminary study using OTU-based Procrustes analysis of microbiomes (47), the 6 pairs of cecal-fecal samples from mice that were fed an AIN-93G diet or an AIN-93G diet supplemented with 5% inulin showed a modest cecal-fecal correlation (P = 0.0445) (unpublished observation from the author's laboratory). The consistency among these results supports the conclusion that fecal samples provide reasonably good representation at the genus level for the response of large intestinal microbiome to fermentable fiber.

Studies in animal models reveal some gaps in our knowledge of dietary regulation of the intestinal microbiome. A few recent OTU-based studies included the diversity analysis of the microbiome. Although supplementation with sugar beet pulp plus inulin was found to increase intestinal microbiome diversity if added to a diet with low fermentable fiber (48), the addition of purified fiber to a plant-based diet did not seem to have the same effect (37, 49). In fact, dietary supplementation with purified fiber tends to decrease microbiome diversity (45, 49, 50). It is not clear whether the changes in diversity as well as prevalence were due to preferential changes in the rate of growth or in the loss of certain bacteria. Understanding the basis of the observed microbiome changes requires information on microbial density and total load. This information was lacking in the diversity studies discussed above.

Microbial density of cecal or fecal samples was measured in some other studies where microbial diversity was not analyzed (Table 1) (30, 33, 35, 38, 39, 41, 42, 51, 52). When fermentable fiber was added to a diet lacking well-fermented fiber, microbial density generally increased (33, 38, 51). In contrast, the addition of fermentable fiber to a plant-based or fiber-rich diet did not increase microbial density, probably because basal microbial density was already high (30, 33, 35, 38, 39, 41, 42, 51, 52). One study weighed the total cecum content and thus allowed the calculation of the total cecal microbial load. Although fiber addition to a high-fat diet led to a decrease in the microbial density of cecal samples in that study, the total microbial load in the cecum increased (30). In this case it seems that a preferential expansion of Bifidobacterium, Prevotella, and Roseburia led to the changes in the composition of the microbiome. Although fecal samples have been used most frequently for microbiome analysis, no studies have reported the total 24-h fecal bacterial load. Such measurements could provide information on the daily bacterial turnover in the large intestine.

An important question that still needs to be addressed is the time-dependent effect of fiber. There was only one study where fecal samples were cultured to quantify Lactobacillus along a 28-d fiber supplementation time course (44). The results suggest the presence of adaptive changes: an increase in Lactobacillus that peaks at 2–3 wk of fiber supplementation followed by gradual decrease to the control level by 28 d. No information is available on the presence or absence of adaptive changes among other members of the microbiome, or on microbial diversity, density, or load.

Intestinal microbiome changes due to increased fiber intake in human studies

Overall, human studies lack dietary control prior to or even during trials. However, if the anti-inflammatory effect of fiber is related to its ability to change the intestinal microbiome, we would expect to see similar effects of fiber in the human intestinal microbiome as is observed in animal studies. Table 2 summarizes human studies that have provided fiber intake information. As with the animal studies in Table 1, human studies with potential confounding factors were excluded, including human subjects with diseases, studies without dietary information, and studies with fiber supplements that also introduced (53) non-fiber nutrients.

TABLE 2.

Summary of changes in the intestinal microbiome after increasing the intake of fermentable fiber in human subjects of various age groups1

Effect of fiber on microbiome
Type Cohort2 (n) Treatment3 Duration4 Increase5 Decrease5 Diversity Density Ref6
Observational study
 Cross-sectional on fibers from different sources 58 ± 13 y old (82) Self-administered FFQ followed by USDA nutrient database Before nongastrointestinal or oncologic surgeryF ClostridiaF (fruit, vegetable) BifidobacteriumA (bean) Porphyromona-daceaeB (bean) NA NA (176)S
Clinical Trial
 Controlled 64% carbohydrate diet supplement 21–48 y old (8) 15 g FOS/d replacing sucrose in biscuits 45 d, FOS on days 16–30F Bifidobacterium A Bacteroides B ClostridiaFFusobacteriumFU NA (56)C
 Controlled 58–64% carbohydrate diet supplement 20–34 y old (11) 4 g FOS/d as supplement in tablet and drink 42 d total, FOS on days 7–32F Bifidobacterium A NA NA (57)C
 Supplement Fiber: 35–72 y old (14); Control: 31–81 y old (15) Fiber group given a mixture of 2.5 g inulin and 2.5 g FOS supplement 3 times/d 14 d fiber then 1 d colonoscopy preparation dietCO Bifidobacterium A Eubacterium F Lactobacillus F NA NA (54)C
 Randomized double-blind placebo-controlled 18–45 y old pregnant women: fiber (17); no (16) 3 g GOS/FOS 9:1 mix or placebo (6 g maltodextrin), 3 times/d in drink From 24 to 37 wk of pregnancyF Bifidobacterium A NA NA (58)F,P
 Randomized double-blind placebo-controlled crossover 64–79 y old (41) 5.5 g GOS/d or placebo (maltodextrin) in water 70 d each with 28-d washout in betweenF Bifidobacterium A Clostridium F Eubacterium F Lactobacillus F Enterococcus F Bacteroides B Desulfovibrio P E. coli P NA (59)F
 Randomized double-blind placebo-controlled supplement Healthy formula-fed infant: fiber (22); no fiber (24) Infant formula with or without 6 g GOS/FOS 9:1 mix/L 6 mo (from birth to 6 mo old)F Bifidobacterium A NA NA NA (177)F
 Randomized double-blind placebo-controlled crossover 20–42 y old (31) 10 g very-long-chain inulin/d or placebo (maltodextrin) in water 14 d low-pre- and -probiotic diet and then 21 d each with 21-d washout in betweenF Bifidobacterium A Lactobacillus F Bacteroides-Prevotella B NA (60)F
 Supplement 21 y old (17) 10 g inulin 2 times/d 28 dF Bifidobacterium A NA NA NA (178) G,P
 Blind supplement 19–50 y old (18) 0, 2.5, 5, 10 g GOS/d in chocolate chew (sugar and corn syrup in the control chew) 21 d each in rising dose, then 14-d washoutF Bifidobacterium A Bacteroides B NA (61,62) G,P,S
 Randomized double-blind placebo-controlled crossover Men aged 28 ± 4 y (20) 0 or 21 g/d polydextrose or soluble corn fiber in 3 snack bars 21 d each with no washoutF ClostridiaceaeFDialisterFFaecalibacteriumFLactobacillusF Bifidobacterium A Dorea F Eubacterium F RuminococcusF NA NA (179)S
 Randomized double-blind placebo-controlled Healthy formula-fed infant: Fiber (53) No fiber (55) Infant formula with or without 8 g FOS and inulin 1:1 mix/L 3 mo (from birth to 3 mo old)F NA Bacteroides B EnterobacteriaceaeP NA (63)P
 Randomized double-blind placebo-controlled crossover 18–65 y old with mean BMI >30 (45) 5.5 g GOS/d or placebo (maltodextrin) in water 84 d each with 28-d washout in betweenF Bifidobacterium A Bacteroides B Desulfovibrio P NA (64)F
 Controlled 53% carbohydrate diet randomized double-blind crossover 12–15 y old (24) 0 or 12 g soluble corn fiber supplement/d in 2 meals 21 d each separated by 7-d washoutF ClostridialesFParabacteriodesB RuminococcaceaeF Coprococcus F Enterococcus F NA (53)S
 Randomized dose-response double-blind crossover Females aged 11–14 y (28) 0, 10 or 20 g soluble corn fiber supplement/d, ½ in muffin, ½ in drink (maltodextrin placebo) 28 d each separated by 28-d washoutF Bifidobacterium A Dialister F LachnospiraceaeFParabacteriodesB Anaerostipes F Dorea F RuminococcaceaeF NA (55)S
 Randomized dose-response double-blind placebo-controlled 19–56 y old (10/group, total 100) 0, 5, 10, 20 g HMOs/d or placebo (glucose) as breakfast drink 14 dF Bifidobacterium A (10 and 20 g/d groups) NA NA NA (180)S
 Supplement University undergraduate students 48 g raw potato starch/d (50% resistant starch) in 2 meal drinks 7 d after 3-d acclimationF Bifidobacterium A NA NA NA (181)S
 Randomized dose-response double-blind 18–50 y old (8/group, total 24) 8, 14, 21 g soluble corn fiber/d in 2 beverages 14 dF Bifidobacterium A (8 g/d group) NA NA (65)F
 Randomized double-blind placebo-controlled 45–70 y old, BMI 28–40 (44) 15 g GOS/d or placebo (maltodextrin) in 3 meal drinks 84 dF Bacteroides B Bifidobacterium A Prevotella B NA NA NA (182)CH
1

BMI is given in kg/m2. FOS, fructo-oligosaccharide; GOS, galacto-oligosaccharide; HMO, human milk oligosaccharides, 2′-O-fucosyllactose, lacto-N-neotetraose, or mix of the 2; NA, information not available; —, no effect; ↑, increase; ↓, decrease.

2

Cohorts included both genders unless indicated otherwise.

3

In observational study, this column shows fiber intake measurement.

4

In observational study, this column shows the study population. The superscript abbreviation shown after the study duration description indicates the sample type used: CO, colon content collected during colonoscopy; F, feces.

5

List of bacteria follows alphabetical order. The superscript abbreviation shown after each indicates the phylum it belongs to: A, Actinobacteria; B, Bacteroidetes; F, Firmicutes; FU, Fusobacteria; P, Proteobacteria.

6

The letters shown after the reference number indicate the method used for microbial analysis. C: CFU determined by plating; CH, human intestinal tract chip; F, 16S rRNA-based FISH (fluorescence in-situ hybridization) analysis; G, 16S-rRNA-based denaturing gradient gel electrophoresis; P, quantitative PCR; S, 16S-rRNA-based sequencing and operational taxonomic unit analysis.

The 3 measures of microbiome-related observations (prevalence, diversity, and density) follow the same definitions as were used in Table 1. Included in Table 2 are results of a range of studies from cross-sectional to the most stringent randomized, double-blind, placebo-controlled crossover study design. The subjects in Table 2 had a wide range of ages and were given different fermentable fibers for durations ranging from 14 d to 6 mo. With the exception of one study where samples were collected during colonoscopy, all other studies utilized fecal samples (Table 2). Overall, the effects of fermentable fiber on the intestinal microbiome are remarkably similar between the animal studies summarized in Table 1 and the human studies summarized in Table 2.

Most studies in Table 2 found an increase in the genera of Bifidobacterium and Lactobacillus upon fiber supplementation while the relative presence of the phyla of Bacteroidetes and Proteobacteria decreased in some studies. This pattern of changes was also observed in the single study analyzing colonoscopy samples (54). In the 2 studies that included observations on microbiome diversity, diversity was either unchanged or increased by fiber supplementation (53, 55). Fiber supplements also either increased or had no effect on microbial density (54, 56–65). No human studies determined the total microbial load and thus it is not possible to conclude whether the microbiome alterations were the result of preferential growth among some genera.

Some adult fiber supplement studies assessed the intestinal microbiome at >1 time point. Consistent effects among several time points ≤16 wk were reported (57, 59, 62, 64). In a multigenerational study using germ-free mice colonized with human intestinal microbiomes via oral gavage of feces, dietary fiber consistently increased the presence of Clostridiales and decreased the presence of Bacteroidales based on an OTU analysis (66).

Two studies examined the dose-dependent effect of fiber. A stepwise increase in the dose of galacto-oligosaccharide from 0 to 10 g/d led to a gradual increase in Bifidobacterium and a gradual decrease in Bacteroides in some individuals (62). Similar to many other biological processes, the effect of fiber on the microbiome was saturatable. Doubling the soluble corn fiber supplement from 10 to 20g/d for 1 mo did not lead to much additional change in the microbiome (55).

In the only study that collected colonic content during colonscopy (54), proximal and distal colonic samples had similar microbiome changes upon fiber supplementation. This is not surprising based on the similarity between cecal and fecal microbiomes in response to fiber supplementation in animal studies (Table 1).

Changes in the nutrient environment can explain other microbiome changes

A variety of factors such as nonfiber dietary component, development, and aging, as well as disease and medication, have also been reported to change the intestinal microbiome. Could those factors cause changes in the colonic nutrient environment? If so, could the nutrient environmental changes explain the observed microbiome changes? Table 3 is a summary of some additional microbiome observations and the possible role of the nutrient environment in each case.

TABLE 3.

Summary of observations on the intestinal microbiome changes that can at least be partially explained by the intestinal nutrient environment1

Host Method Observations on the intestinal microbiome (ref) Intestinal nutrient environment as a factor (ref)
Dietary pattern
 M S Feeding salmon cartilage proteoglycan led to alteration in the intestinal microbiome (68) Proteoglycan can be used by intestinal commensal as a substrate (183) and thus its ingestion can affect the microbiome
 M S Fruit or nut supplementation led to microbiome changes such as increases in Bifidobacterium and diversity (67, 69) Fruits and nuts are good dietary sources of fermentable fibers and other prebiotic compounds (67, 184)
 H,M,P S High-fat diet led to changes such as the depletion of Bacteroides and Bifidobacterium and a decrease in diversity (67, 70, 71) High-fat diet led to lower intake of fermentable fiber and thus a lower fiber presence in the intestine (185). It can also increase the unabsorbed fat in the colon (186)
Development and aging
 H A, P Fraternal twins shared developmental changes (75) Twins likely given the same breast milk, formula, and solid food
 H P, S Breastfed and formula-fed infants did not have consistent differences in intestinal microbiome from study to study (72–74) The composition of breast milk and infant formula can be different from study to study so the impact on the intestinal microbiome varied
 H S Infant gut microbiome developed features of the adult microbiome upon switching to table food (187) Infant microbiome had functional genes for the metabolism of polysaccharide in table food prior to the diet switching (187). Table food introduction likely leads to the preferential expansion of those species that have already colonized in the infant gut
 H S Different birth delivery modes led to transient but not long-term difference in microbiome (23, 26, 188) The effect of delivery mode was overridden by later-life dietary factors which then contribute to the interindividual variations (23)
 H S Elderly in long-term care facility has less microbiome diversity and the composition correlates with residence location (76) Residents in each long-term care facility have limited but quite uniform food choices and the food choices could be different between facilities (189)
Disease and medication
 H C, G, P, S, T Patients with IBD have altered intestinal microbiome such as a decrease in Bacteroidetes and Firmicutes and higher γ-Proteobacteria (190–194) IBD could lead to different dietary patterns (195) and lower fruit and fiber intake (196, 197). Although dietary protein is usually well digested and absorbed in the small intestine, host mucin and cellular protein at the site of tissue inflammation/damage could serve as unique substrates for microbes (78, 81)
 H M, S Patients using PPIs showed decreased diversity and higher presence of oral bacteria (23, 198) PPI users have different dietary pattern (199). Higher gastric luminal pH in PPI users could spare orally ingested microbes and thus a have better chance for orally ingested microbes to reach the colon (85,87)
1

Hosts included both sexes. A, microarray; C, bacterial culturing; G, 16S-rRNA-based denaturing gradient gel electrophoresis; H, human; IBD, inflammatory bowel disease; M (method), megagenomic sequencing analysis; M (host), mouse; P (method), qPCR; P (host), primate; PPI, proton pump inhibitor; ref, reference; S, 16S rRNA sequencing and operational taxonomic unit analysis; T, terminal restriction fragment length polymorphism.

The first factor examined in Table 3 is dietary pattern. The first 3 mouse-feeding studies (67–69) included macromolecules that cannot be digested and absorbed in small intestine. These macromolecules would have appeared in the large intestine as microbial nutrients and thus affected microbiome composition. The increase in both the prevalence of Bifidobacterium and the microbiome diversity upon fruit or nut ingestion is similar to the outcome of fiber feeding shown in Tables 1 and 2. This is consistent with the fact that fruits and nuts are 2 known sources of plant fiber. Based on the consistent outcome among fiber, fruit, and nut studies, we would have expected to see a decrease in the prevalence of Bifidobacterium and microbiome diversity upon switching to a low-fiber diet. This was indeed observed in several high-fat (low-fiber) diet studies (67, 70, 71).

Relevant observations were also made in several infant development and elderly studies (Table 3). Although no detailed dietary information was available, the outcome can also be at least partly explained by nutrient availability to the microbes in the large intestine. The composition of breast milk is expected to vary among women. In addition, different preparations of infant formula have different compositions. As a result, fecal Bifidobacterium content was not consistently higher in breastfed infants in these studies (72–74). In contrast, infant twins (identical or fraternal) living in the same household with similar dietary exposure have similar microbiomes (75). Elders in nursing homes have reduced exposure to environmental microbes because of their restricted mobility. This limitation would also reduce their exposure to a wide variety of food and thus encourage the development of similar and lower-diversity intestinal microbiomes within the same facility (76).

Medical conditions or the use of medications to treat these conditions could indirectly change the intestinal microbiome due to alterations in the diet as well as intestinal environment as described in the last part of Table 3. Inflammation can lead to tissue damage and the excessive presence of mucin and endogenous cellular components in the lumen (77). These cellular components are chemically different from plant-based fiber and serve as a carbon source for certain intestinal microorganisms (78–82). In patients using proton pump inhibitors, increased stomach pH can reduce protein digestion in the stomach and small intestine (83, 84). The appearance of this undigested dietary protein in the colon then changes the large intestinal nutrient environment (85) and thus can affect the microbiome. Excessive protein intake in normal individuals was also found to affect the intestinal microbiome (86). Increased stomach pH also allows a better survival of oral bacteria passing through on their way to the lower gastrointestinal tract (87).

In vitro studies on the potential mechanisms leading to changes in bacterial population following fiber feeding

Do changes in the microbiome always involve the acquisition of new bacterial strains from the environment? In studying infection by toxin-producing Clostridium difficile, it was concluded that the pathologic strains were initially present in the healthy intestine as minority members of the community and expanded when the intestinal environment became favorable (88, 89). The finding suggests that microbiome changes in response to fiber could also be the results of preferential population expansion. This is consistent with the observation that fermentable fiber supplementation leads to similar intestinal microbial responses in humans and animals, although these host organisms reside in different environments (Tables 1 and 2).

Two possible factors contribute to the preferential expansion and thus higher prevalence of Bifidobacteria and Lactobacillus upon fiber feeding. One factor may be that their genomes evolved to utilize fermentable fibers (90, 91). Bifidobacterium possesses unique gene clusters that enable the catabolism of glycans and glyco-conjugates (92). The ability of Lactobacillus to use plant materials and its niche adaptability have recently been reviewed (93). In contrast, Proteobacteria such as E. coli ferment simple sugars and amino acids preferentially (94). Some members of Fusobacterium are predominantly nonsaccharolytic (82). Desulfovibrio specializes in the reduction of sulfur found in protein and animal mucopolysaccharides (95, 96). Thus dietary supplementation with complex fermentable fiber does not give E. coli, Fusobacterium, or Desulfovibrio a growth advantage. In fact, a decrease in E. coli, Fusobacterium, and Desulfovibrio were observed in the fecal samples of some human subjects after increased fiber intake (Table 2).

The second factor contributing to the preferential expansion of Bifidobacteria and Lactobacillus is an interaction among bacterial species. The increased prevalence of Bifidobacteria and Lactobacillus upon fiber feeding may further reduce the colonization by Proteobacteria. There may be direct competition in colonization as shown in a germ-free mouse study using an artificial bacterial community to restrict Vibrio cholera colonization (97). In the literature, bacterial metabolites such as SCFAs have often been considered as growth regulators of the intestinal microbiome (98). Some probiotic culture supernatants were found to have bactericidal activity against intestinal E. coli (99), although these supernatants failed to reduce the adhesion of E. coli to intestinal epithelial cells and enhanced the biofilm formation of E. coli.

Evidence that fiber-induced microbiome changes exhibit anti-inflammatory activity

The anti-inflammatory activity of structurally different fermentable fibers has been reviewed (9). If the microbiome change upon fiber feeding contributes to the anti-inflammatory activity, we would expect similar microbiome changes to show anti-inflammatory activity in the absence of fermentable fiber. Indeed, in cell studies, anti-inflammatory activity of probiotic Bifidobacterium and Lactobacillus was observed in both macrophage (100–102) and intestinal cells (103, 104). However, the relation between so-called “beneficial bacteria” and anti-inflammatory activity is not unique. Even E. coli M17 and Saccharomyces boulardii were found to have anti-inflammatory activity in cultured macrophage and dendritic cells, respectively (105, 106).

The bacterial families and genera that were enriched upon fermentable fiber supplementation (Tables 1 and 2) overlap with those found in commercial probiotics (107). To delineate the mechanisms behind the anti-inflammatory activity of fermentable fiber, the effects of probiotics are reviewed below. Relevant publications are divided into 2 sets based on the nature of the control diet. The first set includes those using a fermentable fiber-containing diet such as a normal human diet or commercial plant-based rodent chow. The second set includes those using purified standard rodent diets (AIN-76 or AIN-93) which contain only poorly fermented cellulose. Some studies even compared the anti-inflammatory effects of prebiotics (fermentable fiber), probiotics (purified bacteria), and synbiotics (prebiotics plus probiotics).

Anti-inflammatory activity was consistently found in probiotic studies done in the presence of fermentable fiber. A chow-fed neonatal mouse study examined the effects of pre-, pro-, and synbiotic oral supplementation on pathogen-mediated intestinal inflammation (108). Both prebiotic inulin and Lactobacillus acidophilus had anti-inflammatory activity and synbiotic treatment led to a further reduction of inflammation. Probiotic Bifidobacterium bifidum PRL2010 (109), Lactobacillus reuteri (110), or Lactobacillus casei BL23 (111) reduced chemical-induced colitis or lesions in chow-fed mice. Chow-fed rats also showed reduced inflammation following chemical-induced colitis when probiotic E. coli Nissle 1917 was given (112). Nissle 1917 similarly showed anti-inflammatory effects in chow-fed mice subjected to lipopolysaccharide-induced sepsis (112). Another probiotic strain, Lactobacillus fermentum, also reduced lipopolysaccharide-induced inflammatory responses in chow-fed mice (113). In a trial of ulcerative colitis patients, synbiotic therapy (Bifidobacterium longum plus inulin) led to reduced inflammation (114). The anti-inflammatory effect of Lactobacillus can also be observed in chow-fed IL10-knockout mice that were prone to colitis (100, 115) and chow-fed rats with gastric lesions (116, 117). A diet containing high amounts of poorly fermented cellulose (30%) also showed a microbial growth-dependent anti-inflammatory effect in mouse models (118).

In contrast, in studies involving purified diets with only 5% cellulose, no anti-inflammatory effects of probiotics were observed. Synbiotic supplementation of AIN-76 diet with probiotic Bifidobacterium lactis and prebiotic-resistant starch resulted in protection against chemical-induced colon carcinogenesis in rats (119). This effect was not observed when the AIN-76 diet was supplemented with B. lactis alone (119). Resistant starch alone resulted in limited protection (119). A similar rat model was used to demonstrate the antitumorigenic activity of prebiotic inulin and a probiotic mix (Lactobacillus rhamnosus and B. lactis) in a modified AIN-76 diet (120). While inulin by itself offered some protection, probiotics by themselves had no effect (120). In rats given an AIN76-based high-fat and low-cellulose diet, inulin and synbiotic supplementations both showed anti-inflammatory activity but probiotic supplementation alone (L. rhamnosus and B. lactis in combination) had no effect (121). In a mouse study using AIN-93 diet, substituting standard casein or whey protein with ADM soy protein isolate resulted in anti-inflammatory activity (122). Because the soy protein isolate contains a significant amount of dietary fiber, fiber may have contributed to the anti-inflammatory effect observed in this study. Adding probiotic L. rhamnosus GG to the soy diet did not lead to further reduction of chemical-induced colon inflammation. L. rhamnosus GG, by itself, also did not have anti-inflammatory effect in an AIN-93-based diet (122). In vitro under sterile conditions, pure fiber showed little protection against the fungal toxin-induced inflammatory responses of cultured intestinal cells (123).

The contrasting outcomes of in vivo probiotic supplementation studies in the presence and absence of fermentable fiber as summarized above provide some insights into the anti-inflammatory activity of fermentable fiber. First, the ability of fermentable fiber to promote the growth of bacteria is important as probiotic supplementation of a purified diet with 5% poorly fermented cellulose had no effect. When indicators of bacterial growth such as total intestinal content of SCFAs or deconjugation of primary bile salts were measured, these indicators only increased in the presence of fermentable fiber (119, 124). Probiotics by themselves had no effect on SCFAs or bile salts (119, 124). Second, different probiotics all have similar protective effects. This observation suggests that the anti-inflammatory and antitumorigenic effects observed were not unique to a particular species or even genus of bacteria. In fact, even commercially cultivated Gram-negative E. coli showed protective effects (112).

The importance of total bacterial load in the colon

Recent sequencing-based intestinal microbiome analyses have made significant discoveries pertaining to microbial composition and diversity (125–128). These 2 areas have thus become a focus of health impact. However, unique composition and diversity may not be the only important factors in a healthy microbiome. Equally healthy populations were found to have different microbial composition and diversity (125, 126). As reviewed above, anti-inflammatory effects can be observed by the supplementation of different probiotics as long as a favorable growth condition, i.e., abundance of fermentable fiber, is available. In vitro analysis has directly linked glycan substrate degradation by Bacteroides to polysaccharide capsule biosynthesis (129). Thus, it is possible that simply having a higher total commensal microbial load in the colon is important for health. Unfortunately, very little information is available on this, perhaps because of practical difficulties in quantifying total microbial load, especially in human studies.

Several pieces of indirect evidence support the importance of total commensal microbial load in health promotion. Intestinal microbial losses seen after acute secretory diarrhea were similar to those seen in V. cholera infection (130). Antibiotic treatments that lead to the disruption of the intestinal microbiome (131, 132) are a major cause of recurrent C. difficile infection (133). Microbiome transplantation using feces from healthy individuals can cure C. difficile infection with no specific requirement for a particular microbial composition or diversity (134–136).

Some limited evidence also supports the hypothesis that the anti-inflammatory activity of fiber may come from its ability to increase the intestinal commensal microbial load. One rodent study in Table 1 found an increased total microbial load (total bacterial DNA in daily feces) after dietary supplementation with the fermentable fiber, arabinoxylan (30). Three other rodent studies in Table 1 (33, 38, 51) and 2 human studies in Table 2 (57, 61) found an increased fecal bacterial density (bacteria DNA/g feces) after feeding the fermentable fibers, fructo-oligosaccharide, galacto-oligosaccharide, or guar gum. The above 4 types of fermentable fibers were all found to have the anti-inflammatory activity (42, 50, 108, 120, 121, 137, 138).

To further test the physiologic importance of total commensal microbial load in the large intestine, future studies on fiber and microbiome need to include the measurement of 24-h fecal microbial output and mean fecal microbial DNA density.

The possible role of bacterial metabolites in the anti-inflammatory activity of fermentable fiber

If active microbial growth is needed for the anti-inflammatory effect of fermentable fiber as summarized above, could certain microbial metabolites contribute to this anti-inflammatory activity? Since the anti-inflammatory effects of fiber were also observed in nongastrointestinal tissues (9, 139), microbial metabolites that circulate throughout the body could be important. Intestinal microbes have unique as well as shared metabolic pathways with their hosts. A wide range of chemicals have been both predicted computationally and isolated from intestinal microbiomes (140, 141). These bacterial metabolites can be detected in the host blood and urine (142, 143). The possible anti-inflammatory activities of many of these bacterial metabolites have been studied using cultured cells or animal models (144, 145).

Fermentable fiber can contribute directly to the production of these metabolites by serving as substrates or indirectly by altering the microbiome and thus affecting the degradation of nonfiber. In this review, the focus is on bacterial metabolites that are known to be affected by fiber and have established molecular targets for their physiologic activity. The list of such metabolites may expand in the future since the physiologic significance of bacterial metabolites for the host is still being actively examined.

SCFAs are the best-studied bacterial metabolites. They can be made from fermentable fiber by many commensal microbial species and probiotics. In fact, the plasma concentration of SCFAs is a general indicator of overall intestinal microbial fermentation (51, 119, 146, 147). When the concentrations of plasma SCFAs were measured, studies that showed an anti-inflammatory effect of probiotics had higher concentrations of plasma SCFAs independent of the microbial genus and fermentable fiber source (50, 119, 120). At the molecular level, SCFAs activate several G-protein-coupled cell membrane receptors and inhibit several histone deacetylases (148–152). The immune modulatory role of SCFAs in the intestine has been directly demonstrated in mouse models and appears to be mediated through the molecular targets mentioned above (153, 154). SCFAs also reduce the severity of transplantation-induced graft-versus-host disease (154). Additionally, they reduce the level of proinflammatory cytokines and the severity of colitis in a regulatory T cell–dependent manner (153).

Other organic acids produced during fiber fermentation have also been subjects of interest. For example, fecal and plasma lactate concentrations rose when rats were given fermentable fiber (155–158). Organic acids can also serve as ligands of some G-protein-coupled cell membrane receptors (159). However, the increase in organic acids often coincides with the increase in SCFAs after fiber feeding. In addition, host enzymes using nonfiber nutrients can produce organic acids. Hypothesis-driven studies are needed before the physiologic importance of these bacteria-produced organic acids can be concluded.

The metabolites of bile salts may also contribute to the anti-inflammatory activity of fiber. The wide spectrum of biological activities of bile acids has recently been reviewed (160, 161). Primary bile salts are made in the liver and secreted into the small intestine. After serving their function in the absorption of dietary fat, taurine- and glycine-conjugated bile salts mostly return to the liver through transporter-mediated enterohepatic cycling in the terminal ileum (162). In the colon, leftover bile salts can be deconjugated by bacterial bile salt hydrolase found mainly in Gram-positive intestinal bacteria including the probiotics Lactobacillus and Bifidobacterium (161, 163, 164). In a mouse study, dietary inulin or inulin plus Bifidobacterium, but not Bifidobacterium by itself, can promote bile salt deconjugation (124).

The deconjugation reaction, by removing the growth-promoting bile salts and generating the growth-inhibiting free bile acids, can directly limit the growth of C. difficile, (165, 166). C. difficile expansion is a leading cause of diarrhea during antibiotic treatment. Bile salt deconjugation also allows the conversion of hepatic primary bile acids to secondary bile acids by bacterial enzymes (162). Secondary bile acids have higher affinity for bile acid receptors including nuclear farnesoid X receptor (FXR)α and the cell membrane G-protein-coupled receptor TGR5 (160, 161). Bile acids additionally have affinity for the nuclear pregnane X receptor (PXR), which also binds to a wide spectrum of other endogenous and exogenous chemicals (167). The PXR gene has FXR binding sites. Feeding mice with bile acid led to an FXR-dependent induction of PXR expression (168). Several lines of evidences support an anti-inflammatory activity of bile acids in the liver and intestine. Bile acids showed anti-inflammatory activity in cell studies (160, 161). Patients with inflammatory bowel diseases showed altered bile acid profiles (160, 161). Finally, mice with a FXRα or TGR5 gene knockout suffered from increased inflammation in the liver and intestine (160). However, in vivo rescue experiments using bile acids have not been done.

Colonic bacteria can degrade dietary or endogenous tryptophan to unique indoles, including skatole (169). These metabolites can bind to the nuclear aryl hydrocarbon receptor (AHR). AHR activation or gene knockout can promote or suppress inflammatory responses and have been linked to beneficial or harmful biological effects, respectively (169, 170). Two recent studies examined the effect of dietary fiber on tryptophan catabolism in the intestine. Fiber-rich soybean husk supplementation to dogs led to a change in the intestinal microbiome with an increase in fecal SCFAs and a decrease in fecal indole and skatole (157). SCFAs and indole are likely produced by different intestinal microbes that use different carbon sources (171). Patients with end-stage renal disease and restricted fruit and vegetable intake exhibited the expansion of indole-forming microbes and reduction of SCFA-producing microbes (171). Because AHR has a wide spectrum of ligands, the contribution of bacteria-produced tryptophan metabolites likely depends on other environmental and dietary conditions. The physiologic importance of tryptophan metabolites is thus far from conclusive.

Conclusions

Fiber is a required nutrient with a US Dietary Reference Intake of ∼30 g/d for adults. In developed countries, ingestion of fiber is generally low relative to recommended amounts (172). A US National Health and Nutrition Examination Survey found that only 10.3% of adults have sufficient fiber intake and the average intake is <50% of the daily requirement (173). The anti-inflammatory activity of fiber has been studied extensively (9). In this review, the ability of various fermentable fibers to increase Bifidobacteria and Lactobacillus and likely the total microbial load in the intestine is hypothesized as the mechanism leading to the anti-inflammatory activity of fermentable fiber. Diets containing only poorly fermentable fiber lead to a reduced fecal microbial content and cannot support the growth of probiotics such as Bifidobacteria and Lactobacillus. These low–fermentable fiber diets also fail to show an anti-inflammatory effect. The emerging nutritional importance of fermentable fiber supports the need to promote fiber ingestion and a specific recommendation for a daily fermentable fiber requirement.

Acknowledgments

The author thanks Wei Zheng and Kairui Wang at the University at Buffalo for the technical assistance that led to the unpublished observations. The author also thanks Christine E Campbell and Charlotte Lindqvist at the University at Buffalo for critical comments and editing of the manuscript, and Yijun Sun at the University at Buffalo for discussions during the development of the manuscript. The sole author had responsibility for all parts of the manuscript.

Notes

Supported by the University at Buffalo.

Author disclosure: no conflicts of interest.

Abbreviations used:

AHR

aryl hydrocarbon receptor

FXR

farnesoid X receptor

OTU

operational taxonomic unit

PXR

pregnane X receptor

References

  • 1. Jones J. CODEX-aligned dietary fiber definitions help to bridge the ‘fiber gap’. Nutr J 2014;13:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Gibson G, Roberfroid M. Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J Nutr 1995;125:1401–12. [DOI] [PubMed] [Google Scholar]
  • 3. Bourquin L, Titgemeyer E, Fahey GJ, Garleb K. Fermentation of dietary fibre by human colonic bacteria: disappearance of, short-chain fatty acid production from, and potential water-holding capacity of, various substrates. Scand J Gastroenterol 1993;28:249–55. [DOI] [PubMed] [Google Scholar]
  • 4. Roberfroid M. Prebiotics: the concept revisited. J Nutr 2007;137(Suppl): S830–7. [DOI] [PubMed] [Google Scholar]
  • 5. Sangwan V, Tomar S, Singh R, Singh A, Ali B. Galactooligosaccharides: novel components of designer foods. J Food Sci 2011;76:R103–11. [DOI] [PubMed] [Google Scholar]
  • 6. Connell A. Natural fiber and bowel dysfunction. Am J Clin Nutr 1976;29:1427–31. [DOI] [PubMed] [Google Scholar]
  • 7. Cummings J, Hill M, Jenkins D, Pearson J, Wiggins H. Changes in fecal composition and colonic function due to cereal fiber. Am J Clin Nutr 1976;29:1468–73. [DOI] [PubMed] [Google Scholar]
  • 8. Institute of Medicine. Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein, and amino acids. Washington (DC): National Academies Press; 2005. [Google Scholar]
  • 9. Kuo S-M. The interplay between fiber and the intestinal microbiome in the inflammatory response. Adv Nutr 2013;4:16–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Elinav E, Nowarski R, Thaiss C, Hu B, Jin C, Flavell R. Inflammation- induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer 2013;13:759–71. [DOI] [PubMed] [Google Scholar]
  • 11. Damen M, Popa C, Netea M, Dinarello C, Joosten L. Interleukin-32 in chronic inflammatory conditions is associated with a higher risk of cardiovascular diseases. Atherosclerosis 2017;264:83–91. [DOI] [PubMed] [Google Scholar]
  • 12. Tommasin S, Giannì C, De Giglio L, Pantano P. Neuroimaging techniques to assess inflammation in multiple sclerosis. Neuroscience 2017; [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 13. Lynch M, Ahern T, Sweeney C, Malara A, Tobin A, O'Shea D, Kirby B. Adipokines, psoriasis, systemic inflammation, and endothelial dysfunction. Int J Dermatol 2017;56:1103–18. [DOI] [PubMed] [Google Scholar]
  • 14. Tesch G. Diabetic nephropathy - is this an immune disorder? Clin Sci (Lond) 2017;131:2183–99. [DOI] [PubMed] [Google Scholar]
  • 15. Ni J, Wu G, Albenberg L, Tomov V. Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol 2017;14:573–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Pinzani M, Luong T. Pathogenesis of biliary fibrosis. Biochim Biophys Acta 2017;[Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 17. Thompson G, Trexler P. Gastrointestinal structure and function in germ-free or gnotobiotic animals. Gut 1971;12:230–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Rajilić-Stojanović M, de Vos W. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol Rev 2014;38:996–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Eckburg P, Bik E, Bernstein C, Purdom E, Dethlefsen L, Sargent M, Gill S, Nelson K, Relman D. Diversity of the human intestinal microbial flora. Science 2005;308:1635–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Gill S, Pop M, Deboy R, Eckburg P, Turnbaugh P, Samuel B, Gordon J, Relman D, Fraser-Liggett C, Nelson K. Metagenomic analysis of the human distal gut microbiome. Science 2006;312:1355–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. HMP Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder M, Valles-Colomer M, Vandeputte D, et al. . Population-level analysis of gut microbiome variation. Science 2016;352:560–4. [DOI] [PubMed] [Google Scholar]
  • 23. Zhernakova A, Kurilshikov A, Bonder M, Tigchelaar E, Schirmer M, Vatanen T, Mujagic Z, Vila A, Falony G, Vieira-Silva S, et al. . Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 2016;352:565–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Dubos R. The microbiota of the gastrointestinal tract. Gastroenterology 1966;51:868–74. [PubMed] [Google Scholar]
  • 25. Jiménez E, Fernández L, Marín M, Martín R, Odriozola J, Nueno-Palop C, Narbad A, Olivares M, Xaus J, Rodríguez J. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by caesarean section. Curr Microbiol 2005;51:270–4. [DOI] [PubMed] [Google Scholar]
  • 26. Dominguez-Bello M, Costello E, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci U S A 2010;107:11971–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Mulder I, Schmidt B, Stokes C, Lewis M, Bailey M, Aminov R, Prosser J, Gill B, Pluske J, Mayer C, et al. . Environmentally-acquired bacteria influence microbial diversity and natural innate immune responses at gut surfaces. BMC Biol 2009;7:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Serino M, Luche E, Gres S, Baylac A, Bergé M, Cenac C, Waget A, Klopp P, Iacovoni J, Klopp C, et al. . Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota. Gut 2012;61:543–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Menon R, Watson S, Thomas L, Allred C, Dabney A, Azcarate-Peril M, Sturino J. Diet complexity and estrogen receptor β status affect the composition of the murine intestinal microbiota. Appl Environ Microbiol 2013;79:5763–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Neyrinck A, Possemiers S, Druart C, Van de Wiele T, De Backer F, Cani P, Larondelle Y, Delzenne N. Prebiotic effects of wheat arabinoxylan related to the increase in bifidobacteria, Roseburia and Bacteroides/Prevotella in diet-induced obese mice. PLoS One 2011;6:e20944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Sommer F, Bäckhed F. Know your neighbor: microbiota and host epithelial cells interact locally to control intestinal function and physiology. Bioessays 2016;38:455–64. [DOI] [PubMed] [Google Scholar]
  • 32. Albenberg L, Esipova T, Judge C, Bittinger K, Chen J, Laughlin A, Grunberg S, Baldassano R, Lewis J, Li H, et al. . Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota in humans and mice. Gastroenterology 2014;147:1055–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Howard M, Gordon D, Garleb K, Kerley M. Dietary fructooligosaccharide, xylooligosaccharide and gum arabic have variable effects on cecal and colonic microbiota and epithelial cell proliferation in mice and rats. J Nutr 1995;125:2604–9. [DOI] [PubMed] [Google Scholar]
  • 34. Patterson J, Yasuda K, Welch R, Miller D, Lei X. Supplemental dietary inulin of variable chain lengths alters intestinal bacterial populations in young pigs. J Nutr 2010;140:2158–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Chen H, Mao X, He J, Yu B, Huang Z, Yu J, Zheng P, Chen D. Dietary fibre affects intestinal mucosal barrier function and regulates intestinal bacteria in weaning piglets. Br J Nutr 2013;10:1837–48. [DOI] [PubMed] [Google Scholar]
  • 36. Barszcz M, Taciak M, Skomiał J. The effects of inulin, dried Jerusalem artichoke tuber and a multispecies probiotic preparation on microbiota ecology and immune status of the large intestine in young pigs. Arch Anim Nutr 2016;70:278–92. [DOI] [PubMed] [Google Scholar]
  • 37. Everard A, Lazarevic V, Gaïa N, Johansson M, Ståhlman M, Backhed F, Delzenne N, Schrenzel J, François P, Cani P. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J 2014;8:2116–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Hernández-Hernández O, Marín-Manzano M, Rubio L, Moreno F, Sanz M, Clemente A. Monomer and linkage type of galacto-oligosaccharides affect their resistance to ileal digestion and prebiotic properties in rats. J Nutr 2012;42:1232–9. [DOI] [PubMed] [Google Scholar]
  • 39. Alizadeh A, Akbari P, Difilippo E, Schols H, Ulfman L, Schoterman M, Garssen J, Fink-Gremmels J, Braber S. The piglet as a model for studying dietary components in infant diets: effects of galacto-oligosaccharides on intestinal functions. Br J Nutr 2016;115:605–18. [DOI] [PubMed] [Google Scholar]
  • 40. Paul H, Bomhof M, Vogel H, Reimer R. Diet-induced changes in maternal gut microbiota and metabolomic profiles influence programming of offspring obesity risk in rats. Sci Rep 2016;6:20683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Che L, Chen H, Yu B, He J, Zheng P, Mao X, Yu J, Huang Z, Chen D. Long-term intake of pea fiber affects colonic barrier function, bacterial and transcriptional profile in pig model. Nutr Cancer 2014;66:388–99. [DOI] [PubMed] [Google Scholar]
  • 42. Chen H, Chen D, Qin W, Liu Y, Che L, Huang Z, Luo Y, Zhang Q, Lin D, Liu Y, et al. . Wheat bran components modulate intestinal bacteria and gene expression of barrier function relevant proteins in a piglet model. Int J Food Sci Nutr 2017;68:65–72. [DOI] [PubMed] [Google Scholar]
  • 43. Kieffer D, Piccolo B, Marco M, Kim E, Goodson M, Keenan M, Dunn T, Knudsen K, Martin R, Adams S. Mice fed a high-fat diet supplemented with resistant starch display marked shifts in the liver metabolome concurrent with altered gut bacteria. J Nutr 2016;146:2476–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Henderson A, Kumar A, Barnett B, Dow S, Ryan E. Consumption of rice bran increases mucosal immunoglobulin A concentrations and numbers of intestinal Lactobacillus spp. J Med Food 2012;15:469–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Liu T, Cephas K, Holscher H, Kerr K, Mangian H, Tappenden K, Swanson K. Nondigestible fructans alter gastrointestinal barrier function, gene expression, histomorphology, and the microbiota profiles of diet-Induced obese C57BL/6J mice. J Nutr 2016;146:949–56. [DOI] [PubMed] [Google Scholar]
  • 46. Higashimura Y, Naito Y, Takagi T, Uchiyama K, Mizushima K, Ushiroda C, Ohnogi H, Kudo Y, Yasui M, Inui S, et al. . Protective effect of agaro-oligosaccharides on gut dysbiosis and colon tumorigenesis in high-fat diet-fed mice. Am J Physiol Gastrointest Liver Physiol 2016;310:G367–75. [DOI] [PubMed] [Google Scholar]
  • 47. Zheng W, Tsompana M, Ruscitto A, Sharma A, Genco R, Sun Y, Buck M. An accurate and efficient experimental approach for characterization of the complex oral microbiota. Microbiome 2015;3:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Konstantinov S, Zhu W, Williams B, Tamminga S, Vos W, Akkermans A. Effect of fermentable carbohydrates on piglet faecal bacterial communities as revealed by denaturing gradient gel electrophoresis analysis of 16S ribosomal DNA. FEMS Microbiol Ecol 2003;43:225–35. [DOI] [PubMed] [Google Scholar]
  • 49. Umu Ö, Frank J, Fangel J, Oostindjer M, da Silva C, Bolhuis E, Bosch G, Willats W, Pope P, Diep D. Resistant starch diet induces change in the swine microbiome and a predominance of beneficial bacterial populations. Microbiome 2015;3:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Ghaffarzadegan T, Marungruang N, Fåk F, Nyman M. Molecular properties of guar gum and pectin modify cecal bile acids, microbiota, and plasma lipopolysaccharide-binding protein in rats. PLoS One 2016;11:e0157427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Zumbrun S, Melton-Celsa A, Smith M, Gilbreath J, Merrell D, O'Brien A. Dietary choice affects Shiga toxin-producing Escherichia coli (STEC) O157:H7 colonization and disease. Proc Natl Acad Sci U S A 2013;110:E2126–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bai G, Ni K, Tsuruta T, Nishino N. Dietary casein and soy protein isolate modulate the effects of raffinose and fructooligosaccharides on the composition and fermentation of gut microbiota in rats. J Food Sci 2016;81:H2093–8. [DOI] [PubMed] [Google Scholar]
  • 53. Whisner C, Martin B, Nakatsu C, McCabe G, McCabe L, Peacock M, Weaver C. Soluble maize fibre affects short-term calcium absorption in adolescent boys and girls: a randomised controlled trial using dual stable isotopic tracers. Br J Nutr 2014;112:446–56. [DOI] [PubMed] [Google Scholar]
  • 54. Langlands S, Hopkins M, Coleman N, Cummings J. Prebiotic carbohydrates modify the mucosa associated microflora of the human large bowel. Gut 2004;53:1610–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Whisner C, Martin B, Nakatsu C, Story J, MacDonald-Clarke C, McCabe L, McCabe G, Weaver C. Soluble corn fiber increases calcium absorption associated with shifts in the gut microbiome: a randomized dose-response trial in free-living pubertal females. J Nutr 2016;146:1298–306. [DOI] [PubMed] [Google Scholar]
  • 56. Gibson G, Beatty E, Wang X, Cummings J. Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology 1995;108:975–82. [DOI] [PubMed] [Google Scholar]
  • 57. Buddington R, Williams C, Chen S, Witherly S. Dietary supplement of neosugar alters the fecal flora and decreases activities of some reductive enzymes in human subjects. Am J Clin Nutr 1996;63:709–16. [DOI] [PubMed] [Google Scholar]
  • 58. Shadid R, Haarman M, Knol J, Theis W, Beermann C, Rjosk-Dendorfer D, Schendel D, Koletzko B, Krauss-Etschmann S. Effects of galactooligosaccharide and long-chain fructooligosaccharide supplementation during pregnancy on maternal and neonatal microbiota and immunity—a randomized, double-blind, placebo-controlled study. Am J Clin Nutr 2007;86:1426–37. [DOI] [PubMed] [Google Scholar]
  • 59. Vulevic J, Drakoularakou A, Yaqoob P, Tzortzis G, Gibson G. Modulation of the fecal microflora profile and immune function by a novel trans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers. Am J Clin Nutr 2008;88:1438–46. [DOI] [PubMed] [Google Scholar]
  • 60. Costabile A, Kolida S, Klinder A, Gietl E, Bäuerlein M, Frohberg C, Landschütze V, Gibson G. A double-blind, placebo-controlled, cross-over study to establish the bifidogenic effect of a very-long-chain inulin extracted from globe artichoke (Cynara scolymus) in healthy human subjects. Br J Nutr 2010;104:1007–17. [DOI] [PubMed] [Google Scholar]
  • 61. Davis L, Martínez I, Walter J, Hutkins R. A dose dependent impact of prebiotic galactooligosaccharides on the intestinal microbiota of healthy adults. Int J Food Microbiol 2010;144:285–92. [DOI] [PubMed] [Google Scholar]
  • 62. Davis L, Martínez I, Walter J, Goin C, Hutkins R. Barcoded pyrosequencing reveals that consumption of galactooligosaccharides results in a highly specific bifidogenic response in humans. PLoS One 2011;6:e25200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Closa-Monasterolo R, Gispert-Llaurado M, Luque V, Ferre N, Rubio-Torrents C, Zaragoza-Jordana M, Escribano J. Safety and efficacy of inulin and oligofructose supplementation in infant formula: results from a randomized clinical trial. Clin Nutr 2013;32:918–27. [DOI] [PubMed] [Google Scholar]
  • 64. Vulevic J, Juric A, Tzortzis G, Gibson G. A mixture of trans-galactooligosaccharides reduces markers of metabolic syndrome and modulates the fecal microbiota and immune function of overweight adults. J Nutr 2013;143:324–31. [DOI] [PubMed] [Google Scholar]
  • 65. Costabile A, Deaville E, Morales A, Gibson G. Prebiotic potential of a maize-based soluble fibre and impact of dose on the human gut microbiota. PLoS One 2016;11:e0144457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Sonnenburg E, Smits S, Tikhonov M, Higginbottom S, Wingreen N, Sonnenburg J. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016;529:212–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Ojo B, El-Rassi G, Payton M, Perkins-Veazie P, Clarke S, Smith B, Lucas E. Mango supplementation modulates gut microbial dysbiosis and short-chain fatty acid production independent of body weight reduction in C57BL/6 mice fed a high-fat diet. J Nutr 2016;146:1483–91. [DOI] [PubMed] [Google Scholar]
  • 68. Asano K, Yoshimura S, Nakane A. Alteration of intestinal microbiota in mice orally administered with salmon cartilage proteoglycan, a prophylactic agent. PLoS One 2013;8:e75008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Nakanishi M, Chen Y, Qendro V, Miyamoto S, Weinstock E, Weinstock G, Rosenberg D. Effects of walnut consumption on colon carcinogenesis and microbial community structure. Cancer Prev Res 2016;9:692–703. [DOI] [PubMed] [Google Scholar]
  • 70. Ma J, Prince A, Bader D, Hu M, Ganu R, Baquero K, Blundell P, Alan Harris R, Frias A, Grove K, et al. . High-fat maternal diet during pregnancy persistently alters the offspring microbiome in a primate model. Nat Commun 2014;5:3889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Chu D, Antony K, Ma J, Prince A, Showalter L, Moller M, Aagaard K. The early infant gut microbiome varies in association with a maternal high-fat diet. Genome Med 2016;8:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Ward T, Hosid S, Ioshikhes I, Altosaar I. Human milk metagenome: a functional capacity analysis. BMC Microbiol 2013;13:116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wang M, Li M, Wu S, Lebrilla C, Chapkin R, Ivanov I, Donovan S. Fecal microbiota composition of breast-fed infants is correlated with human milk oligosaccharides consumed. J Pediatr Gastroenterol Nutr 2015;60:825–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Penders J, Vink C, Driessen C, London N, Thijs C, Stobberingh E. Quantification of Bifidobacterium spp., Escherichia coli and Clostridium difficile in faecal samples of breast-fed and formula-fed infants by real-time PCR. FEMS Microbiol Lett 2005;243:141–7. [DOI] [PubMed] [Google Scholar]
  • 75. Palmer C, Bik E, DiGiulio D, Relman D, Brown P. Development of the human infant intestinal microbiota. PLoS Biol 2007;5:e177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Claesson M, Jeffery I, Conde S, Power S, O'Connor E, Cusack S, Harris H, Coakley M, Lakshminarayanan B, O'Sullivan O, et al. . Gut microbiota composition correlates with diet and health in the elderly. Nature 2012;488:178–84. [DOI] [PubMed] [Google Scholar]
  • 77. Johansson M, Hansson G. Immunological aspects of intestinal mucus and mucins. Nat Rev Immunol 2016;16:639–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Macfarlane S, Woodmansey E, Macfarlane G. Colonization of mucin by human intestinal bacteria and establishment of biofilm communities in a two-stage continuous culture system. Appl Environ Microbiol 2005;71:7483–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Marcobal A, Barboza M, Sonnenburg E, Pudlo N, Martens E, Desai P, Lebrilla C, Weimer B, Mills D, German J, et al. . Bacteroides in the infant gut consume milk oligosaccharides via mucus-utilization pathways. Cell Host Microbe 2011;10:507–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Zitomersky N, Atkinson B, Franklin S, Mitchell P, Snapper S, Comstock L, Bousvaros A. Characterization of adherent bacteroidales from intestinal biopsies of children and young adults with inflammatory bowel disease. PLoS One 2013;8:e63686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Khalil N, Walton G, Gibson G, Tuohy K, Andrews S. In vitro batch cultures of gut microbiota from healthy and ulcerative colitis (UC) subjects suggest that sulphate-reducing bacteria levels are raised in UC and by a protein-rich diet. Int J Food Sci Nutr 2014;65:79–88. [DOI] [PubMed] [Google Scholar]
  • 82. Salyers A. Energy sources of major intestinal fermentative anaerobes. Am J Clin Nutr 1979;32:158–63. [DOI] [PubMed] [Google Scholar]
  • 83. Chu S, Schubert M. Gastric secretion. Curr Opin Gastroenterol 2013;29:636–41. [DOI] [PubMed] [Google Scholar]
  • 84. Sanaka M, Yamamoto T, Kuyama Y. Effects of proton pump inhibitors on gastric emptying: a systematic review. Dig Dis Sci 2010;55:2431–40. [DOI] [PubMed] [Google Scholar]
  • 85. Evenepoel P, Claus D, Geypens B, Maes B, Hiele M, Rutgeerts P, Ghoos Y. Evidence for impaired assimilation and increased colonic fermentation of protein, related to gastric acid suppression therapy. Aliment Pharmacol Ther 1998;12:1011–9. [DOI] [PubMed] [Google Scholar]
  • 86. Rist V, Weiss E, Eklund M, Mosenthin R. Impact of dietary protein on microbiota composition and activity in the gastrointestinal tract of piglets in relation to gut health: a review. Animal 2013;7:1067–78. [DOI] [PubMed] [Google Scholar]
  • 87. Schubert M. Functional anatomy and physiology of gastric secretion. Curr Opin Gastroenterol 2015;31:479–85. [DOI] [PubMed] [Google Scholar]
  • 88. Adlerberth I, Huang H, Lindberg E, Åberg N, Hesselmar B, Saalman R, Nord C, Wold A, Weintraub A. Toxin-producing Clostridium difficile strains as long-term gut colonizers in healthy infants. J Clin Microbiol 2014;52:173–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Semenyuk E, Poroyko V, Johnston P, Jones S, Knight K, Gerding D, Driks A. Analysis of bacterial communities during Clostridium difficile infection in the mouse. Infect Immun 2015;83:4383–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Kim J, Jeong H, Yu D, Choi S, Hur C, Park M, Yoon S, Kim D, Ji G, Park H, et al. . Genome sequence of the probiotic bacterium Bifidobacterium animalis subsp. lactis AD011. J Bacteriol 2009;191:678–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Suryavanshi M, Paul D, Doijad S, Bhute S, Hingamire T, Gune R, Shouche Y. Draft genome sequence of Lactobacillus plantarum strains E2C2 and E2C5 isolated from human stool culture. Stand Genomic Sci 2017;12:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Schell M, Karmirantzou M, Snel B, Vilanova D, Berger B, Pessi G, Zwahlen M, Desiere F, Bork P, Delley M, et al. . The genome sequence of Bifidobacterium longum reflects its adaptation to the human gastrointestinal tract. Proc Natl Acad Sci U S A 2002;99:14422–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Stefanovic E, Fitzgerald G, McAuliffe O. Advances in the genomics and metabolomics of dairy lactobacilli: a review. Food Microbiol 2017;61:33–49. [DOI] [PubMed] [Google Scholar]
  • 94. Walter J, Ley R. The human gut microbiome: ecology and recent evolutionary changes. Annu Rev Microbiol 2011;65:411–29. [DOI] [PubMed] [Google Scholar]
  • 95. Barton L, Fauque G. Biochemistry, physiology and biotechnology of sulfate-reducing bacteria. Adv Appl Microbiol 2009;68:41–98. [DOI] [PubMed] [Google Scholar]
  • 96. Singh S, Lin H. Hydrogen sulfide in physiology and diseases of the digestive tract. Microorganisms 2015;3:866–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Hsiao A, Ahmed A, Subramanian S, Griffin N, Drewry L, Petri WJ, Haque R, Ahmed T, Gordon J. Members of the human gut microbiota involved in recovery from Vibrio cholerae infection. Nature 2014;515:423–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke J, Topping D, Suzuki T, et al. . Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011;469:543–7. [DOI] [PubMed] [Google Scholar]
  • 99. Miyazaki Y, Kamiya S, Hanawa T, Fukuda M, Kawakami H, Takahashi H, Yokota H. Effect of probiotic bacterial strains of Lactobacillus, Bifidobacterium, and Enterococcus on enteroaggregative Escherichia coli. J Infect Chemother 2010;16:10–8. [DOI] [PubMed] [Google Scholar]
  • 100. Peña J, Rogers A, Ge Z, Ng V, Li S, Fox J, Versalovic J. Probiotic Lactobacillus spp. diminish Helicobacter hepaticus-induced inflammatory bowel disease in interleukin-10-deficient mice. Infect Immun 2005;73:912–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Matsumoto M, Benno Y. Anti-inflammatory metabolite production in the gut from the consumption of probiotic yogurt containing Bifidobacterium animalis subsp. lactis LKM512. Biosci Biotechnol Biochem 2006;70:1287–92. [DOI] [PubMed] [Google Scholar]
  • 102. Okada Y, Tsuzuki Y, Hokari R, Komoto S, Kurihara C, Kawaguchi A, Nagao S, Miura S. Anti-inflammatory effects of the genus Bifidobacterium on macrophages by modification of phospho-I kappaB and SOCS gene expression. Int J Exp Pathol 2009;90:131–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Carey C, Kostrzynska M. Lactic acid bacteria and bifidobacteria attenuate the proinflammatory response in intestinal epithelial cells induced by Salmonella enterica serovar Typhimurium. Can J Microbiol 2013;59:9–17. [DOI] [PubMed] [Google Scholar]
  • 104. Huang I, Lin I, Liu P, Cheng M, Liu Y, Hsieh Y, Chen J, Chen C, Chang H, Shu C. Lactobacillus acidophilus attenuates Salmonella-induced intestinal inflammation via TGF-β signaling. BMC Microbiol 2015;15:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Fitzpatrick L, Small J, Hoerr R, Bostwick E, Maines L, Koltun W. In vitro and in vivo effects of the probiotic Escherichia coli strain M-17: immunomodulation and attenuation of murine colitis. Br J Nutr 2008;100:530–41. [DOI] [PubMed] [Google Scholar]
  • 106. Thomas S, Przesdzing I, Metzke D, Schmitz J, Radbruch A, Baumgart D. Saccharomyces boulardii inhibits lipopolysaccharide-induced activation of human dendritic cells and T cell proliferation. Clin Exp Immunol 2009;156:78–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Syngai G, Gopi R, Bharali R, Dey S, Lakshmanan G, Ahmed G. Probiotics - the versatile functional food ingredients. J Food Sci Technol 2016;53:921–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Foye O, Huang I, Chiou C, Walker W, Shi H. Early administration of probiotic Lactobacillus acidophilus and/or prebiotic inulin attenuates pathogen-mediated intestinal inflammation and Smad 7 cell signaling. FEMS Immunol Med Microbiol 2012;65:467–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Duranti S, Gaiani F, Mancabelli L, Milani C, Grandi A, Bolchi A, Santoni A, Lugli G, Ferrario C, Mangifesta M, et al. . Elucidating the gut microbiome of ulcerative colitis: bifidobacteria as novel microbial biomarkers. FEMS Microbiol Ecol 2016;92:[Epub]. [DOI] [PubMed] [Google Scholar]
  • 110. Ahl D, Liu H, Schreiber O, Roos S, Phillipson M, Holm L. Lactobacillus reuteri increases mucus thickness and ameliorates dextran sulphate sodium-induced colitis in mice. Acta Physiol 2016;217:300–10. [DOI] [PubMed] [Google Scholar]
  • 111. Lenoir M, Del Carmen S, Cortes-Perez N, Lozano-Ojalvo D, Muñoz-Provencio D, Chain F, Langella P, de Moreno de LeBlanc A, LeBlanc J, Bermúdez-Humarán L. Lactobacillus casei BL23 regulates Treg and Th17 T-cell populations and reduces DMH-associated colorectal cancer. J Gastroenterol 2016;51:862–73. [DOI] [PubMed] [Google Scholar]
  • 112. Arribas B, Rodríguez-Cabezas M, Camuesco D, Comalada M, Bailón E, Utrilla P, Nieto A, Concha A, Zarzuelo A, Gálvez J. A probiotic strain of Escherichia coli, Nissle 1917, given orally exerts local and systemic anti-inflammatory effects in lipopolysaccharide-induced sepsis in mice. Br J Pharmacol 2009;157:1024–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Arribas B, Rodríguez-Cabezas M, Comalada M, Bailón E, Camuesco D, Olivares M, Xaus J, Zarzuelo A, Gálvez J. Evaluation of the preventative effects exerted by Lactobacillus fermentum in an experimental model of septic shock induced in mice. Br J Nutr 2009;101:51–8. [DOI] [PubMed] [Google Scholar]
  • 114. Furrie E, Macfarlane S, Kennedy A, Cummings J, Walsh S, O'neil D, Macfarlane G. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut 2005;54:242–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Madsen K, Doyle J, Jewell L, Tavernini M, Fedorak R. Lactobacillus species prevents colitis in interleukin 10 gene-deficient mice. Gastroenterology 1999;116:1107–14. [DOI] [PubMed] [Google Scholar]
  • 116. Uchida M, Kurakazu K. Yogurt containing Lactobacillus gasseri OLL2716 exerts gastroprotective action against acute gastric lesion and antral ulcer in rats. J Pharmacol Sci 2004;96:84–90. [DOI] [PubMed] [Google Scholar]
  • 117. Uchida M, Shimizu K, Kurakazu K. Yogurt containing Lactobacillus gasseri OLL 2716 (LG21 yogurt) accelerated the healing of acetic acid-induced gastric ulcer in rats. Biosci Biotechnol Biochem 2010;74:1891–4. [DOI] [PubMed] [Google Scholar]
  • 118. Morowitz M, Di Caro V, Pang D, Cummings J, Firek B, Rogers M, Ranganathan S, Clark R, Aneja R. Dietary supplementation with nonfermentable fiber alters the gut microbiota and confers protection in murine models of sepsis. Crit Care Med 2017;45:e516–e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Le Leu R, Hu Y, Brown I, Woodman R, Young G. Synbiotic intervention of Bifidobacterium lactis and resistant starch protects against colorectal cancer development in rats. Carcinogenesis 2010;31:246–51. [DOI] [PubMed] [Google Scholar]
  • 120. Femia A, Luceri C, Dolara P, Giannini A, Biggeri A, Salvadori M, Clune Y, Collins K, Paglierani M, Caderni G. Antitumorigenic activity of the prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis on azoxymethane-induced colon carcinogenesis in rats. Carcinogenesis 2002;23:1953–60. [DOI] [PubMed] [Google Scholar]
  • 121. Roller M, Rechkemmer G, Watzl B. Prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis modulates intestinal immune functions in rats. J Nutr 2004;134:153–6. [DOI] [PubMed] [Google Scholar]
  • 122. Jiang H, Przybyszewski J, Mitra D, Becker C, Brehm-Stecher B, Tentinger A, MacDonald R. Soy protein diet, but not Lactobacillus rhamnosus GG, decreases mucin-1, trefoil factor-3, and tumor necrosis factor-α in colon of dextran sodium sulfate-treated C57BL/6 mice. J Nutr 2011;141:1239–46. [DOI] [PubMed] [Google Scholar]
  • 123. Akbari P, Fink-Gremmels J, Willems R, Difilippo E, Schols H, Schoterman M, Garssen J, Braber S. Characterizing microbiota-independent effects of oligosaccharides on intestinal epithelial cells: insight into the role of structure and size: structure-activity relationships of non-digestible oligosaccharides. Eur J Nutr 2017;56:1919–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Kuo S-M, Merhige PM, Hagey LR. The effect of dietary prebiotics and probiotics on body weight, large intestine indices, and fecal bile acid profile in wildtype and IL10-/- mice. PLoS One 2013;8:e60270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet J, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A 2010;107:14691–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Lin A, Bik E, Costello E, Dethlefsen L, Haque R, Relman D, Singh U. Distinct distal gut microbiome diversity and composition in healthy children from Bangladesh and the United States. PLoS One 2013;8:e53838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Karlsson F, Tremaroli V, Nookaew I, Bergström G, Behre C, Fagerberg B, Nielsen J, Bäckhed F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013;498:99–103. [DOI] [PubMed] [Google Scholar]
  • 128. Ridaura V, Faith J, Rey F, Cheng J, Duncan A, Kau A, Griffin N, Lombard V, Henrissat B, Bain J, et al. . Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science 2013;341:1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Martens E, Roth R, Heuser J, Gordon J. Coordinate regulation of glycan degradation and polysaccharide capsule biosynthesis by a prominent human gut symbiont. J Biol Chem 2009;284:18445–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. David L, Weil A, Ryan E, Calderwood S, Harris J, Chowdhury F, Begum Y, Qadri F, LaRocque R, Turnbaugh P. Gut microbial succession follows acute secretory diarrhea in humans. MBio 2015;6:e00381–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Dethlefsen L, Relman D. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 2011;108 Suppl 1:S4554–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Macfarlane S. Antibiotic treatments and microbes in the gut. Environ Microbiol 2014;16:919–24. [DOI] [PubMed] [Google Scholar]
  • 133. Razik R, Rumman A, Bahreini Z, McGeer A, Nguyen G. Recurrence of Clostridium difficile infection in patients with inflammatory bowel disease: the RECIDIVISM study. Am J Gastroenterol 2016;111:1141–6. [DOI] [PubMed] [Google Scholar]
  • 134. Shahinas D, Silverman M, Sittler T, Chiu C, Kim P, Allen-Vercoe E, Weese S, Wong A, Low D, Pillai D. Toward an understanding of changes in diversity associated with fecal microbiome transplantation based on 16S rRNA gene deep sequencing. MBio 2012;3:e00338–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Staley C, Hamilton M, Vaughn B, Graiziger C, Newman K, Kabage A, Sadowsky M, Khoruts A. Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study. Am J Gastroenterol 2017;112:940–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Satokari R, Fuentes S, Mattila E, Jalanka J, de Vos W, Arkkila P. Fecal transplantation treatment of antibiotic-induced, noninfectious colitis and long-term microbiota follow-up. Case Rep Med 2014;2014:913867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Morel F, Dai Q, Ni J, Thomas D, Parnet P, Fança-Berthon P. α-Galacto-oligosaccharides dose-dependently reduce appetite and decrease inflammation in overweight adults. J Nutr 2015;145:2052–9. [DOI] [PubMed] [Google Scholar]
  • 138. Bouchaud G, Castan L, Chesné J, Braza F, Aubert P, Neunlist M, Magnan A, Bodinier M. Maternal exposure to GOS/inulin mixture prevents food allergies and promotes tolerance in offspring in mice. Allergy 2016;71:68–76. [DOI] [PubMed] [Google Scholar]
  • 139. Trompette A, Gollwitzer E, Yadava K, Sichelstiel A, Sprenger N, Ngom-Bru C, Blanchard C, Junt T, Nicod L, Harris N, et al. . Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat Med 2014;20:159–66. [DOI] [PubMed] [Google Scholar]
  • 140. Nicholson J, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science 2012;336:1262–7. [DOI] [PubMed] [Google Scholar]
  • 141. Donia M, Fischbach M. HUMAN MICROBIOTA. Small molecules from the human microbiota. Science 2015;349:1254766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Zhang M, et al. . Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A 2008;195:2117–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Wikoff W, Anfora A, Liu J, Schultz P, Lesley S, Peters E, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A 2009;106:3698–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Levy M, Thaiss C, Elinav E. Metabolites: messengers between the microbiota and the immune system. Genes Dev 2016;30:1589–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Blander J, Longman R, Iliev I, Sonnenberg G, Artis D. Regulation of inflammation by microbiota interactions with the host. Nat Immunol 2017;18:851–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Bugaut M. Occurrence, absorption and metabolism of short chain fatty acids in the digestive tract of mammals. Comp Biochem Physiol B 1987;86:439–72. [DOI] [PubMed] [Google Scholar]
  • 147. Cummings J, Macfarlane G. Role of intestinal bacteria in nutrient metabolism. JPEN J Parenter Enteral Nutr 1997;21:357–65. [DOI] [PubMed] [Google Scholar]
  • 148. Tazoe H, Otomo Y, Karaki S, Kato I, Fukami Y, Terasaki M, Kuwahara A. Expression of short-chain fatty acid receptor GPR41 in the human colon. Biomed Res 2009;30:149–56. [DOI] [PubMed] [Google Scholar]
  • 149. Yajima T, Inoue R, Yajima M, Tsuruta T, Karaki S, Hira T, Kuwahara A. The G-protein on cholesterol-rich membrane microdomains mediates mucosal sensing of short-chain fatty acid and secretory response in rat colon. Acta Physiol 2011;203:381–9. [DOI] [PubMed] [Google Scholar]
  • 150. Natarajan N, Hori D, Flavahan S, Steppan J, Flavahan N, Berkowitz D, Pluznick J. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G-protein coupled receptor 41. Physiol Genomics 2016;48:826–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Tan J, McKenzie C, Potamitis M, Thorburn A, Mackay C, Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol 2014;121:91–119. [DOI] [PubMed] [Google Scholar]
  • 152. Koh A, De Vadder F, Kovatcheva-Datchary P, Bäckhed F. From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites. Cell 2016;165:1332–45. [DOI] [PubMed] [Google Scholar]
  • 153. Smith P, Howitt M, Panikov N, Michaud M, Gallini C, Bohlooly-Y M, Glickman J, Garrett W.. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341:569–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Mathewson N, Jenq R, Mathew A, Koenigsknecht M, Hanash A, Toubai T, Oravecz-Wilson K, Wu S, Sun Y, Rossi C, et al. . Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease. Nat Immunol 2016;17:505–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Ramirez-Farias C, Slezak K, Fuller Z, Duncan A, Holtrop G, Louis P. Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii. Br J Nutr 2009;101:541–50. [DOI] [PubMed] [Google Scholar]
  • 156. Liu G, Xiao L, Fang T, Cai Y, Jia G, Zhao H, Wang J, Chen X, Wu C. Pea fiber and wheat bran fiber show distinct metabolic profiles in rats as investigated by a 1H NMR-based metabolomic approach. PLoS One 2014;9:e115561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Myint H, Iwahashi Y, Koike S, Kobayashi Y. Effect of soybean husk supplementation on the fecal fermentation metabolites and microbiota of dogs. Anim Sci J 2017; [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 158. Kröger S, Vahjen W, Zentek J. Influence of lignocellulose and low or high levels of sugar beet pulp on nutrient digestibility and the fecal microbiota in dogs. J Anim Sci 2017;95:1598–605. [DOI] [PubMed] [Google Scholar]
  • 159. Ristic B, Bhutia Y, Ganapathy V. Cell-surface G-protein-coupled receptors for tumor-associated metabolites: a direct link to mitochondrial dysfunction in cancer. Biochim Biophys Acta 2017;1868:246–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Martinot E, Sèdes L, Baptissart M, Lobaccaro J, Caira F, Beaudoin C, Volle D. Bile acids and their receptors. Mol Aspects Med 2017;56:2–9. [DOI] [PubMed] [Google Scholar]
  • 161. Long S, Gahan C, Joyce S. Interactions between gut bacteria and bile in health and disease. Mol Aspects Med 2017;56:54–65. [DOI] [PubMed] [Google Scholar]
  • 162. Hofmann A, Hagey L. Bile acids: chemistry, pathochemistry, biology, pathobiology, and therapeutics. Cell Mol Life Sci 2008;65:2461–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Begley M, Hill C, Gahan C. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol 2006;72:1729–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Ridlon J, Kang D, Hylemon P. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 2006;47:241–59. [DOI] [PubMed] [Google Scholar]
  • 165. Sorg J, Sonenshein A. Bile salts and glycine as cogerminants for Clostridium difficile spores. J Bacteriol 2008;190:2505–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Sorg J, Sonenshein A. Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. J Bacteriol 2010;192:4983–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Kliewer S, Goodwin B, Willson T. The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocr Rev 2002;23:687–702. [DOI] [PubMed] [Google Scholar]
  • 168. Jung D, Mangelsdorf D, Meyer U. Pregnane X receptor is a target of farnesoid X receptor. J Biol Chem 2006;281:19081–91. [DOI] [PubMed] [Google Scholar]
  • 169. Hubbard T, Murray I, Perdew G. Indole and tryptophan metabolism: endogenous and dietary routes to Ah receptor activation. Drug Metab Dispos 2015;43:1522–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Lawrence B, Vorderstrasse B. New insights into the aryl hydrocarbon receptor as a modulator of host responses to infection. Semin Immunopathol 2013;35:615–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Wong J, Piceno Y, DeSantis T, Pahl M, Andersen G, Vaziri N. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am J Nephrol 2014;39:230–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Blumfield M, Hure A, Macdonald-Wicks L, Smith R, Collins C. Systematic review and meta-analysis of energy and macronutrient intakes during pregnancy in developed countries. Nutr Rev 2012;70:322–36. [DOI] [PubMed] [Google Scholar]
  • 173. McGill C, Birkett A, Fulgonii Iii V. Healthy Eating Index-2010 and food groups consumed by US adults who meet or exceed fiber intake recommendations NHANES 2001–2010. Food Nutr Res 2016;60:29977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Miyazato S, Kishimoto Y, Takahashi K, Kaminogawa S, Hosono A. Continuous intake of resistant maltodextrin enhanced intestinal immune response through changes in the intestinal environment in mice. Biosci Microbiota Food Health 2016;35:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Nakata T, Kyoui D, Takahashi H, Kimura B, Kuda T. Inhibitory effects of soybean oligosaccharides and water-soluble soybean fibre on formation of putrefactive compounds from soy protein by gut microbiota. Int J Biol Macromol 2017;97:173–80. [DOI] [PubMed] [Google Scholar]
  • 176. Dominianni C, Sinha R, Goedert J, Pei Z, Yang L, Hayes R, Ahn J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS One 2015;10:e0124599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Scholtens P, Alliet P, Raes M, Alles M, Kroes H, Boehm G, Knippels L, Knol J, Vandenplas Y. Fecal secretory immunoglobulin A is increased in healthy infants who receive a formula with short-chain galacto-oligosaccharides and long-chain fructo-oligosaccharides. J Nutr 2008;138:1141–7. [DOI] [PubMed] [Google Scholar]
  • 178. Joossens M, Huys G, Van Steen K, Cnockaert M, Vermeire S, Rutgeerts P, Verbeke K, Vandamme P, De Preter V. High-throughput method for comparative analysis of denaturing gradient gel electrophoresis profiles from human fecal samples reveals significant increases in two bifidobacterial species after inulin-type prebiotic intake. FEMS Microbiol Ecol 2011;75:343–9. [DOI] [PubMed] [Google Scholar]
  • 179. Hooda S, Boler B, Serao M, Brulc J, Staeger M, Boileau T, Dowd S, Fahey GJ, Swanson K. 454 pyrosequencing reveals a shift in fecal microbiota of healthy adult men consuming polydextrose or soluble corn fiber. J Nutr 2012;142:1259–65. [DOI] [PubMed] [Google Scholar]
  • 180. Elison E, Vigsnaes L, Rindom Krogsgaard L, Rasmussen J, Sørensen N, McConnell B, Hennet T, Sommer M, Bytzer P. Oral supplementation of healthy adults with 2'-O-fucosyllactose and lacto-N-neotetraose is well tolerated and shifts the intestinal microbiota. Br J Nutr 2016;16:1356–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Venkataraman A, Sieber J, Schmidt A, Waldron C, Theis K, Schmidt T. Variable responses of human microbiomes to dietary supplementation with resistant starch. Microbiome 2016;4:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Canfora E, van der Beek C, Hermes G, Goossens G, Jocken J, Holst J, van Eijk H, Venema K, Smidt H, Zoetendal E, et al. . Supplementation of diet with galacto-oligosaccharides increases Bifidobacteria, but not insulin sensitivity, in obese prediabetic individuals. Gastroenterology 2017;153:87–97. [DOI] [PubMed] [Google Scholar]
  • 183. Kuritza A, Salyers A. Digestion of proteoglycan by Bacteroides thetaiotaomicron. J Bacteriol 1983;153:1180–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Lamuela-Raventos R, St Onge M. Prebiotic nut compounds and human microbiota. Crit Rev Food Sci Nutr 2017;57:3154–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Johnson L, Mander A, Jones L, Emmett P, Jebb S. Energy-dense, low-fiber, high-fat dietary pattern is associated with increased fatness in childhood. Am J Clin Nutr 2008;87:846–54. [DOI] [PubMed] [Google Scholar]
  • 186. Porsgaard T, Høy C. Lymphatic transport in rats of several dietary fats differing in fatty acid profile and triacylglycerol structure. J Nutr 2000;130:1619–24. [DOI] [PubMed] [Google Scholar]
  • 187. Koenig J, Spor A, Scalfone N, Fricker A, Stombaugh J, Knight R, Angenent L, Ley R. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci U S A 2011;108 Suppl 1:4578–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Liu D, Yu J, Li L, Ai Q, Feng J, Song C, Li H. Bacterial community structure associated with elective cesarean section versus vaginal delivery in chinese newborns. J Pediatr Gastroenterol Nutr 2015;60:240–6. [DOI] [PubMed] [Google Scholar]
  • 189. Pezzana A, Cereda E, Avagnina P, Malfi G, Paiola E, Frighi Z, Capizzi I, Sgnaolin E, Amerio ML. Nutritional care needs in elderly residents of long-term care institutions: potential implications for policies. J Nutr Health Aging 2015;19:947–54. [DOI] [PubMed] [Google Scholar]
  • 190. Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, Vandamme P, Vermeire S. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut 2011;60:631–7. [DOI] [PubMed] [Google Scholar]
  • 191. Elson C, Alexander K. Host-microbiota interactions in the intestine. Dig Dis 2015;33:131–6. [DOI] [PubMed] [Google Scholar]
  • 192. Frank D, St Amand A, Feldman R, Boedeker E, Harpaz N, Pace N. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A 2007;104:13780–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Li Q, Wang C, Tang C, Li N, Li J. Molecular-phylogenetic characterization of the microbiota in ulcerated and non-ulcerated regions in the patients with Crohn's disease. PLoS One 2012;7:e34939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Nemoto H, Kataoka K, Ishikawa H, Ikata K, Arimochi H, Iwasaki T, Ohnishi Y, Kuwahara T, Yasutomo K. Reduced diversity and imbalance of fecal microbiota in patients with ulcerative colitis. Dig Dis Sci 2012;57:2955–64. [DOI] [PubMed] [Google Scholar]
  • 195. Ballegaard M, Bjergstrøm A, Brøndum S, Hylander E, Jensen L, Ladefoged K. Self-reported food intolerance in chronic inflammatory bowel disease. Scand J Gastroenterol 1997;32:569–71. [DOI] [PubMed] [Google Scholar]
  • 196. Russel M, Engels L, Muris J, Limonard C, Volovics A, Brummer R, Stockbrügger R. Modern life' in the epidemiology of inflammatory bowel disease: a case-control study with special emphasis on nutritional factors. Eur J Gastroenterol Hepatol 1998;10:243–9. [DOI] [PubMed] [Google Scholar]
  • 197. Brotherton C, Taylor A. Dietary fiber information for individuals with Crohn disease: reports of gastrointestinal effects. Gastroenterol Nurs 2013;36:320–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Imhann F, Bonder M, Vich Vila A, Fu J, Mujagic Z, Vork L, Tigchelaar E, Jankipersadsing S, Cenit M, Harmsen H, et al. . Proton pump inhibitors affect the gut microbiome. Gut 2016;65:740–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Yamamichi N, Mochizuki S, Asada-Hirayama I, Mikami-Matsuda R, Shimamoto T, Konno-Shimizu M, Takahashi Y, Takeuchi C, Niimi K, Ono S, et al. . Lifestyle factors affecting gastroesophageal reflux disease symptoms: a cross-sectional study of healthy 19864 adults using FSSG scores. BMC Med 2012;10:45. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Developments in Nutrition are provided here courtesy of American Society for Nutrition

RESOURCES