FUTURE DIRECTIONS

AND

METHODS

FOR

IBD RESEARCH

Functional Impacts of the Intestinal Microbiome in the Pathogenesis of Inflammatory Bowel Disease Jennifer Li, BSc,* James Butcher, BSc,* David Mack, MD,† and Alain Stintzi, PhD*

Abstract: The human intestinal microbiome plays a critical role in human health and disease, including the pathogenesis of inflammatory bowel disease (IBD). Numerous studies have identified altered bacterial diversity and abundance at varying taxonomic levels through biopsies and fecal samples of patients with IBD and diseased model animals. However, inconsistent observations regarding the microbial compositions of such patients have hindered the efforts in assessing the etiological role of specific bacterial species in the pathophysiology of IBD. These observations highlight the importance of minimizing the confounding factors associated with IBD and the need for a standardized methodology to analyze well-defined microbial sampling sources in early IBD diagnosis. Furthermore, establishing the linkage between microbiota compositions with their function within the host system can provide new insights on the pathogenesis of IBD. Such research has been greatly facilitated by technological advances that include functional metagenomics coupled with proteomic and metabolomic profiling. This review provides updates on the composition of the microbiome in IBD and emphasizes microbiota dysbiosis-involved mechanisms. We highlight functional roles of specific bacterial groups in the development and management of IBD. Functional analyses of the microbiome may be the key to understanding the role of microbiota in the development and chronicity of IBD and reveal new strategies for therapeutic intervention. (Inflamm Bowel Dis 2015;21:139–153) Key Words: Crohn’s disease, ulcerative colitis, intestinal microbiota, dysbiosis

T

he human intestinal microbiome consists of a complex ecosystem of microbial cells that play a critical role in human health. Under normal physiological conditions, this dynamic population acts in symbiosis with its host. Consequently, a rift within this mutualistic relationship is thought to manifest into various disorders, including inflammatory bowel disease (IBD).1–3 IBD is largely defined by 2 chronic inflammatory disorders, Crohn’s disease (CD) and ulcerative colitis (UC), which are characterized by chronic persistent inflammation of the intestinal mucosa lining the intestinal tract. The inflammation during CD extends deeper into the bowel wall than UC.4 The etiopathogenesis of IBD has been conceptualized as occurring in genetically susceptible individuals after a triggering event that Received for publication June 30, 2014; Accepted August 6, 2014. From the *Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada; and † Department of Pediatrics and CHEO Research Institute, University of Ottawa, Ottawa, ON, Canada. A. Stintzi and D. Mack are funded by the Government of Canada through Genome Canada and the Ontario Genomics Institute (OGI-067), CIHR grant number GPH-129340, CIHR grant number MOP-114872, and the Ontario Ministry of Economic Development and Innovation (REG1-4450). D. Mack and A. Stintzi also acknowledge funding from the 3C Foundation of Canada and the Crohn’s and Colitis Canada (CCC). The authors have no conflicts of interest to disclose. Reprints: David Mack, MD, Department of Pediatrics, CHEO, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada (e-mail: [email protected]); or Alain Stintzi, PhD, Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology; University of Ottawa, 451 Smyth Rd., Ottawa, ON, Canada, K1H 8M5 (e-mail: [email protected]). Copyright © 2014 Crohn’s & Colitis Foundation of America, Inc. DOI 10.1097/MIB.0000000000000215 Published online 21 September 2014.

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

generates an aberrant host immune response, and increasing evidence suggests that the intestinal microbiota is an important contributor to the disease.1,2,5–8 In recent years, numerous studies have been conducted to characterize the intestinal microbiota in patients with IBD. These studies have been particularly facilitated by methodological advances in high-throughput sequencing, metagenomics, proteomics, and metabolomic profiling.3,9–11 The integration of this knowledge into hypothesis-driven research using animal models can help define the etiological role or causal effect of the intestinal microbiota and/or microbial metabolic pathways in the development of intestinal inflammation. To date, a wealth of information has been generated regarding the altered microbiota composition, or dysbiosis, at different taxonomic levels that manifest in patients with IBD and animal models for IBD. Although no microbe has yet been identified as a sole causative agent for IBD, the specific changes observed within the microbiome during IBD have been proposed for use as biomarkers for IBD diagnosis, for predicting disease outcomes and for monitoring patient responses to therapeutic interventions.3,8,12–14 Lingering questions still remain on whether potential biomarker microbes are actively involved in the clinical features seen in IBD or whether they are simply bystanders that thrive in the inflamed milieu present during active IBD. Given the numerous confounding factors that affect microbial species distribution and abundance, it is not surprising that various studies have reported not only highly variable but also inconsistent observations on the compositional changes of microbiota that result from IBD. However, an emerging paradigm postulates that the metabolic functions fulfilled by the gut microbiota are more www.ibdjournal.org |

139

Li et al

relevant than the specific microbes that are fulfilling these roles and that identifying the dysregulation of these metabolic processes will yield greater insight into disease progression than simply identifying specific microbes that are present or absent. As a result, functional predictions from altered microbiota are required to address this knowledge gap. Researchers in the current -omics era have explored combinatorial approaches such as metagenomics, metatranscriptomics, metaproteomics, and metabolomics to assess the composition, gene expression, protein activity, and metabolic potential of the gut microbiota15–19; these interdisciplinary approaches should provide powerful tools to help define the structure, dynamics, and function of gut microbiota. This article is aimed at highlighting studies on altered microbiota with an emphasis on linking these changes in microbiota composition with functional changes that occur during IBD. These studies provide insights on the role of microbial dysbiosis and various microbial metabolic pathways that may contribute to the pathophysiology of IBD. Further unraveling the potential metabolic roles of the microbiota in IBD should lead to a better understanding of the root causes of IBD and may provide alternative therapies that target the diseased microbiota (as opposed to the diseased host) and enhance disease management.

GASTROINTESTINAL MICROBIOTA: COMPOSTION, FUNCTION, AND DISEASE The human gastrointestinal tract is colonized by a diverse population of more than 1000 bacterial species along with archaea, eukaryotes, fungi, and viruses.20–24 The combined microbial metagenome of .5 million genes is more than 100 times larger than its host, which is consistent with our ever-increasing understanding of the diverse functions that the microbiota contribute in normal human physiology and during disease pathology.23,25,26 Under normal physiological conditions, the gut microbes provide mutualistic benefits to their host with major contributions in metabolic, protective, and trophic functions.27–29 The diversity and composition of a healthy human microbiome for specific individuals is profoundly affected by diet, host genetics, early microbial exposures, and environment. Extensive sampling of the human microbiome through oral and stool samples has revealed diverse microbial communities at the family level, which are strongly influenced by the habitat in which they are found.30 However, studies indicate that the functional repertoire of the gut microbiome is relatively more stable than the compositional diversity,30 which highlights the importance of examining the human microbiome in diseased populations from a functionality point of view. There are pronounced temporal variations within a normal individual’s microbiome even in short timespans.31 However, there is general consensus that one’s microbiome does become more stable and more similar to the general population as one enters adulthood; this stability then declines in the elderly.32–35 Although the diversity of bacterial species varies greatly between individuals, recent studies have identified common core subsets within the microbiome that are relatively stable throughout

140

| www.ibdjournal.org

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

large populations and could even last for decades.12,36 A recently published article has narrowed the 1000 bacterial species of the gastrointestinal microbiome into a long-term core of approximately 100 species.36 Many species within this subset are considered early colonizers that likely originated from immediate family members.36 The Firmicutes and Bacteroidetes phyla (which begin to colonize in infants within the first week of birth) later represent the majority of the intestinal microbiota in adults with Bacteroides as the most abundant, yet most variable, genus.12,37–40 An in vivo study characterized the chronological establishment of healthy microbiota in mice.41 Like the human microbiome, the Firmicutes and Bacteroidetes phyla also dominate the mouse gut microbial ecosystem.41 Nevertheless, at the family level, different studies showed variable results, suggesting the importance of analyzing microbiota at the lower phylogenetic levels.41 It is interesting to note that lowabundance organisms provide specialized functions that are beneficial to the host12; thus, even small shifts in microbial composition may have a measurable impact on one’s health. The impacts of the intestinal microbiome have been studied in many aspects of health and disease. A study assessing the intestinal microbiome composition in the elderly clearly showed a correlation between microbial composition, diet, and health status (including clinical measures of frailty, comorbidity, nutritional status, markers of inflammation, and with metabolites) suggesting a role for dietdriven microbiota changes.32 Interestingly, a metagenomic-wide association study characterizing the intestinal microbiome of individuals with type 2 diabetes revealed 6000 microbial markers for classifying type 2 diabetes.42 The gut microbiota also interacts intensely with the host immune system through the intestinal mucosal surface.21 Curiously, it has also been established that the gut microbiota can interact with distant organs, such as the brain.43 Altogether, these reports strongly emphasize the critical role of gut microbiota in human health through host–microorganism functional interactions.

MICROBIOME IN IBD Intestinal dysbiosis (specifically a compositional imbalance of commensal bacteria) is the central characteristic of the gut microbiota in IBD and has been considerably documented.8,13,44–46 Copious studies have compared the distinct microbial compositions of healthy individuals and patients with IBD through 16S rRNA gene analysis.47–49 Such studies have generally used biopsies (for mucosa-associated microbiota) or fecal samples to determine specific bacterial alterations in patients with IBD.47–49 Moreover, researchers have uncovered remarkable dissimilarity in the intestinal microbiota of healthy individuals and patients with IBD from fecal samples.50–52 This inconsistency makes it challenging to assess the clinical significance of species-specific variability because the composition of the gut microbiota is also affected by age, diet, smoking, geography, and condition of health (Fig. 1).53,54 Nevertheless, there are certain bacterial species or phylotypes that are found more frequently in either healthy or diseased populations. A common feature of the intestinal microbiome in IBD is the reduced abundance of several types of bacteria, particularly

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Functional Impacts of the Intestinal Microbiome

FIGURE 1. The interplay between host microbiota dysbiosis, genetic status and environmental factors contribute to the pathogenesis of IBD.

members of the Firmicutes and Bacteroidetes phyla.13,45,46 In fact, species richness (a-diversity) as a whole is seen to decrease in patients with IBD.46 Losses of the core microbiome and temporal stability at differing functional levels have also been identified.46,50,55–60 Although some researchers have also reported similar microbiota alterations in biopsy and fecal samples from patients with IBD,61 others have documented differences related to sources of samples.13 A recent investigation studying the intestinal microbiome in the largest pediatric inception, treatment-naive CD cohort to date has convincingly revealed the importance of using ileal or rectal samples as compared with fecal sources, suggesting that the assessment of rectal mucosal-associated microbiome offers a unique potential for convenient and early diagnosis of CD.13 Despite the fact that species-driven dysbiosis in CD and UC can be different,51,62–65 overall species diversity is generally reduced in both CD and UC compared with healthy populations.51,66,67 However, these changes are often inconsistent and occasionally even conflicting among different studies as further assessed in the next section below. Hence, it is imperative that researchers consider the sampling time, sample source (e.g., ileum, rectum, feces, etc), and patient disease status when reporting results and drawing conclusions. Additionally, pharmacological treatment or other therapeutic interventions are also potential confounders in studying the composition of the microbiota in IBD and should also be taken into account.49 Considering the complexity of microbial compositions, an ecological analytical approach has been used to identify modules of interacting bacteria that represent quantitative reproducible features of microbial compositions.68 This study has revealed the presence of 5 microbial modules in endoscopic lavage samples from patients with IBD

and healthy controls; 2 of these modules showed a distinct metabolic functionality and were reciprocally associated with IBD.68 These 2 modules could act as potential indicators to better characterize IBD from both a compositional and functional perspective and thus aid in IBD disease management. It has long been known that the host’s genetic makeup also affects their vulnerability towards IBD.6 Although some genetic risk factors are specific to either CD or UC, there are a group of common genes between the 2 conditions as well. Although CD and UC are both associated with genomic regions that implicate products of genes involved in leukocyte trafficking, there is evidence for association patterns that are distinct between CD and UC.69 CD-predominant associations include nucleotide-binding oligomerization domain 2 (NOD2) and genes that regulate autophagy.69–72 Interestingly, Cuthbert et al72 determined that homozygotes and compound heterozygotes of the NOD2 mutation have .20-fold increased risk to CD. NOD2 recognizes bacterial peptidoglycan (muramyl dipeptide) and is required for the production of several intestinal antimicrobial peptides that provide protective immunity to the host.73 NOD2 mutations lead to a loss of function, which increases susceptibility to CD.74 NOD2 is also known to interact with autophagy-related protein 16-like 1 (ATG16L1) or immunity-related GTPase M (IRGM) for autophagic clearance of intracellular pathogens throughout the body.75 Thus, when NOD2 is mutated, ATG16L1 or IRGM alone are insufficient for the induction the autophagic response, which, in turn, can exacerbate microbial dysbiosis. Similarly, ATG16L12/2 mice also displayed an increased risk for CD.76 In contrast, extracellular matrix protein 1 is a susceptibility locus specific for UC.77 Extracellular matrix protein 1 is expressed in the intestinal tracts and activates NF-kB, an important immune response regulator.77 www.ibdjournal.org |

141

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Li et al

Genes involved in the unfolded protein response are also associated with IBD.75 The Il102/2, Il22/2, Il10r22/2Tgfbr22/2, and Tbx212/2Rag22/2 genotypes act as risk factors that promote the development of spontaneous colitis in mice.78 For instance, many patients diagnosed with early-onset IBD have mutations affecting the interleukin-10 (IL-10) receptor. Because IL-10 is an anti-inflammatory cytokine, disruption of this pathway yields a hyperinflammatory immune response within the intestine.79

COMPOSITIONAL CHANGES OF SPECIES-SPECIFIC BACTERIA IN IBD The intestinal microbiota signatures of patients with IBD often differ from healthy populations. The compositional shifts that differ between the groups include reduced microbial diversity and beneficial commensal species, as well as increased levels of potential pathobionts.13,51,67,80 Still, these compositional changes have evidently been complex. Table 1 provides a list of speciesspecific bacterial abundance changes (increased or decreased) in patients with CD and UC; although, as discussed above, there are conflicting reports for several bacteria. The dominant gut microbe, Bacteroides, can both positively and negatively affect the host.133 In certain circumstances, individual species of Bacteroides may be reduced, whereas the overall genus is enriched in patients with IBD.14,67 A study on the spatial organization and composition of the mucosal biota of patients with IBD indicated Bacteroides fragilis biofilm as a main feature of IBD.134 Recent studies continue to reveal significant abundance changes of several bacteria including Bdellovibrio bacteriovorus, Erysipelotrichales, Pasteurellaceae, and Veillonellaceae in patients with CD (Table 1).13,106 Specifically, reduced B. bacteriovorus occurs along the intestine in patients with IBD from the duodenum to rectum with the most dramatic decrease in the ileum.106 Patients with UC have higher bacterial densities associated with biopsies than patients with CD.62 Reduced Faecalibacterium and Bifidobacterium as well as increased Bacteroides were found in patients with active CD from multiple IBD research centers.86 The reduced levels of F. prausnitzii were found to be inversely related to the severity of UC.64 Furthermore, the reduction of F. prausnitzii indicated a higher risk for postoperative recurrence of ileal CD.135 In addition, characterizing the mucosa-associated microbiota of twin pairs discordant or concordant for CD revealed a reduced abundance of F. prausnitzii and elevated abundance of Escherichia coli in patients with CD.53 A consistent reduction of F. prausnitzii was observed in fecal samples of patients with UC during remission, particularly those with a history of frequent relapse, whereas maintenance of remission was associated with F. prausnitzii increase.102 However, despite these multiple reports of reduced levels of F. prausnitzii, Hansen et al51 detected increased F. prausnitzii in de novo pediatric CD patients at the onset of disease but not in UC. In another study, comparing twins with CD, patients with colonic CD and ileal CD exhibited an increase and decrease in F. prausnitzii, respectively.52 The differences between these studies can likely be attributed to differences between sampling

142

| www.ibdjournal.org

sources and times (e.g., colonic mucosa versus stool, de novo versus established IBD). Again, these comparisons highlight the importance of a standardized sampling methodology to minimize confounding factors and to facilitate comparability between studies. Patients with CD also showed a lower relative abundance of Bacteroides uniformis and higher relative abundances of Bacteroides ovatus and Bacteriodes vulgatus.66 Reduced bacterial populations within the Clostridium Cluster IV and XIVA groups were observed in fecal samples from CD and UC patients, respectively.98,136 However, a recent case-control study has suggested that reduced numbers of the Clostridium Cluster IV group occurs in both CD and UC.101 Another dominant species, Roseburia hominis, also appears to be reduced significantly in patients with UC and correlates inversely to UC disease activity and duration.64 The abundances of F. prausnitzii and R. hominis parallel with each other, suggesting that these microbes may potentially define UC dysbiosis and could thus be used to manage the microbial balance in patients with UC by the administration of probiotics or prebiotics.64 Interestingly, these bacterial populations do not directly correlate with the reduced levels of short-chain fatty acid (SCFA) metabolites that are known to regulate inflammation, which again suggests that compositional analysis alone may not be sufficient enough to characterize IBD.64 A study on the fecal microbiota of patients with UC identified 8 distinct clusters, 7 of which distinguish patients with UC from the single healthy individual cluster.137 Furthermore, these groups consist of various unclassified bacteria, which include Eubacterium, Fusobacterium, Ruminococcus, Gammaproteobacteria, Bacteroides, and Lactobacillus.137 Altogether, these bacteria seem to be able to distinguish patients with UC from those of healthy individuals and thus could be potential IBD biomarkers. Pathogenic bacteria such as Escherichia coli (particularly adherent invasive E. coli [AIEC]), Rhodococcus spp., Shigella spp., and Stenotrophomonas maltophilia are increasingly observed in patients with IBD.87,122,123,131,138 Analysis of fecal samples from pediatric IBD, revealed that E. coli was positively and Bacteroides, negatively, correlated with age in patients with CD, whereas patients with UC showed the inverse.65 Additionally, patients with IBD showed an increased urogenital biofilm of Gardnerella vaginalis, a key pathogen in bacterial vaginosis.139 It should be noted that patients with IBD can suffer from various infections associated with opportunistic pathogens; and thus, the increased levels of these pathogenic bacteria may be due to the secondary infections unrelated to IBD pathogenesis.140,141 The overall incidence of IBD is rapidly increasing worldwide.142 Although IBD is the most commonly diagnosed disease in young adults (20–30 yr old), it can manifest at any age. Alarmingly, epidemiological studies have shown that the incidence of IBD onset in children is increasing.73,143 These results are quite intriguing because the microbiome is known to change immensely over the course of aging.144 The microbiome of a newborn infant is much less complex than that of an adult, with birth-route, type of feeds, and genetics as the major influences on the initial microbial abundance and diversity.3 The microbiome stabilizes as one becomes a young adult, at which life-stage, the environment has a larger influence on

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Functional Impacts of the Intestinal Microbiome

TABLE 1. Abundance Changes of Gut Microbiota Identified in Biopsy and/or Fecal Samples of Patients with IBDa Bacterial Taxa Phylum/Class

Order, Family, Genus or Species

CD

UC

References

Actinobacteria

(Not specified) Bifidobacterium B. bifidum Mycobacterium avium paratuberculosis Bacteroides, B. ovatus, B. vulgatus Bacteroidale, Bacteriodes or B. uniformis Odoribacter Parabacteroides Prevotella Enterococcus Lactobacillus Leuconostocaceae Listeria Streptococcaceae Clostridiales Clostridium difficile Clostridium coccoides Clostridium leptum Eubacterium rectale Faecalibacterium prausnitzii Lachnospiraceae Phascolarctobacterium Roseburia or R. hominis Ruminococcaceae Ruminococcus gnavus/R. torques Erysipelotrichales Veillonellaceae Fusobacteriaceae Bdellovibrio bacteriovorus Desulfovibrio Campylobacter Helicobacter Enterobacteriaceae Escherichia coli Escherichia coli (AIEC) Pasteurellacaeae Shigella Stenotrophomonas maltophilia (Not specified) (Not specified) (Not specified) Akkermansia Methanosphaera stadtmanae

+ +/2 2 + + 2

+ +

Refs. 46,68,81,82 Ref. 49 Ref. 49 Refs. 83–85 Refs. 66,86–88 Refs. 13,66,67,81,82,89,90 Refs. 17 Refs. 91 Refs. 47,92,93 Refs. 89,94 Ref. 49 Ref. 17 Ref. 89 Ref. 61 Refs. 47,87 Refs. 82,89,95–97 Refs. 98–100 Refs. 98,100,101 Refs. 94,99 Refs. 45,46,49,52,53,64,81,86,89,94,100,102–104 Refs. 46,61,91 Ref. 17 Refs. 17,52,64,100,104 Refs. 46,89,91 Refs. 52,92,105 Ref. 13 Ref. 13 Refs. 13,68,91 Ref. 106 Refs. 107–110 Refs. 80,97,111–114 Refs. 114–119 Refs. 13,47,52,61,88,114,120,121 Refs. 49,53,87,89,94,121 Refs. 92,114,122–130 Ref. 13 Refs. 87,89 Ref. 131 Refs. 46,47,68,81,82,94,103,114 Ref. 68 Ref. 68 Refs. 14,90,105 Ref. 132

Bacteroidetes

Firmicutes/Bacilli

Firmicutes/Clostridia

Firmicutes/Erysiplotrichi Firmicutes/Negativicutes Fusobacteria Proteobacteria/Deltaproteobacteria Proteobacteria/Epsiloproteobacteria Proteobacteria/Gammaproteobacteria

Proteobacteria Tenericutes Verrucomicrobia Euryarchaeota (Archaea)

2 + + +

2 2 +

2 + 2 +/2 +/¼ 2 2 +/2 2 2 2 + 2 + + 2 +/¼ +/¼ + + + + + + + + + 2 +

2 + +/¼ 2 2 2/¼ 2 2 2 +

+ + +/¼ +/¼ + + +/¼

+ + + 2 +

a The plus (+) and minus (2) signs denote a reported increase or decrease in abundance for the relevant microbiota respectively. The equal sign (¼) represents that no difference in abundance was found in that relevant study.

www.ibdjournal.org |

143

Li et al

its composition.3 Age-specific taxonomic changes have been identified in the intestinal microbiome.54 A meta-analysis of human microbiota from multiple studies has shown an age-related fecal bacterial gradient with a transition from communities enriched in Bifidobacteriaceae, Clostridiaceae, Enterococcaceae, Enterobacteriaceae, Lactobacillaceae, and Streptococcaceae in early age to communities enriched in Bacteroidaceae Lachnospiraceae, Prevotellaceae, and Ruminococcaceae in adults.145 Among all of the compositional changes of the microbiome during IBD, it is particularly important to highlight the dysbiosis caused by pathobionts. Pathobionts are distinguishable from pathogens as they only become harmful to predisposed individuals when an environmental stimulus is triggered.146,147 Helicobacter hepaticus is such a pathobiont causing colitis in immunocompromised mice.148,149 Yet, the type VI secretion system of H. hepaticus has been found to induce anti-inflammatory gene expression within intestinal epithelial cells, thus promoting a balance between host colonization and intestinal inflammation.72 Escherichia coli, such as AIEC, is another important pathobiont that may play a role in IBD development150 (see details below in section Antiinflammatory Effects of Microbiota: Function and Mechanisms). The expression of the mannose-binding FimH adhesin is critical for the role of AIEC in IBD.151,152 AIEC strains contain point mutations in FimH that cause a pathoadaptive alteration which enhances intestinal inflammatory response.153 AIEC also alters the microbiota composition in susceptible mice by elevating the production of bioactive lipopolysaccharide (LPS) and flagellin.154 Other relevant pathobionts include Clostridium difficile, Klebsiella pneumoniae, Proteus mirabilis, and Prevotellaceae.155 Nevertheless, given that the microbiota exhibits important control over pathobionts and pathogens,156 interventions can be applied to prevent the etiological effect of pathobionts. For example, the microbial molecule polysaccharide A (which is produced by Bacteroides fragilis) has been demonstrated to prevent H. hepaticus-induced experimental IBD in animal models, likely by inducing IL-10 production and subsequent suppression of colitis.149

ANTI-INFLAMMATORY EFFECTS OF MICROBIOTA: FUNCTION AND MECHANISMS The anti-inflammatory properties of the intestinal microbiota involve host–microbiota interactions that use various metabolic pathways and the immune system. An early study showed that the development of spontaneous colitis and activation of the immune system requires the presence of enteric bacteria.157 Metagenomic computational assessment of the intestinal microbiome identified an overall shift associated with IBD that was seen at both the microbial gene-level and metabolic network-level.3 Such studies demonstrate the importance of evaluating changes in microbial functionality to understand their effects on IBD pathogenesis. Functional signatures of the CD microbiota from stool samples include alterations in bacterial carbohydrate metabolism, bacteria–host interactions, and host-secreted enzymes compared with healthy controls.158 These signatures could be used as potential targets for biomarkers

144

| www.ibdjournal.org

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

and therapeutic interventions. We have an increased appreciation for the potential role of specific bacteria in the etiopathogenesis of IBD. This section highlights some key microbial functions and mechanisms that may influence IBD disease progression.

Metabolite-producing Microbiota The metabolic functions of the intestinal microbiota greatly influence the intestinal environment and thus, the host’s health.2 Changes in microbial composition can lead to an imbalance in metabolites production which in turn might be linked to IBD pathogenesis. Various bacteria, including Faecalibacterium, produce SCFAs, such as acetate, propionate, and butyrate.159,160 Acetates are a substrate for butyrate production, whereas butyrate is a major energy source for colonic epithelial cells; thus, butyrate-producing bacteria play an important role in epithelial barrier integrity.159,160 Indeed, Machiels et al64 found a decrease in butyrate-producing bacteria and a reduction of acetate and proprionate levels in fecal samples from patients with UC. Additionally, a study using an in vitro simulator of the human intestinal microbial ecosystem found lowered SCFA and butyrate production by the luminal communities present in patients with UC.99 The abundance of known SCFA producers (Clostridium Clusters IV and XIVA, F. prausnitzii, and Roseburia spp.) and genes for butyryl-CoA:acetate CoA transferase (responsible for butyrate production) were also reduced.99 Diet greatly influences the composition of the microbiome and the metabolites that are produced by the bacterial species. Both long- and short-term dietary styles have been linked to microbial enterotypes. For example, Bacteroides species increase with protein and animal-fat rich diets and Prevotella increases with carbohydrate rich diets.161 Increases in animal-based diets result in an increased abundance of bile-tolerant bacteria such as Bacteroidetes (Alistipes and Bacteroides) and Proteobacteria (Bilophila) and a decreased abundance of Firmicutes (Roseburia, Eubacterium rectale and Ruminococcus bromii).162 Bile acid (e.g., taurocholic acid) production increases as well in animal-based diets, affecting microbial composition by promoting the expansion of pathogens such as sulphite-reducing Bilophila wadsworthia, which increase microbiota dysbiosis perturbing immune homeostasis.163 Tryptophan may be also involved in the etiopathogenesis of IBD. Interleukin-27 (IL-27) is a mediator of gut epithelial barrier protection and is elevated during IBD.164 The increase of IL-27 impacts the expression of hundreds of genes including indoleamine 2,3dioxygenase. Indoleamine 2,3-dioxygenase is an anti-inflammatory and antibacterial protein that is involved in initiating tryptophan catabolism.165 IL-27–induced indoleamine 2,3-dioxygenase leads to microbiome dysbiosis by inhibiting the growth of the intestinal bacteria through local tryptophan depletion.164 Interestingly, tryptophan reduction is involved in inflammation throughout the human body; deficiencies in tryptophan hydroxylase-1 (which converts tryptophan to serotonin) are known to intensify neuroinflammation.166

Microbiota Disruption of the Intestinal Barrier The intestinal barrier is composed of a single layer of epithelial cells linked through tight junctions and a mucus shield

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

with strong surface hydrophobicity.167 This cell layer is composed of various cell types, including goblet and Paneth cells, that produce mucin and antimicrobial peptides (e.g., defensins, cathelicidins, and lysozymes), respectively. Additionally, goblet and Paneth cells provide strong barrier functions that control the equilibrium between tolerance to microbes and non-self-antigens.168,169 Consequently, disruption of epithelial barrier integrity has been implicated in IBD pathogenesis through a wide range of specific mechanisms encompassing structural, metabolic, and innate immune pathways.170 Barrier defects include increased permeability of the mucus to water and various small or large molecules that can elicit immune response either as a cause or a consequence of IBD. Various bacteria produce hemolysins, which damage host cell membranes. Hemolysin secreted by Escherichia coli increases the initial lesions in the colon mucosa (dubbed as “focal leaks”), which in turn reduces transepithelial electrical resistance and increased macromolecular uptake.171,172 Mucosal samples between patients with IBD and healthy controls show similar levels of a-hemolysin-producing E. coli. However, patients with active UC display 10-fold higher levels of a-hemolysin, suggesting a role of a-hemolysin in disruption of the epithelial barrier, again highlighting the importance of measuring the metabolic capacity of the microbiota rather than simply its composition.172 Bacterial proteases are also known to affect intestinal integrity. Gelatinase, a metalloproteinase produced by Enterococcus faecalis, disrupts the epithelial barrier and increases inflammation in mice that are genetically susceptible to IBD.173 Enterococcal isolates from IBD show higher frequencies of virulence genes including gelE that encodes for gelatinase.174 Similar to other pathogenic species such as Campylobacter, Escherichia coli, and Shigella species, certain Helicobacter species produce cytolethal distending toxins,175 which cause cell cycle arrest, chromatin fragmentation, and apoptosis and are speculated to adversely influence IBD development.176 The gut microbiota composition of IL-10–deficient mice of different research facilities has been found to differ, and this variation was associated with differing susceptibility to colitis induced by Helicobacter hepaticus.177 Indeed, increased prevalence of enterohepatic Helicobacter species in patients with CD and UC have been observed,115 although there are conflicting reports whether Helicobacters (specifically Helicobacter pylori) contribute to IBD progression.178 As discussed above, such discrepancies can likely be attributed to different sampling methodologies. Adherent Lactobacillus spp. can induce upregulation of extracellular mucin secretion and thus protect the gut mucosal surface against the adherence of enteropathogens.179 Intriguingly, frequent use of nonantibiotic non-steroidal anti-inflammatory agents was also found to be a potential risk factor for IBD180 because non-steroidal anti-inflammatory agents are well known to disrupt mucosal integrity along the intestinal tract30 and might impact the microbiota composition.

Adherent Invasive E. coli The colonization of AIEC is frequently observed in patients with IBD and, its role in IBD seems to be multifactorial (Table 1). Mucosa-associated AIEC strains with a 54 kb polyketide synthase

Functional Impacts of the Intestinal Microbiome

genotoxic island were identified at a significantly higher rate in IBD and colorectal cancer patients.181 The polyketide synthase island encodes the multienzymatic machinery involved in the synthesis of colibactin (a peptide–polyketide hybrid genotoxin), and colibactin was involved in inducing tumorigenesis.181 In addition, defects in Paneth cell function or an autophagy deficiency of the host can affect one’s ability to control AIEC infection. Patients with IBD exhibit abnormal expression of carcinoembryonic antigen-related cell adhesion molecule 6 in the ileum, which can also promote AIEC colonization.182,183 IBD-derived AIEC produces pili variants that enhance their adhesion to ileal epithelial cells.124,153 AIEC are known to induce the expression of claudin-2, a pore-forming tight junction protein, and the production of proinflammatory cytokines by infected macrophages.124,125,184 AIEC also induces inflammation by upregulating gut epithelial cell microRNAs that reduce autophagy and whose perturbation is known to be associated with autoimmune diseases like IBD.146,185 Together with genetic factors, IBD-associated AIEC also modulates the ubiquitin proteasome system by decreasing the accumulation of polyubiquitin conjugates, increasing 26S proteasome activities and reducing protein levels of the NF-kB de-ubiquitinase CYLD in the intestine, resulting in aberrant NF-kB activation present during IBD pathogenesis.126 A study on transient AIEC colonization in mice revealed that mice lacking Toll-like receptor 5, which recognizes the presence of bacterial flagellin, are prone to inflammation development.186 This colonization is correlated with microbiota compositional alterations and elevated levels of bioactive LPS and flagellin, implicating an additional role for AIEC in IBD by altering the microbiota composition through a flagellin-dependant manner.186 Furthermore, a higher abundance of the Clostridia class (notably Lachnospiraceae family) was observed, which produces flagellins that are immunodominant antigens in several T-cell–mediated colitis models and CD.186,187 Thus, microbiota alterations are likely involved in promoting the chronic inflammatory process. A recent study also found that a high-fat highsugar diet resulted in microbiota dysbiosis in mice with altered host barrier function; this dysbiosis favored AIEC colonization.92 In agreement with these results, dietary supplementation with the polysaccharide maltodextrin-induced type 1 pili expression in Escherichia coli enhanced AIEC adhesion through type 1 pili-dependent mechanism and enhanced biofilm formation.188

Microbiota in Immune Response Although microbiota–host immune system interactions have been clearly shown to play a major role in IBD, these interactions will be only briefly discussed in this review. Readers are encouraged to read the following reviews for a more detailed discussion of these interactions.40,44,69 Instead, several key reports that demonstrate the direct link between IBD immune response and the intestinal microbiota are highlighted. Reduced levels of F. prausnitzii in IBD can be explained by the anti-inflammatory properties of this species that function by partly blocking NF-kB activation and IL-8 production in IBD animal models and thus improving the dysbiosis seen in www.ibdjournal.org |

145

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Li et al

chemically induced mouse models of colitis.135 Bacteriodes fragilis, a major commensal, produces antigens, such as polysaccharide A, that induce the conversion of CD4+ T cells into FoxP3 regulatory T cells, which in turn produce the IL-10 cytokines that dampen the inflammatory response.149,189,190 In addition, Lactobacillus casei/paracasei species produce a bacterial protease, lactocepin, which selectively degrades the lymphocyte-recruiting chemokine IP-10.191 IP-10 is a proinflammatory chemokine that has been implicated in IBD; thus, its degradation by lactocepin can partially explain the beneficial effects from Lactobacillus spp.191,192 Patients with CD also show a dysregulated response to microbial DNA that is mediated through upregulation of IL-17A, a proinflammatory cytokine.193 The microbiota can also produce bacterial peptidoglycans that can activate the host’s nucleotide-binding oligomerization domain-containing protein 2 (NOD2), a major player implicated in IBD pathogenesis and stimulate the release of proinflammatory cytokines, autophagy, and antigen presentation.6 LPS is a key outer membrane component of Gram-negative bacteria such as Enterobacteriaceae, and its endotoxicity contributes to T-cell mediated colitis development in mice.194 Yet, mutated LPS with low endotoxicity is associated with mucosal immune homeostasis and prevents colitis with fecal samples displaying higher Bacteroidetes and lower Enterobacteriaceae abundances.194

a role for oxygen in intestinal dysbiosis.4 In support of this, there seems to be a shift from anaerobiosis in the healthy state to dysanaerobiosis in IBD with an elevated oxygen level in the gut.199 However, the potential use of antioxidants or probiotics to decrease oxygen levels for the management/treatment of IBD remains to be fully tested. Interestingly, in contrast to this hypothesis, another potentially microbiota-related metabolic disease, type 2 diabetes, has been recently hypothesized to be a redox disease, where the lack of oxygen could be a trigger for type 2 diabetes.200 These findings also support further study into the contribution of oxygen to IBD. Indeed, Walujkar et al201 recently observed both microbial dysbiosis and dysanaerobiosis in patients with UC.

MICROBIOTA-ASSOCIATED INTERVENTIONS FOR IBD Because there is no identified cure for IBD, disease control is a primary focus for therapeutics. Microbiota-involved pathogenesis in IBD has prompted the investigation on the effect of various interventions that specifically target the microbiome in IBD disease progression. The impacts from the administration of antibiotics, probiotics, prebiotics, or dietary supplementation are among the best studied interventions that are currently in use clinically.

Antibiotics Role of the Inflammasome Colonic microbiota can be regulated by host cytoplasmic multiprotein complexes, known as the inflammasome, which constitutes one of the nucleotide-binding oligomerization domain protein-like receptors and provides a regulatory sensing system within the colon.195 NLRP6 suppresses colitogenic microbiota and increases the abundance of Prevotella through IL-18; genetic deletion of NLRP6 has an adverse impact on microbial composition by causing a shift towards a proinflammatory environment that may contribute to IBD initiation and maintenance.93 Treatment with metronidazole and ciprofloxacin led to a reduction of Prevotella species and an ameliorated, anticolitogenic microbiota.93 These results provide evidence showing a perturbation in inflammasome pathway, which could potentially be considered a risk factor for IBD. Prolonged IBD increases the risk in developing colorectal cancer.196 Hence, impact of NLRP6 deficiency on microbiota composition through IL-18 was also found to lead to an enhanced tendency for inflammation-induced colorectal tumorigenesis.93 Additionally, NLRP6 also negatively regulates innate immunity and host defense against pathogenic bacteria, such as Listeria monocytogenes, Salmonella typhimurium, and Escherichia coli.197 Microbiota-induced inflammation driven by chemokine production promotes epithelial cell proliferation through local activation of the IL-6 pathway.198

Role of Oxygen and Reactive Oxygen Species (Hypothesis) The decrease of obligate anaerobes of the Firmicutes phylum and increase in facultative anaerobes in colitic models suggests

146

| www.ibdjournal.org

A meta-analysis of placebo-controlled trials supports the long-term (a median of 6 mo) treatment of CD with nitroimidazole or clofazimine.202 This can be explained by the spectrum activity of nitroimidazole against anaerobes and the immunosuppressive effects displayed by clofazimine. However, a 2-year combination therapy with clarithromycin, rifabutin, and clofazimine for CD did not show sustainable benefits; there was only short-term improvement that was attributed to the nonspecific antibacterial activity of these drugs.203 Another pilot clinical trial conducted in patients with CD suggested that remission and beneficial response were more frequently seen in patients treated with ciprofloxacin for 10 weeks as compared with metronidazol treatment and placebo.204 However, another meta-analysis of randomized controlled trials did not reveal evidence of the benefit of antibiotics in patients with CD.205 The conflicting results seen in various studies could be readily illustrated by the fact that therapeutic doses of antibiotics can alter the overall gut microbiota composition and function because antibiotics are generally nondiscriminant in their targets.206 Although older patients with IBD seem to receive limited benefits from antibiotics, antibiotic treatment at early stages in life (especially in newborns) seems to be a risk factor for the development of IBD. Various retrospective studies have showed an association between pediatric IBD development and antibiotic use,207–209 with one study suggesting a strong association of 3 cephalosporin purchases with CD (but not UC).209 Additionally, Gevers et al13 were able to assess a small subset of the patients with CD that were on antibiotics during sample collection, allowing a comparison between the microbiome in patients with CD

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Functional Impacts of the Intestinal Microbiome

TABLE 2. Microbiota Dysbiosis-involved Functional Alterations of Gut Microbiome: Examples of Plausible, Causal or Contributory Mechanisms in the Pathogenesis of IBD Bacteria Taxa Firmicutes (including species of Eubacterium, Faecalibacterium, Roseburia, and Phascolarctobacterium) Bifidobacterium, Faecalibacterium prausnitzii Bifidobacterium, Bacteroides, Clostridium, Enterobacter, Lactobacillus Faecalibacterium prausnitzii Clostridia, Bacteroides Firmicutes/Clostridia (Ruminococcus torques) Firmicutes (Lactobacillus acidophilus) Firmicutes (Lactobacillus or L. casei/ paracasei) Firmicute (Lactobacillus plantarum) Pathogens (e.g., Enterobacteriaceae)

AIEC

Enterococci Pathogens (e.g., Helicobacter) Pathogens (e.g., E. coli, Campylobacter and Helicobacter) Proteobacteria (e.g., Bilophila, Desulfovibrio) Proteobacteria (e.g., Bdellovibrio bacteriovorus)

Functional Considerations Maintain metabolic symbiosis associated with production of SCFAs (e.g., acetate, propionate, and butyrate) that affect colonic pH, the growth of pathogens, and energy supply to the colonic epithelial cells Decrease glycan-degrading enzymes Promote the production of host mucosal glycans Produce choline metabolites that modulate lipid metabolism and glucose homeostasis Produce bile acid metabolites to facilitate lipid absorption, barrier function, and energy homeostasis Block NF-kB activation and IL-8 production Provide antigens that induce regulatory T cells to generate antiinflammatory cytokines to protect commensal gut microbiota Degrade mucin Reduce platelet-activating factor-induced NF-kB activation and IL-8 production in gut epithelial cells Promote mucin secretion Produce lactocepin protease that selectively degrades proinflammatory chemokines Produce an extracellular immunomodulatory peptide mediating microbiota/host interaction Produce mucinases to hydrolyze mucus Produce toxins Produce hemolysin Produce lipopolysaccharide Produce flagellin, a dominant antigen in IBD Manipulate ubiquitination and deubiquitination pathways Epithelial barrier disruption with microbial invasion, aberrant immune response with increased T-helper 17 cells, and cytokine production (e.g., IL-27) Alter microbiota composition Produce hemolysin Disrupt gut barrier Survive inside macrophages and induce production of proinflammatory cytokines by infected macrophages Modulate ubiquitin proteasome system Upregulate microRNAs in gut epithelial cells to reduce autophagy Produce bacterial proteases (e.g., gelatinase) Affect metabolite pathways including tryptophan metabolism Produce cytolethal distending toxins Produce hydrogen sulfide Prey other Gram-negative bacteria for survival and act as a population balancer Reduce pathogen colonization

References Refs. 2,119,159, 218,219

Refs. 2,159

Ref. 2 Ref. 135 Refs. 189,220 Ref. 92 Ref. 221 Refs. 179,191,192

Ref. 222 Refs. 126,164,186, 187,194,222–226

Refs. 124– 126,153,185,186

Refs. 173,174,227 Refs. 119,228 Refs. 175,176, 229–231 Refs. 163,177 Refs. 106,232

www.ibdjournal.org |

147

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Li et al

with and without antibiotic exposure. A strong effect was observed on the microbial composition, and exposure to antibiotics generally amplified the dysbiosis described in their treatment-naive inception cohort of pediatric patients.13 These results indicate that the effects of antibiotics on the microbiota composition during the period of commensal gut microbiota establishment in newborns may result in the establishment of microbiota’s that are susceptible to developing IBD later in life.

Probiotics Probiotics comprises a number of strains of microbes and offer a method to potentially alter the intestinal microbiome.210,211 Probiotic usage by patients with IBD is common (e.g., in 40%– 50% of patients with IBD in 2 separate survey-based studies conducted in the United Kingdom).212,213 Various randomized trials have been conducted to understand the capacity of probiotics as induction, maintenance, and postoperative prophylaxis in IBD.210 Current probiotics have shown no benefit in any aspect of CD therapy, whereas there remains some advocacy for their use in mild UC.210 One highly concentrated probiotic mixture, VSL#3, contains 8 strains of Bifidobacterium, Lactobacillus, or Streptococcus spp. and has received attention for its beneficial role in the management of chronic recurrent pouchitis192,210,214–216 that follows in patients undergoing ileoanal anastomosis and J-Pouch creation for patients with UC who failed conventional medical therapies. Another study showed that giving daily doses of Lactobacillus spp. (2.5 · 109 cfu/g) to patients with UC with pouchitis for 2 months could restore the mucosal barrier against Escherichia coli although there were no significant differences in the mucosal microbiota compositions.217 The beneficial effects of Lactobacillus spp. on the amelioration of IBD are indeed mechanistically supported as their metabolites or products can reduce proinflammatory cytokines (Table 2).179,191,192,221 Nevertheless, because of the increased abundance of Bidfidobacterium and Lactobacillus species observed in patients with active CD, cautions against probiotic use have also been suggested.49 Altogether, it is still unclear whether the increase of apparently beneficial microbes is a secondary phenomenon in these patients with CD.

Prebiotics Prebiotics, by definition, are compounds that change the composition and/or activity of the intestinal microbiota in a manner that is beneficial to the host.233 Inulin and oligofructose are 2 prebiotics, which promote the growth of indigenous Bifidobacterium and Lactobacillus spp.234 Patients with CD receiving oligofructoseenriched inulin showed elevated levels of acetaldehyde and butyrate among the analyzed fecal metabolites.235 In chemically induced colitic rats, difructose dianhydride-enriched caramels exhibit effects, such as promoting beneficial colon microbiota, increasing the levels of SCFAs and inhibiting the production of proinflammatory cytokines.236 Similarly, other prebiotics, such as cellobiose and rice fiber also possess anti-inflammatory effects by modulating the colonic environment and reducing proinflammatory cytokines in experimental colitis.237,238 Although prebiotics seem to be promising as

148

| www.ibdjournal.org

a potential treatments, their effects on modifying gut microbiota may vary between healthy individuals and patients with IBD.239 This concern is consistent with the current caution in not supporting the use of prebiotics in the dietary management of CD adults.240

Diet Both long- and short-term diet modifications are known to profoundly influence the structure and activity of the intestinal microbiome.161,162 Observational studies of high-fat and sugar (“Western”) diets show a remarkable increase of Escherichia coli populations (including AIEC as discussed above)92 and a decreased abundance of sulphite-reducing pathogens that exacerbate dysbiosis.163 Additionally, experimental studies have demonstrated the effect of dietary supplementation on animal colitis. Dietary fiber (cellulose) supplementation in the early lives of mice stimulated noteworthy shifts in the mice’s gut microbiota with decreased abundance of Coribacteriaceae and increased levels of Peptostreptococcaceae and Clostridiaceae. This was associated with reduced gut inflammation in adult mice.241 Oral administration of the anti-inflammatory peptide pyroglutamyl leucine (an enzymatic hydrolysate of wheat gluten) in mice improved dextran sulfate sodium–induced colitis with increased and decreased abundance of Bacteroidetes and Firmicutes, respectively.242 Aberrant DNA methylation is a purported risk factor for IBD.243,244 In keeping with this hypothesis, animal studies have shown that transient prenatal or maternal exposure to diets supplemented with methyl donor micronutrients induced a colitogenic microbiome and augmented colitis in both the maternal mice and their offspring.243,244 In another study assessing functional behavior of microbes and dietary interventions, a set of mouse experiments were performed inoculating intestinal microbes isolated from children suffering from kwashiorkor (a condition characterized by severe protein–calorie deprivation). Germ-free mice transplanted with the intestinal microbiota of infants with kwashiorkor gained weight with an interventional diet but returned to losing weight when fed a typical diet.245 Thus, recurrence of disease inducing functionality by the microbiota with dietary resumption speaks to the resiliency of the functions provided by these microbes. Persistence will need to be considered because we address intestinal microbiome interventions as treatment for IBD. Taken together, these studies do align with the dietary management of IBD as currently advocated.240,246

CONCLUSIONS IBD is a multifactorial disorder that is suspected to be highly influenced by the functional characteristics of one’s intestinal microbiota. Extensive and expansive analyses through large cohort studies have been aided by recent technological advances to expand our knowledge of altered gut microbiota composition and function in IBD. Nevertheless, many studies have only targeted the higher taxonomic levels of bacteria; and thus, a shift to identify specific families, genera, or species can broaden our understanding of their functional role(s) in the pathogenesis of IBD. Functional dissection of the microbiome and thus

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

elucidation of relevant mechanisms pose a significant challenge because of the complex relationship within and among the microbiota, host, and environment. Elucidating the functional parameters that govern these relationships will likely prove as far more important than phenotypic characterization of the intestinal microbiota. New technologies coupled with both compositional and functional strategies may contribute in defining the role of specific bacterial species and their relevant metabolic influences. Such studies could reveal whether the microbiome plays an etiological or contributing role in the onset or progression of this complex disease and thus, revolutionize our understanding of IBD.

REFERENCES 1. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–124. 2. Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–1267. 3. 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;109:594–599. 4. Podolsky DK. Inflammatory bowel disease (1). N Engl J Med. 1991;325: 928–937. 5. Abraham C, Cho JH. Inflammatory bowel disease. N Engl J Med. 2009; 361:2066–2078. 6. Khor B, Gardet A, Xavier RJ. Genetics and pathogenesis of inflammatory bowel disease. Nature. 2011;474:307–317. 7. Ng SC, Bernstein CN, Vatn MH, et al. Geographical variability and environmental risk factors in inflammatory bowel disease. Gut. 2013; 62:630–649. 8. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448:427–434. 9. Fraher MH, O’Toole PW, Quigley EM. Techniques used to characterize the gut microbiota: a guide for the clinician. Nat Rev Gastroenterol Hepatol. 2012;9:312–322. 10. Maccaferri S, Biagi E, Brigidi P. Metagenomics: key to human gut microbiota. Dig Dis. 2011;29:525–530. 11. Yau Y, Leong RW, Zeng M, et al. Proteomics and metabolomics in inflammatory bowel disease. J Gastroenterol Hepatol. 2013;28: 1076–1086. 12. Arumugam M, Raes J, Pelletier E, et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174–180. 13. Gevers D, Kugathasan S, Denson LA, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392. 14. Berry D, Reinisch W. Intestinal microbiota: a source of novel biomarkers in inflammatory bowel diseases? Best Pract Res Clin Gastroenterol. 2013;27:47–58. 15. Lepage P, Leclerc MC, Joossens M, et al. A metagenomic insight into our gut’s microbiome. Gut. 2013;62:146–158. 16. Meyer F, Trimble WL, Chang EB, et al. Functional predictions from inference and observation in sequence-based inflammatory bowel disease research. Genome Biol. 2012;13:169. 17. Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79. 18. Ridaura VK, Faith JJ, Rey FE, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341: 1241214. 19. Theriot CM, Koenigsknecht MJ, Carlson PE Jr, et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome increase susceptibility to Clostridium difficile infection. Nat Commun. 2014;5:3114. 20. Hooper LV, Gordon JI. Commensal host-bacterial relationships in the gut. Science. 2001;292:1115–1118. 21. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273.

Functional Impacts of the Intestinal Microbiome

22. Marchesi J, Shanahan F. The normal intestinal microbiota. Curr Opin Infect Dis. 2007;20:508–513. 23. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 2006;124:837–848. 24. Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology. 2008;134:577–594. 25. Fava F, Danese S. Intestinal microbiota in inflammatory bowel disease: friend of foe? World J Gastroenterol. 2011;17:557–566. 26. Sommer F, Backhed F. The gut microbiota–masters of host development and physiology. Nat Rev Microbiol. 2013;11:227–238. 27. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet. 2003; 361:512–519. 28. Hooper LV. Do symbiotic bacteria subvert host immunity? Nat Rev Microbiol. 2009;7:367–374. 29. Frick JS, Autenrieth IB. The gut microflora and its variety of roles in health and disease. Curr Top Microbiol Immunol. 2013;358:273–289. 30. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. 31. Caporaso JG, Lauber CL, Costello EK, et al. Moving pictures of the human microbiome. Genome Biol. 2011;12:R50. 32. Claesson MJ, Jeffery IB, Conde S, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178–184. 33. Dominguez-Bello MG, Blaser MJ, Ley RE, et al. Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology. 2011;140:1713–1719. 34. Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–230. 35. Biagi E, Nylund L, Candela M, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PloS One. 2010;5:e10667. 36. Faith JJ, Guruge JL, Charbonneau M, et al. The long-term stability of the human gut microbiota. Science. 2013;341:1237439. 37. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638. 38. Ley RE, Backhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–11075. 39. Morelli L. Postnatal development of intestinal microflora as influenced by infant nutrition. J Nutr. 2008;138:1791S–1795S. 40. Mackie RI, Sghir A, Gaskins HR. Developmental microbial ecology of the neonatal gastrointestinal tract. Am J Clin Nutr. 1999;69: 1035S–1045S. 41. Claus SP, Ellero SL, Berger B, et al. Colonization-induced host-gut microbial metabolic interaction. MBio. 2011;2:e00271–e00210. 42. Qin J, Li Y, Cai Z, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. 43. Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol. 2012;10:735–742. 44. Manichanh C, Borruel N, Casellas F, et al. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9:599–608. 45. Frank DN, St Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A. 2007;104:13780–13785. 46. Lepage P, Hasler R, Spehlmann ME, et al. Twin study indicates loss of interaction between microbiota and mucosa of patients with ulcerative colitis. Gastroenterology. 2011;141:227–236. 47. Kleessen B, Kroesen AJ, Buhr HJ, et al. Mucosal and invading bacteria in patients with inflammatory bowel disease compared with controls. Scand J Gastroenterol. 2002;37:1034–1041. 48. Lepage P, Seksik P, Sutren M, et al. Biodiversity of the mucosa-associated microbiota is stable along the distal digestive tract in healthy individuals and patients with IBD. Inflamm Bowel Dis. 2005;11:473–480. 49. Wang W, Chen L, Zhou R, et al. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J Clin Microbiol. 2014;52:398–406. 50. Walker AW, Ince J, Duncan SH, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5: 220–230. 51. Hansen R, Russell RK, Reiff C, et al. Microbiota of de-novo pediatric IBD: increased Faecalibacterium prausnitzii and reduced bacterial diversity in Crohn’s but not in ulcerative colitis. Am J Gastroenterol. 2012; 107:1913–1922. www.ibdjournal.org |

149

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

Li et al

52. Willing BP, Dicksved J, Halfvarson J, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139:1844–1854.e1841. 53. Willing B, Halfvarson J, Dicksved J, et al. Twin studies reveal specific imbalances in the mucosa-associated microbiota of patients with ileal Crohn’s disease. Inflamm Bowel Dis. 2009;15:653–660. 54. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–227. 55. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. 56. Tap J, Mondot S, Levenez F, et al. Towards the human intestinal microbiota phylogenetic core. Environ Microbiol. 2009;11:2574–2584. 57. Sobhani I, Tap J, Roudot-Thoraval F, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PloS One. 2011;6:e16393. 58. Kolmeder CA, de Been M, Nikkila J, et al. Comparative metaproteomics and diversity analysis of human intestinal microbiota testifies for its temporal stability and expression of core functions. PloS One. 2012;7: e29913. 59. Rajilic-Stojanovic M, Heilig HG, Tims S, et al. Long-term monitoring of the human intestinal microbiota composition. Environ Microbiol. [published online ahead of print October 15, 2012]. doi: 10.1111/1462-2920.12023. 60. Krych L, Hansen CH, Hansen AK, et al. Quantitatively different, yet qualitatively alike: a meta-analysis of the mouse core gut microbiome with a view towards the human gut microbiome. PloS One. 2013;8:e62578. 61. Papa E, Docktor M, Smillie C, et al. Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PloS One. 2012;7:e39242. 62. Bibiloni R, Mangold M, Madsen KL, et al. The bacteriology of biopsies differs between newly diagnosed, untreated, Crohn’s disease and ulcerative colitis patients. J Med Microbiol. 2006;55:1141–1149. 63. Andoh A, Imaeda H, Aomatsu T, et al. Comparison of the fecal microbiota profiles between ulcerative colitis and Crohn’s disease using terminal restriction fragment length polymorphism analysis. J Gastroenterol. 2011; 46:479–486. 64. Machiels K, Joossens M, Sabino J, et al. A decrease of the butyrateproducing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63:1275–1283. 65. Nwosu FC, Thorkildsen LT, Avershina E, et al. Age-dependent fecal bacterial correlation to inflammatory bowel disease for newly diagnosed untreated children. Gastroenterol Res Pract. 2013;2013:302398. 66. Dicksved J, Halfvarson J, Rosenquist M, et al. Molecular analysis of the gut microbiota of identical twins with Crohn’s disease. ISME J. 2008;2:716–727. 67. Zitomersky NL, Atkinson BJ, Franklin SW, et al. Characterization of adherent bacteroidales from intestinal biopsies of children and young adults with inflammatory bowel disease. PloS One. 2013;8:e63686. 68. Tong M, Li X, Wegener Parfrey L, et al. A modular organization of the human intestinal mucosal microbiota and its association with inflammatory bowel disease. PloS One. 2013;8:e80702. 69. Cho JH, Brant SR. Recent insights into the genetics of inflammatory bowel disease. Gastroenterology. 2011;140:1704–1712. 70. Albuquerque MC, Aleixo AL, Benchimol EI, et al. The IFN-gamma +874T/A gene polymorphism is associated with retinochoroiditis toxoplasmosis susceptibility. Mem Inst Oswaldo Cruz. 2009;104:451–455. 71. Ward LM, Rauch F, Matzinger MA, et al. Iliac bone histomorphometry in children with newly diagnosed inflammatory bowel disease. Osteoporosis Int. 2010;21:331–337. 72. Cuthbert AP, Fisher SA, Mirza MM, et al. The contribution of NOD2 gene mutations to the risk and site of disease in inflammatory bowel disease. Gastroenterology. 2002;122:867–874. 73. Benchimol EI, Guttmann A, Griffiths AM, et al. Increasing incidence of paediatric inflammatory bowel disease in Ontario, Canada: evidence from health administrative data. Gut. 2009;58:1490–1497. 74. Lopes FR, Silva JC, Benchimol M, et al. The protist Trichomonas vaginalis harbors multiple lineages of transcriptionally active Mutator-like elements. BMC Genomics. 2009;10:330. 75. Pereira-Neves A, Benchimol M. Tritrichomonas foetus: budding from multinucleated pseudocysts. Protist. 2009;160:536–551. 76. Benchimol M. Hydrogenosomes under microscopy. Tissue Cell. 2009; 41:151–168. 77. Montravers P, Lepape A, Dubreuil L, et al. Clinical and microbiological profiles of community-acquired and nosocomial intra-abdominal infec-

150

| www.ibdjournal.org

78. 79. 80.

81.

82.

83. 84.

85.

86. 87.

88. 89. 90.

91.

92.

93. 94.

95.

96. 97.

98. 99.

100.

tions: results of the French prospective, observational EBIIA study. J Antimicrob Chemother. 2009;63:785–794. Seow CH, Benchimol EI, Steinhart AH, et al. Budesonide for Crohn’s disease. Expert Opin Drug Metab Toxicol. 2009;5:971–979. Tai E, Benchimol S. TRIMming p53 for ubiquitination. Proc Natl Acad Sci U S A. 2009;106:11431–11432. Hansen R, Berry SH, Mukhopadhya I, et al. The microaerophilic microbiota of de-novo paediatric inflammatory bowel disease: the BISCUIT study. PloS One. 2013;8:e58825. Frank DN, Robertson CE, Hamm CM, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm Bowel Dis. 2011; 17:179–184. Li E, Hamm CM, Gulati AS, et al. Inflammatory bowel diseases phenotype, C. difficile and NOD2 genotype are associated with shifts in human ileum associated microbial composition. PloS One. 2012;7:e26284. Sanderson JD, Moss MT, Tizard ML, et al. Mycobacterium paratuberculosis DNA in Crohn’s disease tissue. Gut. 1992;33:890–896. Bull TJ, McMinn EJ, Sidi-Boumedine K, et al. Detection and verification of Mycobacterium avium subsp. paratuberculosis in fresh ileocolonic mucosal biopsy specimens from individuals with and without Crohn’s disease. J Clin Microbiol. 2003;41:2915–2923. Scanu AM, Bull TJ, Cannas S, et al. Mycobacterium avium subspecies paratuberculosis infection in cases of irritable bowel syndrome and comparison with Crohn’s disease and Johne’s disease: common neural and immune pathogenicities. J Clin Microbiol. 2007;45:3883–3890. Andoh A, Kuzuoka H, Tsujikawa T, et al. Multicenter analysis of fecal microbiota profiles in Japanese patients with Crohn’s disease. J Gastroenterol. 2012;47:1298–1307. Thorkildsen LT, Nwosu FC, Avershina E, et al. Dominant fecal microbiota in newly diagnosed untreated inflammatory bowel disease patients. Gastroenterol Res Pract. 2013;2013:636785. Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora in inflammatory bowel disease. Gastroenterology. 2002;122:44–54. Kang S, Denman SE, Morrison M, et al. Dysbiosis of fecal microbiota in Crohn’s disease patients as revealed by a custom phylogenetic microarray. Inflamm Bowel Dis. 2010;16:2034–2042. Kang CS, Ban M, Choi EJ, et al. Extracellular vesicles derived from gut microbiota, especially Akkermansia muciniphila, protect the progression of dextran sulfate sodium-induced colitis. PloS One. 2013;8:e76520. Walker AW, Sanderson JD, Churcher C, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2011;11:7. Martinez-Medina M, Denizot J, Dreux N, et al. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut. 2014;63:116–124. Elinav E, Strowig T, Kau AL, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145:745–757. Mondot S, Kang S, Furet JP, et al. Highlighting new phylogenetic specificities of Crohn’s disease microbiota. Inflamm Bowel Dis. 2011;17: 185–192. Clayton EM, Rea MC, Shanahan F, et al. The vexed relationship between Clostridium difficile and inflammatory bowel disease: an assessment of carriage in an outpatient setting among patients in remission. Am J Gastroenterol. 2009;104:1162–1169. Berg AM, Kelly CP, Farraye FA. Clostridium difficile infection in the inflammatory bowel disease patient. Inflamm Bowel Dis. 2013;19:194–204. Masclee GM, Penders J, Jonkers DM, et al. Is clostridium difficile associated with relapse of inflammatory bowel disease? results from a retrospective and prospective cohort study in the Netherlands. Inflamm Bowel Dis. 2013;19:2125–2131. Sokol H, Seksik P, Rigottier-Gois L, et al. Specificities of the fecal microbiota in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:106–111. Vermeiren J, Van den Abbeele P, Laukens D, et al. Decreased colonization of fecal Clostridium coccoides/Eubacterium rectale species from ulcerative colitis patients in an in vitro dynamic gut model with mucin environment. FEMS Microbiol Ecol. 2012;79:685–696. Kumari R, Ahuja V, Paul J. Fluctuations in butyrate-producing bacteria in ulcerative colitis patients of North India. World J Gastroenterol. 2013; 19:3404–3414.

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

101. Kabeerdoss J, Sankaran V, Pugazhendhi S, et al. Clostridium leptum group bacteria abundance and diversity in the fecal microbiota of patients with inflammatory bowel disease: a case-control study in India. BMC Gastroenterol. 2013;13:20. 102. Varela E, Manichanh C, Gallart M, et al. Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis. Aliment Pharmacol Ther. 2013;38:151–161. 103. McLaughlin SD, Walker AW, Churcher C, et al. The bacteriology of pouchitis: a molecular phylogenetic analysis using 16S rRNA gene cloning and sequencing. Ann Surg. 2010;252:90–98. 104. Angelberger S, Reinisch W, Makristathis A, et al. Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation. Am J Gastroenterol. 2013;108:1620–1630. 105. Png CW, Linden SK, Gilshenan KS, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105:2420–2428. 106. Iebba V, Santangelo F, Totino V, et al. Higher prevalence and abundance of Bdellovibrio bacteriovorus in the human gut of healthy subjects. PloS One. 2013;8:e61608. 107. Loubinoux J, Bronowicki JP, Pereira IA, et al. Sulfate-reducing bacteria in human feces and their association with inflammatory bowel diseases. FEMS Microbiol Ecol. 2002;40:107–112. 108. Jia W, Whitehead RN, Griffiths L, et al. Diversity and distribution of sulphate-reducing bacteria in human faeces from healthy subjects and patients with inflammatory bowel disease. FEMS Immunol Med Microbiol. 2012;65:55–68. 109. Hakansson A, Tormo-Badia N, Baridi A, et al. Immunological alteration and changes of gut microbiota after dextran sulfate sodium (DSS) administration in mice. Clin Exp Med. [published online ahead of print January 11, 2014]. doi: 10.1007/s10238-013-0270-5. 110. Lennon G, Balfe A, Bambury N, et al. Correlations between colonic crypt mucin chemotype, inflammatory grade and Desulfovibrio species in ulcerative colitis. Colorectal Dis. 2014;16:O161–O169. 111. Man SM, Zhang L, Day AS, et al. Campylobacter concisus and other Campylobacter species in children with newly diagnosed Crohn’s disease. Inflamm Bowel Dis. 2010;16:1008–1016. 112. Man SM. The clinical importance of emerging Campylobacter species. Nat Rev Gastroenterol Hepatol. 2011;8:669–685. 113. Mahendran V, Riordan SM, Grimm MC, et al. Prevalence of Campylobacter species in adult Crohn’s disease and the preferential colonization sites of Campylobacter species in the human intestine. PloS One. 2011;6:e25417. 114. Mukhopadhya I, Hansen R, El-Omar EM, et al. IBD-what role do Proteobacteria play? Nat Rev Gastroenterol Hepatol. 2012;9:219–230. 115. Bohr UR, Glasbrenner B, Primus A, et al. Identification of enterohepatic Helicobacter species in patients suffering from inflammatory bowel disease. J Clin Microbiol. 2004;42:2766–2768. 116. Man SM, Zhang L, Day AS, et al. Detection of enterohepatic and gastric helicobacter species in fecal specimens of children with Crohn’s disease. Helicobacter. 2008;13:234–238. 117. Thomson JM, Hansen R, Berry SH, et al. Enterohepatic helicobacter in ulcerative colitis: potential pathogenic entities? PloS One. 2011;6:e17184. 118. Simone DA, Zhang X, Li J, et al. Comparison of responses of primate spinothalamic tract neurons to pruritic and algogenic stimuli. J Neurophysiol. 2004;91:213–222. 119. Lu K, Knutson CG, Wishnok JS, et al. Serum metabolomics in a Helicobacter hepaticus mouse model of inflammatory bowel disease reveal important changes in the microbiome, serum peptides, and intermediary metabolism. J Proteome Res. 2012;11:4916–4926. 120. Seksik P, Rigottier-Gois L, Gramet G, et al. Alterations of the dominant faecal bacterial groups in patients with Crohn’s disease of the colon. Gut. 2003;52:237–242. 121. Kotlowski R, Bernstein CN, Sepehri S, et al. High prevalence of Escherichia coli belonging to the B2+D phylogenetic group in inflammatory bowel disease. Gut. 2007;56:669–675. 122. Darfeuille-Michaud A, Neut C, Barnich N, et al. Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn’s disease. Gastroenterology. 1998;115:1405–1413. 123. Martinez-Medina M, Aldeguer X, Gonzalez-Huix F, et al. Abnormal microbiota composition in the ileocolonic mucosa of Crohn’s disease patients as revealed by polymerase chain reaction-denaturing gradient gel electrophoresis. Inflamm Bowel Dis. 2006;12:1136–1145.

Functional Impacts of the Intestinal Microbiome

124. Denizot J, Sivignon A, Barreau F, et al. Adherent-invasive Escherichia coli induce claudin-2 expression and barrier defect in CEABAC10 mice and Crohn’s disease patients. Inflamm Bowel Dis. 2012;18: 294–304. 125. Negroni A, Costanzo M, Vitali R, et al. Characterization of adherentinvasive Escherichia coli isolated from pediatric patients with inflammatory bowel disease. Inflamm Bowel Dis. 2012;18:913–924. 126. Cleynen I, Vazeille E, Artieda M, et al. Genetic and microbial factors modulating the ubiquitin proteasome system in inflammatory bowel disease. Gut. 2014;63:1265–1274. 127. Darfeuille-Michaud A, Boudeau J, Bulois P, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004;127:412–421. 128. Martinez-Medina M, Aldeguer X, Lopez-Siles M, et al. Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn’s disease. Inflamm Bowel Dis. 2009;15:872–882. 129. Baumgart M, Dogan B, Rishniw M, et al. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s disease involving the ileum. ISME J. 2007;1:403–418. 130. Thomazini CM, Samegima DA, Rodrigues MA, et al. High prevalence of aggregative adherent Escherichia coli strains in the mucosa-associated microbiota of patients with inflammatory bowel diseases. Int J Med Microbiol. 2011;301:475–479. 131. Knosel T, Schewe C, Petersen N, et al. Prevalence of infectious pathogens in Crohn’s disease. Pathol Res Pract. 2009;205:223–230. 132. Blais Lecours P, Marsolais D, Cormier Y, et al. Increased prevalence of Methanosphaera stadtmanae in inflammatory bowel diseases. PloS One. 2014;9:e87734. 133. Wexler HM. Bacteroides: the good, the bad, and the nitty-gritty. Clin Microbiol Rev. 2007;20:593–621. 134. Swidsinski A, Weber J, Loening-Baucke V, et al. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol. 2005;43:3380–3389. 135. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008; 105:16731–16736. 136. Collins MD, Lawson PA, Willems A, et al. The phylogeny of the genus Clostridium: proposal of five new genera and eleven new species combinations. Int J Syst Bacteriol. 1994;44:812–826. 137. Andoh A, Sakata S, Koizumi Y, et al. Terminal restriction fragment length polymorphism analysis of the diversity of fecal microbiota in patients with ulcerative colitis. Inflamm Bowel Dis. 2007;13:955–962. 138. Sasaki M, Klapproth JM. The role of bacteria in the pathogenesis of ulcerative colitis. J Signal Transduct. 2012;2012:704953. 139. Schilling J, Loening-Baucke V, Dorffel Y. Increased Gardnerella vaginalis urogenital biofilm in inflammatory bowel disease. J Crohns Colitis. 2014;8:543–549. 140. Dave M, Purohit T, Razonable R, et al. Opportunistic infections due to inflammatory bowel disease therapy. Inflamm Bowel Dis. 2014;20: 196–212. 141. Rahier JF, Magro F, Abreu C, et al. Second European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease. J Crohns Colitis. 2014;8:443–468. 142. Molodecky NA, Soon IS, Rabi DM, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46–54.e42; quiz e30. 143. Benchimol EI, Fortinsky KJ, Gozdyra P, et al. Epidemiology of pediatric inflammatory bowel disease: a systematic review of international trends. Inflamm Bowel Dis. 2011;17:423–439. 144. Midlej V, Benchimol M. Giardia lamblia behavior during encystment: how morphological changes in shape occur. Parasitol Int. 2009;58:72–80. 145. Saland JM, Shneider BL, Bromberg JS, et al. Successful split liverkidney transplant for factor H associated hemolytic uremic syndrome. Clin J Am Soc Nephrol. 2009;4:201–206. 146. Patel KK, Stappenbeck TS. Autophagy and intestinal homeostasis. Annu Rev Physiol. 2013;75:241–262. 147. Goldschmidt R, Benchimol J, Chor Maio M. The cardiological perspective: Francisco Laranja and research on Chagas’ disease. Interview by www.ibdjournal.org |

151

Li et al

148. 149. 150. 151.

152.

153.

154.

155.

156. 157.

158. 159.

160. 161. 162. 163.

164.

165. 166.

167. 168. 169. 170. 171.

152

Rose Goldschmidt, Jaime Benchimol, Marcos Chor Maio [in Portuguese]. Hist Cienc Saude Manguinhos. 2009;16(suppl 1):95–114. Benchimol EI, Walsh CM, Ling SC. Early diagnosis of neonatal cholestatic jaundice: test at 2 weeks. Can Fam Physician. 2009;55: 1184–1192. Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–625. Benchimol JL. Editor’s note. Hist Cienc Saude Manguinhos. 2009;16: 847–849. Chevalier N, Carrier P, Piche M, et al. Adrenocortical incidentaloma with uncertain prognosis associated with an inadequately treated congenital adrenal hyperplasia. Ann Endocrinol. 2010;71:56–59. Huang HL, Yeh WC, Lai MZ, et al. Impaired TNFalpha-induced A20 expression in E1A/Ras-transformed cells. Br J Cancer. 2009;101: 1555–1564. Dreux N, Denizot J, Martinez-Medina M, et al. Point mutations in FimH adhesin of Crohn’s disease-associated adherent-invasive Escherichia coli enhance intestinal inflammatory response. PLoS Pathogens. 2013;9: e1003141. Komiyama A, Hultin M, Nasstrom K, et al. Soft tissue conditions and marginal bone changes around immediately loaded implants inserted in edentate jaws following computer guided treatment planning and flapless surgery: a ./¼1-year clinical follow-up study. Clin Implant Dent Relat Res. 2012;14:157–169. Pillois X, Phuong Do Thi N, Reynaud A, et al. TaqIB polymorphism in cholesterol ester transfer protein (CETP) gene predicts future cardiovascular death in patients experiencing an acute coronary syndrome. Clin Chem Lab Med. 2009;47:1039–1046. Midlej V, Benchimol M. Trichomonas vaginalis kills and eats–evidence for phagocytic activity as a cytopathic effect. Parasitology. 2010;137:65–76. Sellon RK, Tonkonogy S, Schultz M, et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun. 1998;66: 5224–5231. Erickson AR, Cantarel BL, Lamendella R, et al. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PloS One. 2012;7:e49138. Mahowald MA, Rey FE, Seedorf H, et al. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proc Natl Acad Sci U S A. 2009;106:5859–5864. Wong JM, de Souza R, Kendall CW, et al. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006;40:235–243. Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–108. David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. Devkota S, Wang Y, Musch MW, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature. 2012;487:104–108. Diegelmann J, Olszak T, Goke B, et al. A novel role for interleukin-27 (IL-27) as mediator of intestinal epithelial barrier protection mediated via differential signal transducer and activator of transcription (STAT) protein signaling and induction of antibacterial and anti-inflammatory proteins. J Biol Chem. 2012;287:286–298. Ciorba MA. Indoleamine 2,3 dioxygenase in intestinal disease. Curr Opin Gastroenterol. 2013;29:146–152. Nowak EC, de Vries VC, Wasiuk A, et al. Tryptophan hydroxylase-1 regulates immune tolerance and inflammation. J Exp Med. 2012;209: 2127–2135. Mack DR, Neumann AW, Policova Z, et al. Surface hydrophobicity of the intestinal tract. Am J Physiol. 1992;262:G171–G177. Antoni L, Nuding S, Weller D, et al. Human colonic mucus is a reservoir for antimicrobial peptides. J Crohns Colitis. 2013;7:e652–e664. Meisch JP, Nishimura M, Vogel RM, et al. Human beta-defensin 3 peptide is increased and redistributed in Crohn’s ileitis. Inflamm Bowel Dis. 2013; 19:942–953. Van Limbergen J, Wilson DC, Satsangi J. The genetics of Crohn’s disease. Annu Rev Genomics Hum Genet. 2009;10:89–116. Troeger H, Richter JF, Beutin L, et al. Escherichia coli alpha-haemolysin induces focal leaks in colonic epithelium: a novel mechanism of bacterial translocation. Cell Microbiol. 2007;9:2530–2540. | www.ibdjournal.org

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

172. Bucker R, Schulz E, Gunzel D, et al. Alpha-Haemolysin of Escherichia coli in IBD: a potentiator of inflammatory activity in the colon. Gut. [published online ahead of print February 17, 2014]. doi: 10.1136/gutjnl2013-306099. 173. Steck N, Mueller K, Schemann M, et al. Bacterial proteases in IBD and IBS. Gut. 2012;61:1610–1618. 174. Golinska E, Tomusiak A, Gosiewski T, et al. Virulence factors of Enterococcus strains isolated from patients with inflammatory bowel disease. World J Gastroenterol. 2013;19:3562–3572. 175. Young VB, Chien CC, Knox KA, et al. Cytolethal distending toxin in avian and human isolates of Helicobacter pullorum. J Infect Dis. 2000; 182:620–623. 176. Smith JL, Bayles DO. The contribution of cytolethal distending toxin to bacterial pathogenesis. Crit Rev Microbiol. 2006;32:227–248. 177. Yang I, Eibach D, Kops F, et al. Intestinal microbiota composition of interleukin-10 deficient C57BL/6J mice and susceptibility to Helicobacter hepaticus-induced colitis. PloS One. 2013;8:e70783. 178. Bell SJ, Chisholm SA, Owen RJ, et al. Evaluation of Helicobacter species in inflammatory bowel disease. Aliment Pharmacol Ther. 2003;18:481–486. 179. Mack DR, Ahrne S, Hyde L, et al. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut. 2003;52:827–833. 180. Ananthakrishnan AN, Higuchi LM, Huang ES, et al. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for Crohn disease and ulcerative colitis: a cohort study. Ann Intern Med. 2012;156:350–359. 181. Arthur JC, Perez-Chanona E, Muhlbauer M, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 2012; 338:120–123. 182. Barnich N, Carvalho FA, Glasser AL, et al. CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J Clin Invest. 2007;117:1566–1574. 183. Tchoupa AK, Schuhmacher T, Hauck CR. Signaling by epithelial members of the CEACAM family—mucosal docking sites for pathogenic bacteria. Cell Commun Signal. 2014;12:27. 184. Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140:1720–1728. 185. Nguyen HT, Dalmasso G, Muller S, et al. Crohn’s disease-associated adherent invasive Escherichia coli modulate levels of microRNAs in intestinal epithelial cells to reduce autophagy. Gastroenterology. 2014;146:508–519. 186. Chassaing B, Koren O, Carvalho FA, et al. AIEC pathobiont instigates chronic colitis in susceptible hosts by altering microbiota composition. Gut. 2014;63:1069–1080. 187. Duck LW, Walter MR, Novak J, et al. Isolation of flagellated bacteria implicated in Crohn’s disease. Inflamm Bowel Dis. 2007;13:1191–1201. 188. Nickerson KP, McDonald C. Crohn’s disease-associated adherent-invasive Escherichia coli adhesion is enhanced by exposure to the ubiquitous dietary polysaccharide maltodextrin. PloS One. 2012;7:e52132. 189. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A. 2010;107:12204–12209. 190. Belkaid Y, Tarbell K. Regulatory T cells in the control of hostmicroorganism interactions. Annu Rev Immunol. 2009;27:551–589. 191. von Schillde MA, Hormannsperger G, Weiher M, et al. Lactocepin secreted by Lactobacillus exerts anti-inflammatory effects by selectively degrading proinflammatory chemokines. Cell Host Microbe. 2012;11:387–396. 192. Hormannsperger G, von Schillde MA, Haller D. Lactocepin as a protective microbial structure in the context of IBD. Gut Microbes. 2013;4:152–157. 193. Hotte NS, Salim SY, Tso RH, et al. Patients with inflammatory bowel disease exhibit dysregulated responses to microbial DNA. PloS One. 2012;7:e37932. 194. Gronbach K, Flade I, Holst O, et al. Endotoxicity of lipopolysaccharide as a determinant of T-cell-mediated colitis induction in mice. Gastroenterology. 2014;146:765–775. 195. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140:821–832. 196. Sebastian S, Hernandez V, Myrelid P, et al. Colorectal cancer in inflammatory bowel disease: results of the 3rd ECCO pathogenesis scientific workshop (I). J Crohns Colitis. 2014;8:5–18. 197. Anand PK, Malireddi RK, Lukens JR, et al. NLRP6 negatively regulates innate immunity and host defence against bacterial pathogens. Nature. 2012;488:389–393.

Inflamm Bowel Dis  Volume 21, Number 1, January 2015

198. Hu B, Elinav E, Huber S, et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc Natl Acad Sci U S A. 2013;110:9862–9867. 199. Rigottier-Gois L. Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis. ISME J. 2013;7:1256–1261. 200. Watson JD. Type 2 diabetes as a redox disease. Lancet. 2014;383:841–843. 201. Walujkar SA, Dhotre DP, Marathe NP, et al. Characterization of bacterial community shift in human Ulcerative Colitis patients revealed by Illumina based 16S rRNA gene amplicon sequencing. Gut Pathog. 2014;6:22. 202. Feller M, Huwiler K, Schoepfer A, et al. Long-term antibiotic treatment for Crohn’s disease: systematic review and meta-analysis of placebocontrolled trials. Clin Infect Dis. 2010;50:473–480. 203. Selby W, Pavli P, Crotty B, et al. Two-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn’s disease. Gastroenterology. 2007;132:2313–2319. 204. Thia KT, Mahadevan U, Feagan BG, et al. Ciprofloxacin or metronidazole for the treatment of perianal fistulas in patients with Crohn’s disease: a randomized, double-blind, placebo-controlled pilot study. Inflamm Bowel Dis. 2009;15:17–24. 205. Huang J, Liao C, Wu L, et al. A meta-analysis of randomized controlled trials comparing antibacterial therapy with placebo in Crohn’s disease. Colorectal Dis. 2011;13:617–626. 206. Perez-Cobas AE, Gosalbes MJ, Friedrichs A, et al. Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut. 2013;62: 1591–1601. 207. Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. Am J Gastroenterol. 2010;105:2687–2692. 208. Hviid A, Svanstrom H, Frisch M. Antibiotic use and inflammatory bowel diseases in childhood. Gut. 2011;60:49–54. 209. Virta L, Auvinen A, Helenius H, et al. 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;175:775–784. 210. Mack DR. Probiotics in inflammatory bowel diseases and associated conditions. Nutrients. 2011;3:245–264. 211. Sartor RB. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology. 2004;126:1620–1633. 212. Hedin CR, Mullard M, Sharratt E, et al. Probiotic and prebiotic use in patients with inflammatory bowel disease: a case-control study. Inflamm Bowel Dis. 2010;16:2099–2108. 213. Agathou CL, Beales IL. Factors associated with the use of probiotics in patients with inflammatory bowel disease. F1000Res. 2013;2:69. 214. Bibiloni R, Fedorak RN, Tannock GW, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100:1539–1546. 215. Miele E, Pascarella F, Giannetti E, et al. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol. 2009;104:437–443. 216. Jonkers D, Penders J, Masclee A, et al. Probiotics in the management of inflammatory bowel disease: a systematic review of intervention studies in adult patients. Drugs. 2012;72:803–823. 217. Persborn M, Gerritsen J, Wallon C, et al. The effects of probiotics on barrier function and mucosal pouch microbiota during maintenance treatment for severe pouchitis in patients with ulcerative colitis. Aliment Pharmacol Ther. 2013;38:772–783. 218. Duncan SH, Hold GL, Barcenilla A, et al. Roseburia intestinalis sp. nov., a novel saccharolytic, butyrate-producing bacterium from human faeces. Int J Syst Evol Microbiol. 2002;52:1615–1620. 219. Watanabe Y, Nagai F, Morotomi M. Characterization of Phascolarctobacterium succinatutens sp. nov., an asaccharolytic, succinate-utilizing bacterium isolated from human feces. Appl Environ Microbiol. 2012;78:511–518. 220. Nagano Y, Itoh K, Honda K. The induction of Treg cells by gutindigenous Clostridium. Curr Opin Immunol. 2012;24:392–397. 221. Borthakur A, Bhattacharyya S, Kumar A, et al. Lactobacillus acidophilus alleviates platelet-activating factor-induced inflammatory responses in human intestinal epithelial cells. PloS One. 2013;8:e75664. 222. Bernardo D, Sanchez B, Al-Hassi HO, et al. Microbiota/host crosstalk biomarkers: regulatory response of human intestinal dendritic cells exposed to Lactobacillus extracellular encrypted peptide. PloS One. 2012;7:e36262.

Functional Impacts of the Intestinal Microbiome

223. Angot A, Vergunst A, Genin S, et al. Exploitation of eukaryotic ubiquitin signaling pathways by effectors translocated by bacterial type III and type IV secretion systems. PLoS Pathogens. 2007;3:e3. 224. Hetzenecker AM, Seidl MC, Kosovac K, et al. Downregulation of the ubiquitin-proteasome system in normal colonic macrophages and reinduction in inflammatory bowel disease. Digestion. 2012;86:34–47. 225. Lodes MJ, Cong Y, Elson CO, et al. Bacterial flagellin is a dominant antigen in Crohn disease. J Clin Invest. 2004;113:1296–1306. 226. Vijay-Kumar M, Gewirtz AT. Role of flagellin in Crohn’s disease: emblematic of the progress and enigmas in understanding inflammatory bowel disease. Inflamm Bowel Dis. 2009;15:789–795. 227. Steck N, Hoffmann M, Sava IG, et al. Enterococcus faecalis metalloprotease compromises epithelial barrier and contributes to intestinal inflammation. Gastroenterology. 2011;141:959–971. 228. Hashimoto T, Perlot T, Rehman A, et al. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature. 2012; 487:477–481. 229. Fox JG, Rogers AB, Whary MT, et al. Gastroenteritis in NF-kappaBdeficient mice is produced with wild-type Camplyobacter jejuni but not with C. jejuni lacking cytolethal distending toxin despite persistent colonization with both strains. Infect Immun. 2004;72:1116–1125. 230. Pratt JS, Sachen KL, Wood HD, et al. Modulation of host immune responses by the cytolethal distending toxin of Helicobacter hepaticus. Infect Immun. 2006;74:4496–4504. 231. Ge Z, Rogers AB, Feng Y, et al. Bacterial cytolethal distending toxin promotes the development of dysplasia in a model of microbially induced hepatocarcinogenesis. Cell Microbiol. 2007;9:2070–2080. 232. Atterbury RJ, Hobley L, Till R, et al. Effects of orally administered Bdellovibrio bacteriovorus on the well-being and Salmonella colonization of young chicks. Appl Environ Microbiol. 2011;77:5794–5803. 233. Roberfroid M. Prebiotics: the concept revisited. J Nutr. 2007;137: 830S–837S. 234. Guarner F. Prebiotics in inflammatory bowel diseases. Br J Nutr. 2007; 98(suppl 1):S85–S89. 235. De Preter V, Joossens M, Ballet V, et al. Metabolic profiling of the impact of oligofructose-enriched inulin in Crohn’s disease patients: a doubleblinded randomized controlled trial. Clin Transl Gastroenterol. 2013;4:e30. 236. Arribas B, Suarez-Pereira E, Ortiz Mellet C, et al. Di-D-fructose dianhydride-enriched caramels: effect on colon microbiota, inflammation, and tissue damage in trinitrobenzenesulfonic acid-induced colitic rats. J Agric Food Chem. 2010;58:6476–6484. 237. Nishimura T, Andoh A, Hashimoto T, et al. Cellobiose prevents the development of dextran sulfate sodium (DSS)-Induced experimental colitis. J Clin Biochem Nutr. 2010;46:105–110. 238. Komiyama Y, Andoh A, Fujiwara D, et al. New prebiotics from rice bran ameliorate inflammation in murine colitis models through the modulation of intestinal homeostasis and the mucosal immune system. Scand J Gastroenterol. 2011;46:40–52. 239. Whelan K. Mechanisms and effectiveness of prebiotics in modifying the gastrointestinal microbiota for the management of digestive disorders. Proc Nutr Soc. 2013;72:288–298. 240. Lee J, Allen R, Ashley S, et al. British Dietetic Association evidencebased guidelines for the dietary management of Crohn’s disease in adults. J Hum Nutr Diet. 2014;27:207–218. 241. Nagy-Szakal D, Hollister EB, Luna RA, et al. Cellulose supplementation early in life ameliorates colitis in adult mice. PloS One. 2013;8:e56685. 242. Wada S, Sato K, Ohta R, et al. Ingestion of low dose pyroglutamyl leucine improves dextran sulfate sodium-induced colitis and intestinal microbiota in mice. J Agric Food Chem. 2013;61:8807–8813. 243. Mir SA, Nagy-Szakal D, Dowd SE, et al. Prenatal methyl-donor supplementation augments colitis in young adult mice. PloS One. 2013;8: e73162. 244. Schaible TD, Harris RA, Dowd SE, et al. Maternal methyl-donor supplementation induces prolonged murine offspring colitis susceptibility in association with mucosal epigenetic and microbiomic changes. Hum Mol Genet. 2011;20:1687–1696. 245. Smith MI, Yatsunenko T, Manary MJ, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science. 2013;339:548–554. 246. Olendzki BC, Silverstein TD, Persuitte GM, et al. An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report. Nutr J. 2014;13:5.

www.ibdjournal.org |

153

Functional impacts of the intestinal microbiome in the pathogenesis of inflammatory bowel disease.

: The human intestinal microbiome plays a critical role in human health and disease, including the pathogenesis of inflammatory bowel disease (IBD). N...
1MB Sizes 0 Downloads 5 Views