Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jan 1.
Published in final edited form as: Transl Res. 2016 Jun 14;179:38–48. doi: 10.1016/j.trsl.2016.06.002

The Gut Microbiota and Inflammatory Bowel Diseases

Jun Miyoshi 1, Eugene B Chang 1
PMCID: PMC5156589  NIHMSID: NIHMS795511  PMID: 27371886

Abstract

Inflammatory bowel diseases (IBD) are chronic diseases of unclear etiology that affect over 1 million individuals in the United States and over 2.5 million people in Europe(1). However, they are also expanding globally, affecting populations in Asia, South America, and the Middle East as they become more industrialized. These diseases are believed to arise from the convergence of genetic, environmental, and microbial factors that trigger aberrant immune and tissue responses, resulting in intestinal inflammation. Advances in cultivation-independent investigations, experimental models, and bioinformatics approaches have improved our understanding of the role of gut microbiota in IBD. However, determining and understanding the functional consequences of gut dysbiosis and altered host-microbiota interactions in IBD remain a challenge due to the limits of current experimental models and difficulty in establishing causal links in human-based investigations. Continued development of new methodologies and improvements in clinical study design are needed to better understand the interplay of genetic, microbial, and immunological factors in IBD. This knowledge can then be applied clinically to improve therapeutic strategies and outcomes for IBD.

Introduction

Inflammatory bowel diseases (IBD) are chronic disorders of unclear typify “Western” disorders, i.e. diseases that were previously uncommon, but now are emerging with alarming frequency over the past century as diet, environment, and social norms have changed in industrialized countries(1). IBD are heterogeneous diseases that likely arise from the convergence of genetic, microbial, and environmental factors. They fall into two main clinical phenotypes, ulcerative colitis (UC) and Crohn’s disease (CD), which were first described in the 19th century and 20th century, respectively(2). UC is characterized by mucosal inflammation in colon, beginning in the rectum and extending proximally in a continuous and circumferential fashion. In contrast, CD can affect any part of the GI tract and its lesions are focal and transmural, progressing to deep ulcerations that can result in development of fistulas, abscesses and strictures. Both types of IBD often involve chronic relapsing that can be medically managed, but may require surgical intervention in severe cases.

A role for gut microbes in IBD has been suspected since the early descriptions of potential infectious agents, but no single agent has been proven to cause IBD(2). Many IBD genetic risk variants identified by genome wide association studies, for example, are known or implicated to be involved in mediating host responses to gut microbiota. This has raised the possibility that members of the commensal gut microbiota play a role in the etiopathogenesis of IBD, particularly under conditions that might induce virulence properties. Furthermore, recent advances in experimental models, technology, and computational platforms have shed light on the structure and function of complex microbial communities and host-microbe interactions, creating a framework for understanding potential disease-causing mechanisms.

IBD is clearly associated with intestinal dysbiosis, i.e. the imbalance in the structures and/or functions of gut microbiota that disrupts host-microbe and immune homeostasis(37). However, many questions remain unanswered. Is intestinal dysbiosis a cause or consequence of IBD? Are these diseases caused by an aberrant host response to normal gut microbiota and/or to they arise from potentially disease-causing organisms (pathobionts) which are acquired or are part of the commensal microbiota? Are IBD-associated changes in gut microbiota constant or continuously changing throughout the course of disease? How do the environmental, dietary, medications, host, and other factors affect the gut microbiota and IBD development? Are interventions that attempt to correct gut dysbosis in IBD effective? If not, why? These questions will be addressed by this review, but for many, the answers await further study and inquiry.

Host-genetic factors affecting the development of IBD

Many studies of the genetic basis of IBD, including genome-wide association studies (GWAS), have revealed key insights and improved our understanding of IBD pathogenesis. From the over 200 single-nucleotide polymorphisms (SNPs) associated with increased risk for IBD(8), several pathways and genes have been identified that may either affect the way gut microbes are assembled or how the host is impacted by alterations in the gut microbiome. The latter include functions such as intestinal barrier function, wound healing, autophagy, apoptosis, immune activation, and stress responses(913).

Nucleotide-binding oligomerization domain-containing protein 2 (NOD2, also known as CARD15) was the first gene of the IBD susceptibility genes to be identified that encodes an intracellular receptor for the bacterial peptidoglycan muramyl dipeptide (MDP)(14, 15). The discovery of NOD2 and other IBD susceptibility genes (e.g. ATG16L1, IRGM, CARD9, and IL23R) underscores the importance of innate and adaptive host immune responses to gut microbiota. Furthermore, mutations that potentially alter the function or expression of these genes can affect the gut microbiome in ways that disrupt host-microbe interactions to set the stage for the onset of disease(16, 17). Nod2-deficient mice, for instance, exhibit significantly increased representation by Bacteroides, Firmicutes, and Bacilli in their terminal ileum, and decreased ability to clear the pathogenic bacteria, Helicobacter hepaticus(18). In addition, CD and UC patients with NOD2 risk alleles (Leu1007fs, R702W, or G908R) often have decreased Clostridium Groups XIVa and IV and increased Actinobacteria and Proteobacteria(19). Knights et al. analyzed intestinal biopsy samples from IBD patients and reported the association between NOD2 risk allele count and increased relative abundance of Enterobacteriaceae. They also suggested that other genetic risk loci (e.g. TNFSF15 and IL12B) appeared to affect the taxonomic structure of the microbiome(20). A study in healthy individuals showed that the IRGM mutation (rs11747270 SNP, a CD-risk locus) is associated with increased presence of Prevotella, but negatively correlated with the presence of Bacteroides(21). In turn, gut microbiota appear to affect Nod2 gene expression, because germ free mice have less Nod2 expression in the terminal ileum an effect can be restored by conventionalizing the mice with gut microbiota(18).

Not all forms of IBD are equally impacted by changes in gut microbiota. Cases of very early-onset, aggressive IBD, where disease develops soon after birth and before maturation of a gut microbiota, are likely caused by dominant genetic (monogenic) defects(22). For example, IL-10 is known as one of major anti-inflammatory cytokines and the deficiency of IL-10 and IL-10 receptor genes are associated with very early-onset IBD(2325). Similarly, genetic variants of components of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, required for generation of reactive oxygen species and phagosome function, have been reported in early onset cases(26). Variants in tetratricopeptide repeat domain 7 (TTC7A) were also found in patients with severe very early-onset IBD, possibly causing impaired epithelial barrier function.(27)

In the majority of cases, however, genetic susceptibility is insufficient to cause IBD. Even among monozygotic (identical) twins, where the concordance for CD between is between 35% and 58% and for UC between 16% to 18.5%(2830), the penetrance is not 100%. Thus, non-genetic factors, such as environment and gut microbiota, are important contributors to the pathogenesis of IBD. The T300A variant in ATG16L1 is associated with ileal CD and with increased endoplasmic reticulum stress in Paneth cell IL-1β production in response to MDP. However, this gene variant is common in the general population and most subjects carrying it do not develop IBD(31). In addition, mice hypomorphic for the Atg16l1 mutation that have demonstrated abnormalities of Paneth cells are prone to the development of dextran sodium sulfate (DSS)-induced intestinal inflammation only when concomitantly infected with the replicative murine norovirus(32).

Environmental factors affecting gut microbiota

In the past century, there has been a remarkable increase in incidence and prevalence of IBD on a global scale, especially in industrialized countries where dramatic changes in hygienic conditions, diet, environment, and lifestyle have taken place(33). Over this short evolutionary time scale, the increase in IBD cannot be attributed to genetic drift, and are more likely caused by environmental and societal factors. In this regard, the ever increasing and promiscuous use of antibiotics may have a contributory role by disturbing the gut microbiota. In genetically prone individuals, alterations in critical host-microbe interactions could promote the eventual development of IBD. A cohort study among Danish children from 1995 to 2003 showed a greater relative risk of IBD in antibiotic users compared with non-users(34). A study among children born in 1994–2008 in Finland revealed the risk of pediatric Crohn's disease increased with the number of antibiotic purchases from birth(35). A retrospective cohort study in the United Kingdom showed that childhood exposure to anaerobe-targeting antibiotics is associated with IBD development(36). A recent meta-analysis also concluded that the exposure to antibiotics is associated with increased risk for CD but not UC, especially amongst children(37).

Rapid changes in environmental factors are also potential contributors to collective shifts of intestinal microbiomes of human populations. A recent population-based cohort study in Canada revealed that there was increased risk of IBD with younger age at time of immigration in individuals from regions with lower prevalence of IBD(38). A cohort study of Danes born in 1973–2008 showed that Cesarean sections moderately increased rates of IBD with onset in childhood(39). Blaser and colleagues showed that mice exposed to sub-therapeutic, low-dose antibiotic therapy after weaning exhibited increased adiposity, changes in hepatic lipid metabolism, and altered immune balance that were associated with significant changes in gut microbial membership and function. This antibiotic treatment increased levels of colonic short-chain fatty acids (SCFAs), products of microbial fermentation that have many bioactive functions affecting the host. They also showed that the transient exposure to antibiotics early in life was sufficient to induce long-lasting effects on body composition. Their observations suggested that the early life exposure to antibiotics impact gut microbiota in ways that can have long-term effects on metabolism and development(40, 41). Deshmukh et al. showed that the antibiotic treatment for pregnant dams cause the alteration of the gut microbiota in their neonates and it was associated with decreased numbers of circulating and bone marrow neutrophils and granulocyte/macrophage-restricted progenitor cells in the bone marrow and increased susceptibility to sepsis of the neonates(42). Finally, Maurice et al. reported that short-term exposure to other medications, such as digoxin and phenacetin, affects the physiology and assemblage of the gut microbiota, which can ultimately change the host ability to metabolize medications(43).

Changes in diet and dietary habits are among the largest shifts that occur with increased industrialization and westernization. In addition to increased daily caloric consumption, diets are higher in fat and refined sugars, and lower in complex carbohydrates and fiber. A controlled-feeding study showed that the composition of gut microbiota is dramatically changed by a high-fat/low-fiber compared to a low-fat/high-fiber diet(44). African children on a high-fiber, plant-based diet exhibit a vastly different gut microbial community than their European counterparts whose dietary intake consists of a diets rich in sugar, fat, and protein(45). A study in wild type and IL-10-deficient mice showed that different types of dietary fat had different effects on gut microbiota. A diet rich in saturated milk fat, for instance, caused shifts in the gut microbiota that favored blooms of Proteobacteria, particularly Bilophila wadsworthia, compared to isocaloric, isonitrogenous diets high in polyunsaturated (predominantly n-6) or lard fat. Diet-induced blooms of B. wadworthia increased the incidence and severity of spontaneous colitis in genetically susceptibile IL10-deficient mice(46). In addition, David et al. demonstrated in human subjects that an animal-based diet rapidly changed the composition of the gut microbiota and lead to increased levels of bile-tolerant organisms (Alistipes, Bilophila and Bacteroides) including B. wadworthia and decreases in Firmicutesthat metabolize plant polysaccharides, such as Roseburia, Eubacterium rectale, and Ruminococcus bromii.(47)

Improvements in hygienic practices and environment conditions over the past century have also been implicated in contributing to the increase incidence and prevalence of IBD. The “hygiene hypothesis” propones that reduction of important microbial cues necessary to educate the host immune system results in decreased gut microbial diversity and altered host-microbe interactions that can promote disease in susceptible hosts(48). A study in India showed that CD was positively associated with urban living and safe drinking water and negatively with the presence of cattle in the home compound(49). Chung et al. colonized germ free mice with mouse or human microbiota and demonstrated that human microbiota were not sufficient for the proper development of the intestinal immune system due to host-microbe species mismatch. As a consequence, mice colonized with human microbiota were more susceptible to Salmonella infection than the mice with mouse microbiota(50). In addition, it has been shown that the maternal microbiota during gestation plays a role in the development of innate immunity in mouse pups, making them less prone to microbial infection and immune activation by microbial signals(51).

Intestinal dysbiosis in IBD patients

Many changes in both structure (composition) and function of gut microbiota are associated with active IBD. Seksik et al. demonstrated that the biodiversity of the microbiota was still high but that Enterobacteria were increased in fecal samples of CD patients(3). Gophna et al. found that increased Proteobacteria and Bacteroidetes and decreased Clostridia in biopsy samples from CD patients(6). Frank et al. reported that resected GI tract tissue samples from CD and UC patients showed depletion of members of Bacteroidetes and Lachnospiraceae (group IV and XIVa Clostridia)(7). Ott et al. and Manicanh et al. found decreased diversity of the gut microbiome in CD and UC using biopsy samples and fecal samples(4, 5). Sokol et al. demonstrated a lower proportion of F. prausnitzii in resected ileal specimens were associated with endoscopic recurrence after surgery in CD patients. They also showed that the anti-inflammatory potential of this species and the decrease in fecal samples from active IBD patients(52, 53). Machiels et al. reported a reduced abundance of butyrate-producing Roseburia hominis and Faecalibacterium prausnitzii in fecal samples of UC patients(54). Gevers et al. analyzed the mucosa-associated microbiota in ileal and rectal biopsy samples from newly-diagnosed pediatric CD patients and found increased abundance in Enterobacteriaceae, Pasteurellacaea, Veillonellaceae, and Fusobacteriaceae, and decreased abundance in Erysipelotrichales, Bacteroidales, and Clostridias(55). In assessing all these findings, the sample type and sites from where they were taken (e.g. fecal or surgical sample, remission or inflamed condition, or which portion of GI tract) have to be considered. The composition and abundance of both luminal and mucosa-associated microbiota vary along the GI tract(56, 57) and can be significantly impacted by intestinal inflammation in a number of different ways. Mucosal-associated microbiota can differ significantly from their luminal counterparts, the latter most likely being the major contributors to fecal microbiota. Thus, the fecal sampling commonly used by many IBD studies may not accurately reflect regional changes in gut microbes, particularly those that are taking place at the mucosal surface at sites of inflammation. Two other challenges to the understanding the role of gut microbes in IBD are related to the bioinformatics analysis of microbial community structure and function. Most methods for analysis do not provide sufficient resolution to go beyond the genus level(58) and are unable to determine if IBD is caused or contributed to by IBD-related pathobionts or pathogens. However, several new computational platforms have recently been developed that now allow investigators to examine operational taxonomic units at an extremely precise level by utilizing very subtle variations among 16S ribosomal RNA gene sequences(59, 60). For metagenomic and metatranscriptomic analysis to assess functional profiles of microbial communities, an advanced analysis and visualization platform for ‘omics data (Anvi’o) now provides an assembly-based metagenomic workflow that integrates, analyzes, and displays sequencing datasets of diverse origins to address a wide variety of questions(61).

Other important members of the gut microbiota that may be relevant to IBD should be mentioned, although they are poorly understood and investigated. Bacteriophage are viruses that infect and replicate within bacteria. They are extremely diverse and abundant, and appear to be as prevalent in the gut as bacteria(62). Temperate phages are bacteriophages which can be either lytic or lysogenic, the former representing virulent phage, whereas the latter integrate into the bacterial genome to be dormant until induction, capable of altering bacterial function and fitness. Bacteriophage also represent an important form of horizontal gene transfer within bacterial populations, the relevance of which in IBD is undetermined. Lepage et al. analyzed virus-like particles (VLPs) in colonic mucosal biopsy samples from CD patients and healthy individuals and detected significantly more bacteriophage in CD patients than in healthy controls. Among samples from CD patients, ulcerated mucosa is less colonized by bacteriophages than non-ulcerated mucosa. They also reported that each individual seemed to harbor one dominant phage family(63). Another study of viral communities in the gut showed a lower diversity of the virome but larger variability among fecal samples from CD patients compared with healthy controls(64). Currently, incomplete inventories of gut phage, technical limitations, and the lack of experimental models prevent us from achieving a more complete annotation of the healthy and IBD virome and in understanding the functional insights of these findings.

Fungi and Archaea are two other groups of the gut microbiota that are poorly understood. Intestinal fungal communities have been shown to interact with the host immune system through the innate immune receptor Dectin-1(65). Dectin-1 recognizes β-1,3-glucans in the fungal cell wall and activates intracellular signals through CARD9 leading to inflammatory cytokine production and induction of Th17 responses. Interestingly, variants in CARD9 are strongly associated with Crohn’s disease and ulcerative colitis. In addition, a SNP in the Dectin-1 gene appears to be associated with medically-refractory UC(65). Ott et al. reported that fungal diversity increased in colonic biopsy samples from CD patients compared with healthy controls(66). Li et al. analyzed ileal mucosal specimens and fecal samples from a small patient cohort and showed that fungal diversity, particularly among Candida albicans, Aspergillus clavatus, and C. neoformans. increased in CD patients. They also showed differences in the fungal composition in mucosal samples with and without inflammation(67). On the other hand, Chehoud et al. found a lower diversity of fungal microbiota, although two lineages annotated as Candida appeared significantly greater in fecal samples from IBD patients(68). Lewis et al. examined pediatric subjects and showed that five fungal taxa detected in samples (Saccharomyces cerevisiae, Clavispora lusitaniae, Cyberlindnera jadinii, Candida albicans and Kluyveromyces marxianus) were positively associated with CD(69). Interestingly, anti-Saccharomyces cerevisiae antibodies (ASCA), which are believed to be a response primarily to C. albicans, have been used as a serological marker of ileal CD. C. albicans was also found to be more abundant in stool samples from CD patients than from healthy controls(70, 71). Even fewer studies have investigated Archaea in IBD. Scanlan et al. found that methanogens are less abundant in fecal samples from IBD patients, possibly impacting methane production and metabolism of the gut microbiota(72). Another study has shown that Archaea are less abundant in children compared to adults and there was no significant differences between pediatric IBD patients and healthy children(68). Similarly, a study of pediatric subjects for the genome of Methanobrevibacter smithii, which is the primarily representative of Archaea in the gut, demonstrated that most of children had little or no detectable colonization and there was no significant difference between CD patients and healthy controls(69).

Microbes, mechanisms, and mediators of intestinal dysbiosis that contribute to the pathogenesis of IBD

Intestinal dysbiosis can contribute to the pathogenesis of IBD either by loss of health-promoting or gain of disease-promoting microbes. Mazmanian et al. reported that the human symbiont Bacteroides fragilis protect mice from Helicobacter hepaticus-induced inflammation through the action of polysaccharide A (PSA). PSA suppressed the production of pro-inflammatory cytokine IL-17 and induced IL-10-producing CD4+ T cells(73). Fujiya et al. demonstrated that the quorum-sensing pentapeptide, competence and sporulation factor, of Bacillus subtilis is selectively taken up by the membrane organic cation transporter-2 of intestinal epithelial cells, resulting in activation of key survival pathways such as heat shock proteins, Akt, and MAPK(74). Several studies also show that soluble factors produced by probiotics and gut microbes have anti-inflammatory and cytoprotecitve effects(75, 76).

Short chain fatty acids (SCFAs), which are prodigiously produced by intestinal bacterial fermentation of dietary fiber, appear to be important mediators in health. Their levels are often significantly decreased in IBD which may be a key factor that compromises intestinal and immune homeostasis(54, 77). Maslowski et al. showed that the binding of G-protein-coupled receptor 43 (GPR43, also known as FFAR2) by SCFAs is necessary for the resolution of inflammatory responses(78). Atarashi et al. reported that SCFA-producing bacterial strains in Clostridia clusters IV, XIVa, XVII from a healthy human fecal sample induced colonic regulatory T (Treg)cell differentiation, expansion, and function(79, 80). Furthermore, Smith et al. showed that SCFAs regulate the size and function of the colonic Treg pool in a GPR43-dependent manner(81). Thus, inadequate development and function of Treg cells, due to low fiber diet- or dysbiosis-induced decreases in SCFA production can contribute to the severity and chronicity of IBD.

In contrast, the emergence of disease-promoting bacteria (pathobionts and pathogens) that promote immune activation and/or produce factors that negatively affect the host can also potentially contribute the pathogenesis of IBD. For example, increased sulfur-reducing, bile-tolerant bacteria and production of hydrogen sulfide (H2S) have been reported in UC patients(82, 83). Many of these types of bacteria have strong anti-oxidant capabilities, which provides a selective advantage to survive in the harsh milieu of intestinal inflammation. H2S has also be shown to be genotoxic, modulating the expression of genes involved in cell-cycle progression and triggering inflammatory and DNA repair responses(84). H2S can also impair the oxidation of butyrate(85). The expression of thiosulfate sulfur transferase, one of two enzymes responsible for detoxification of H2S, has been found to be significantly decreased in active UC, but restored to normal after successful treatment(86). Several reports have implicated Enterobacteriaceae in IBD, largely due to increased observed surges in abundance(3, 6, 87). Two Enterobacteriaceae species, K. pneumoniae and P. mirabilis, isolated from TRUC mice (mice deficient in innate and adaptive immunity that develop spontaneous UC-like inflammation) are able to cause colitis in wild type mice in concert with the endogenous microbiota(88, 89). Winter et al. showed that host-asociated nitrate, derived as a by-product of the inflammatory response, provides increased fitness of Escherichia coli to outcompete other bacteria that require fermentation substrates(90). A recent study showed that immunoglobulin A (IgA)-coated intestinal microbes from IBD patients promoted susceptibility to DSS colitis in gnotobiotic mice(91). Another study of undernutrition and enteropathy reported that gut bacteria, particularly Enterobacteriaceae, that promote host IgA responses may interact with other consortium members to produce enteropathy(92). On the other hand, Moon et al. showed that phenotype variations in a same mouse strain at different institutions may be a result of gut microbiota that can degrade the IgA secretory component and IgA itself, causing decreased levels of luminal and fecal IgA(93). They also demonstrated these IgA-low mice exhibited greater susceptibility to DSS-induced colitis than their IgA-high counterparts. Taken together, these findings highlight the complex interplay between gut microbiota and luminal IgA responses in the pathogenesis of intestinal inflammation.

Investigations of bacterial transcriptomes have provided additional insights into microbial virulence and fitness under conditions of inflammation-induced stress. E. coli NC101 from IL-10−/− mice with colitis showed upregulation of the stress response operon for small heat shock proteins IbpA and IbpB that confer protection from oxidative stress. Increases in IbpA and Ibp was also implicated in reducing host inflammatory responses(94). Other studies of adherent-invasive E. coli (AIEC) showed that specific FimH mutations were associated with CD and UC patients(95, 96). FimH facilitates AIEC binding to the mucosa and increased abundance of AIEC has been reported in IBD. Collectively, studies to date have shown that the inflammatory environment induces gene expression and function of certain bacteria in ways that allow them to survive, adapt, and contribute to the pathogenesis of IBD.

Several attempts have been made to characterize and understand the functional properties of gut microbial communities in IBD. Morgan et al. assessed microbial functions based on reference genomes of well represented taxa of fecal samples and intestinal biopsies from IBD patients and healthy subjects, validating some of their findings with shotgun metagenomics(97). They found that 12% of the analyzed functional pathways and 2% of genera changed in IBD patients, suggesting that functional inferences might be more informative than changes in microbial composition. Increases in glycolysis and carbohydrate transport/metabolism and decreases in lipid metabolism and catabolism modules were found, possibly indicating alterations of energy metabolism in microbial communities. In addition, increased representation of genes involved in secretion systems associated with cell wall degradation and exotoxins as well as microbial adherence/invasion were also observed in ileal CD. Greenblum et al integrated the metagenomic data with an in silico systems analysis of metabolic networks(98). They concluded that much of the microbial enzyme-level variation associated with IBD seemed to relate to changes in how the gut microbiome interacts with the host gut environment rather than alterations in core metabolic processes. In the analysis of newly-diagnosed, treatment-naive pediatric CD patients, Gevers et al. employed the PICRUSt algorithm to predict the microbial function with mucosal biopsy samples and also performed shotgun metagenomics sequencing on a subset of fecal samples(55). PICRUSt showed a loss in basic biosynthesis associated with reductions in Bacteroides and Clostridia and a switch towards a pathobiont-like auxotrophy associated with increases in aerobic or aerotolerant taxa, such as Proteobacteria and Pasteurellaceae. From their shotgun metagenomics analysis, they concluded that the bacterial species increased in CD contributed to the glycerophospholipid and lipopolysaccharide metabolism pathways thought to cause inflammation and that increased phosphonoacetate hydrolase expression would enhance the ability to utilize novel carbon and phosphate sources.

Challenges to microbiome studies in IBD

Over the past decade, major advances have been made in understanding the role of the intestinal microbiota in IBD pathogenesis. However, it still remains unclear whether IBD-associated dysbiosis is causative, contributory, or consequential to the disease. In all likelihood, all three possibilities occur. The inability to identify with certainty causative microbial organisms is due to limitations in technology, bioinformatics, and clinical study design. Most studies of IBD dysbiosis, for example, have been performed after the onset of disease and interpreted without consideration of clinical metadata other than the clinical phenotype (CD or UC) and location of disease. Most microbial data have been acquired from stool samples, which may not reflect the more disease-relevant changes in regional and mucosa-associated microbiota. The presence of mucosal immune activation and inflammation also exert selective pressures, favoring microorganisms that have the ability to survive and flourish under these harsh conditions and impairing the fitness of most commensal microbes. Thus, the dysbiosis described by most cross-sectional studies is mostly a consequence of the disease process. That said, many of the members of the IBD-associated microbiota are pro-inflammatory which helps sustain the conditions favorable to them, and, at the same time, promote the chronicity and severity of IBD. To identify causative microbes, i.e. those responsible for the initial development of IBD, is difficult, because studies ideally should be done before the onset of disease, which, in most cases, is not possible because we are unable to determine who will develop IBD. The analysis of IBD-associated dysbiotic microbial communities is also limited by the commonly used computational platforms for 16S rRNA gene and metagenomic/metatranscriptome analysis, the former unable to go beyond the genus level and the latter by incomplete inventories of microbial genome/genes to provide insight into community function. However, higher resolution bioinformatics tools are now becoming available that can provide strain- and genome-level information which ultimately may reveal key insights into the pathogenesis of IBD (Minimum Entropy Decomposition and Anvi’o) (5961).

Cross-sectional studies of gut microbiota in IBD subjects have provided a plethora of descriptive information that often is difficult to interpret, mostly because of large inherent variations in individual gut microbiota and the heterogeneity of study subjects. Future studies should involve a longitudinal design that can be performed on fewer patients because each patient would serve as their own control. Moreover, clinical information collected at each point can be used to interpret corresponding changes in gut microbiota and host functions. These metadata also provide a basis for developing leads that can be followed up experimentally and to develop more informative measures to stratify IBD patients that can help physicians achieve more precise diagnosis, treatment, and better outcomes.

For all its limitations, research using animal models of IBD still represents our best opportunity (currently) to discover the mechanisms of pathogenesis. The use of gnotobiotic animal models, for example, provides the surrogate measure to assess the function of specific microbial communities or evaluate individual strains in the context of a controlled environment, genetics, diet, and disease. 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 gut microbiota(99) or IBD patient populations(100) will contribute important knowledge of bioactive molecules that could translate into therapeutic innovations for inflammatory disease.

The pitfalls, potential, and promise of microbiome-base interventions in IBD

If IBD is caused by the convergence of host genetics, environment, and gut microbes, there is a strong rationale for controlling the environment (e.g. smoking, diet, etc) and in restoring a healthy state to the gut microbiota, as we currently have no measures to correct genetic anomalies. Several microbiome-based interventions have been studied in IBD, including pre-, pro-, and postbiotics. With the exception of a few circumstances, few of these agents have shown promise or efficacy(101, 102). The lack of success of these agents is not surprising, given the rather simplistic view on which these approaches are based, i.e. restoring “good” microbes by either sheer mass or through changing conditions that mostly favor fermentative microbes that produce SCFAs. Many of these preparations contain microbial strains that may not be indigenous to the gut or were selected on limited or no evidentiary basis. The notion that these microbes can take hold, supercede the endogenous microbiota of IBD patients, and survive in a hostile inflammatory environment is tenuous. Moreover, it is unlikely that “one size fits all”, given the heterogeneity of IBD patients and their microbiota.

Recently, fecal microbiota transplantation (FMT) has attracted considerable attention as a treatment option for IBD. The rationale for FMT is based on the notion that the donor microbiota has been naturally selected under conditions that promote health. The transplant of this “microbial organ” to a host recipient would replace the functions of the diseased microbial organ, thus restoring intestinal and immune homeostasis. While highly effective for the treatment of Clostridium difficile colitis, its efficacy in IBD remains controversial(103). Interestingly, in studies of FMT for UC patients, Angelberger et al.(104) and Rossen et al.(105) reported that only responders showed successful colonization of donor-derived phylotypes at 12 weeks after FMT, which may be indicative of how difficult it is to “transplant” another person’s microbial organ into an IBD host. Many factors may therefore have to be taken into consideration to increase the success of FMT in IBD, such as the genetic background of donor and recipient, environmental factors, disease states, and the inherent resilience of the recipient’s microbiotia to change, which may be difficult to replace because these organisms have been already selected for their ability to survive and flourish in an inflammatory condition. Other factors should also be taken under consideration as well, such as long-term complications and unintended consequences of FMT. A recent study by Chehoud et al. looked at a single healthy adult donor and three pediatric ulcerative colitis recipient patients who received multiple FMT treatments over the course of 6–12 weeks(106). While the initial response to FMT appeared promising, all three subject eventually relapsed and had to be place on immunomodulators. However, the study also found the transfer of bacteriophage populations and their genetic elements from donor to recipients that persisted. The long-term clinical significance of these findings remains uncertain, but they underscore the importance of developing better tools, more studies at the clinical and experimental level, and longer-term follow-up. Without these pieces of information, we are ill-equipped to fully define the microbiota which in a practical sense impacts the success and failure of FMT.

Today’s therapeutic goal for IBD is to induce and maintain remission, however, the ultimate goals should focus on prevention and cure. With regard to the microbiome, we have the opportunity to gain more knowledge that can be leveraged to achieve these goals.

Acknowledgments

The authors received funding from the NIH grants P30 DK42086, R01 DK097268, T32 DK07074, the Kenneth Rainin Foundation, the Helmsley Charitable Trust, and from the Gastrointestinal Research Foundation of Chicago (to Eugene B. Chang). We thank Joeli Brinkman for her help in editing the manuscript.

Abbreviations

IBD

inflammatory bowel diseases

GI tract

gastrointestinal tract

UC

ulcerative colitis

CD

Crohn’s disease

GWAS

genome-wide association study

SNP

single-nucleotide polymorphism

NADPH

nicotinamide adenine dinucleotide phosphate

MDP

muramyl dipeptide

DSS

dextran sodium sulfate

SCFA

short-chain fatty acids

VLP

virus-like particle

ASCA

anti-Saccharomyces cerevisiae antibody

PSA

polysaccharide A

GPR43

G-protein-coupled receptor 43

Treg

regulatory T cell

IgA

immunoglobulin A

AIEC

adherent-invasive Escherichia coli

FMT

fecal microbiota transplantation

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures

The authors have received research grant from Tsumura & Co. to study natural products.

References

  • 1.Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015;12(12):720–727. doi: 10.1038/nrgastro.2015.150. [DOI] [PubMed] [Google Scholar]
  • 2.Kirsner JB. Historical aspects of inflammatory bowel disease. J Clin Gastroenterol. 1988;10(3):286–297. doi: 10.1097/00004836-198806000-00012. [DOI] [PubMed] [Google Scholar]
  • 3.Seksik P, Rigottier-Gois L, Gramet G, Sutren M, Pochart P, Marteau P, et al. Alterations of the dominant faecal bacterial groups in patients with Crohn's disease of the colon. Gut. 2003;52(2):237–242. doi: 10.1136/gut.52.2.237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ott SJ, Musfeldt M, Wenderoth DF, Hampe J, Brant O, Folsch UR, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53(5):685–693. doi: 10.1136/gut.2003.025403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Manichanh C, Rigottier-Gois L, Bonnaud E, Gloux K, Pelletier E, Frangeul L, et al. Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach. Gut. 2006;55(2):205–211. doi: 10.1136/gut.2005.073817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gophna U, Sommerfeld K, Gophna S, Doolittle WF, Veldhuyzen van Zanten SJ. Differences between tissue-associated intestinal microfloras of patients with Crohn's disease and ulcerative colitis. J Clin Microbiol. 2006;44(11):4136–4141. doi: 10.1128/JCM.01004-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104(34):13780–13785. doi: 10.1073/pnas.0706625104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979–986. doi: 10.1038/ng.3359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Liu TC, Stappenbeck TS. Genetics and Pathogenesis of Inflammatory Bowel Disease. Annu Rev Pathol. 2016 doi: 10.1146/annurev-pathol-012615-044152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Van Limbergen J, Radford-Smith G, Satsangi J. Advances in IBD genetics. Nat Rev Gastroenterol Hepatol. 2014;11(6):372–385. doi: 10.1038/nrgastro.2014.27. [DOI] [PubMed] [Google Scholar]
  • 11.Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–124. doi: 10.1038/nature11582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Anderson CA, Boucher G, Lees CW, Franke A, D'Amato M, Taylor KD, et al. Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet. 2011;43(3):246–252. doi: 10.1038/ng.764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Franke A, McGovern DP, Barrett JC, Wang K, Radford-Smith GL, Ahmad T, et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat Genet. 2010;42(12):1118–1125. doi: 10.1038/ng.717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001;411(6837):603–606. doi: 10.1038/35079114. [DOI] [PubMed] [Google Scholar]
  • 15.Hugot JP, Chamaillard M, Zouali H, Lesage S, Cezard JP, Belaiche J, et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease. Nature. 2001;411(6837):599–603. doi: 10.1038/35079107. [DOI] [PubMed] [Google Scholar]
  • 16.Cadwell K, Liu JY, Brown SL, Miyoshi H, Loh J, Lennerz JK, et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature. 2008;456(7219):259–263. doi: 10.1038/nature07416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kaser A, Blumberg RS. Autophagy, microbial sensing, endoplasmic reticulum stress, and epithelial function in inflammatory bowel disease. Gastroenterology. 2011;140(6):1738–1747. doi: 10.1053/j.gastro.2011.02.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci U S A. 2009;106(37):15813–15818. doi: 10.1073/pnas.0907722106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Frank DN, Robertson CE, Hamm CM, Kpadeh Z, Zhang T, Chen H, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm Bowel Dis. 2011;17(1):179–184. doi: 10.1002/ibd.21339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Knights D, Silverberg MS, Weersma RK, Gevers D, Dijkstra G, Huang H, et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014;6(12):107. doi: 10.1186/s13073-014-0107-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Quince C, Lundin EE, Andreasson AN, Greco D, Rafter J, Talley NJ, et al. The impact of Crohn's disease genes on healthy human gut microbiota: a pilot study. Gut. 2013;62(6):952–954. doi: 10.1136/gutjnl-2012-304214. [DOI] [PubMed] [Google Scholar]
  • 22.Uhlig HH, Schwerd T, Koletzko S, Shah N, Kammermeier J, Elkadri A, et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology. 2014;147(5):990–1007. e3. doi: 10.1053/j.gastro.2014.07.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schaffer AA, Noyan F, et al. Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 2009;361(21):2033–2045. doi: 10.1056/NEJMoa0907206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Shah N, Kammermeier J, Elawad M, Glocker EO. Interleukin-10 and interleukin-10-receptor defects in inflammatory bowel disease. Curr Allergy Asthma Rep. 2012;12(5):373–379. doi: 10.1007/s11882-012-0286-z. [DOI] [PubMed] [Google Scholar]
  • 25.Kotlarz D, Beier R, Murugan D, Diestelhorst J, Jensen O, Boztug K, et al. Loss of interleukin-10 signaling and infantile inflammatory bowel disease: implications for diagnosis and therapy. Gastroenterology. 2012;143(2):347–355. doi: 10.1053/j.gastro.2012.04.045. [DOI] [PubMed] [Google Scholar]
  • 26.Dhillon SS, Fattouh R, Elkadri A, Xu W, Murchie R, Walters T, et al. Variants in nicotinamide adenine dinucleotide phosphate oxidase complex components determine susceptibility to very early onset inflammatory bowel disease. Gastroenterology. 2014;147(3):680–689. e2. doi: 10.1053/j.gastro.2014.06.005. [DOI] [PubMed] [Google Scholar]
  • 27.Avitzur Y, Guo C, Mastropaolo LA, Bahrami E, Chen H, Zhao Z, et al. Mutations in tetratricopeptide repeat domain 7A result in a severe form of very early onset inflammatory bowel disease. Gastroenterology. 2014;146(4):1028–1039. doi: 10.1053/j.gastro.2014.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Orholm M, Binder V, Sorensen TI, Rasmussen LP, Kyvik KO. Concordance of inflammatory bowel disease among Danish twins. Results of a nationwide study. Scand J Gastroenterol. 2000;35(10):1075–1081. doi: 10.1080/003655200451207. [DOI] [PubMed] [Google Scholar]
  • 29.Halfvarson J, Bodin L, Tysk C, Lindberg E, Jarnerot G. Inflammatory bowel disease in a Swedish twin cohort: a long-term follow-up of concordance and clinical characteristics. Gastroenterology. 2003;124(7):1767–1773. doi: 10.1016/s0016-5085(03)00385-8. [DOI] [PubMed] [Google Scholar]
  • 30.Spehlmann ME, Begun AZ, Burghardt J, Lepage P, Raedler A, Schreiber S. Epidemiology of inflammatory bowel disease in a German twin cohort: results of a nationwide study. Inflamm Bowel Dis. 2008;14(7):968–976. doi: 10.1002/ibd.20380. [DOI] [PubMed] [Google Scholar]
  • 31.Prescott NJ, Fisher SA, Franke A, Hampe J, Onnie CM, Soars D, et al. A nonsynonymous SNP in ATG16L1 predisposes to ileal Crohn's disease and is independent of CARD15 and IBD5. Gastroenterology. 2007;132(5):1665–1671. doi: 10.1053/j.gastro.2007.03.034. [DOI] [PubMed] [Google Scholar]
  • 32.Cadwell K, Patel KK, Maloney NS, Liu TC, Ng AC, Storer CE, et al. Virus-plus-susceptibility gene interaction determines Crohn's disease gene Atg16L1 phenotypes in intestine. Cell. 2010;141(7):1135–1145. doi: 10.1016/j.cell.2010.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46–54. e42. doi: 10.1053/j.gastro.2011.10.001. quiz e30. [DOI] [PubMed] [Google Scholar]
  • 34.Hviid A, Svanstrom H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut. 2011;60(1):49–54. doi: 10.1136/gut.2010.219683. [DOI] [PubMed] [Google Scholar]
  • 35.Virta L, Auvinen A, Helenius H, Huovinen P, Kolho KL. Association of repeated exposure to antibiotics with the development of pediatric Crohn's disease--a nationwide, register-based finnish case-control study. Am J Epidemiol. 2012;175(8):775–784. doi: 10.1093/aje/kwr400. [DOI] [PubMed] [Google Scholar]
  • 36.Kronman MP, Zaoutis TE, Haynes K, Feng R, Coffin SE. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics. 2012;130(4):e794–e803. doi: 10.1542/peds.2011-3886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ungaro R, Bernstein CN, Gearry R, Hviid A, Kolho KL, Kronman MP, et al. Antibiotics associated with increased risk of new-onset Crohn's disease but not ulcerative colitis: a meta-analysis. Am J Gastroenterol. 2014;109(11):1728–1738. doi: 10.1038/ajg.2014.246. [DOI] [PubMed] [Google Scholar]
  • 38.Benchimol EI, Mack DR, Guttmann A, Nguyen GC, To T, Mojaverian N, et al. Inflammatory bowel disease in immigrants to Canada and their children: a population-based cohort study. Am J Gastroenterol. 2015;110(4):553–563. doi: 10.1038/ajg.2015.52. [DOI] [PubMed] [Google Scholar]
  • 39.Bager P, Simonsen J, Nielsen NM, Frisch M. Cesarean section and offspring's risk of inflammatory bowel disease: a national cohort study. Inflamm Bowel Dis. 2012;18(5):857–862. doi: 10.1002/ibd.21805. [DOI] [PubMed] [Google Scholar]
  • 40.Cho I, Yamanishi S, Cox L, Methe BA, Zavadil J, Li K, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488(7413):621–626. doi: 10.1038/nature11400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158(4):705–721. doi: 10.1016/j.cell.2014.05.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Deshmukh HS, Liu Y, Menkiti OR, Mei J, Dai N, O'Leary CE, et al. The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat Med. 2014;20(5):524–530. doi: 10.1038/nm.3542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Maurice CF, Haiser HJ, Turnbaugh PJ. Xenobiotics shape the physiology and gene expression of the active human gut microbiome. Cell. 2013;152(1–2):39–50. doi: 10.1016/j.cell.2012.10.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334(6052):105–108. doi: 10.1126/science.1208344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107(33):14691–14696. doi: 10.1073/pnas.1005963107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Devkota S, Wang Y, Musch MW, Leone V, Fehlner-Peach H, Nadimpalli A, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature. 2012;487(7405):104–108. doi: 10.1038/nature11225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505(7484):559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wills-Karp M, Santeliz J, Karp CL. The germless theory of allergic disease: revisiting the hygiene hypothesis. Nat Rev Immunol. 2001;1(1):69–75. doi: 10.1038/35095579. [DOI] [PubMed] [Google Scholar]
  • 49.Pugazhendhi S, Sahu MK, Subramanian V, Pulimood A, Ramakrishna BS. Environmental factors associated with Crohn's disease in India. Indian J Gastroenterol. 2011;30(6):264–269. doi: 10.1007/s12664-011-0145-1. [DOI] [PubMed] [Google Scholar]
  • 50.Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012;149(7):1578–1593. doi: 10.1016/j.cell.2012.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gomez de Aguero M, Ganal-Vonarburg SC, Fuhrer T, Rupp S, Uchimura Y, Li H, et al. The maternal microbiota drives early postnatal innate immune development. Science. 2016;351(6279):1296–1302. doi: 10.1126/science.aad2571. [DOI] [PubMed] [Google Scholar]
  • 52.Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, 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(43):16731–16736. doi: 10.1073/pnas.0804812105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009;15(8):1183–1189. doi: 10.1002/ibd.20903. [DOI] [PubMed] [Google Scholar]
  • 54.Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63(8):1275–1283. doi: 10.1136/gutjnl-2013-304833. [DOI] [PubMed] [Google Scholar]
  • 55.Gevers D, Kugathasan S, Denson LA, Vazquez-Baeza Y, Van Treuren W, Ren B, et al. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe. 2014;15(3):382–392. doi: 10.1016/j.chom.2014.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gu S, Chen D, Zhang JN, Lv X, Wang K, Duan LP, et al. Bacterial community mapping of the mouse gastrointestinal tract. PLoS One. 2013;8(10):e74957. doi: 10.1371/journal.pone.0074957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Peterson DA, Frank DN, Pace NR, Gordon JI. Metagenomic approaches for defining the pathogenesis of inflammatory bowel diseases. Cell Host Microbe. 2008;3(6):417–427. doi: 10.1016/j.chom.2008.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Schloss PD, Westcott SL. Assessing and improving methods used in operational taxonomic unit-based approaches for 16S rRNA gene sequence analysis. Appl Environ Microbiol. 2011;77(10):3219–3226. doi: 10.1128/AEM.02810-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Eren AM, Maignien L, Sul WJ, Murphy LG, Grim SL, Morrison HG, et al. Oligotyping: Differentiating between closely related microbial taxa using 16S rRNA gene data. Methods Ecol Evol. 2013;4(12) doi: 10.1111/2041-210X.12114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Eren AM, Morrison HG, Lescault PJ, Reveillaud J, Vineis JH, Sogin ML. Minimum entropy decomposition: unsupervised oligotyping for sensitive partitioning of high-throughput marker gene sequences. ISME J. 2015;9(4):968–979. doi: 10.1038/ismej.2014.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Eren AM, Esen OC, Quince C, Vineis JH, Morrison HG, Sogin ML, et al. Anvi'o: an advanced analysis and visualization platform for 'omics data. PeerJ. 2015;3:e1319. doi: 10.7717/peerj.1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wang W, Jovel J, Halloran B, Wine E, Patterson J, Ford G, et al. Metagenomic analysis of microbiome in colon tissue from subjects with inflammatory bowel diseases reveals interplay of viruses and bacteria. Inflamm Bowel Dis. 2015;21(6):1419–1427. doi: 10.1097/MIB.0000000000000344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lepage P, Colombet J, Marteau P, Sime-Ngando T, Dore J, Leclerc M. Dysbiosis in inflammatory bowel disease: a role for bacteriophages? Gut. 2008;57(3):424–425. doi: 10.1136/gut.2007.134668. [DOI] [PubMed] [Google Scholar]
  • 64.Perez-Brocal V, Garcia-Lopez R, Vazquez-Castellanos JF, Nos P, Beltran B, Latorre A, et al. Study of the viral and microbial communities associated with Crohn's disease: a metagenomic approach. Clin Transl Gastroenterol. 2013;4:e36. doi: 10.1038/ctg.2013.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Iliev ID, Funari VA, Taylor KD, Nguyen Q, Reyes CN, Strom SP, et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science. 2012;336(6086):1314–1317. doi: 10.1126/science.1221789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ott SJ, Kuhbacher T, Musfeldt M, Rosenstiel P, Hellmig S, Rehman A, et al. Fungi and inflammatory bowel diseases: Alterations of composition and diversity. Scand J Gastroenterol. 2008;43(7):831–841. doi: 10.1080/00365520801935434. [DOI] [PubMed] [Google Scholar]
  • 67.Li Q, Wang C, Tang C, He Q, Li N, Li J. Dysbiosis of gut fungal microbiota is associated with mucosal inflammation in Crohn's disease. J Clin Gastroenterol. 2014;48(6):513–523. doi: 10.1097/MCG.0000000000000035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chehoud C, Albenberg LG, Judge C, Hoffmann C, Grunberg S, Bittinger K, et al. Fungal Signature in the Gut Microbiota of Pediatric Patients With Inflammatory Bowel Disease. Inflamm Bowel Dis. 2015;21(8):1948–1956. doi: 10.1097/MIB.0000000000000454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lewis JD, Chen EZ, Baldassano RN, Otley AR, Griffiths AM, Lee D, et al. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn's Disease. Cell Host Microbe. 2015;18(4):489–500. doi: 10.1016/j.chom.2015.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Standaert-Vitse A, Jouault T, Vandewalle P, Mille C, Seddik M, Sendid B, et al. Candida albicans is an immunogen for anti-Saccharomyces cerevisiae antibody markers of Crohn's disease. Gastroenterology. 2006;130(6):1764–1775. doi: 10.1053/j.gastro.2006.02.009. [DOI] [PubMed] [Google Scholar]
  • 71.Standaert-Vitse A, Sendid B, Joossens M, Francois N, Vandewalle-El Khoury P, Branche J, et al. Candida albicans colonization and ASCA in familial Crohn's disease. The American journal of gastroenterology. 2009;104(7):1745–1753. doi: 10.1038/ajg.2009.225. [DOI] [PubMed] [Google Scholar]
  • 72.Scanlan PD, Shanahan F, Marchesi JR. Human methanogen diversity and incidence in healthy and diseased colonic groups using mcrA gene analysis. BMC Microbiol. 2008;8:79. doi: 10.1186/1471-2180-8-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453(7195):620–625. doi: 10.1038/nature07008. [DOI] [PubMed] [Google Scholar]
  • 74.Fujiya M, Musch MW, Nakagawa Y, Hu S, Alverdy J, Kohgo Y, et al. The Bacillus subtilis quorum-sensing molecule CSF contributes to intestinal homeostasis via OCTN2, a host cell membrane transporter. Cell Host Microbe. 2007;1(4):299–308. doi: 10.1016/j.chom.2007.05.004. [DOI] [PubMed] [Google Scholar]
  • 75.Petrof EO, Kojima K, Ropeleski MJ, Musch MW, Tao Y, De Simone C, et al. Probiotics inhibit nuclear factor-kappaB and induce heat shock proteins in colonic epithelial cells through proteasome inhibition. Gastroenterology. 2004;127(5):1474–1487. doi: 10.1053/j.gastro.2004.09.001. [DOI] [PubMed] [Google Scholar]
  • 76.Yan F, Cao H, Cover TL, Washington MK, Shi Y, Liu L, et al. Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J Clin Invest. 2011;121(6):2242–2253. doi: 10.1172/JCI44031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Vital M, Penton CR, Wang Q, Young VB, Antonopoulos DA, Sogin ML, et al. A gene-targeted approach to investigate the intestinal butyrate-producing bacterial community. Microbiome. 2013;1(1):8. doi: 10.1186/2049-2618-1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461(7268):1282–1286. doi: 10.1038/nature08530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331(6015):337–341. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500(7461):232–236. doi: 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
  • 81.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573. doi: 10.1126/science.1241165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Roediger WE, Moore J, Babidge W. Colonic sulfide in pathogenesis and treatment of ulcerative colitis. Dig Dis Sci. 1997;42(8):1571–1579. doi: 10.1023/a:1018851723920. [DOI] [PubMed] [Google Scholar]
  • 83.Levine J, Ellis CJ, Furne JK, Springfield J, Levitt MD. Fecal hydrogen sulfide production in ulcerative colitis. Am J Gastroenterol. 1998;93(1):83–87. doi: 10.1111/j.1572-0241.1998.083_c.x. [DOI] [PubMed] [Google Scholar]
  • 84.Attene-Ramos MS, Nava GM, Muellner MG, Wagner ED, Plewa MJ, Gaskins HR. DNA damage and toxicogenomic analyses of hydrogen sulfide in human intestinal epithelial FHs 74 Int cells. Environ Mol Mutagen. 2010;51(4):304–314. doi: 10.1002/em.20546. [DOI] [PubMed] [Google Scholar]
  • 85.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(1–2):117–124. doi: 10.1023/a:1006838231432. [DOI] [PubMed] [Google Scholar]
  • 86.De Preter V, Arijs I, Windey K, Vanhove W, Vermeire S, Schuit F, et al. Decreased mucosal sulfide detoxification is related to an impaired butyrate oxidation in ulcerative colitis. Inflamm Bowel Dis. 2012;18(12):2371–2380. doi: 10.1002/ibd.22949. [DOI] [PubMed] [Google Scholar]
  • 87.Giaffer MH, Holdsworth CD, Duerden BI. The assessment of faecal flora in patients with inflammatory bowel disease by a simplified bacteriological technique. J Med Microbiol. 1991;35(4):238–243. doi: 10.1099/00222615-35-4-238. [DOI] [PubMed] [Google Scholar]
  • 88.Garrett WS, Lord GM, Punit S, Lugo-Villarino G, Mazmanian SK, Ito S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell. 2007;131(1):33–45. doi: 10.1016/j.cell.2007.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Garrett WS, Gallini CA, Yatsunenko T, Michaud M, DuBois A, Delaney ML, et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe. 2010;8(3):292–300. doi: 10.1016/j.chom.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Winter SE, Winter MG, Xavier MN, Thiennimitr P, Poon V, Keestra AM, et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science. 2013;339(6120):708–711. doi: 10.1126/science.1232467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell. 2014;158(5):1000–1010. doi: 10.1016/j.cell.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kau AL, Planer JD, Liu J, Rao S, Yatsunenko T, Trehan I, et al. Functional characterization of IgA-targeted bacterial taxa from undernourished Malawian children that produce diet-dependent enteropathy. Sci Transl Med. 2015;7(276):276ra24. doi: 10.1126/scitranslmed.aaa4877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Moon C, Baldridge MT, Wallace MA, Burnham CA, Virgin HW, Stappenbeck TS. Vertically transmitted faecal IgA levels determine extra-chromosomal phenotypic variation. Nature. 2015;521(7550):90–93. doi: 10.1038/nature14139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Patwa LG, Fan TJ, Tchaptchet S, Liu Y, Lussier YA, Sartor RB, et al. Chronic intestinal inflammation induces stress-response genes in commensal Escherichia coli. Gastroenterology. 2011;141(5):1842–1851. e1–e10. doi: 10.1053/j.gastro.2011.06.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Iebba V, Conte MP, Lepanto MS, Di Nardo G, Santangelo F, Aloi M, et al. Microevolution in fimH gene of mucosa-associated Escherichia coli strains isolated from pediatric patients with inflammatory bowel disease. Infect Immun. 2012;80(4):1408–1417. doi: 10.1128/IAI.06181-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dreux N, Denizot J, Martinez-Medina M, Mellmann A, Billig M, Kisiela D, et al. Point mutations in FimH adhesin of Crohn's disease-associated adherent-invasive Escherichia coli enhance intestinal inflammatory response. PLoS Pathog. 2013;9(1):e1003141. doi: 10.1371/journal.ppat.1003141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Morgan XC, Tickle TL, Sokol H, Gevers D, Devaney KL, Ward DV, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13(9):R79. doi: 10.1186/gb-2012-13-9-r79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Greenblum S, Turnbaugh PJ, Borenstein E. Metagenomic systems biology of the human gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease. Proc Natl Acad Sci U S A. 2012;109(2):594–599. doi: 10.1073/pnas.1116053109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Smirnov KS, Maier TV, Walker A, Heinzmann SS, Forcisi S, Martinez I, et al. Challenges of metabolomics in human gut microbiota research. Int J Med Microbiol. 2016 doi: 10.1016/j.ijmm.2016.03.006. [DOI] [PubMed] [Google Scholar]
  • 100.Sands BE. Biomarkers of Inflammation in Inflammatory Bowel Disease. Gastroenterology. 2015;149(5):1275–1285. e2. doi: 10.1053/j.gastro.2015.07.003. [DOI] [PubMed] [Google Scholar]
  • 101.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(1):21–35. doi: 10.1097/01.MIB.0000437495.30052.be. [DOI] [PubMed] [Google Scholar]
  • 102.Bourreille A, Cadiot G, Le Dreau G, Laharie D, Beaugerie L, Dupas JL, et al. Saccharomyces boulardii does not prevent relapse of Crohn's disease. Clin Gastroenterol Hepatol. 2013;11(8):982–987. doi: 10.1016/j.cgh.2013.02.021. [DOI] [PubMed] [Google Scholar]
  • 103.Rossen NG, MacDonald JK, de Vries EM, D'Haens GR, de Vos WM, Zoetendal EG, et al. Fecal microbiota transplantation as novel therapy in gastroenterology: A systematic review. World J Gastroenterol. 2015;21(17):5359–5371. doi: 10.3748/wjg.v21.i17.5359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Angelberger S, Reinisch W, Makristathis A, Lichtenberger C, Dejaco C, Papay P, et al. Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation. Am J Gastroenterol. 2013;108(10):1620–1630. doi: 10.1038/ajg.2013.257. [DOI] [PubMed] [Google Scholar]
  • 105.Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, Duflou A, et al. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology. 2015;149(1):110–118. e4. doi: 10.1053/j.gastro.2015.03.045. [DOI] [PubMed] [Google Scholar]
  • 106.Chehoud C, Dryga A, Hwang Y, Nagy-Szakal D, Hollister EB, Luna RA, et al. Transfer of Viral Communities between Human Individuals during Fecal Microbiota Transplantation. MBio. 2016;7(2) doi: 10.1128/mBio.00322-16. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES