366
Views
135
CrossRef citations to date
0
Altmetric
Review

The microbiota in inflammatory bowel disease: current and therapeutic insights

, &
Pages 63-73 | Published online: 10 Jun 2017

Abstract

Inflammatory bowel disease is a heterogeneous group of chronic disorders that result from the interaction of the intestinal immune system with the gut microbiome. Until recently, most investigative efforts and therapeutic breakthroughs were centered on understanding and manipulating the altered mucosal immune response that characterizes these diseases. However, more recent studies have highlighted the important role of environmental factors, and in particular the microbiota, in disease onset and disease exacerbation. Advances in genomic sequencing technology and bioinformatics have facilitated an explosion of investigative inquiries into the composition and function of the intestinal microbiome in health and disease and have advanced our understanding of the interplay between the gut microbiota and the host immune system. The gut microbiome is dynamic and changes with age and in response to diet, antibiotics and other environmental factors, and these alterations in the microbiome contribute to disease onset and exacerbation. Strategies to manipulate the microbiome through diet, probiotics, antibiotics or fecal microbiota transplantation may potentially be used therapeutically to influence modulate disease activity. This review will characterize the factors involved in the development of the intestinal microbiome and will describe the typical alterations in the microbiota that are characteristic of inflammatory bowel disease. Additionally, this manuscript will summarize the early but promising literature on the role of the gut microbiota in the pathogenesis of inflammatory bowel disease with implications for utilizing this data for diagnostic or therapeutic application in the clinical management of patients with these diseases.

Introduction

Inflammatory bowel disease (IBD) represents a heterogeneous group of chronic immune-mediated inflammatory diseases affecting the gastrointestinal tract. There are two primary phenotypes of IBD, ulcerative colitis (UC) and Crohn’s disease (CD). UC is characterized by chronic contiguous and circumferential mucosal inflammation extending proximally from the rectum, but is isolated to the colon. In comparison, the stereotypical inflammation seen in CD is patchy and transmural, and may affect any part of the gastrointestinal tract. Although the etiology of IBD is incompletely understood, recent studies support the hypothesis that IBD results from a complex interplay of genetics, immune dysregulation and environmental triggers that may exert their effect through alterations of the intestinal microbiota.

Currently, IBD affects an estimated 1.4 million individuals in the USA, with a significant increase in incidence over the past decades. This has spurred the hypothesis that environmental factors play a critical role in the pathogenesis of IBD.Citation1,Citation2 Further evidence of environmental impact on disease development is seen in the discordant incidence of IBD in industrialized compared with non-industrialized countries, as well as the rising incidence of IBD in countries that are undergoing demographic and economic development.Citation3,Citation4 Similarly, children emigrating from countries with low IBD prevalence to countries with high IBD prevalence assume the same risk of developing IBD as their peers residing in high IBD prevalence areas for many generations.Citation5 While genes play a clear role in the pathogenesis of IBD, with over 200 genes predisposing to IBD discovered so far, the critical role of environment is further highlighted by monozygotic twin studies, where concordance of IBD among monozygotic twin pairs is <50% as well as the fact that there is incomplete penetrance of gene abnormalities predisposing to IBD in the general population.Citation6Citation8

Current theories of IBD pathogenesis postulate that pathologic alterations in the intestinal microbiome trigger an aberrant mucosal immune response in genetically predisposed individuals, leading to the development of chronic intestinal inflammation. These pathologic alterations in gut microbial composition seen in IBD are referred to as intestinal “dysbiosis.” Research suggests that perturbations in the gut microbiome are an essential factor triggering inflammation in IBD rather than merely a consequence of the chronic inflammation.Citation9 Abundant evidence supports the integral role the intestinal microbiome plays in the pathogenesis of IBD, including: 1) a characteristic dysbiosis is often observed in individuals with CD, UC and pouchitis,Citation10Citation16 2) fecal stream diversion improves disease activity in CD, whereas reinfusion of fecal contents results in recurrent inflammation,Citation17 3) the majority of IBD susceptibility genetic polymorphisms are associated with host mucosal barrier function and are involved in host–microbiome interactions,Citation18Citation23 4) antibiotics and probiotics have been shown to be effective for induction or maintenance of remission in IBD,Citation24Citation26 5) depletion of commensal microbes can result in impaired mucosal healing, chronic mucosal inflammation and colitis,Citation27 and 6) germ-free animals do not develop colitis without introduction of fecal bacteria to induce inflammation.Citation28

While advances in bioinformatics, genomics and experimental models of IBD have identified how environmental factors such as age, diet and antibiotic exposure contribute to the development of dysbiosis and aberrant gut microbial–host immunologic interactions, many questions remain. The following review aims to: 1) describe factors in the development of the intestinal microbiome, 2) define features of intestinal dysbiosis in IBD, and, 3) explore how current knowledge may lead to the development of therapeutic interventions by harnessing the microbiome in the treatment of IBD.

The microbiome in health: factors that influence development, alteration and maintenance of structure and function

The gut microbiota is the largest and most diverse community of microbes in the human body. The intestinal microbiota, or microorganism population of the intestine, constitutes only a fraction of the complexity of the intestinal microbiome, which includes diverse array of microbial genes and gene products of the microbiota. During times of good health, the intestinal microbiome acts symbiotically to produce vitamins, repress expansion of pathologic organisms and facilitate digestion of dietary substrates, all the while in constant contact with the host immune system.Citation29 Additionally, the microbiome contributes to gut epithelial cell renewal and enteric immune system development. This diverse community of bacteria, fungi, bacteriophages, and archaea exist in colonies of varying density throughout the gastrointestinal tract, with the highest microbial density reaching 1012 cells/g of luminal contents in the colon.Citation30 Attempts have been made to delineate what constitutes a “healthy” microbial composition of the intestinal microbiome, with varying and somewhat disparate results. The difficulty in defining a “healthy” microbiome is based upon the complexity and variations found in the fecal microbiome, with more than 1000 potential bacterial species able to colonize the human intestines. Despite the large variation found in an individual’s microbiome, the majority of species (>90%) belong to the Bacteroidetes and Firmicutes phyla.Citation31,Citation32

The composition of the gut microbiota changes over time. An individual’s intestinal microbiome is more malleable in infancy and early childhood, assumes more stability and similarity to the general population in adulthood and subsequently loses diversity in the elderly.Citation12,Citation33 During infancy, the gut microbiome is typically minimally complex and is influenced by birth route and diet.Citation34Citation36 Method of delivery (cesarean section versus vaginal delivery) differentially affects the neonatal intestinal microbiota composition, with vaginal and fecal flora colonizing the newborn gut if born vaginally and typical skin flora if born via cesarean section.Citation37 A Danish cohort study of infants born between 1973 and 2008 demonstrated that cesarean sections were associated with a moderately increased risk of IBD; however, this finding was recently disputed by Bernstein et al, who observed that patients with IBD were not more likely to have been born via cesarean section than controls or their siblings without IBD.Citation38,Citation39

Infant diet also impacts the composition of the intestinal microbiome. Exclusively breastfed infants have been found to have increased numbers of taxa from Actinobacteria whereas formula-fed infants have higher levels of γ-Proteobacteria.Citation40,Citation41 Additionally, formula-fed infants demonstrate more “adult”-like patterns of intestinal microbiota. Interestingly, intestinal microbiota of breast milk-fed infants are significantly less diverse than formula-fed infants, but their microbial genes demonstrated more robust interactions with host immune system, metabolism and biosynthesis.Citation42 There is a reduced risk of development of IBD among infants who were breastfed.Citation43 The cessation of breastfeeding and associated reduction in passage of maternal IgA induces changes in the microbiome characteristic of an adult microbiome, including increased prominence of Firmicutes and Bateriodetes; however, peak microbial diversity and microbiome stability is often not reached until adulthood.Citation44Citation46

During childhood and adulthood, diet appears to have a major influence on microbial composition and function.Citation47 A controlled study examining the effects of diet on the microbiome demonstrated that the composition of gut microbiota is changed dramatically by increases in dietary fat and decreases in dietary fiber compared with low-fat/high-fiber diets.Citation48 African children on high-fiber vegetarian diets demonstrate different gut microbiomes than their European peers, whose diets are typified by high sugar, fat and animal protein.Citation49 Diets rich in animal fats have also been shown to favor increased density of bile-tolerant organisms, including Bilophila wadsworthia.Citation47 Interleukin (IL)-10 knockout mice fed diets rich in milk-derived saturated fat favored colonization with Proteobacteria (specifically B. wadsworthia), compared with diets high in polyunsaturated fat. Interestingly, these IL-10 knockout mice fed diets high in saturated milk fat demonstrated increased incidence and severity of spontaneous colitis, providing a potential mechanistic link between the observed association between the proliferation of Westernized diets and increased incidence of IBD globally.Citation50

Similarly, low vitamin D has been identified as a potential risk factor for the development of IBD.Citation51 Vitamin D deficiency may contribute to intestinal inflammation through multiple effects and alters the gut microbiome.Citation52 Alternatively, supplementation with vitamin D has been shown to alter the intestinal microbiome by increasing alpha diversity (species richness).Citation53 While abundant data support dietary influences as having a dramatic, either pro-inflammatory or anti-inflammatory effect on the intestinal microbiome, further research is required to delineate the mechanism by which specific dietary alterations are useful in the treatment of IBD.

Alterations in intestinal microbiome implicated in the development of IBD

While IBD is clearly associated with intestinal dysbiosis, no single microbe or microbial milieu has been proven causal. Intestinal dysbiosis may contribute to the pathogenesis of IBD by loss of “health-promoting” or potential gain of “pathobionts.” Pathobionts are distinguishable from bacterial pathogens in that they only become pathologic in the setting of a specific environmental stimulus in genetically susceptible individuals. With recent advances in genetic sequencing and functional microbial analysis, many studies have been able to identify intestinal dysbioses that are present in patients with IBD. While there are somewhat disparate results, common themes among studies support the finding of a generalized reduction in biodiversity (alpha diversity) as well as decreased representation of several specific taxa, including Firmicutes and Bacteroidetes, among individuals with IBD.Citation10Citation16 Additionally, specific taxonomic shifts have been associated with IBD, including a relative increase in the abundance of Enterobacteriaceae, including Escherichia coli and Fusobacterium.Citation54Citation56 Newly diagnosed, treatment-naïve patients provide an ideal human study population in which to observe the potentially pathologic intestinal dysbioses that occur in IBD. Interestingly, in a large cohort of newly diagnosed, treatment-naïve children with CD, ileal and rectal biopsy samples demonstrated an increased abundance of Enterobacteriaceae, Pasteurellaceae, Veillonellaceae, and Fusobacteriaceae and decreased abundance of Bacteroidales and Clostridia.Citation13 Fite et al demonstrated that not only do mucosa-associated microbes differ between those with active colonic inflammation (UC) and those with normal mucosa, there appears to be longitudinal variation in mucosal bacterial populations in UC that is associated with disease severity.Citation57 Moreover, fecal microbiota composition in active UC is consistent across geographic location, age and gender, and IBD-associated microbiota appears to be stable during remission among those with UC.Citation58

Although fewer studies have examined the role of fungi specifically in the propagation of inflammation in IBD, fungi are a ubiquitous component of the intestinal microbiome. In CD, intestinal fungal communities have increased diversity in colonic and ileal biopsy samples compared with healthy controls.Citation59,Citation60 In pediatric patients with CD, five fungal taxa were found to be associated with CD, including: Saccharomyces cerevisiae, Calvispora lusitaniae, Cyberlindnera jadinii, Candida albicans, and Kluyveromyces marxianus.Citation61 In addition, fungal microbial components are utilized in the diagnosis and prognosis of CD; specifically anti-saccharomyces cerevisiae antibodies (ASCA), which react with a yeast cell wall polysaccharide, serve as a serological marker for ileal CD.Citation1 A recent study demonstrated increased abundance of Candida tropicalis in CD patients compared with their unaffected relatives, and this correlated with ASCA titers and abundance of Serratia marcescens and E. coli in biofilms.Citation62 Like fungi, bacteriophages are an understudied population of the intestinal microbiome. Colonic mucosal biopsy samples from patients with CD had significantly more bacteriophage than healthy controls.Citation63,Citation64 However, given the relative paucity of data examining the functional role of fungi, viruses and archaea in the intestinal microbiome, further studies are required to develop a deeper understanding of how changes in these populations may result in IBD.

Interestingly, studies have demonstrated a reduced diversity of intestinal microbiota in inflamed versus normal intestinal mucosa within the same patient, suggesting that host immune factors play a role in maintaining or reacting to mucosal inflammation.Citation65 It has long been suspected that an individual’s genotype contributes to overall susceptibility in developing IBD. Studies evaluating specific genetic variants have highlighted the interplay between the gut immune system and intestinal microbiota. The first gene identified to confer susceptibility in developing CD was nucleotide-binding oligomerization domain containing 2 (NOD2).Citation66 Under normal circumstances, NOD2 is primarily expressed in Paneth cells, the role of which in the intestinal mucosa is to produce antimicrobial defensins. The product of NOD2 stimulates host immune response upon recognition of microbial cell wall components. Patients with NOD2 mutations have been found to have decreased IL-10, an important anti-inflammatory cytokine. Additionally, those with NOD2 mutations have been found to harbor increased intestinal mucosal adherent bacteria.Citation10,Citation67 In mice, intestinal microbiota affects expression of NOD2.Citation68 NOD2-deficient mice demonstrate significant increases in Bacteroides, Firmicutes, and Bacilli in their terminal ileum, and decreased ability to clear pathogenic Helicobacter hepaticus.Citation68 Patients with NOD2 risk alleles have decreased clostridium groups XIVa and IV and increased Actinobacteria, Proteobacteria and Enterobacteriaceae.Citation10 Since the initial identification of NOD2 and associated risk alleles, over 160 genetic loci have been identified to modulate the risk of IBD.Citation6,Citation19,Citation69 Furthermore, genetic analyses have linked specific loci associated with a dysregulated intestinal immune response to commensal microbes with the development of IBD.Citation6

Analogous to the dynamic relationship between mucosal bacterial milieu and the host immune system, fungal microbiome communities interact with host immune cells via the innate immune receptor, Dectin-1.Citation70 Dectin-1 recognizes fungal cell wall glucans and activates intracellular signals through the CARD9 receptor, leading to production of inflammatory cytokines and subsequent induction of Th17 responses.Citation70 Variants in CARD9 and Decin-1 are associated with CD and UC, specifically, mutations in Dectin-1 (CLEC7A) are associated with medically refractory UC.Citation69,Citation70

Environmental factors affecting the intestinal microbiome and the development of IBD

While antibiotics may have a therapeutic role in established IBD, several studies have shown that antibiotic exposure prior to the development of IBD is associated with incident IBD. In a retrospective cohort of Danish children, a greater relative risk of developing IBD was observed among children exposed to antibiotics.Citation71 Likewise, a Finnish study demonstrated the risk of pediatric-onset CD increased with the number of purchased courses of antibiotics from birth.Citation72 A large retrospective cohort study from the UK demonstrated that exposure to antibiotics, particularly anaerobic antibiotics, during childhood was associated with a relative rate of increase in the development of IBD by 84%.Citation73 In a recent meta-analysis, exposure to antibiotics during childhood was shown to be associated with increased risk of CD but not UC.Citation74 Together, these findings suggest that early life and repeated exposures of antibiotics may result in sustained, potentially detrimental effects on the intestinal microbiome and could contribute to the pathogenesis of IBD.

As previously mentioned, diet has a significant effect on the fecal microbiome. With this in mind, several large longitudinal studies have demonstrated an association between reduced risk of IBD and a diet high in fruits and vegetables, and an elevated risk of IBD in those who consumed diets rich in animal fats and refined sugars.Citation4 In addition, Western diets high in fat have been demonstrated to increase risk of IBD; specifically, consumption of a high ratio of omega-6 fatty acids (pro-inflammatory) to omega-3 fatty acids (anti-inflammatory) has been associated with an increased incidence of UC.Citation75,Citation76 In mice, a diet high in n-6 polyunsaturated fatty acids exacerbated colitis and resulted in enrichment of the intestinal microbiome with pro-inflammatory Enterobacteriaceae and Clostridia spp.Citation77

The intestinal microbiome as a diagnostic and therapeutic tool

While earlier studies focused on species characterization of the intestinal microbiome in health versus disease, advances in metagenomics and metabolomics have introduced the importance of understanding the functional properties of the intestinal microbiome in IBD. Specifically, such functional studies have highlighted differences in: carbohydrate and lipid transport/metabolism, cell wall degradation, formation of exotoxins, and microbial adherence and invasion in IBD versus in health.Citation1 In defining the functional role of the intestinal microbial community in disease and health, there is a directive to harness this knowledge to restore health to a diseased microbiome through diet, prebiotics, probiotics, antibiotics and/or fecal microbiota transplantation (FMT).

Diet has an immense impact on gut microbiome development and diversity, and has been well studied as a potential therapy for IBD. Perhaps the largest body of literature on use of diet as a targeted IBD therapy evaluates the efficacy and microbial changes associated with exclusive enteral nutrition (EEN). EEN is a complete exclusion diet, where patients receive 100% of their daily calorie intake from formula rather than table foods. EEN may be completed with intact protein formula, semi-elemental or elemental formula. Studies employing metagenomics have demonstrated alterations in the intestinal microbiome before and after treatment with EEN.Citation61 EEN has been shown to induce alterations in the microbiome as early as 1 week after initiation. Interestingly, the intestinal microbiome in these individuals did not resemble that of healthy individuals. Similar results were demonstrated by Gerasimidis et al in a prospective, case–control study evaluating changes in fecal microbial diversity and metabolic activity of 15 children with CD treated with EEN.Citation78 Soon after initiation, and throughout treatment with EEN, fecal microbiota diversity decreased, as did concentrations of previously identified commensal microbes. These observations correlated with an improvement in clinical disease activity and reduction in inflammatory markers.Citation78,Citation79 Interestingly, these differences in microbiome were more pronounced in study subjects who responded to EEN compared with those who did not, potentially identifying biomarkers of disease phenotype and aiding future selection of individual patients who are more likely to respond to particular therapies. Analogous changes to the structure of the intestinal microbiome were not demonstrated in children on partial enteral nutrition (PEN; a diet composed of partial formula and table foods) despite receiving a similar volume of enteral formula, suggesting that the changes to the microbiome seen with EEN may stem from exclusion of table foods.Citation80

EEN in CD has been shown to improve clinical symptoms, mucosal healing and nutritional status in children, with clinical remission rates equivalent to systemic corticosteroids.Citation81 In a randomized controlled trial evaluating the efficacy of steroids versus EEN for treatment of CD, both treatments equally improved clinical symptoms as well as serum inflammatory markers, but EEN resulted in significantly better mucosal healing.Citation81 Similar clinical results have not been reproduced in adults, as EEN resulted in lower rates of clinical response than steroids, although this may be a question of compliance versus efficacy.Citation80

While EEN for induction of remission is clearly beneficial for children with CD, it is often less feasible for long-term maintenance of remission. When combined with a regular diet, even 50% of calories from EEN reduce the rate of CD relapse by 50%.Citation82 However, effectiveness at clinical and mucosal remission is greatest when 100% EEN is employed, rather than PEN.Citation83,Citation84 Given that many patients feel EEN to be prohibitively restrictive, there have been efforts to identify whole food diets that may confer similar therapeutic benefit.

The specific carbohydrate diet (SCD) is one such exclusion diet that appears to have a positive effect in IBD. The SCD diet was developed by Dr Sydney Haas, a pediatrician in the 1930s who developed the diet as a treatment for celiac disease. This diet removes all grains, including gluten. It was the primary treatment for celiac disease prior to the discovery of gluten as the offending antigen. The SCD diet was then popularized by Elaine Gottschall as treatment for IBD after her daughter’s UC was successfully treated with the diet. The diet also removes sweeteners except for honey, most processed foods and all milk products, except for hard cheeses and yogurt fermented >24 hours. In an online survey of 417 IBD patients (47% CD and 43% UC) who used the SCD as treatment, 33% reported remission at 2 months after initiation of the SCD, and 42% at both 6 and 12 months. For those reporting clinical remission, 13% reported time to achieve remission of <2 weeks, 17% reported 2 weeks to a month, 36% reported 1–3 months, and 34% reported >3 months. For individuals who reported reaching remission, 47% of individuals reported associated improvement in abnormal laboratory values.Citation85 Suskind et al reported clinical remission in seven children with active CD after initiation of the SCD diet without use of concomitant treatment.Citation86 In addition to clinical remission, these children demonstrated normalization/improvement inflammatory markers, including fecal calprotectin. Cohen et al reported clinical and mucosal improvements, as documented with capsule endoscopy in seven children with CD who used SCD for 52 weeks.Citation87 In addition to the aforementioned studies, case series including over 75 patients have shown clinical and laboratory improvement in both CD and UC patients on SCD.Citation88,Citation89

In a recent prospective multicenter study of SCD in pediatric patients with mild-to-moderate CD or UC, 12 pediatric patients (aged 10–17 years) were followed for 12 weeks. Dietary therapy was associated with clinical remission in eight patients and was ineffective for two patients, while two individuals were unable to maintain the diet. At 12 weeks, the mean C-reactive protein decreased from 24.1±22.3 mg/L to 7.1±0.4 mg/L in the Seattle cohort (nL<8.0 mg/L) and from 20.7±10.9 mg/L to 4.8±4.5 mg/L in the Atlanta cohort (nL<4.9 mg/L). Concomitant with clinical and laboratory improvements, significant changes in microbial composition occurred with the dietary change.Citation90

Other exclusion diets have been used for reduction of inflammation and symptom improvement in IBD. The Crohn’s Disease Exclusion Diet (CDED), designed and evaluated by Dr Arie Levine, is based on the hypothesis that the efficacy of EEN is dependent on exclusion of dietary components that may potentially influence intestinal permeability or promotion of a pro-inflammatory microbiome. Foods specifically excluded from the diet include: gluten, dairy, gluten-free baked goods, animal fat, processed meats, products containing emulsifiers and all canned or processed foods. In a prospective cohort of pediatric and adult patients with mild-to-moderate CD treated with 50% PEN and the CDED, Sigall-Boneh et al, showed this exclusion diet was successful in achieving induction of clinical remission, including a reduction in inflammatory markers.Citation91 Additionally, a small subgroup of seven patients treated with the CDED alone also achieved clinical remission.Citation91 Similarly, Riordan et al demonstrated that in adults with active CD, after induction of elemental diet, those who excluded trigger foods had prolonged clinical remission and improvement in serum inflammatory markers compared with those who consumed a regular diet.Citation92 Foods most frequently excluded owing to “intolerances” included: cereals, dairy products and yeast. Similarly, Rajendran et al found that IgG4-targeted exclusion diets for adults with active CD resulted in symptomatic improvement and reduction in serum inflammatory markers.Citation93 In a small, prospective controlled study of adults with CD in clinical remission, Chiba et al demonstrated that a lacto-ovo-vegetarian diet resulted in reduction in relapse and prolongation of time to relapse versus a standard omnivorous diet.Citation94 While these exclusion diets hold promise for induction or maintenance of remission in CD, further research is necessary to characterize their effects on mucosal healing and alterations in microbiome.

Other diets have not been as well studied in IBD; however, low-lactose diets have been shown to reduce clinical symptoms of IBD. Similarly, patients with IBD report symptom improvement on low FODMAP (Low Fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) diet, but there is paucity of data that supports a reduction in intestinal mucosal inflammation.Citation95,Citation96 Low FODMAP diets have been found to reduce potentially favorable bacterial species within the colon, particularly Faecalibacterium prausnitzii and reduced production of butyrate.Citation97 However, these alterations in the microbiome have unclear clinical consequences. Other restriction diets such as the vegan and vegetarian diets are popular among some patient populations. Zimmer et al demonstrated alterations in the ratios of Bacteroides spp., Bifidobacterium supp., and Enterobacteriaceae in patients adherent to vegan and vegetarian diets.Citation98 Further studies are required before these restriction diets can be recommended for the treatment of IBD.

The mechanism of action in reduction of clinical symptoms and intestinal inflammation with EEN and exclusionary diets remains unclear. Restriction diets may act through exclusion of certain substrates that modulate the intestinal microbiome, or may be related to the exclusion of food additives, including emulsifiers that may modulate the intestinal microbiome and/or directly affect the immune system through alterations in the mucus barrier as well as the epithelial lining.Citation80 Murine models of colitis (IL-10 knockout mice) fed common commercial food emulsifiers, carboxymethylcellulose and polysorbate-80 demonstrated thinning of the colonic mucosal layer, invasion of bacteria into the lamina propria, altered microbiome and worsening colitis.Citation99 Nickerson et al has demonstrated parallels between the increasing dietary prevalence of maltodextrin (MDX), a common starch-based food additive, and rising incidence of CD. Through a series of experiments to uncover potential mechanisms, Nickerson et al demonstrated that exposure to MDX enhances mucosal adhesion and biofilm formation by E. coli, including adherent-invasive E, coli, and increases viability of intracellular Salmonella in mucosal macrophages and epithelial cells. Using this evidence, they hypothesize that MDX may contribute to the development of IBD by priming the intestinal mucosal to be more sensitive to epithelial damage due to a reduction in epithelial antimicrobial defense mechanisms.Citation100 In sum, while there are promising preliminary data supporting the use of exclusion diets in the treatment of IBD, additional studies are required to draw definitive conclusions regarding how exclusion diets influence the intestinal microbiome composition and function and how these changes may result in long-term clinical and histopathological remission.

FMT is being actively explored as a means of altering the intestinal microbiome for therapeutic benefit in IBD. The rationale supporting FMT as a putative therapeutic modality includes the observation that active IBD is characterized by reduced colonic microbial diversity and the hypothesis that “healthy” donor microbiota may restore homeostasis to the aberrant microbiome and host immune response. Initial interest in utilizing FMT for IBD stemmed from the great success of FMT in treating recurrent or refractory Clostridium difficile infections, another disorder characterized by loss of diversity in the microbiome.Citation101 Unfortunately, results of clinical studies exploring FMT as a therapy for IBD have been less robust.Citation69,Citation102,Citation103 Although some smaller studies have shown potential efficacy of FMT in UC, two larger randomized placebo control trials of FMT in UC demonstrated little to no efficacy.Citation104Citation106 There are currently insufficient data to support the use of FMT for treatment of CD, but given the small number of studies and small patient sample sizes, additional studies are required to draw definitive conclusions on the efficacy of FMT for induction or maintenance of remission in IBD.

Antibiotics have long been utilized to manage IBD. Primarily, antibiotics have been employed as an adjunctive treatment in specific clinical scenarios, such as perianal and intra-abdominal abscesses, fistula and toxic megacolon.Citation24,Citation26 However, certain antibiotics such as ciprofloxacin, metronidazole, and rifaximin with their broad-spectrum antimicrobial coverage have been explored as a primary therapy. In theory, antibiotics may alter the clinical course of IBD by several mechanisms, including: decreasing luminal bacteria concentrations, altering the microbial composition to potentially favor the establishment of beneficial bacteria, and decreasing bacterial tissue invasion.Citation26 Clinically, treatment with enteral antibiotics has been shown to reduce intestinal inflammation and has been efficacious in treating mucosal inflammation in CD and pouchitis.Citation24Citation26,Citation107 However, their use as maintenance therapy in CD is less clear. In a randomized, placebo-controlled, blinded study of 213 adults with active CD, a combination of clarithromycin, rifabutin and clofazimine resulted in short-term improvement in remission, but offered no benefit for prevention of long-term relapse.Citation108 A meta-analysis evaluating randomized controlled trials in which antibiotic therapy was compared with placebo for the treatment of IBD demonstrated some benefit of using antibiotics to treat IBD. A total of 10 studies were included in the evaluation of antibiotics in CD and found a pooled odds ratio (OR) for clinical improvement of 1.35 (95% confidence interval [CI], 1.16–1.58) for the use of antibiotic therapy compared with placebo. For patients with UC, a total of nine studies included in the analysis demonstrated a pooled OR of 2.17 (95% CI: 1.54–3.05) in favor of antibiotic therapy.Citation109 Despite mounting evidence, additional randomized controlled trials are required to guide decisions regarding the specific clinical roles of antibiotics in the treatment of IBD.

Several studies have evaluated probiotics, both in induction and maintenance of remission in IBD. Probiotics, as a whole, have not demonstrated efficacy in the treatment of CD; however, there may be some role for the use of specific probiotics in the management of mild-to-moderate active UC or recurrent pouchitis after ileoanal anastomosis.Citation110 VSL#3 is a highly-concentrated probiotic preparation that contains eight different types of bacteria within the Lactobacillus, Streptococcus and Bifidobacterium spp. In placebo-controlled trials, VSL#3 was shown to prevent recurrence of pouchitis.Citation111,Citation112 While data exist to support the limited utility of certain probiotics in the treatment of IBD, further studies are required to delineate the anti-inflammatory mechanism of probiotics in IBD.Citation113

Prebiotics are compounds that change the structure or metabolome of the intestinal microbiota. Inulin and oligofructose are two prebiotics that have been shown to promote the growth of beneficial Bifidobacterium and Lactobacillus spp. both in humans and in rats.Citation114,Citation115 Additionally, cellobiose and rice fiber are dietary-fiber sourced prebiotics that have been shown to reduce pro-infammatory cytokines in experimental models of colitis.Citation116 Few studies have examined the beneficial clinical effects of prebiotics in active IBD. Benjamin et al in a randomized placebo-controlled study of adults with active CD evaluated changes in disease activity index after administration of fructo-oligosaccharides (FOS). Despite some changes to immunoloregulation of dendritic cells observed in those receiving FOS, no significant clinical improvements were seen, nor were there significant changes to the fecal microbiome between the groups at baseline or after the 4-week intervention.Citation117

Fermentable fiber, another form of prebiotic, is metabolized by colonic bacteria to short chain fatty acids (SCFA),that is, acetate, propionate and butyrate, which are known to modulate cell proliferation, histone acetylation, gene expression and immune response.Citation118 SCFA-producing bacterial strains in Clostridia clusters IV, XIVa and XVII from healthy individuals induce colonic regulatory T cells (Tregs) differentiation, expansion and function.Citation119 SCFA produced by Clostridia spp have been found to induce CD4+ T regulatory cells (Treg) and reduce colitis in mouse models.Citation69,Citation120 In adults with IBD, fecal samples demonstrated reduced concentrations of butyrate and acetate compared with healthy controls, suggesting that SCFA may play some protective role in the prevention of intestinal inflammation.Citation121 In animal models of IBD, supplementation with soluble fiber has been shown to reduce intestinal inflammation by increasing the production of SCFA and altering the intestinal microbiome.Citation122,Citation123 In humans, dietary fiber may reduce the risk of flare in patients with CD, although additional studies are required to recommend fiber supplementation as an anti-inflammatory therapy in IBD.Citation124

Future studies

Defining the characteristics of “disease” and “health” in intestinal microbiota is only the first step in ascertaining the pathogenesis of IBD. To date, specific profiles of intestinal microbiota have not been beneficial as a diagnostic test or biomarker for IBD; and further studies are required to identify whether aspects of the microbiome may be useful in identifying disease phenotype or predicting response to therapy.Citation11,Citation13,Citation14,Citation69 Many earlier studies have been wrought with several challenges in delineating causality when reporting microbial changes associated with IBD. Additionally, robust meta-analysis to draw conclusions have been challenging, given the diversity of microbial sampling and important clinical aspects of disease, including disease phenotype, location of disease, and prior pharmacologic exposures.Citation1 Fortunately, with evolving experimental technologies to assist in the functional characterization of the microbiome, there is promise to move beyond defining the phenotypic footprint of the microbiome in IBD to identify how the microbiome contributes to: 1) the onset and propagation of disease, and potentially more significantly, 2) how we may manipulate the microbiome as a future treatment of IBD.

Disclosure

The authors report no conflicts of interest in this work.

References

  • MiyoshiJChangEBThe gut microbiota and inflammatory bowel diseasesTransl Res2017179384827371886
  • LoftusEVJrClinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influencesGastroenterology200412661504151715168363
  • MolodeckyNASoonISRabiDMIncreasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic reviewGastroenterology201214214654e42 quiz e3022001864
  • AnanthakrishnanANKhaliliHKonijetiGGA prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitisGastroenterology2013145597097723912083
  • PonderALongMDA clinical review of recent findings in the epidemiology of inflammatory bowel diseaseClinEpidemiol20135237247
  • KosticADXavierRJGeversDThe microbiome in inflammatory bowel disease: current status and the future aheadGastroenterology201414661489149924560869
  • HalmeLPaavola-SakkiPTurunenULappalainenMFarkkilaMKontulaKFamily and twin studies in inflammatory bowel diseaseWorld J Gastroenterol200612233668367216773682
  • YazdanyarSKamstrupPRTybjaerg-HansenANordestgaardBGPenetrance of NOD2/CARD15 genetic variants in the general populationCMAJ2010182766166520371648
  • HansenJJSartorRBTherapeutic manipulation of the microbiome in IBD: current results and future approachesCurr Treat Options Gastroenterol201513110512025595930
  • FrankDNRobertsonCEHammCMDisease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseasesInflamm Bowel Dis201117117918420839241
  • MorganXCTickleTLSokolHDysfunction of the intestinal microbiome in inflammatory bowel disease and treatmentGenome Biol2012139R7923013615
  • LiJButcherJMackDStintziAFunctional impacts of the intestinal microbiome in the pathogenesis of inflammatory bowel diseaseInflamm Bowel Dis201521113915325248007
  • GeversDKugathasanSDensonLAThe treatment-naive microbiome in new-onset Crohn’s diseaseCell Host Microbe201415338239224629344
  • FrankDNSt AmandALFeldmanRABoedekerECHarpazNPaceNRMolecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseasesProcNatlAcadSci U S A2007104341378013785
  • OttSJMusfeldtMWenderothDFReduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel diseaseGut200453568569315082587
  • ManichanhCRigottier-GoisLBonnaudEReduced diversity of faecalmicrobiota in Crohn’s disease revealed by a metagenomic approachGut200655220521116188921
  • FicheraAMcCormackRRubinMAHurstRDMichelassiFLong-term outcome of surgically treated Crohn’s colitis: a prospective studyDis Colon Rectum200548596396915785882
  • LeeYKMazmanianSKHas the microbiota played a critical role in the evolution of the adaptive immune system?Science201033060121768177321205662
  • JostinsLRipkeSWeersmaRKHost-microbe interactions have shaped the genetic architecture of inflammatory bowel diseaseNature2012491742211912423128233
  • McGovernDPJonesMRTaylorKDInternational IBD Genetics ConsortiumFucosyltransferase 2 (FUT2) non-secretor status is associated with Crohn’s diseaseHum Mol Genet201019173468347620570966
  • RauschPRehmanAKunzelSColonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (Secretor) genotypeProcNatlAcadSci U S A2011108471903019035
  • ElinavEStrowigTKauALNLRP6 inflammasome regulates colonic microbial ecology and risk for colitisCell2011145574575721565393
  • DheerRSantaolallaRDaviesJMIntestinal epithelial toll-like receptor 4 signaling affects epithelial function and colonic microbiota and promotes a risk for transmissible colitisInfect Immun201684379881026755160
  • SartorRBTherapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebioticsGastroenterology200412661620163315168372
  • RietdijkSTD’HaensGRRecent developments in the treatment of inflammatory bowel diseaseJ Dig Dis201314628228723419117
  • PerencevichMBurakoffRUse of antibiotics in the treatment of inflammatory bowel diseaseInflamm Bowel Dis200612765166416804403
  • SalehMTrinchieriGInnate immune mechanisms of colitis and colitis-associated colorectal cancerNat Rev Immunol201111192021151034
  • KennedyRJHoperMDeodharKErwinPJKirkSJGardinerKRInterleukin 10-deficient colitis: new similarities to human inflammatory bowel diseaseBr J Surg200087101346135111044159
  • Human Microbiome Project CStructure, function and diversity of the healthy human microbiomeNature2012486740220721422699609
  • DaveMHigginsPDMiddhaSRiouxKPThe human gut microbiome: current knowledge, challenges, and future directionsTransl Res2012160424625722683238
  • QinJLiRRaesJA human gut microbial gene catalogue established by metagenomic sequencingNature20104647285596520203603
  • EckburgPBBikEMBernsteinCNDiversity of the human intestinal microbial floraScience200530857281635163815831718
  • ClaessonMJJefferyIBCondeSGut microbiota composition correlates with diet and health in the elderlyNature2012488741017818422797518
  • Dominguez-BelloMGBlaserMJLeyREKnightRDevelopment of the human gastrointestinal microbiota and insights from high-throughput sequencingGastroenterology201114061713171921530737
  • YatsunenkoTReyFEManaryMJHuman gut microbiome viewed across age and geographyNature2012486740222222722699611
  • LimESZhouYZhaoGEarly life dynamics of the human gut virome and bacterial microbiome in infantsNat Med201521101228123426366711
  • JakobssonHEAbrahamssonTRJenmalmMCDecreased gut microbiota diversity, delayed Bacteroidetescolonisation and reduced Th1 responses in infants delivered by caesarean sectionGut201463455956623926244
  • BagerPSimonsenJNielsenNMFrischMCesarean section and offspring’s risk of inflammatory bowel disease: a national cohort studyInflamm Bowel Dis201218585786221739532
  • BernsteinCNBanerjeeATargownikLECesarean section delivery is not a risk factor for development of inflammatory bowel disease: apopulation-based analysisClinGastroenterolHepatol20161415057
  • BezirtzoglouETsiotsiasAWellingGWMicrobiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH)Anaerobe201117647848221497661
  • PendersJThijsCVinkCFactors influencing the composition of the intestinal microbiota in early infancyPediatrics2006118251152116882802
  • PraveenPJordanFPriamiCMorineMJThe role of breast-feeding in infant immune system: a systems perspective on the intestinal microbiomeMicrobiome201534126399409
  • BarclayARRussellRKWilsonMLGilmourWHSatsangiJWilsonDCSystematic review: the role of breastfeeding in the development of pediatric inflammatory bowel diseaseJ Pediatr2009155342142619464699
  • BackhedFRoswallJPengYDynamics and stabilization of the human gut microbiome during the first year of lifeCell Host Microbe201517569070325974306
  • PlanerJDPengYKauALDevelopment of the gut microbiota and mucosal IgA responses in twins and gnotobiotic miceNature2016534760626326627279225
  • LozuponeCAStombaughJIGordonJIJanssonJKKnightRDiversity, stability and resilience of the human gut microbiotaNature2012489741522023022972295
  • DavidLAMauriceCFCarmodyRNDiet rapidly and reproducibly alters the human gut microbiomeNature2014505748455956324336217
  • WuGDChenJHoffmannCLinking long-term dietary patterns with gut microbial enterotypesScience2011334605210510821885731
  • De FilippoCCavalieriDDi PaolaMImpact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural AfricaProcNatlAcadSci U S A2010107331469114696
  • DevkotaSWangYMuschMWDietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− miceNature2012487740510410822722865
  • AnanthakrishnanANKhaliliHHiguchiLMHigher predicted vitamin D status is associated with reduced risk of Crohn’s diseaseGastroenterology2012142348248922155183
  • GargMRosellaOLubelJSGibsonPRAssociation of circulating vitamin D concentrations with intestinal but not systemic inflammation in inflammatory bowel diseaseInflamm Bowel Dis201319122634264324105392
  • BashirMPrietlBTauschmannMEffects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tractEur J Nutr20165541479148926130323
  • LuppCRobertsonMLWickhamMEHost-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of EnterobacteriaceaeCell Host Microbe20072211912918005726
  • Darfeuille-MichaudABoudeauJBuloisPHigh prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s diseaseGastroenterology2004127241242115300573
  • OhkusaTSatoNOgiharaTMoritaKOgawaMOkayasuIFusobacteriumvarium localized in the colonic mucosa of patients with ulcerative colitis stimulates species-specific antibodyJ GastroenterolHepatol2002178849853
  • FiteAMacfarlaneSFurrieELongitudinal analyses of gut mucosal microbiotas in ulcerative colitis in relation to patient age and disease severity and durationJ ClinMicrobiol2013513849856
  • Rajilic-StojanovicMShanahanFGuarnerFde VosWMPhylogenetic analysis of dysbiosis in ulcerative colitis during remissionInflamm Bowel Dis201319348148823385241
  • OttSJKuhbacherTMusfeldtMFungi and inflammatory bowel diseases: alterations of composition and diversityScand J Gastroenterol200843783184118584522
  • LiQWangCTangCHeQLiNLiJDysbiosis of gut fungal microbiota is associated with mucosal inflammation in Crohn’s diseaseJ ClinGastroenterol2014486513523
  • LewisJDChenEZBaldassanoRNInflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn’sdiseaseCell Host Microbe201518448950026468751
  • HoarauGMukherjeePKGower-RousseauCBacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn’s DiseaseMBio201675 pii: e01250-16
  • LepagePColombetJMarteauPSime-NgandoTDoréJLeclercMDysbiosis in inflammatory bowel disease: a role for bacteriophages?Gut200857342442518268057
  • NormanJMHandleySABaldridgeMTDisease-specific alterations in the enteric virome in inflammatory bowel diseaseCell2015160344746025619688
  • SepehriSKotlowskiRBernsteinCNKrauseDOMicrobial diversity of inflamed and noninflamed gut biopsy tissues in inflammatory bowel diseaseInflamm Bowel Dis200713667568317262808
  • OguraYBonenDKInoharaNA frameshift mutation in NOD2 associated with susceptibility to Crohn’s diseaseNature2001411683760360611385577
  • PhilpottDJGirardinSECrohn’s disease-associated Nod2 mutants reduce IL10 transcriptionNat Immunol200910545545719381138
  • Petnicki-OcwiejaTHrncirTLiuYJNod2 is required for the regulation of commensal microbiota in the intestineProcNatlAcadSci U S A2009106371581315818
  • XavierRJMicrobiota as therapeutic targetsDig Dis201634555856527331403
  • IlievIDFunariVATaylorKDInteractions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitisScience201233660861314131722674328
  • HviidASvanstromHFrischMAntibiotic use and inflammatory bowel diseases in childhoodGut2011601495420966024
  • VirtaLAuvinenAHeleniusHHuovinenPKolhoKLAssociation of repeated exposure to antibiotics with the development of pediatric Crohn’s disease–a nationwide, register-based finnish case-control studyAm J Epidemiol2012175877578422366379
  • KronmanMPZaoutisTEHaynesKFengRCoffinSEAntibiotic exposure and IBD development among children: a population-based cohort studyPediatrics20121304e794e80323008454
  • UngaroRBernsteinCNGearryRAntibiotics associated with increased risk of new-onset Crohn’s disease but not ulcerative colitis: a meta-analysisAm J Gastroenterol2014109111728173825223575
  • Investigators IBDiESTjonnelandAOvervadKLinoleic acid, a dietary n-6 polyunsaturated fatty acid, and the aetiology of ulcerative colitis: a nested case-control study within a European prospective cohort studyGut200958121606161119628674
  • AnanthakrishnanANKhaliliHKonijetiGGLong-term intake of dietary fat and risk of ulcerative colitis and Crohn’s diseaseGut201463577678423828881
  • GhoshSDeCoffeDBrownKFish oil attenuates omega-6 polyunsaturated fatty acid-induced dysbiosis and infectious colitis but impairs LPS dephosphorylation activity causing sepsisPLoS One201382e5546823405155
  • GerasimidisKBertzMHanskeLDecline in presumptively protective gut bacterial species and metabolites are paradoxically associated with disease improvement in pediatric Crohn’s disease during enteral nutritionInflamm Bowel Dis201420586187124651582
  • SokolHSeksikPFuretJPLow counts of Faecalibacteriumprausnitzii in colitis microbiotaInflamm Bowel Dis20091581183118919235886
  • LewisJDAbreuMTDiet as a trigger or therapy for inflammatory bowel diseasesGastroenterology20171522398414.e627793606
  • BorrelliOCordischiLCirulliMPolymeric diet alone versus corticosteroids in the treatment of active pediatric Crohn’s disease: a randomized controlled open-label trialClinGastroenterolHepatol200646744753
  • TakagiSUtsunomiyaKKuriyamaSEffectiveness of an ‘half elemental diet’ as maintenance therapy for Crohn’s disease: a randomized-controlled trialAliment PharmacolTher200624913331340
  • GroverZMuirRLewindonPExclusive enteral nutrition induces early clinical, mucosal and transmural remission in paediatric Crohn’s diseaseJ Gastroenterol201449463864523636735
  • LeeDBaldassanoRNOtleyARComparative effectiveness of nutritional and biological therapy in North American Children with active Crohn’sdiseaseInflamm Bowel Dis20152181786179325970545
  • SuskindDLWahbehGCohenSAPatients perceive clinical benefit with the specific carbohydrate diet for inflammatory bowel diseaseDig Dis Sci201661113255326027638834
  • SuskindDLWahbehGGregoryNVendettuoliHChristieDNutritional therapy in pediatric Crohn disease: the specific carbohydrate dietJ PediatrGastroenterolNutr20145818791
  • CohenSAGoldBDOlivaSClinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn diseaseJ PediatrGastroenterolNutr2014594516521
  • ObihCWahbehGLeeDSpecific carbohydrate diet for pediatric inflammatory bowel disease in clinical practice within an academic IBD centerNutrition201632441842526655069
  • KakodkarSFarooquiAJMikolaitisSLMutluEAThe specific carbohydrate diet for inflammatory bowel disease: a case seriesJ AcadNutr Diet2015115812261232
  • SuskindDLCohenSABrittnacherMJClinical and fecal microbial changes with diet therapy in active inflammatory bowel diseaseJ ClinGastroenterol Epub20161227
  • Sigall-BonehRPfeffer-GikTSegalIZangenTBoazMLevineAPartial enteral nutrition with a Crohn’s disease exclusion diet is effective for induction of remission in children and young adults with Crohn’s diseaseInflamm Bowel Dis20142081353136024983973
  • RiordanAMHunterJOCowanRETreatment of active Crohn’s disease by exclusion diet: East Anglian multicentre controlled trialLancet19933428880113111347901473
  • RajendranNKumarDFood-specific IgG4-guided exclusion diets improve symptoms in Crohn’s disease: a pilot studyColorectal Dis20111391009101320626437
  • ChibaMAbeTTsudaHLifestyle-related disease in Crohn’s disease: relapse prevention by a semi-vegetarian dietWorld J Gastroenterol201016202484249520503448
  • GearryRBIrvingPMBarrettJSNathanDMShepherdSJGibsonPRReduction of dietary poorly absorbed short-chain carbohydrates (FODMAPs) improves abdominal symptoms in patients with inflammatory bowel disease-a pilot studyJ Crohns Colitis20093181421172242
  • PrinceACMyersCEJoyceTIrvingPLomerMWhelanKFermentable carbohydrate restriction (Low FODMAP Diet) in clinical practice improves functional gastrointestinal symptoms in patients with inflammatory bowel diseaseInflamm Bowel Dis20162251129113626914438
  • HalmosEPChristophersenCTBirdARShepherdSJMuirJGGibsonPRConsistent prebiotic effect on gut microbiotawith altered FODMAP intake in patients with Crohn’sdisease: a randomised, controlled cross-over trial of well-defined dietsClinTranslGastroenterol20167e164
  • ZimmerJLangeBFrickJSA vegan or vegetarian diet substantially alters the human colonic faecal microbiotaEur J ClinNutr20126615360
  • ChassaingBKorenOGoodrichJKDietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndromeNature20155197541929625731162
  • NickersonKPChaninRMcDonaldCDeregulation of intestinal anti-microbial defense by the dietary additive, maltodextrinGut Microbes201561788325738413
  • van NoodEVriezeANieuwdorpMDuodenal infusion of donor feces for recurrent Clostridium difficileN Engl J Med2013368540741523323867
  • IaniroGBibboSScaldaferriFGasbarriniACammarotaGFecal microbiota transplantation in inflammatory bowel disease: beyond the excitementMedicine (Baltimore)20149319e9725340496
  • KumpPKGrochenigHPLacknerSAlteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitisInflamm Bowel Dis201319102155216523899544
  • KundeSPhamABonczykSSafety, tolerability, and clinical response after fecal transplantation in children and young adults with ulcerative colitisJ PediatrGastroenterolNutr2013566597601
  • MoayyediPSuretteMGKimPTFecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trialGastroenterology20151491102109 e625857665
  • RossenNGFuentesSvan der SpekMJFindings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitisGastroenterology20151491110118.e425836986
  • CasellasFBorruelNPapoMAntiinflammatory effects of enterically coated amoxicillin-clavulanic acid in active ulcerative colitisInflamm Bowel Dis199841159552221
  • SelbyWPavliPCrottyBAntibiotics in Crohn’s Disease Study GroupTwo-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn’s diseaseGastroenterology200713272313231917570206
  • WangSLWangZRYangCQMeta-analysis of broad-spectrum antibiotic therapy in patients with active inflammatory bowel diseaseExpTher Med20124610511056
  • MackDRProbiotics in inflammatory bowel diseases and associated conditionsNutrients20113224526422254095
  • GionchettiPRizzelloFVenturiAOral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trialGastroenterology2000119230530910930365
  • MimuraTRizzelloFHelwigUOnce daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitisGut200453110811414684584
  • MatsuokaKKanaiTThe gut microbiota and inflammatory bowel diseaseSeminImmunopathol20153714755
  • GuarnerFPrebiotics in inflammatory bowel diseasesBr J Nutr200798Suppl 1S85S8917922967
  • ArribasBSuarez-PereiraEOrtiz MelletCDi-D-fructose dianhydride-enriched caramels: effect on colon microbiota, inflammation, and tissue damage in trinitrobenzenesulfonic acid-induced colitic ratsJ Agric Food Chem201058106476648420423151
  • NishimuraTAndohAHashimotoTKoboriATsujikawaTFujiyamaYCellobiose prevents the development of dextran sulfate sodium (DSS)-induced experimental colitisJ ClinBiochemNutr2010462105110
  • BenjaminJLHedinCRKoutsoumpasARandomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn’s diseaseGut201160792392921262918
  • KimSKimJHParkBOKwakYSPerspectives on the therapeutic potential of short-chain fatty acid receptorsBMB Rep201447317317824499669
  • AtarashiKTanoueTShimaTInduction of colonic regulatory T cells by indigenous Clostridium speciesScience2011331601533734121205640
  • SmithPMHowittMRPanikovNThe microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasisScience2013341614556957323828891
  • Huda-FaujanNAbdulamirASFatimahABThe impact of the level of the intestinal short chain fatty acids in inflammatory bowel disease patients versus healthy subjectsOpen Biochem J20104535820563285
  • KolevaPTValchevaRSSunXGänzleMGDielemanLAInulin and fructo-oligosaccharides have divergent effects on colitis and commensal microbiota in HLA-B27 transgenic ratsBr J Nutr201210891633164322243836
  • JooEYamaneSHamasakiAEnteral supplement enriched with glutamine, fiber, and oligosaccharide attenuates experimental colitis in miceNutrition201329354955523274091
  • BrothertonCSMartinCALongMDKappelmanMDSandlerRSAvoidance of fiber is associated with greater risk of Crohn’s disease flare in a 6-month periodClin Gastroenterol Hepatol20161481130113626748217