Abstract
The rising incidence of inflammatory bowel diseases in recent decades has notably paralleled changing lifestyle habits in Western nations, which are now making their way into more traditional societies. Diet plays a key role in IBD pathogenesis, and there is a growing appreciation that the interaction between diet and microbes in a susceptible person contributes significantly to the onset of disease. In this review, we examine what is known about dietary and microbial factors that promote IBD. We summarize recent findings regarding the effects of diet in IBD epidemiology from prospective population cohort studies, as well as new insights into IBD-associated dysbiosis. Microbial metabolism of dietary components can influence the epithelial barrier and the mucosal immune system, and understanding how these interactions generate or suppress inflammation will be a significant focus of IBD research. Our knowledge of dietary and microbial risk factors for IBD provides important considerations for developing therapeutic approaches through dietary modification or re-shaping the microbiota. We conclude by calling for increased sophistication in designing studies on the role of diet and microbes in IBD pathogenesis and disease resolution in order to accelerate progress in response to the growing challenge posed by these complex disorders.
Keywords: Diet, IBD, microbiota, pathogenesis, microbial metabolism
1 Introduction
Inflammatory bowel diseases (IBD), which include Crohn’s disease (CD) and ulcerative colitis (UC), present a significant global health challenge. The worldwide incidence rates for CD and UC have risen in recent decades [1]. As with many other complex diseases of immunity, epidemiological trends in the occurrence of IBD suggest a link to Western culture and lifestyles. The highest incidence and prevalence of IBD is found in the United States and Europe [1]. IBD are expanding rapidly in Asia, particularly in Japan, India, and the Middle East, where Westernization has progressed significantly [2]. Migrants who come into contact with Western societies appear to be at high risk of developing IBD, even after they return to their home countries [3].
Genetic and environmental factors play a critical role in IBD pathogenesis. Genetic analyses of IBD have identified over 200 loci associated with UC or CD risk [4–6]. Meanwhile, a variety of environmental and behavioral factors are linked to IBD pathogenesis, including diet, smoking, stress, sleep patterns, hygiene, and antibiotic use [7]. The intestinal microbiota represents another major contributor to disease risk [8]. Unraveling the complex network of interactions underlying IBD pathogenesis is essential for making advances in the diagnosis and treatment of these disorders that could curtail their global expansion.
There is a great deal of interest in the link between diet, the intestinal microbiota, and IBD [9–11]. Diet plays important roles in intestinal function and immune activity. It also shapes the intestinal microbiota community structure [12] and influences community function by acting as a source of material for microbial metabolism [13]. Both diet and the microbiota are seen as potential targets for therapeutic intervention in IBD. In this Review, we will summarize recent advances in our knowledge of how diet and the microbiota, acting independently or in concert, contribute to IBD risk. We will also discuss these findings in the context of efforts to develop IBD treatment focused on manipulating diet or the microbiota.
2 Diet as a risk factor in IBD
The ability of specific foods or dietary patterns to influence the risk of CD and UC has been extensively studied. Early investigations relied on retrospective case-control studies; while these studies assessed a broad range of dietary factors, a number of reports produced conflicting results on the benefit or detriment of certain foods, and many were unable to produce statistically significant results due to small sample size [9, 14]. In recent years, prospective studies of large population cohorts in North America and Europe (Table 1) have attempted to resolve previous discrepancies and controversies regarding dietary effects of macronutrient composition on IBD risk, but with varying success.
Table 1.
Diet factor | Sample features | Findings | Reference |
---|---|---|---|
A. European Prospective Investigation into Cancer and Nutrition (EPIC) (nested case-control studies) | |||
Dietary pattern | Cohort size=366,351; UC: cases=256, controls=1022; CD: cases=117, controls=468 | “Sugar and soft drinks” diet increased risk in UC cases diagnosed >2 years after diet survey (Highest vs lowest quintile: IRR 1.68, 95% CI 1.00–2.82, p=0.02). Low vegetable intake enhanced risk associated with high sugar and soft drink intake. No pattern associated with CD. Mediterranean diet not associated with reduced risk for UC or CD. | Racine et al. 2016 [20] |
Carbohydrates | Cohort size=401,326; UC: cases=244, controls=976; CD: cases=110, controls=440 | No associations between UC or CD risk and total carbohydrate, sugar, or starch intake | Chan et al. 2014 [21] |
Fatty acids | Cohort size=229,702; CD: cases=73, controls=292 | DHA intake inversely associated with CD risk (trend across quintiles: OR 0.54, 95% CI 0.30–0.99, p=0.04). No effect seen for intake of EPA, alpha-linolenic acid, linoleic acid, or oleic acid | Chan et al. 2014b [25] |
Omega-3 PUFA | Cohort size=25,639; UC: cases=22, controls=91 | Association between DHA intake and reduced UC risk (trend across tertiles: OR 0.43, 95% CI 0.22–0.86, p=0.02). Similar protective trends were observed for increasing intake of EPA (p=0.06) and total omega-3 PUFA (p=0.10). | John et al. 2010 [26] |
Fatty acids | Cohort size=203,193; UC: cases=126, controls=504 | Increased linoleic intake associated with UC risk (trend across quartiles: OR 1.32, 95% CI 1.04–1.66, p=0.02). DHA intake associated with decreased UC risk (trend: OR 0.59, 95% CI 0.37–0.94, p=0.03). No association between EPA, alpha-linolenic acid, or oleic acid and UC risk. | Tjonneland et al. 2009 [27] |
Various macronutrients and micronutrients | Cohort size=260,686; UC: cases=139, controls=556 | Trend suggestive of elevated risk from increased total PUFA intake (trend across quartiles: OR 1.19, 95% CI 0.99–1.43, p=0.07). No other macronutrient or micronutrient showed association with altered risk of UC. | Hart et al. 2008 [24] |
B. Nurses’ Health Study (cohort studies) | |||
Dietary fat | Cohort size=170,805 women followed over 26 years; cases: UC=338, CD=269 | Higher intake ratio of omega-3/omega-6 PUFA associated with reduced incidence of UC (trend across quintiles: HR 0.69, 95% CI 0.49–0.98, p=0.03). Trend of inverse association between total omega-3 intake and UC risk (HR 0.72, 95% CI 0.51–1.02, p=0.13). Trend of positive correlation between trans-unsaturated fat intake and UC risk (HR 1.34, 95% CI 0.94–1.46, p=0.07). No associations between dietary fat intake and CD incidence observed. | Ananthakrishnan et al. 2014 [19] |
Dietary fiber | Cohort size=170,776 women followed over 26 years; cases: UC=338, CD=269 | Higher intake of dietary fiber associated with lower incidence of CD (Highest vs. lowest quintile: HR 0.59, 95% CI 0.39–0.90). A strong negative correlation to CD risk was ascribed to fruit intake, and a weaker negative correlation to vegetable intake. No effects of dietary fiber on UC risk were observed. | Ananthankrishnan et al. 2013 [30] |
C. Other | |||
Animal protein | E3N (France): cohort size=67,581 women, mean follow-up time=10.4 years; cases: UC=44, CD=30 | Intake of total protein, but not total carbohydrate or fat, associated with higher risk of both UC (p=0.06) and CD (p=0.04). Higher intake of animal protein, particularly meat and fish, were positively associated with IBD risk. | Jantchou et al. 2010 [17] |
Several lines of evidence point to aspects of the typical Western diet that may promote the development of IBD. The contemporary diet of many Westerners contains high quantities of animal protein and fat, sugar, and processed foods, and low amounts of plant-based foods [12, 15, 16]. Increased meat consumption has been associated with increased risk of both CD and UC [16, 17]. The connection between fat intake and IBD is more tenuous: a systematic review of 19 diet studies claimed that high total fat intake was associated with elevated risk of CD and UC [16], but this claim has been challenged elsewhere [18], and recent prospective studies of dietary patterns in two large cohorts found no association between total fat consumption and risk of CD or UC [19, 20]. Likewise, total sugar intake, also typically elevated in Western diets, appeared to have no effect on IBD risk in a prospective cohort [21]. However, a more recent prospective analysis of dietary patterns demonstrated that a diet marked by high sugar and soft drink consumption plus low vegetable intake was associated with increased UC risk for patients diagnosed more than 2 years after dietary assessment was completed [20].
Intake of polyunsaturated fatty acids (PUFA) was positively associated with increased risk of CD in case-control studies of adult and pediatric IBD patients [16, 22–24]. However, more recent data from prospective cohort studies has suggested that omega-3 PUFA may lower risk of CD [25] or have no effect on CD risk [19]. Omega-3 PUFA may also have a minor protective effect in the context of UC risk [19, 26]. Conversely, omega-6 intake has been positively associated with CD [16, 22, 23] and UC [27], although data from a recent prospective study does not support this connection [19]. Interestingly, a prospective analysis of a multinational European cohort concluded that IBD risk was not altered by consumption of a Mediterranean diet, which is high in omega-3 PUFA [20]. The effects of omega-3:omega-6 PUFA ratios on disease susceptibility may be modulated by genetic variants in fatty acid metabolism genes such as CYP4F3 and FADS2 [28]. This finding points to the need for further studies of how diet-gene interactions contribute to IBD risk.
Fruit and vegetable intake has been associated with decreased risk of CD [16, 23, 29, 30]. There is some evidence that vegetable intake also protects from UC [16], although recent prospective cohort studies disagree on this point [20, 30]. Interpreting low fruit and vegetable intake as causative for IBD may be confounded by the fact that patients may avoid these foods when intestinal symptoms arise [18].
Animal studies have generally supported the notion that fat intake plays a crucial role in the etiology of intestinal inflammation. Mice fed a diet with 40% of energy from fat, meant to represent the typical fat content of a Western diet, developed colitis more rapidly after dextran sodium sulfate (DSS) treatment than chow-fed mice that consumed 20% of calories from fat [31]. In the Il-10−/− mouse, which is genetically susceptible to intestinal inflammation, long-term consumption of a diet high in milk fat increased the penetrance of the colitis phenotype from 30% to 60% [32]. However, it is difficult to establish a direct link between this observation and human IBD risk, since dairy intake is not associated with IBD risk in analyses of patient dietary habits [16, 20]. Data concerning the effects of PUFA intake on intestinal inflammation in rodent models have been more difficult to interpret. A diet supplemented with omega-3 fatty acids protected against colitis in a CD45RBhi adoptive transfer model of colitis [33]. After induction of colitis by 2,4,6-trinitrobenzene sulfonic acid (TNBS), rats consuming an omega-3-rich diet responded better to 5-aminosalicylic acid therapy than those receiving a control diet [34]. However, omega-3 fatty acids, but not omega-6 fatty acids or saturated fats, were found to aggravate DSS colitis in another study [35]. The lack of agreement among these findings may reflect genetic variation of fatty acid metabolism among different mouse strains, differential mechanisms of rodent colitis, or the timing of receiving the diet (prior to inflammation vs. during inflammation).
Artificial food additives prevalent in Western diets may promote intestinal inflammation by interfering with barrier function in the gut. Emulsifiers are detergent-like molecules used in processed foods, particularly those with high fat content, such as ice cream. Treatment of intestinal epithelial monolayers and isolated Peyer’s patches in vitro with low concentrations of the emulsifying agent polysorbate-80 (P80) enhanced the ability of bacteria to translocate across the barrier [36]. Diets with P80 or another emulsifier, carboxymethylcellulose (CMC), compromised mucosal integrity in mice, resulting in greater proximity of luminal microbes to the epithelium and chronic low-grade intestinal inflammation. In Il-10−/− or Tlr5−/− mice, emulsifier intake increased the incidence of colitis. These agents also changed the microbial profile of the mouse intestine, creating blooms of mucus-degrading bacteria such as Ruminococcus gnavus and Akkermansia muciniphila, and expansions of pro-inflammatory Proteobacteria [37]. While a direct role for emulsifier action on host physiology or microbes has not been investigated in human IBD, these results support the idea that contemporary changes in the constitution of our diets can predispose individuals to intestinal inflammation [38]. Currently there are no controlled epidemiological studies that have investigated a potential association between emulsifier consumption and IBD risk in patient populations. However, there is a rough correlation between emulsifier consumption in a given country and that country’s rate of CD incidence [38]. Because processed foods containing emulsifiers may also contain high quantities of fats and sugars, dietary assessment studies of IBD patient populations may not be adequately suited to build a strong case for the effect of emulsifiers on IBD risk. A possible approach may involve randomized controlled trials of IBD patients to see if limiting exposure to emulsifiers affects induction or maintenance of remission. One such proposal involves testing the effect of introducing emulsifiers into enteral nutrition therapy for CD patients and observing their remission rate in comparison to unexposed controls [38].
3 The gut microbiota and IBD
The microbial community assemblage (microbiota) of the gastrointestinal tract plays numerous critical roles in human physiology and metabolism, leading to the suggestion that the microbiota comprise a virtual super-organ [39]. Among its functions include: extracting indigestible nutrients from food that are otherwise inaccessible to humans; vitamin synthesis; promoting intestinal homeostasis by regulating secretions and motility; and educating the immune system to develop mucosal tolerance. Host-microbe interactions are central to the pathogenesis of IBD. Many of the genes associated with IBD risk, such as NOD2 [40, 41], involve innate immunity and sensing of microbial components. Variants in these genes may create dysfunctional immune responses to the normally tolerated microbes in the intestine.
Intestinal dysbiosis, or a deviation from optimal homeostasis and metabolism of the gut microbiota, is a hallmark of IBD. IBD patients have lower diversity in their gut microbiota [42, 43]. There is often a significant loss of bacteria that produce short-chain fatty acids (SCFA), such as Roseburia and Phascolarctobacterium [44, 45]. The butyrate producer Faecalibacterium prausnitzii is dramatically less abundant in CD patients, illustrating a loss of microbial metabolic capacity [44–46]. The loss of anti-inflammatory SCFA produced by these bacteria may also contribute to the pathophysiology of intestinal inflammation. Pathogenic bacteria that attach to the mucosa, such as adherent-invasive Escherichia coli [47] and Fusobacterium species [48], are found in greater quantities in IBD microbiomes. Attachment of these bacteria could stimulate immune responses that exacerbate IBD.
For much of the past century, investigating microbes has depended upon culture-based methods, limiting the study of many heretofore-uncultivable microbial strains from the intestine. With the advent of deep sequencing technologies, it has become possible to catalog the full complement of microbial genes in the intestine [49, 50]. Metagenomic analysis of IBD microbiomes demonstrates that, while only 2% of bacterial genera show significant changes in abundance relative to healthy controls, 12% of genetic networks comprising unique metabolic pathways were significantly increased or decreased [44]. These included basic metabolic modules such as amino acid biosynthesis and carbohydrate metabolism. Some pathways underwent disease-specific changes. For instance, glutathione metabolism genes were more abundant in UC, which could be a bacterial protection mechanism against reactive oxygen species produced in the inflamed gut. In ileal CD, SCFA metabolism genes were underrepresented, while type II secretion systems, such as those used by pathogenic E. coli, became more abundant. It remains unclear if these changes are causal in IBD etiology, or simply microbes reacting to the development of a hostile, inflamed gut environment.
Emerging studies suggest that microbes are not just passive actors in IBD pathogenesis. A wealth of evidence supports the notion that changes in the activity of the gut microbiota can push a susceptible host toward disease. In recent years, the concept of “pathobionts” has emerged in the IBD field. Pathobionts are commensal organisms that can become harmful to the host in the proper environmental milieu [51]. Devkota et al. [32] showed how diet can trigger pathobiont expansion in the context of intestinal inflammation. Mice fed milk fat-enriched diets released a large quantity of taurocholic acid, a bile acid, into the intestine. This promoted a bloom of the sulfite-reducing bacterium, Bilophila wadsworthia, which was associated with increased incidence of colitis in Il10−/− animals. Interestingly, oral gavage of taurocholic acid alone was sufficient to recapitulate both the increased incidence of colitis and the B. wadsworthia outgrowth in these animals. Although the precise mechanism of how Bilophila could promote inflammation was unclear, Devkota and colleagues noted that taurocholic acid provides a source of sulfur that B. wadsworthia can convert into hydrogen sulfide, a genotoxic agent that is elevated in human IBD.
The causes of IBD-associated dysbiosis are unclear, but numerous environmental and lifestyle factors may contribute to it. Early life events play key roles in the acquisition of the gut microbiota, and improper microbiota development in the first years of life is associated with later risk for numerous inflammatory conditions [52]. Childhood proximity to pets [53] or livestock [54] is associated with reduced IBD risk, suggesting the importance of exposure to microbes at a young age. Breastfeeding helps the development of the intestinal microbiota in infants, as breast milk contains oligosaccharides that are consumed by intestinal bacteria, as well as secretory IgA, an immune mediator of gut homeostasis [55, 56]. While some population studies have implicated breastfeeding as a protective factor against IBD [53, 57], other studies have found no evidence to this effect [58, 59]. Dysbiosis is also more pronounced following antibiotic use [45].
Diet controls gut microbiota assembly and may play a key role in establishing dysbiosis related to IBD. Numerous studies of the relationship between diet and microbiota in human populations have delineated specific patterns of microbes that correlate with high intake of plant or animal foods. Compared with European children eating a typical Western diet, children in a rural African village subsisting on a primarily vegetarian, fiber-rich diet have a gut microbiota characterized by a higher ratio of phylum Bacteroidetes to phylum Firmicutes, more abundant Prevotella and Xylanibacter species capable of metabolizing plant polysaccharides, and decreased Shigella and Escherichia [60]. Gut microbial communities are thought to form two major “enterotypes” that are linked with long-term dietary patterns: a Bacteroides enterotype is linked with high intake of animal fat and protein, while a Prevotella enterotype is linked with intake of carbohydrates and fiber [15]. Short-term dietary change can have a rapid impact on reshaping the gut microbiota. An animal-based dietary regimen leads to microbial community changes within 24 hours, marked by increases of bile-tolerant Alistipes, Bilophila, and Bacteroides species [61]. Although diet can induce rapid changes in the microbial population, it may not affect enterotypes shaped by long-term dietary preferences [15]. Microbes that are responsive to dietary change have been implicated in IBD. Notably, animal-based diets rich in protein and fat promote the growth of Bilophila and reduce the abundance of SCFA-generating Roseburia [61]. Animal-based diets are also associated with increases in Enterobacteriaceae including Escherichia [60]; pathogenic variants of E. coli are often found near areas of inflammation in CD patients with ileal disease [44, 45, 47, 62].
4 Microbial metabolism: effects on intestinal inflammation and IBD
Apart from the outgrowth of harmful bacteria, IBD-associated dysbiosis is also characterized by a loss of beneficial microbes that metabolize dietary components into pro-homeostatic molecules. Fermentative bacteria in the intestine convert dietary fiber to short-chain fatty acids (SCFA) including acetate, propionate, and butyrate [63]. SCFA play a critical role in immune homeostasis in the intestine by promoting the differentiation of regulatory T cells via upregulation of the FoxP3 transcription factor (Treg) [64, 65]. A possible mechanism for this transcriptional response involves epigenetic activation of the FoxP3 locus in conjunction with the inhibition of histone deacetylase enzymes by butyrate [64]. Activation of the G-protein-coupled receptor GPR43 (or Ffar2) may also mediate the ability of SCFA to modulate immune responses [65, 66]. SCFA production is impaired in IBD, as demonstrated by measurements of decreased fecal SCFA in patients [67], and from bacterial community analyses demonstrating lower abundances of SCFA-producing bacteria such as Faecalibacterium prausnitzii and Roseburia in active IBD [44]. In rodent models of colitis, administration of SCFA [64, 65] or SCFA-producing commensal microbes [46, 68–70] reduced disease severity. It is worth noting that studies involving the effect of SCFA in animal models of IBD typically involving exposure of the test group to SCFA or commensal bacteria before the initiation of disease. Mice fed a high-fiber diet prior to DSS treatment were protected against colitis; however, the same diet had no beneficial effect if consumed after establishment of DSS colitis [71]. Likewise, dietary fiber has not been convincingly shown to improve active IBD in numerous randomized controlled studies [72].
Bile acid signaling is another point of convergence between the host, gut microbes, and dietary factors which can influence health [73]. Bile acids assist in digestion by emulsifying fats so that they can be absorbed in the intestine. They are made from cholesterol in the liver, and prior to their release in the digestive tract they undergo conjugation to glycine or taurine. Deconjugation of bile acids in the intestine occurs via bile salt hydrolase (BSH) enzymes expressed exclusively by bacteria. Bacteria can further modify bile acids through dehydroxylation. The balance of bile acid composition in the intestine is a determinant of microbiota composition [74]. The products of bacterial bile acid metabolism act as ligands for bile acid receptors FXR and TGR5. FXR activation in the intestine promotes anti-inflammatory signaling and barrier function [75]. IBD patients have higher concentrations of sulfated and conjugated bile acids in their stool than healthy controls [76]. Furthermore, IBD-associated dysbiosis affects the abundance of BSH genes in the intestinal microbiome; in particular, there is a significant decrease of Firmicutes-associated BSH genes in CD patients [77]. Increased levels of conjugated bile acids can support the growth of pathobionts such as Bilophila wadsworthia [32]; concomitantly, the increased ratio of conjugated to unconjugated bile acids would result in a loss of the latter’s anti-inflammatory properties through reductions in FXR signaling and increased expression of pro-inflammatory cytokines [75, 76].
Hydrogen sulfide (H2S) is a gaseous molecule whose involvement in IBD pathology is controversial [78, 79]. Higher quantities of H2S can be measured in the feces of UC patients than healthy subjects [80, 81]. H2S inhibits beta-oxidation of SCFAs by colonocytes, depriving them of energy and compromising barrier function [82]. H2S also enhances T cell activation [83](Miller et al. 2012) and acute exposure to H2S causes genomic damage [84]. On the other hand, H2S has been demonstrated to suppress inflammation and promote mucosal healing in animal models of colitis [85–87].
Microbes are an important source of hydrogen sulfide in the gut [78]. Bilophia wadsworthia generates hydrogen sulfide via reduction of taurine [88]. High intake of milk fat promotes delivery of taurine-conjugated bile acid to the intestine, thus providing Bilophila with a taurine source for H2S metabolism. The bloom of Bilophila may induce a T-cell response that drives colitis in a genetically susceptible host [32]. Fusobacterium nucleatum can produce H2S via cysteine degradation [89]. Pathogenic F. nucleatum has been isolated as a component of the dysbiotic IBD gut [44, 90] and may serve as a predictor of disease severity [45].
A further connection between diet, microbial metabolism, and control of inflammation recently emerged from studies of the intestinal function of aryl hydrocarbon receptor (AhR). AhR is a ubiquitously expressed nuclear receptor that activates xenobiotic metabolism genes in the presence of compounds such as dioxin. Unexpectedly, AhR also plays a key role in adaptive immunity by participating in the differentiation and activity of T cells, including Th1, Th2, and Th17. In IBD patients, AhR expression in the intestine is dampened [91], potentially via upregulation of miRNA-124 [92]. This may create conditions of pro-inflammatory immune activation, as well as local sequelae such as the development of fibrosis [93]. An important dietary source of AhR ligands is indole-3-carbinol, a compound found in cruciferous vegetables [94]. It has been speculated that indole-3-carbinol’s effects on AhR may contribute to the protective effect of vegetable consumption in modulating IBD risk [30, 95]. Furthermore, gut microbes can also produce AhR agonists. Feeding mice a tryptophan-rich diet leads to an expansion of Lactobacillus reuterii, which can metabolize tryptophan into indole-3-aldehyde [96]. This compound stimulates AhR-dependent expression of Il-22, which can ameliorate symptoms of DSS colitis. More recently, Lamas et al. [97] demonstrated that the ability of the gut microbiota to produce AhR ligands from tryptophan is linked with host genetic variation in CARD9, an IBD risk gene. Fecal samples from IBD patients carrying copies of the CARD9 risk allele or CARD9−/− mice contain lower levels of tryptophan-derived indole compounds that may function as AhR ligands, and these samples were impaired in their ability to activate AhR [97]. This work reinforces the concept of AhR as a focal point for maintenance of a positive host-microbe relationship in the gut. Moreover, it further underscores the importance of considering gene-diet or gene-microbe interactions when investigating environmental factors contributing to IBD risk.
5 Dietary and microbial approaches to IBD therapy: advances and challenges
Therapeutic manipulation of diet [10, 11] or the microbiome [9, 98, 99] is highly sought after as an adjunctive or replacement treatment strategy. Although much is known about dietary and microbial factors associated with IBD risk, strategies that aim to correct these factors by supplying a particular dietary element or restoring the microbial community are not always effective. This has been seen with studies of omega-3 supplementation in CD, which demonstrated no benefit in maintenance of remission [100, 101]. Certain dietary interventions, then, may be better if used preventatively, rather than therapeutically or in maintenance of remission. However, without effective means for determining who is at risk of developing IBD, designing dietary strategies aimed at IBD prevention will be difficult.
The goal of IBD treatment, which used to focus primarily on symptom management, has advanced in recent years to include long-term remission with evidence of tissue and systemic recovery from inflammation, preferably without relying on the use of corticosteroids or immunosuppressants which pose health risks when used for a prolonged period [102]. Mucosal healing, or endoscopic remission, is a prognosticator of extended remission in both UC and CD [103, 104]. Clinical biomarkers that correlate well with increased remission include low fecal calprotectin [105] and low serum C-reactive protein (CRP) [106, 107].
Several dietary intervention strategies have been shown to promote clinical remission and mucosal healing. Exclusive enteral nutrition (EEN) is a dietary replacement therapy used in the treatment of pediatric CD [108]. In prospective, open-label studies of children with CD, EEN has shown the ability to induce mucosal healing and normalize clinical and biochemical markers [109, 110]. However, EEN is less effective than corticosteroids at inducing remission in adult CD patients [108]. Sigall-Boneh et al. [111] investigated the ability of partial enteral nutrition in combination with a CD exclusion diet to induce remission in 47 children and young adults with active CD. After six weeks, remission was achieved in 70% of the cohort, as measured by disease indices and C-reactive protein measures. Chiba et al. [112] examined the effectiveness of a semi-vegetarian diet (SVD) to help maintain remission in a cohort of Japanese CD patients. After 2 years, patients consuming SVD had a lower relapse rate than omnivorous patients, concomitant with reduced CRP levels [112].
Evaluations of dietary interventions have not consistently applied these criteria to define their efficacy. For example, recent studies on the low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet have relied on patient questionnaires regarding disease symptoms, such as stool frequency [113, 114]. While there were significant improvements in symptomatic indices reported by these patients, the conclusion that these treatments are effective ways to achieve long-term health goals requires more thorough investigation into the impact of the low FODMAP diet on clinical and endoscopic disease markers.
Microbiota-targeted approaches have demonstrated some promise for treating IBD [98, 99]. In fecal microbiota transplantation (FMT), microbes from the stool of healthy donors are applied via enema or endoscopic delivery into the diseased intestine. FMT has been successfully used to treat recurrent Clostridium difficile infections [115]. Several early case studies of FMT applied to IBD patients suggested its potential use to treat active disease [116]. A randomized controlled trial published in 2013 that looked at FMT for inducing remission in UC found no significant benefit [117]. However, a more recent RCT concluded that FMT significantly outperformed placebo in inducing remission in UC patients [118].
A number of randomized controlled trials have investigated the effectiveness of probiotics for treating IBD, with mixed results. A 2014 meta-analysis concluded that probiotics were effective as primary or adjunctive therapy in inducing and maintaining remission in UC, but not CD [119]. The probiotic mixture VSL#3 has shown especially promising results as a UC therapy [120–123]. Less consistent benefits have been observed in clinical trials of E. coli Nissle 1917 [124, 125]. A clinical benefit of probiotics in CD has not yet been demonstrated, although in one randomized clinical trial, it was shown that patients taking VSL#3 after bowel resection surgery had lower levels of mucosal pro-inflammatory cytokines than patients receiving a placebo [126]. Dietary approaches to reshaping the microbiota involve the consumption of prebiotics, which supply food substrates that could promote the growth of beneficial bacteria. Randomized controlled trials of prebiotic consumption have demonstrated potential benefits for treating active UC [127, 128], but not for CD [129]. A more promising approach to microbial manipulation for the treatment of CD may lie in the use of “synbiotics” which combine probiotic strains with prebiotic dietary components. Synbiotic therapy using Bifidobacterium longum and an inulin/oligofructosaccharide blend has been shown to improve clinical profiles and reduce inflammatory markers in active CD [130].
Finally, identification of anti-inflammatory metabolites synthesized by gut microbes has raised interest in their potential use as “postbiotic” therapies. Much of this research has yet to progress into human clinical studies, but it will be interesting to follow its development in the coming years. For example, Nakao and colleagues [131] performed a small-molecule screen for AhR agonists from a library of microbe-derived compounds. The top hit was 1,4-dihydroxy-2-naphthoic acid (DHNA), a vitamin K2 precursor made by Propionibacterium freudenreichii, a bacterium that grows on Swiss cheese. Introducing DHNA into the diet of mice for one week protected them from DSS colitis [131]. Because DHNA is available as a nutritional supplement in Japan, the authors proposed follow-up studies on the efficacy of DHNA for reducing IBD risk. While FMT, probiotics, prebiotics, synbiotics, and postbiotics hold promise as future treatment approaches for IBD, successfully establishing these as alternatives to current treatment regimens will require a deeper understanding of the ecological processes underlying IBD-associated dysbiosis. Strategies to introduce beneficial strains of bacteria into the diseased intestine, either singly or as communities, may be hampered by the intrinsic resilience of the IBD microbiota to recolonization, just as microbial communities in healthy people help confer resistance to the establishment of pathogens [132, 133].
6 Prospects
IBD arises through a complex set of interactions between host genetics, gut microbes, the immune system, and environmental influences that include diet. The diseases are highly idiopathic, and the traditional classification of IBD into two phenotypes, CD and UC, likely does not reflect the true range of disease states that produce inflammation of the intestine. Each patient’s variation of IBD reflects a unique combination of contributing factors, intrinsic and extrinsic, continuous and historical; no single factor is likely to be responsible in every case. Addressing the challenges of identifying causal factors in IBD requires systems-level approaches that capture the nature of interactions between the different players in disease pathogenesis. There is still much that can be learned from patient populations, both in cross-sectional case-control studies as well as prospective longitudinal studies, but careful stratification must be performed to identify signals of disease-associated variables. Attention should be paid to dietary patterns, use of antibiotics or other medications, disease location, and so forth.
Dietary strategies to successfully treat IBD and maintain disease remission are an attractive alternative to presently available medical and surgical therapies. In assessing the impact of these approaches, it will be important to compare dietary therapies to our current arsenal of IBD treatments with respect to clinical standards of care. Mucosal healing has been recognized as a key prognosticator of deep remission in IBD [103, 134, 135], and clinicians investigating nutritional treatment of active disease should adopt this assessment criterion, as was done for EEN [136]. As we progress in our understanding of the causes of IBD, we must also remain open to the prospect that dietary regimens may need to be personalized for individuals or subsets of patients based on genetics, immune status, personal history, and other factors. A tailored approach may be useful for treating disease, as well as for achieving the yet-elusive goal of prescribing diets to prevent the onset of IBD in high-risk patients.
Over the past decade, we have made important strides in understanding the role of the intestinal microbiota in IBD pathogenesis. However, we still lack much in terms of mechanistic insight into how microbes contribute to the onset of disease. We propose several approaches to study design that would remedy this deficit of knowledge. Longitudinal studies of patient populations at risk of developing IBD should include microbiome analyses in order to shed light on potential causative events related to microbial ecology. The interpretation of dysbiosis in IBD patients should be undertaken with sufficient appreciation for clinical metadata that helps identify important associations that take place in subsets of patients, which would more accurately reflect the idiopathic nature of IBD.
Future efforts to study IBD-associated dysbiosis should include a greater focus on the mucosa-associated microbiota. The intestinal mucosa and stool are inhabited by distinctive microbial communities [45, 137]. Intestinal microbes residing near the mucosal surface can regulate local immunity via direct contact with host tissues or by diffusible chemical signals [138]. In new-onset CD, the degree of dysbiosis is greater in ileal or rectal mucosal biopsies than in stool; furthermore, mucosal communities from ileal or rectal biopsies outperform fecal communities in predictive models for CD diagnosis [45]. These findings suggest that communities recovered from biopsies may offer greater insight into dysbiotic processes and host-microbes interactions that drive the pathophysiology of IBD. Rectal biopsies, in particular, may offer easy access to high-value microbiome data.
Finally, advances in sequencing technology and computational approaches to analyzing genomic data on microbial communities should be harnessed to go beyond our present focus on taxonomic groups in order to learn more about specific strains of microbes that are likely to possess unique functions that play a role in the development of complex diseases [139].
Research into animal models of IBD still represents our best opportunities to discover the mechanisms of pathogenesis. The use of gnotobiotic mouse models to examine the functions of specific microbial communities in the context of genetics and diet is an area that holds much promise. As the microbiome field moves away from descriptive studies toward a deeper understanding of microbiota function, the importance of metabolomics studies, in tandem with other meta-’omics (genomes, transcriptomes, proteomes) will be paramount to discovering how host and microbes choreograph their interactions in health and disease. Similarly, metabolomics approaches to studying single microbes [140] or IBD patient populations [141] will contribute important knowledge of bioactive molecules that could translate into therapeutic innovations for inflammatory bowel diseases.
Acknowledgments
This work was supported by National Institutes of Health/NIDDK grants R01DK097268, R56DK102872, and T32DK007074 (to E.B.C.), and by an NIDDK center grant to the University of Chicago Digestive Diseases Research Core Center (P30DK42086). The authors thank Dr. Joseph Pierre for helpful discussions regarding the manuscript.
Abbreviations
- AhR
aryl hydrocarbon receptor
- BSH
bile salt hydrolase
- CD
Crohn’s disease
- DHNA
1,4-dihydroxy-2-naphthoic acid
- DSS
dextran sodium sulfate
- EEN
exclusive enteral nutrition
- FXR
farnesoid X receptor
- GWAS
genome-wide association studies
- IBD
inflammatory bowel diseases
- PUFA
polyunsaturated fatty acids
- SCFA
short-chain fatty acids
- TNBS
2,4,6-trinitrobenzene sulfonic acid
- UC
ulcerative colitis
Footnotes
Conflict of interest statement: K.T.D. and E.B.C. have no conflicts to declare.
References
- 1.Molodecky NA, Soon IS, Rabi DM, Ghali WA, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46–54. doi: 10.1053/j.gastro.2011.10.001. [DOI] [PubMed] [Google Scholar]
- 2.Prideaux L, Kamm MA, De Cruz PP, Chan FKL, et al. Inflammatory bowel disease in Asia: a systematic review. J Gastroenterol Hepatol. 2012;27:1266–80. doi: 10.1111/j.1440-1746.2012.07150.x. [DOI] [PubMed] [Google Scholar]
- 3.Barreiro-de Acosta M, Alvarez Castro A, Souto R, Iglesias M, et al. Emigration to western industrialized countries: A risk factor for developing inflammatory bowel disease. J Crohns Colitis. 2011;5:566–569. doi: 10.1016/j.crohns.2011.05.009. [DOI] [PubMed] [Google Scholar]
- 4.Jostins L, Ripke S, Weersma RK, Duerr RH, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–124. doi: 10.1038/nature11582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Huang C, Haritunians T, Okou DT, Cutler DJ, et al. Characterization of genetic loci that affect susceptibility to inflammatory bowel diseases in African Americans. Gastroenterology. 2015;149:1575–1586. doi: 10.1053/j.gastro.2015.07.065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Liu JZ, van Sommeren S, Huang H, Ng SC, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–986. doi: 10.1038/ng.3359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2015;12:205–217. doi: 10.1038/nrgastro.2015.34. [DOI] [PubMed] [Google Scholar]
- 8.Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology. 2014;146:1489–99. doi: 10.1053/j.gastro.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gentschew L, Ferguson LR. Role of nutrition and microbiota in susceptibility to inflammatory bowel diseases. Mol Nutr Food Res. 2012;56:524–535. doi: 10.1002/mnfr.201100630. [DOI] [PubMed] [Google Scholar]
- 10.Leone VA, Cham CM, Chang EB. Diet, gut microbes, and genetics in immune function: can we leverage our current knowledge to achieve better outcomes in inflammatory bowel diseases? Curr Opin Immunol. 2014;31:16–23. doi: 10.1016/j.coi.2014.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lee D, Albenberg L, Compher C, Baldassano R, et al. Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology. 2015;148:1087–1106. doi: 10.1053/j.gastro.2015.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1:6ra14. doi: 10.1126/scitranslmed.3000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nyangale EP, Mottram DS, Gibson GR. Gut microbial activity, implications for health and disease: The potential role of metabolite analysis. J Proteome Res. 2012;11:5573–5585. doi: 10.1021/pr300637d. [DOI] [PubMed] [Google Scholar]
- 14.Chapman-Kiddell CA, Davies PSW, Gillen L, Radford-Smith GL. Role of diet in the development of inflammatory bowel disease. Inflamm Bowel Dis. 2010;16:137–51. doi: 10.1002/ibd.20968. [DOI] [PubMed] [Google Scholar]
- 15.Wu GD, Chen J, Hoffmann C, Bittinger K, et al. Science. 2011;334:105–108. doi: 10.1126/science.1208344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing inflammatory bowel disease: a systematic review of the literature. Am J Gastroenterol. 2011;106:563–573. doi: 10.1038/ajg.2011.44. [DOI] [PubMed] [Google Scholar]
- 17.Jantchou P, Morois S, Clavel-Chapelon F, Boutron-Ruault MC, et al. Animal protein intake and risk of inflammatory bowel disease: The E3N prospective study. Am J Gastroenterol. 2010;105:2195–201. doi: 10.1038/ajg.2010.192. [DOI] [PubMed] [Google Scholar]
- 18.Richman E, Rhodes JM. Review article: evidence-based dietary advice for patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2013;38:1156–71. doi: 10.1111/apt.12500. [DOI] [PubMed] [Google Scholar]
- 19.Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, et al. Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease. Gut. 2014;63:776–784. doi: 10.1136/gutjnl-2013-305304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Racine A, Carbonnel F, Chan SSM, Hart AR, et al. Dietary patterns and risk of inflammatory bowel disease in Europe. Inflamm Bowel Dis. 2016;22:345–354. doi: 10.1097/MIB.0000000000000638. [DOI] [PubMed] [Google Scholar]
- 21.Chan SSM, Luben R, van Schaik F, Oldenburg B, et al. Carbohydrate intake in the etiology of Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis. 2014;20:2013–2021. doi: 10.1097/MIB.0000000000000168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sakamoto N, Kono S, Wakai K, Fukuda Y, et al. Dietary risk factors for inflammatory bowel disease: a multicenter case-control study in Japan. Inflamm Bowel Dis. 2005;11:154–63. doi: 10.1097/00054725-200502000-00009. [DOI] [PubMed] [Google Scholar]
- 23.Amre DK, D’Souza S, Morgan K, Seidman G, et al. Imbalances in dietary consumption of fatty acids, vegetables, and fruits are associated with risk for Crohn’s disease in children. Am J Gastroenterol. 2007;102:2016–25. doi: 10.1111/j.1572-0241.2007.01411.x. [DOI] [PubMed] [Google Scholar]
- 24.Hart AR, Luben R, Olsen A, Tjonneland A, et al. Diet in the aetiology of ulcerative colitis: a European prospective cohort study. Digestion. 2008;77:57–64. doi: 10.1159/000121412. [DOI] [PubMed] [Google Scholar]
- 25.Chan SSM, Luben R, Olsen A, Tjonneland A, et al. Association between high dietary intake of the n-3 polyunsaturated fatty acid docosahexaenoic acid and reduced risk of Crohn’s disease. Aliment Pharmacol Ther. 2014;39:834–842. doi: 10.1111/apt.12670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.John S, Luben R, Shrestha SS, Welch A, et al. Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study. Eur J Gastroenterol Hepatol. 2010;22:602–6. doi: 10.1097/MEG.0b013e3283352d05. [DOI] [PubMed] [Google Scholar]
- 27.Tjonneland A, Overvad K, Bergmann MM, Nagel G, et al. Linoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case-control study within a European prospective cohort study. Gut. 2009;58:1606–11. doi: 10.1136/gut.2008.169078. [DOI] [PubMed] [Google Scholar]
- 28.Costea I, Mack DR, Lemaitre RN, Israel D, et al. Interactions between the dietary polyunsaturated fatty acid ratio and genetic factors determine susceptibility to pediatric Crohn’s disease. Gastroenterology. 2014;146:929–931. doi: 10.1053/j.gastro.2013.12.034. [DOI] [PubMed] [Google Scholar]
- 29.D’Souza S, Levy E, Mack D, Israel D, et al. Dietary patterns and risk for Crohn’s disease in children. Inflamm Bowel Dis. 2008;14:367–73. doi: 10.1002/ibd.20333. [DOI] [PubMed] [Google Scholar]
- 30.Ananthakrishnan AN, Khalili H, Konijeti GG, Higuchi LM, et al. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology. 2013;145:970–977. doi: 10.1053/j.gastro.2013.07.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Van der Logt EMJ, Blokzijl T, Van der Meer R, Faber KN, et al. Westernized high-fat diet accelerates weight loss in dextran sulfate sodium-induced colitis in mice, which is further aggravated by supplementation of heme. J Nutr Biochem. 2013;24:1159–1165. doi: 10.1016/j.jnutbio.2012.09.001. [DOI] [PubMed] [Google Scholar]
- 32.Devkota S, Wang Y, Musch MW, Leone V, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature. 2012;487:104–108. doi: 10.1038/nature11225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Whiting CV, Bland PW, Tarlton JF. Dietary n-3 polyunsaturated fatty acids reduce disease and colonic proinflammatory cytokines in a mouse model of colitis. Inflamm Bowel Dis. 2005;11:340–349. doi: 10.1097/01.mib.0000164016.98913.7c. [DOI] [PubMed] [Google Scholar]
- 34.Mbodji K, Charpentier C, Guérin C, Querec C, et al. Adjunct therapy of n-3 fatty acids to 5-ASA ameliorates inflammatory score and decreases NF-κB in rats with TNBS-induced colitis. J Nutr Biochem. 2013;24:700–705. doi: 10.1016/j.jnutbio.2012.03.022. [DOI] [PubMed] [Google Scholar]
- 35.Matsunaga H, Hokari R, Kurihara C, Okada Y, et al. Omega-3 fatty acids exacerbate DSS-induced colitis through decreased adiponectin in colonic subepithelial myofibroblasts. Inflamm Bowel Dis. 2008;14:1348–1357. doi: 10.1002/ibd.20491. [DOI] [PubMed] [Google Scholar]
- 36.Roberts CL, Keita AV, Duncan SH, O’Kennedy N, et al. Translocation of Crohn’s disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut. 2010;59:1331–1339. doi: 10.1136/gut.2009.195370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chassaing B, Koren O, Goodrich JK, Poole AC, et al. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Nature. 2015;519:92–96. doi: 10.1038/nature14232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Roberts CL, Rushworth SL, Richman E, Rhodes JM. Hypothesis: Increased consumption of emulsifiers as an explanation for the rising incidence of Crohn’s disease. J Crohns Colitis. 2013;7:338–341. doi: 10.1016/j.crohns.2013.01.004. [DOI] [PubMed] [Google Scholar]
- 39.Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, et al. Host-bacterial mutualism in the human intestine. Science. 2005;307:1915–1920. doi: 10.1126/science.1104816. [DOI] [PubMed] [Google Scholar]
- 40.Hugot JP, Chamaillard M, Zouali H, Lesage S, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature. 2001;411:599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
- 41.Ogura Y, Bonen DK, Inohara N, Nicolae DL, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature. 2001;411:603–606. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
- 42.Ott SJ, Musfeldt M, Wenderoth DF, Hampe J, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53:685–93. doi: 10.1136/gut.2003.025403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut. 2006;55:205–211. doi: 10.1136/gut.2005.073817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Morgan XC, Tickle TL, Sokol H, Gevers D, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79. doi: 10.1186/gb-2012-13-9-r79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gevers D, Kugathasan S, Denson LA, Vázquez-Baeza Y, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–92. doi: 10.1016/j.chom.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sokol H, Pigneur B, Watterlot L, Lakhdari O, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731–16736. doi: 10.1073/pnas.0804812105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004;127:412–421. doi: 10.1053/j.gastro.2004.04.061. [DOI] [PubMed] [Google Scholar]
- 48.Ohkusa T, Sato N, Ogihara T, Morita K, et al. Fusobacterium varium localized in the colonic mucosa of patients with ulcerative colitis stimulates species-specific antibody. J Gastroenterol Hepatol. 2002;17:849–853. doi: 10.1046/j.1440-1746.2002.02834.x. [DOI] [PubMed] [Google Scholar]
- 49.Consortium THMP. A framework for human microbiome research. Nature. 2012;486:215–221. doi: 10.1038/nature11209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li J, Jia H, Cai X, Zhong H, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–841. doi: 10.1038/nbt.2942. [DOI] [PubMed] [Google Scholar]
- 51.Chow J, Tang H, Mazmanian SK. Pathobionts of the gastrointestinal microbiota and inflammatory disease. Curr Opin Immunol. 2011;23:473–480. doi: 10.1016/j.coi.2011.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gensollen T, Iyer SS, Kasper DL, Blumberg RS. How colonization by microbiota in early life shapes the immune system. Science (80- ) 2016;352:539–544. doi: 10.1126/science.aad9378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ng SC, Tang W, Leong RW, Chen M, et al. Environmental risk factors in inflammatory bowel disease: a population-based case-control study in Asia-Pacific. Gut. 2015;64:1063–71. doi: 10.1136/gutjnl-2014-307410. [DOI] [PubMed] [Google Scholar]
- 54.Timm S, Svanes C, Janson C, Sigsgaard T, et al. Place of upbringing in early childhood as related to inflammatory bowel diseases in adulthood: a population-based cohort study in Northern Europe. Eur J Epidemiol. 2014;29:429–37. doi: 10.1007/s10654-014-9922-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hennet T, Borsig L. Breastfed at Tiffany’s. Trends Biochem Sci. 2016;41:508–518. doi: 10.1016/j.tibs.2016.02.008. [DOI] [PubMed] [Google Scholar]
- 56.Rogier EW, Frantz AL, Bruno MEC, Wedlund L, et al. Secretory antibodies in breast milk promote long-term intestinal homeostasis by regulating the gut microbiota and host gene expression. Proc Natl Acad Sci U S A. 2014;111:3074–9. doi: 10.1073/pnas.1315792111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Klement E, Cohen RV, Boxman J, Joseph A, et al. Breastfeeding and risk of inflammatory bowel disease: a systematic review with meta-analysis. Am J Clin Nutr. 2004;80:1342–1352. doi: 10.1093/ajcn/80.5.1342. [DOI] [PubMed] [Google Scholar]
- 58.Barclay AR, Russell RK, Wilson ML, Gilmour WH, et al. Systematic review: the role of breastfeeding in the development of pediatric inflammatory bowel disease. J Pediatr. 2009;155:421–6. doi: 10.1016/j.jpeds.2009.03.017. [DOI] [PubMed] [Google Scholar]
- 59.Khalili H, Ananthakrishnan AN, Higuchi LM, Richter JM, et al. Early life factors and risk of inflammatory bowel disease in adulthood. Inflamm Bowel Dis. 2013;19:542–7. doi: 10.1097/MIB.0b013e31828132f8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–14696. doi: 10.1073/pnas.1005963107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.David La, Maurice CF, Carmody RN, Gootenberg DB, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Baumgart M, Dogan B, Rishniw M, Weitzman G, et al. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s disease involving the ileum. ISME J. 2007;1:403–18. doi: 10.1038/ismej.2007.52. [DOI] [PubMed] [Google Scholar]
- 63.Macfarlane S, Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc. 2003;62:67–72. doi: 10.1079/PNS2002207. [DOI] [PubMed] [Google Scholar]
- 64.Furusawa Y, Obata Y, Fukuda S, Endo T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–450. doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
- 65.Smith PM, Howitt MR, Panikov N, Michaud M, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science (80- ) 2013;341:569–574. doi: 10.1126/science.1241165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Maslowski KM, Vieira AT, Ng A, Kranich J, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–1286. doi: 10.1038/nature08530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Treem WR, Ahsan N, Shoup M, Hyams JS. Fecal short-chain fatty acids in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 1994;18:159–164. doi: 10.1097/00005176-199402000-00007. [DOI] [PubMed] [Google Scholar]
- 68.Atarashi K, Tanoue T, Oshima K, Suda W, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232–236. doi: 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
- 69.Martín R, Chain F, Miquel S, Lu J, et al. The commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-induced chronic moderate and severe colitis models. Inflamm Bowel Dis. 2014;20:417–430. doi: 10.1097/01.MIB.0000440815.76627.64. [DOI] [PubMed] [Google Scholar]
- 70.Qiu X, Zhang M, Yang X, Hong N, et al. Faecalibacterium prausnitzii upregulates regulatory T cells and anti-inflammatory cytokines in treating TNBS-induced colitis. J Crohns Colitis. 2013;7:e558–568. doi: 10.1016/j.crohns.2013.04.002. [DOI] [PubMed] [Google Scholar]
- 71.Silveira ALM, Ferreira AVM, de Oliveira MC, Rachid MA, et al. Preventive rather than therapeutic treatment with high fiber diet attenuates clinical and inflammatory markers of acute and chronic DSS-induced colitis in mice. Eur J Nutr. 2015 doi: 10.1007/s00394-015-1068-x. [DOI] [PubMed] [Google Scholar]
- 72.Wedlake L, Slack N, Andreyev HJN, Whelan K. Fiber in the treatment and maintenance of inflammatory bowel disease: a systematic review of randomized controlled trials. Inflamm Bowel Dis. 2014;20:576–586. doi: 10.1097/01.MIB.0000437984.92565.31. [DOI] [PubMed] [Google Scholar]
- 73.Joyce SA, Gahan CGM. Bile acid modifications at the microbe-host interface: potential for nutraceutical and pharmaceutical interventions in host health. Annu Rev Food Sci Technol. 2016 doi: 10.1146/annurev-food-041715-033159. [DOI] [PubMed] [Google Scholar]
- 74.Islam KBMS, Fukiya S, Hagio M, Fujii N, et al. Bile acid is a host factor that regulates the composition of the cecal microbiota in rats. Gastroenterology. 2011;141:1773–1781. doi: 10.1053/j.gastro.2011.07.046. [DOI] [PubMed] [Google Scholar]
- 75.Gadaleta RM, van Erpecum KJ, Oldenburg B, Willemsen ECL, et al. Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease. Gut. 2011;60:463–472. doi: 10.1136/gut.2010.212159. [DOI] [PubMed] [Google Scholar]
- 76.Duboc H, Rajca S, Rainteau D, Benarous D, et al. Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases. Gut. 2013;62:531–539. doi: 10.1136/gutjnl-2012-302578. [DOI] [PubMed] [Google Scholar]
- 77.Ogilvie LA, Jones BV. Dysbiosis modulates capacity for bile acid modification in the gut microbiomes of patients with inflammatory bowel disease: a mechanism and marker of disease? Gut. 2012;61:1642–1643. doi: 10.1136/gutjnl-2012-302137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Carbonero F, Benefiel AC, Alizadeh-Ghamsari AH, Gaskins HR. Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol. 2012;3:448. doi: 10.3389/fphys.2012.00448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wallace JL, Blackler RW, Chan MV, Da Silva GJ, et al. Anti-inflammatory and cytoprotective actions of hydrogen sulfide: translation to therapeutics. Antioxid Redox Signal. 2015;22:398–410. doi: 10.1089/ars.2014.5901. [DOI] [PubMed] [Google Scholar]
- 80.Gibson GR, Cummings JH, Macfarlane GT. Growth and activities of sulphate-reducing bacteria in gut contents of healthy subjects and patients with ulcerative colitis. FEMS Microbiol Ecol. 1991;9:103–111. [Google Scholar]
- 81.Levine J, Ellis CJ, Furne JK, Springfield J, et al. Fecal hydrogen sulfide production in ulcerative colitis. Am J Gastroenterol. 1998;93:83–7. doi: 10.1111/j.1572-0241.1998.083_c.x. [DOI] [PubMed] [Google Scholar]
- 82.Babidge W, Millard S, Roediger W. Sulfides impair short chain fatty acid beta-oxidation at acyl-CoA dehydrogenase level in colonocytes: implications for ulcerative colitis. Mol Cell Biochem. 1998;181:117–24. doi: 10.1023/a:1006838231432. [DOI] [PubMed] [Google Scholar]
- 83.Miller TW, Wang EA, Gould S, Stein EV, et al. Hydrogen sulfide is an endogenous potentiator of T cell activation. J Biol Chem. 2012;287:4211–21. doi: 10.1074/jbc.M111.307819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Attene-Ramos MS, Wagner ED, Plewa MJ, Gaskins HR. Evidence that hydrogen sulfide is a genotoxic agent. Mol Cancer Res. 2006;4:9–14. doi: 10.1158/1541-7786.MCR-05-0126. [DOI] [PubMed] [Google Scholar]
- 85.Wallace JL, Vong L, McKnight W, Dicay M, et al. Endogenous and exogenous hydrogen sulfide promotes resolution of colitis in rats. Gastroenterology. 2009;137:569–78. 578.e1. doi: 10.1053/j.gastro.2009.04.012. [DOI] [PubMed] [Google Scholar]
- 86.Fasolino I, Izzo AA, Clavel T, Romano B, et al. Orally administered allyl sulfides from garlic ameliorate murine colitis. Mol Nutr Food Res. 2015;59:434–42. doi: 10.1002/mnfr.201400347. [DOI] [PubMed] [Google Scholar]
- 87.Motta JP, Flannigan KL, Agbor TA, Beatty JK, et al. Hydrogen sulfide protects from colitis and restores intestinal microbiota biofilm and mucus production. Inflamm Bowel Dis. 2015;21:1006–17. doi: 10.1097/MIB.0000000000000345. [DOI] [PubMed] [Google Scholar]
- 88.Laue H, Denger K, Cook AM. Taurine reduction in anaerobic respiration of Bilophila wadsworthia RZATAU. Appl Environ Microbiol. 1997;63:2016–21. doi: 10.1128/aem.63.5.2016-2021.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Fukamachi H, Nakano Y, Yoshimura M, Koga T. Cloning and characterization of the L-cysteine desulfhydrase gene of Fusobacterium nucleatum. FEMS Microbiol Lett. 2002;215:75–80. doi: 10.1111/j.1574-6968.2002.tb11373.x. [DOI] [PubMed] [Google Scholar]
- 90.Strauss J, Kaplan GG, Beck PL, Rioux K, et al. Invasive potential of gut mucosa-derived Fusobacterium nucleatum positively correlates with IBD status of the host. Inflamm Bowel Dis. 2011;17:1971–8. doi: 10.1002/ibd.21606. [DOI] [PubMed] [Google Scholar]
- 91.Monteleone I, Rizzo A, Sarra M, Sica G, et al. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology. 2011;141:237–248. doi: 10.1053/j.gastro.2011.04.007. [DOI] [PubMed] [Google Scholar]
- 92.Zhao Y, Ma T, Chen W, Chen Y, et al. MicroRNA-124 promotes the intestinal inflammation by targeting AHR in Crohn’s disease. J Crohns Colitis. 2016 doi: 10.1093/ecco-jcc/jjw010. [DOI] [PubMed] [Google Scholar]
- 93.Monteleone I, Zorzi F, Marafini I, Fusco DDi, et al. Aryl hydrocarbon receptor-driven signals inhibit collagen synthesis in the gut. Eur J Immunol. 2016 doi: 10.1002/eji.201445228. [DOI] [PubMed] [Google Scholar]
- 94.Li Y, Innocentin S, Withers DR, Roberts NA, et al. Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell. 2011;147:629–40. doi: 10.1016/j.cell.2011.09.025. [DOI] [PubMed] [Google Scholar]
- 95.Monteleone I, MacDonald TT, Pallone F, Monteleone G. The aryl hydrocarbon receptor in inflammatory bowel disease: linking the environment to disease pathogenesis. Curr Opin Gastroenterol. 2012;28:310–313. doi: 10.1097/MOG.0b013e328352ad69. [DOI] [PubMed] [Google Scholar]
- 96.Zelante T, Iannitti R, Cunha C, DeLuca A, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–385. doi: 10.1016/j.immuni.2013.08.003. [DOI] [PubMed] [Google Scholar]
- 97.Lamas B, Richard ML, Leducq V, Pham H-P, et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat Med. 2016 doi: 10.1038/nm.4102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Serban DE. Microbiota in inflammatory bowel disease pathogenesis and therapy: is it all about diet? Nutr Clin Pract. 2015;30:760–779. doi: 10.1177/0884533615606898. [DOI] [PubMed] [Google Scholar]
- 99.Hansen JJ, Sartor RB. Therapeutic manipulation of the microbiome in IBD: current results and future approaches. Curr Treat Options Gastroenterol. 2015;13:105–120. doi: 10.1007/s11938-014-0042-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Feagan BG, Sandborn WJ, Mittmann U, Bar-Meir S, et al. Omega-3 free fatty acids for the maintenance of remission in Crohn disease: the EPIC Randomized Controlled Trials. JAMA. 2008;299:1690–1697. doi: 10.1001/jama.299.14.1690. [DOI] [PubMed] [Google Scholar]
- 101.Lev-Tzion R, Griffiths AM, Leder O, Turner D. Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2014;2:CD006320. doi: 10.1002/14651858.CD006320.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.D’Haens GR, Sartor RB, Silverberg MS, Petersson J, et al. Future directions in inflammatory bowel disease management. J Crohns Colitis. 2014;8:726–34. doi: 10.1016/j.crohns.2014.02.025. [DOI] [PubMed] [Google Scholar]
- 103.Shah SC, Colombel J-F, Sands BE, Narula N. Mucosal healing is associated with improved long-term outcomes of patients with ulcerative colitis: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2016 doi: 10.1016/j.cgh.2016.01.015. [DOI] [PubMed] [Google Scholar]
- 104.Shah SC, Colombel JF, Sands BE, Narula N. Systematic review with meta-analysis: mucosal healing is associated with improved long-term outcomes in Crohn’s disease. Aliment Pharmacol Ther. 2016;43:317–33. doi: 10.1111/apt.13475. [DOI] [PubMed] [Google Scholar]
- 105.Mooiweer E, Severs M, Schipper MEI, Fidder HH, et al. Low fecal calprotectin predicts sustained clinical remission in inflammatory bowel disease patients: a plea for deep remission. J Crohns Colitis. 2015;9:50–55. doi: 10.1093/ecco-jcc/jju003. [DOI] [PubMed] [Google Scholar]
- 106.Solem CA, Loftus EV, Tremaine WJ, Harmsen WS, et al. Correlation of C-reactive protein with clinical, endoscopic, histologic, and radiographic activity in inflammatory bowel disease. Inflamm Bowel Dis. 2005;11:707–12. doi: 10.1097/01.mib.0000173271.18319.53. [DOI] [PubMed] [Google Scholar]
- 107.Henriksen M, Jahnsen J, Lygren I, Stray N, et al. C-reactive protein: a predictive factor and marker of inflammation in inflammatory bowel disease. Results from a prospective population-based study. Gut. 2008;57:1518–23. doi: 10.1136/gut.2007.146357. [DOI] [PubMed] [Google Scholar]
- 108.Zachos M, Tondeur M, Griffiths AM. Enteral nutritional therapy for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2007:CD000542. doi: 10.1002/14651858.CD000542.pub2. [DOI] [PubMed] [Google Scholar]
- 109.Grover Z, Muir R, Lewindon P. Exclusive enteral nutrition induces early clinical, mucosal and transmural remission in paediatric Crohn’s disease. J Gastroenterol. 2014;49:638–45. doi: 10.1007/s00535-013-0815-0. [DOI] [PubMed] [Google Scholar]
- 110.Grover Z, Burgess C, Muir R, Reilly C, et al. Early Mucosal Healing with Exclusive Enteral Nutrition is Associated with Improved Outcomes in Newly Diagnosed Children with Luminal Crohn’s disease. J Crohns Colitis. 2016 doi: 10.1093/ecco-jcc/jjw075. [DOI] [PubMed] [Google Scholar]
- 111.Sigall-Boneh R, Pfeffer-Gik T, Segal I, Zangen T, et al. Partial enteral nutrition with a Crohn’s disease exclusion diet is effective for induction of remission in children and young adults with Crohn’s disease. Inflamm Bowel Dis. 2014;20:1353–1360. doi: 10.1097/MIB.0000000000000110. [DOI] [PubMed] [Google Scholar]
- 112.Chiba M, Abe T, Tsuda H, Sugawara T, et al. Lifestyle-related disease in Crohn’s disease: relapse prevention by a semi-vegetarian diet. World J Gastroenterol. 2010;16:2484–2495. doi: 10.3748/wjg.v16.i20.2484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Prince AC, Myers CE, Joyce T, Irving P, et al. Fermentable Carbohydrate Restriction (Low FODMAP Diet) in Clinical Practice Improves Functional Gastrointestinal Symptoms in Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22:1129–36. doi: 10.1097/MIB.0000000000000708. [DOI] [PubMed] [Google Scholar]
- 114.Maagaard L, Ankersen DV, Végh Z, Burisch J, et al. Follow-up of patients with functional bowel symptoms treated with a low FODMAP diet. World J Gastroenterol. 2016;22:4009–19. doi: 10.3748/wjg.v22.i15.4009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368:407–415. doi: 10.1056/NEJMoa1205037. [DOI] [PubMed] [Google Scholar]
- 116.Anderson JL, Edney RJ, Whelan K. Systematic review: faecal microbiota transplantation in the management of inflammatory bowel disease. Aliment Pharmacol Ther. 2012;36:503–516. doi: 10.1111/j.1365-2036.2012.05220.x. [DOI] [PubMed] [Google Scholar]
- 117.Kump PK, Gröchenig HP, Lackner S, Trajanoski S, et al. Alteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitis. Inflamm Bowel Dis. 2013;19:2155–2165. doi: 10.1097/MIB.0b013e31829ea325. [DOI] [PubMed] [Google Scholar]
- 118.Moayyedi P, Surette MG, Kim PT, Libertucci J, et al. Fecal microbiota transplantation induces remission in patients With active ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015;149:102–109. doi: 10.1053/j.gastro.2015.04.001. [DOI] [PubMed] [Google Scholar]
- 119.Shen J, Zuo ZX, Mao AP. Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn’s disease, and pouchitis: meta-analysis of randomized controlled trials. Inflamm Bowel Dis. 2014;20:21–35. doi: 10.1097/01.MIB.0000437495.30052.be. [DOI] [PubMed] [Google Scholar]
- 120.Mardini HE, Grigorian AY. Probiotic mix VSL#3 is effective adjunctive therapy for mild to moderately active ulcerative colitis: a meta-analysis. Inflamm Bowel Dis. 2014;20:1562–1567. doi: 10.1097/MIB.0000000000000084. [DOI] [PubMed] [Google Scholar]
- 121.Miele E, Pascarella F, Giannetti E, Quaglietta L, et al. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol. 2009;104:437–443. doi: 10.1038/ajg.2008.118. [DOI] [PubMed] [Google Scholar]
- 122.Sood A, Midha V, Makharia GK, Ahuja V, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7:1202–1209. doi: 10.1016/j.cgh.2009.07.016. [DOI] [PubMed] [Google Scholar]
- 123.Tursi A, Brandimarte G, Papa A, Giglio A, et al. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J Gastroenterol. 2010;105:2218–2227. doi: 10.1038/ajg.2010.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Henker J, Müller S, Laass MW, Schreiner A, et al. Probiotic Escherichia coli Nissle 1917;(EcN: for successful remission maintenance of ulcerative colitis in children and adolescents: an open-label pilot study. Zeitschrift Für Gastroenterol. 2008;46:874–5. doi: 10.1055/s-2008-1027463. [DOI] [PubMed] [Google Scholar]
- 125.Petersen AM, Mirsepasi H, Halkjær SI, Mortensen EM, et al. Ciprofloxacin and probiotic Escherichia coli Nissle add-on treatment in active ulcerative colitis: a double-blind randomized placebo controlled clinical trial. J Crohns Colitis. 2014;8:1498–505. doi: 10.1016/j.crohns.2014.06.001. [DOI] [PubMed] [Google Scholar]
- 126.Fedorak RN, Feagan BG, Hotte N, Leddin D, et al. The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clin Gastroenterol Hepatol. 2015;13:928–935. doi: 10.1016/j.cgh.2014.10.031. [DOI] [PubMed] [Google Scholar]
- 127.Kanauchi O, Suga T, Tochihara M, Hibi T, et al. Treatment of ulcerative colitis by feeding with germinated barley foodstuff: first report of a multicenter open control trial. J Gastroenterol. 2013;37:67–72. doi: 10.1007/BF03326417. [DOI] [PubMed] [Google Scholar]
- 128.Ben-Arye E, Goldin E, Wengrower D, Stamper A, et al. Wheat grass juice in the treatment of active distal ulcerative colitis: a randomized double-blind placebo-controlled trial. Scand J Gastroenterol. 2002;37:444–449. doi: 10.1080/003655202317316088. [DOI] [PubMed] [Google Scholar]
- 129.Benjamin JL, Hedin CRH, Koutsoumpas A, Ng SC, et al. Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s disease. Gut. 2011;60:923–929. doi: 10.1136/gut.2010.232025. [DOI] [PubMed] [Google Scholar]
- 130.Steed H, Macfarlane GT, Blackett KL, Bahrami B, et al. Clinical trial: the microbiological and immunological effects of synbiotic consumption - a randomized double-blind placebo-controlled study in active Crohn’s disease. Aliment Pharmacol Ther. 2010;32:872–883. doi: 10.1111/j.1365-2036.2010.04417.x. [DOI] [PubMed] [Google Scholar]
- 131.Fukumoto S, Toshimitsu T, Matsuoka S, Maruyama A, et al. Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis. Immunol Cell Biol. 2014;92:460–465. doi: 10.1038/icb.2014.2. [DOI] [PubMed] [Google Scholar]
- 132.Candela M, Perna F, Carnevali P, Vitali B, et al. Interaction of probiotic Lactobacillus and Bifidobacterium strains with human intestinal epithelial cells: Adhesion properties, competition against enteropathogens and modulation of IL-8 production. Int J Food Microbiol. 2008;125:286–292. doi: 10.1016/j.ijfoodmicro.2008.04.012. [DOI] [PubMed] [Google Scholar]
- 133.Fukuda S, Toh H, Hase K, Oshima K, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469:543–547. doi: 10.1038/nature09646. [DOI] [PubMed] [Google Scholar]
- 134.Colombel JF, Rutgeerts P, Reinisch W, Esser D, et al. Early mucosal healing with infliximab is associated with improved long-term clinical outcomes in ulcerative colitis. Gastroenterology. 2011;141:1194–1201. doi: 10.1053/j.gastro.2011.06.054. [DOI] [PubMed] [Google Scholar]
- 135.Baert F, Moortgat L, Van Assche G, Caenepeel P, et al. Mucosal healing predicts sustained clinical remission in patients with early-stage Crohn’s disease. Gastroenterology. 2010;138:463–468. doi: 10.1053/j.gastro.2009.09.056. [DOI] [PubMed] [Google Scholar]
- 136.Borrelli O, Cordischi L, Cirulli M, Paganelli M, et al. Polymeric diet alone versus corticosteroids in the treatment of active pediatric Crohn’s disease: a randomized controlled open-label trial. Clin Gastroenterol Hepatol. 2006;4:744–753. doi: 10.1016/j.cgh.2006.03.010. [DOI] [PubMed] [Google Scholar]
- 137.Momozawa Y, Deffontaine V, Louis E, Medrano JF. Characterization of bacteria in biopsies of colon and stools by high throughput sequencing of the V2 region of bacterial 16S rRNA gene in human. PLoS One. 2011;6:e16952. doi: 10.1371/journal.pone.0016952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Belkaid Y, Naik S. Compartmentalized and systemic control of tissue immunity by commensals. Nat Immunol. 2013;14:646–53. doi: 10.1038/ni.2604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Faith JJ, Colombel JF, Gordon JI. Identifying strains that contribute to complex diseases through the study of microbial inheritance. Proc Natl Acad Sci U S A. 2015;112:633–640. doi: 10.1073/pnas.1418781112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Miquel S, Leclerc M, Martin R, Chain F, et al. Identification of metabolic signatures linked to anti-inflammatory effects of Faecalibacterium prausnitzii. MBio. 2015;6:e00300–15. doi: 10.1128/mBio.00300-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jansson J, Willing B, Lucio M, Fekete A, et al. Metabolomics reveals metabolic biomarkers of Crohn’s disease. PLoS One. 2009;4:e6386. doi: 10.1371/journal.pone.0006386. [DOI] [PMC free article] [PubMed] [Google Scholar]