11
THE GUT MICROBIOME AND DISEASE The Microbiome in Inammatory Bowel Disease: Current Status and the Future Ahead 1 Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge; and 2 Gastrointestinal Unit, Center for the Study of Inammatory Bowel Disease, and Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts Studies of the roles of microbial communities in the development of inammatory bowel disease (IBD) have reached an important milestone. A decade of genome-wide association studies and other genetic analyses have linked IBD with loci that implicate an aberrant immune response to the intestinal microbiota. More recently, proling studies of the intestinal microbiome have associated the pathogenesis of IBD with characteristic shifts in the composition of the intestinal microbiota, reinforcing the view that IBD results from altered interactions between intestinal microbes and the mucosal immune system. Enhanced technologies can increase our understanding of the interactions between the host and its resident micro- biota and their respective roles in IBD from both a large- scale pathway view and at the metabolic level. We review important microbiome studies of patients with IBD and describe what we have learned about the mechanisms of intestinal microbiota dysfunction. We describe the recent progress in microbiome research from exploratory 16S- based studies, reporting associations of specic organisms with a disease, to more recent studies that have taken a more nuanced view, addressing the function of the micro- biota by metagenomic and metabolomic methods. Finally, we propose study designs and methodologies for future investigations of the microbiome in patients with inam- matory gut and autoimmune diseases in general. Keywords: Microbiota; Crohns Disease; Ulcerative Colitis; Metagenomics. O ver the past decade, inammatory bowel disease (IBD) has emerged as one of the most studied hu- man conditions linked to the gut microbiota. 1,2 IBD com- prises both Crohns disease (CD) and ulcerative colitis (UC), which together affect more than 3.6 million people. 3 Large- scale studies of human genetics across a total of 75,000 cases and controls have revealed 163 host susceptibility loci to date. 4 These loci are enriched for pathways that interact with environmental factors to modulate intestinal homeo- stasis. 5 The incidence of the disease has been on the rise over the past few decades, further highlighting the role of environmental factors in this disease. IBD was once a very rare disorder and only began to rise dramatically in inci- dence in the second half of the 20th century in North America and Europe, at times doubling every decade; it has expanded into developing countries in the past 2 decades, although there are more cases of UC than CD in the devel- oping world. 6 In addition, several twin studies have shown that the concordance rate for IBD between monozygotic twin pairs is signicantly less than 50%, with the least concordance in CD. 7 IBD is thus a multifaceted disorder in which not only germline genetics and the immune system but also several environmental factors play an important role. 8 One such factor, the gut microbial community, is gaining increasing attention for its inuence on many as- pects of health in general 9 and IBD in particular (Table 1). The gut microbiota, the largest reservoir of microbes in the body, coexists with its host in variable concentrations throughout the gastrointestinal tract, reaching an upper level in the colon of 10 11 or 10 12 cells/g of luminal contents. 10 This community performs a range of useful functions for the host, including digesting substrates inaccessible to host enzymes, educating the immune system, and repressing the growth of harmful microorganisms. 11 The extensive use of low- resolution surveys of the microbial community structure in the past, as well as renewed efforts using next-generation Aleksandar D. Kostic 1 Ramnik J. Xavier 1,2 Dirk Gevers 1 Abbreviations used in this paper: CD, Crohns disease; FMT, fecal microbiota transplantation; IBD, inammatory bowel disease; iCD, ileal Crohns disease; SCFA, short-chain fatty acid; T2DM, type 2 diabetes mellitus; UC, ulcerative colitis. © 2014 by the AGA Institute 0016-5085/$36.00 http://dx.doi.org/10.1053/j.gastro.2014.02.009 Gastroenterology 2014;146:14891499 THE GUT MICROBIOME AND DISEASE

Microbiota & ibd gastroenterology 2014

Embed Size (px)

DESCRIPTION

 

Citation preview

Page 1: Microbiota & ibd gastroenterology 2014

Gastroenterology 2014;146:1489–1499

THE GUT MICROBIOME AND DISEASE

The Microbiome in Inflammatory Bowel Disease: Current Statusand the Future Ahead

1 1,2 1 THEGU

TMICRO

BIOM

EAN

DDISE

ASE

1Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge; and 2Gastrointestinal Unit,Center for the Study of Inflammatory Bowel Disease, and Center for Computational and Integrative Biology, MassachusettsGeneral Hospital, Harvard Medical School, Boston, Massachusetts

Aleksandar D. Kostic Ramnik J. Xavier Dirk Gevers

Studies of the roles of microbial communities in thedevelopment of inflammatory bowel disease (IBD) havereached an important milestone. A decade of genome-wideassociation studies and other genetic analyses have linkedIBD with loci that implicate an aberrant immune responseto the intestinal microbiota. More recently, profilingstudies of the intestinal microbiome have associated thepathogenesis of IBD with characteristic shifts in thecomposition of the intestinal microbiota, reinforcing theview that IBD results from altered interactions betweenintestinal microbes and the mucosal immune system.Enhanced technologies can increase our understanding ofthe interactions between the host and its resident micro-biota and their respective roles in IBD from both a large-scale pathway view and at the metabolic level. We reviewimportant microbiome studies of patients with IBD anddescribe what we have learned about the mechanisms ofintestinal microbiota dysfunction. We describe the recentprogress in microbiome research from exploratory 16S-based studies, reporting associations of specific organismswith a disease, to more recent studies that have taken amore nuanced view, addressing the function of the micro-biota by metagenomic and metabolomic methods. Finally,we propose study designs and methodologies for futureinvestigations of the microbiome in patients with inflam-matory gut and autoimmune diseases in general.

Abbreviations used in this paper: CD, Crohn’s disease; FMT, fecal

Keywords: Microbiota; Crohn’s Disease; Ulcerative Colitis;Metagenomics.

ver the past decade, inflammatory bowel disease

microbiota transplantation; IBD, inflammatory bowel disease; iCD, ilealCrohn’s disease; SCFA, short-chain fatty acid; T2DM, type 2 diabetesmellitus; UC, ulcerative colitis.

© 2014 by the AGA Institute0016-5085/$36.00

http://dx.doi.org/10.1053/j.gastro.2014.02.009

O(IBD) has emerged as one of the most studied hu-man conditions linked to the gut microbiota.1,2 IBD com-prises both Crohn’s disease (CD) and ulcerative colitis (UC),which together affect more than 3.6 million people.3 Large-scale studies of human genetics across a total of 75,000cases and controls have revealed 163 host susceptibility loci

to date.4 These loci are enriched for pathways that interactwith environmental factors to modulate intestinal homeo-stasis.5 The incidence of the disease has been on the riseover the past few decades, further highlighting the role ofenvironmental factors in this disease. IBD was once a veryrare disorder and only began to rise dramatically in inci-dence in the second half of the 20th century in NorthAmerica and Europe, at times doubling every decade; it hasexpanded into developing countries in the past 2 decades,although there are more cases of UC than CD in the devel-oping world.6 In addition, several twin studies have shownthat the concordance rate for IBD between monozygotictwin pairs is significantly less than 50%, with the leastconcordance in CD.7 IBD is thus a multifaceted disorder inwhich not only germline genetics and the immune systembut also several environmental factors play an importantrole.8 One such factor, the gut microbial community, isgaining increasing attention for its influence on many as-pects of health in general9 and IBD in particular (Table 1).

The gut microbiota, the largest reservoir of microbes inthe body, coexists with its host in variable concentrationsthroughout the gastrointestinal tract, reaching an upper levelin the colon of 1011 or 1012 cells/g of luminal contents.10 Thiscommunity performs a range of useful functions for the host,including digesting substrates inaccessible to host enzymes,educating the immune system, and repressing the growthof harmful microorganisms.11 The extensive use of low-resolution surveys of the microbial community structure inthe past, as well as renewed efforts using next-generation

Page 2: Microbiota & ibd gastroenterology 2014

Table 1.Changes in the Microbiome Linked to IBD

Microbial composition Decrease in a diversityDecrease in Bacteroides and FirmicutesIncrease in GammaproteobacteriaPresence of E coli, specifically adherent-

invasive E coliPresence of FusobacteriumDecrease in Clostridia, Ruminococcaceae,

Bifidobacterium, LactobacillusDecrease in F prausnitzii

Microbial function Decrease in SCFAs, butyrateDecrease in butanoate and propanoate

metabolismDecrease in amino acid biosynthesisIncrease in auxotrophyIncrease in amino acid transportIncrease in sulfate transportIncreased oxidative stressIncrease in type II secretion system,

secretion of toxins

1490 Kostic et al Gastroenterology Vol. 146, No. 6

THEGUT

MICROBIOM

EAND

DISEASE

sequencing for a high-resolution description of composition,function, and ecology,12,13 have improved our overall un-derstanding of the role of the gut microbiota in health, aprerequisite for the study of disease-related dysbiosis.Several factors can intervene with microbial gut communitycomposition, including genetics, diet, age, drug treatment,smoking, and potentially many more (Figure 1).14 The rela-tive importance of each of these factors is still unclear, butseveral of them are directly or indirectly linked to the diseasestate.

Figure 1. Factors affecting the stability and complexity of the gumicrobiome, including stability, resilience, and complexity, are inIn the healthy gut, these characteristics contribute to important ptraining of the immune system, and digestion of food to supplfactors are indicated to affect the microbiome throughout microbgenetics, diet, and medication, among others (marked in the grintroduce perturbations affecting the complexity and stability oFeatures of an imbalanced microbiome include, for example, anoxidative stress and inflammation and metabolite production.

Environmental Factors AffectingMicrobiome CompositionDiet

One of the most important environmental factorsaffecting microbial composition is dietary preference,which has been shown to determine microbiome compo-sition throughout mammalian evolution.15 Although nospecific diet has been shown to directly cause, prevent, ortreat IBD, it is important to take interactions betweennutrients and microbiota into account when studying therole of the microbiome in disease. Thus far, only limitedinformation on this topic has been gathered in humans,undoubtedly as a result of the challenge of setting up alarge-scale controlled diet study. Wu et al have shown thatlong-term dietary patterns affect the ratios of Bacteroides,Prevotella, and Firmicutes and that short-term changesmay not have major influences.16 In addition, Zimmer et alhave studied the impact of a strict vegan or vegetariandiet on the microbiota17 and found a significant reductionin Bacteroides species, Bifidobacterium species, and theEnterobacteriaceae, whereas total bacterial load remainedunaltered. Since the Enterobacteriaceae are among thetaxa that are consistently found to be increased in pa-tients with IBD (see the following text), it would be ofvalue to include both short-term and long-term dietarypatterns in future studies of the role of the microbiome inIBD. Given the complexity of dietary effects, includingsuch information will likely only be feasible in a largecohort study.18

t microbiome in health and disease. Key characteristics of thefluenced over time from infancy to adulthood and into old age.hysiological processes such as protection against pathogens,y energy and nutrients including vitamins and SCFAs. Manyiome development and even established assembly, includingay boxes at the top of the figure). Some of these factors canf the microbiome, potentially introducing microbial dysbiosis.increase in gram-negative bacteria linked to an environment of

Page 3: Microbiota & ibd gastroenterology 2014

May 2014 The Microbiome in Inflammatory Bowel Disease 1491

THEGU

TMICRO

BIOM

EAN

DDISE

ASE

AgeThere is an age-related variation in the distribution of

IBD phenotypes, with 3 distinct stages of onset. A peak ageof onset is typically 15 to 30 years of age, with late-onsetcases occurring closer to 60 years of age and early onsetoccurring at younger than 10 years of age. Noticeably, thelatter group has had a significant increase in incidence overthe past decade.19 These stages correspond to phases inwhich the gut microbiota alters its diversity and stability.20

Early life is marked by a microbiome of low complexity andlow stability, one that is more volatile, is affected by thebirth route, and fluctuates with events such as changes indiet (switch from breastfeeding to solid foods), illness, andpuberty.21 It takes until adulthood for the microbialassemblage to reach maximal stability and complexity, withimproved resilience toward perturbations.22 However,decreased stability has been observed in elderly subjects(60 years of age or older).23 Given these different charac-teristics of the microbiome at the 3 distinct stages of diseaseonset, a different role for the microbiome in disease initia-tion and progression should be considered.

IBD Genetics Point to an InterplayBetween the Immune System andMicrobiota in IBD

A potential link between genetics and themicrobiome haslong been suspected. The first identified CD susceptibilitygene was NOD2,24 which stimulates an immune reaction onrecognizing muramyl dipeptide, a cell wall peptidoglycanconstituent of gram-positive and gram-negative bacteria.NOD2 is expressed in Paneth cells, which are located pre-dominantly in the terminal ileum at the base of intestinalcrypts, and produce antimicrobial defensins.25 Therefore, itmay not be surprising that mutations in NOD2 can have sig-nificant effects on the composition of the microbial milieu.Indeed, patients with IBD carrying NOD2 mutations haveincreased numbers of mucosa-adherent bacteria2 anddecreased transcription of the anti-inflammatory cytokineinterleukin 10.26 Patients with IBD carrying NOD2 andATG16L1, an IBD susceptibility gene involved in autophagy,risk alleles have significant alterations in the structure oftheir gut microbiota, including decreased levels of Faecali-bacterium and increased levels of Escherichia.27 Subjectshomozygous for loss-of-function alleles for FUT2 are “non-secretors” who do not express ABO antigen on the gastroin-testinal mucosa and bodily secretions. Nonsecretors are atincreased risk for CD28 and exhibit substantial alterations inthe mucosa-associated microbiota.29 Host genetics may thusplay a strong role in the establishment and shaping of the gutmicrobiota; indeed, monozygotic twins share more similarmicrobiomes than nontwin siblings.30 In regard to the skin, arecent study in patients with primary immunodeficiencyshowed a bidirectional dialogue between the microbiomeand the host immune system. The skin of patients with pri-mary immunodeficiency has an altered population compo-sition compared with immunocompetent subjects, which inturn results in increased susceptibility to infection by altering

the immune response toward pathogens.31 Although thereare currently no genome-wide studies examining the in-teractions between common human genetic variation and thecomposition of the microbial ecosystem, such a study couldbe of great value.5

An Overview of Gut MicrobiomeStudies in IBD

Many IBD susceptibility loci suggest an impairedresponse to microbes in disease, but the causality of thisrelationship is unclear. The pathogenesis of IBD may resultfrom a dysregulation of the mucosal immune system drivinga pathogenic immune response against the commensal gutflora.32 Some studies have shown that the gut microbiota isan essential factor in driving inflammation in IBD1; indeed,short-term treatment with enterically coated antibioticsdramatically reduces intestinal inflammation33 and has beenshown to have some efficacy in IBD and particularly inpouchitis.34 Specifically, rifaximin has shown efficacy inrecent trials in CD.35 Additionally, patients with IBD showmucosal secretion of immunoglobulin G antibodies36 andmucosal T-cell responses against commensal microbiota.37

The dramatic improvements in DNA sequencing tech-nology and analysis over the past decade have set the stagefor investigations of the IBD microbiome. Many studies havefound structural imbalances, or dysbioses, that occur in IBDsince the initial report,38 and a broad pattern has begun toemerge that includes a reduction in biodiversity, a decreasedrepresentation of several taxa within the Firmicutes phylum,and an increase in the Gammaproteobacteria.27,39

Many studies consistently report a decrease in biodi-versity, known as a diversity or species richness inecological terms, which is a measure of the total number ofspecies in a community. There is reduced a diversity in thefecal microbiome in patients with CD compared withhealthy controls,40 which was also found in pairs of mono-zygotic twins discordant for CD.41 This decreased diversityhas been attributed to reduced diversity specifically withinthe Firmicutes phylum42 and has also been associated withtemporal instability in the dominant taxa in both UC andCD.43 There is reduced diversity in inflamed versus non-inflamed tissues even within the same patient, and patientswith CD have lower overall bacterial loads at inflamed re-gions.44 The largest IBD-related microbiome study to date ison new-onset CD in a multicenter pediatric cohort.45 Thisstudy analyzed more than 1000 treatment-naïve samples,which were collected from multiple concurrent gastroin-testinal locations, from patients representing the variety ofdisease phenotypes with respect to location, severity, andbehavior. In addition to a detailed characterization of thespecific organisms either lost or associated with disease,this study indicates that assessing the rectal mucosa–associated microbiome offers unique potential for conve-nient and early diagnosis of CD.

Other nonbacterial members of the microbiota, namelythe fungi, viruses, archaea, and phage, may have a significantrole in gastrointestinal disease46; however, the vast majorityof recent studies of the microbiota are based on 16S

Page 4: Microbiota & ibd gastroenterology 2014

1492 Kostic et al Gastroenterology Vol. 146, No. 6

THEGUT

MICROBIOM

EAND

DISEASE

sequencing, thus largely ignoring these groups of organisms.For example, norovirus infection, in the context of an intactgut microflora and mutated Atg16l1, is required for thedevelopment of CD in a mouse model.47 A number of studiesnote a relationship between fungi and IBD,48 including anoverall increase in fungal diversity in UC and CD.49 Therelationship between these organisms and IBD will no doubtbe explored in more detail in the coming years, becausemicrobiome studies will increasingly be performed by un-biased shotgun sequencing.

Microbes Enriched in IBD MayPotentiate Disease

Specific taxonomic shifts have been reported in IBD(Table 1). The Enterobacteriaceae are increased in relativeabundance both in patients with IBD and in mousemodels.50 Escherichia coli, particularly adherent-invasiveE coli strains, have been isolated from ileal CD (iCD) bi-opsy specimens in culture-based studies51 and are enrichedin patients with UC.52 This enrichment is more pronouncedin mucosal samples than in fecal samples.53 The increase inEnterobacteriaceae may indicate the preference of this cladefor an inflammatory environment. In fact, treatment withmesalamine, an anti-inflammatory drug used in patientswith IBD, decreases intestinal inflammation and is associ-ated with a decrease in Escherichia/Shigella.39,54

In addition to trends seen in the lumen, a number ofstudies have observed a shift in microbes that are attachedto the intestinal mucus layer. The small intestine has asingle mucus layer, whereas the colon has 2 mucus layers: afirmly attached inner mucus layer that is essentially sterileand an outer mucus layer of variable thickness.55 The mucuslayer consists of mucins, trefoil peptides, and secretoryimmunoglobulin A.56 Although host-microbiota interactionsare bidirectional, direct contact with the epithelium islimited by the mucus and the production of antimicrobialfactors such as defensins and RegIII-g.57–59 As long as themucus layer is relatively healthy and intact, microbes willattach to the mucus and generally do not have direct accessto epithelial cells. There is a greater overall density ofattached bacteria on the colonic mucus layer in patientswith UC compared with healthy controls.2 The adherent-invasive E coli pathovar, in particular, is at higher abun-dance in mucosal biopsy specimens from patients with CDcompared with healthy subjects and particularly high in ilealspecimens.60 Adherent-invasive E coli invades epithelialcells and can replicate within macrophages61 and inducegranuloma formation in vitro.62 In fact, E coli has also beenfound at higher levels in granulomas from CD relative toother non-CD granulomas.63

A second group of adherent and invasive bacteria is theFusobacteria. The genus Fusobacterium is a group of gram-negative anaerobes that principally colonize the oral cavitybut can also inhabit the gut. Fusobacterium species havebeen found to be at higher abundance in the colonic mucosaof patients with UC relative to control subjects,64,65 andhuman isolates of Fusobacterium varium have been shownto induce colonic mucosal erosion in mice by rectal enema.66

The invasive ability of human Fusobacterium isolates has apositive correlation with the IBD status of the host,67 sug-gesting that invasive Fusobacterium species may influenceIBD pathology. Intriguingly, Fusobacterium species haverecently been shown to be enriched in tumor vs nonin-volved adjacent tissue in colorectal cancer,68,69 and humanFusobacterium isolates have been shown to directly pro-mote tumorigenesis in a mouse model.70,71 Because IBD isamong the highest risk factors for the development ofcolorectal cancer, Fusobacterium species may represent apotential link between these diseases.

Protective Effects of Microbes in IBDSeveral lines of evidence suggest that specific groups of

gut bacteria may have protective effects against IBD. Forexample, the colitis phenotype after treatment with dextransulfate sodium is more severe in mice that are reared germ-free compared with conventionally reared mice.72 Onemechanism by which the commensal microbiota may protectthe host is colonization resistance, in which commensalsoccupy niches within the host and prevent colonization bypathogens73 and help outcompete pathogenic bacteria.74

(Interestingly, the microbiota can sometimes take on theopposite role and facilitate viral infection.75) Commensalmicrobiota can also have direct functional effects on potentialpathogens (eg, in dampening virulence-related gene expres-sion).76 In addition, the gut microbiota plays a role in shapingthe mucosal immune system. Bacteroides and Clostridiumspecies have been shown to induce the expansion of regula-tory T cells, reducing intestinal inflammation.77 Other mem-bers of the microbiota can attenuate mucosal inflammationby regulating NF-kB activation.78

A number of bacterial species, most notably the Bifido-bacterium, Lactobacillus, and Faecalibacterium genera, mayprotect the host from mucosal inflammation by severalmechanisms, including the down-regulation of inflammatorycytokines79 or stimulation of interleukin 10,80 an anti-inflammatory cytokine. Faecalibacterium prausnitzii, onesuch proposed member of the microbiota with anti-inflammatory properties, is underrepresented in IBD.81

F prausnitzii is depleted in iCD biopsy samples concomi-tant with an increase in E coli abundance,82 and low levels ofmucosa-associated F prausnitzii are associated with a higherrisk of recurrent CD after surgery.80 Conversely, recovery ofF prausnitzii after relapse is associated with maintenance ofclinical remission of UC.83

Several constituents of the gut microbiota fermentdietary fiber, a prebiotic, to produce short-chain fatty acids(SCFAs), which include acetate, propionate, and butyrate.SCFAs are the primary energy source for colonic epithelialcells84 and have recently been shown to induce the expan-sion of colonic regulatory T cells.77,85 The Ruminococcaceae,particularly the butyrate-producing genus Faecalibacte-rium,86 is decreased in IBD, especially in iCD.38,39,42,80,82

Other SCFA-producing bacteria, including Odoribacter andthe Leuconostocaceae, are reduced in UC, and Phasco-larctobacterium and Roseburia are reduced in CD.39 Inter-estingly, the Ruminococcaceae consume hydrogen and

Page 5: Microbiota & ibd gastroenterology 2014

May 2014 The Microbiome in Inflammatory Bowel Disease 1493

THEGU

TMICRO

BIOM

EAN

DDISE

ASE

produce acetate that can be used by Roseburia to producebutyrate,39 and it is therefore consistent that both groupstogether are reduced in IBD.

Functional Composition of the GutMicrobiota in IBD

At the phylogenetic level, there is generally high vari-ability in the human microbiota between and within indi-vidual subjects over time.13 However, the functionalcomposition (ie, the functional potential of the gene contentof the metagenome) of the gut microbiota is strikinglystable.13 Metagenomic approaches may therefore providegreater insight into the function of the gut microbiota indisease than taxonomic profiling87,88; indeed, one such met-agenomics study of the IBD microbiome found that 12% ofmetabolic pathways were significantly different betweenpatients with IBD and healthy controls compared with just2% of genus-level clades.39 Metagenomic andmetaproteomicstudies have confirmed a decrease in butanoate and prop-anoate metabolism genes in iCD39 and lower overall levels ofbutyrate and other SCFAs in iCD,89 consistent with the de-creases in SCFA-producing Firmicutes clades seen in taxo-nomic profiling studies. Another metagenomic trend that hasbeen identified in the IBD microbiome is an increase infunctions characteristic of auxotrophic and pathobiont bac-teria, such as a decrease in biosynthesis of amino acids, andan increase in amino acid transporter genes.39 These bacteriagenerally have a reduced ability to produce their own nu-trients but rather transport them from the environmentbecause they are readily available at sites of inflammationand tissue destruction.39

A number of studies note an increase in sulfate-reducingbacteria, such as Desulfovibrio, in IBD.90 Mesalamine, acommon treatment for patients with IBD, inhibits the pro-duction of fecal sulfide; intriguingly, stool samples frompatients not treated with mesalamine show higher levels ofsulfide.91 Genes involved in the metabolism of the sulfur-containing amino acid cysteine are increased in IBD,particularly in iCD, and there is increased sulfate transportin both UC and CD.39 Saturated fat–derived taurine conju-gates to bile acids, increasing the availability of free sulfurand causing an expansion of the sulfate-reducing pathobiontBilophila wadsworthia, driving colitis in genetically suscep-tible Il10�/� but not wild-type mice.92 The IBD metagenomehas an increased propensity for managing oxidative stress, ahallmark of an inflammatory environment, as indicated byincreased glutathione transport and riboflavin metabolismin UC.39 There is also an increase in type II secretion sys-tems,39 which are involved in the secretion of toxins, and anincrease in bacterial genes with virulence-related func-tions89 in patients with CD, indicative of a shift toward aninflammation-promoting microbiome.

The Gut Microbiota inRelated Diseases

A number of parallels can be drawn between IBD andrelated metabolic diseases such as type 2 diabetes mellitus

(T2DM) and obesity. For example, there is an overalldecrease in diversity in obesity at both the phylogeneticlevel (ie, a reduced number of distinct species)30 and themetagenomic gene count level (ie, a reduced number ofdistinct genes).93 Major shifts in clade abundances include areduction of the Firmicutes and Clostridia in T2DM94 and asignificant increase in the Firmicutes to Bacteroidetes ratioin obesity in mice95; however, it is less clear whether thisshift also holds true in human obesity.30,96 There is adecrease in Bifidobacterium species in obesity and T2DM,96

and Bifidobacterium is reduced in children who becomeoverweight,97 suggesting that it may act as a predictivefactor. As in IBD, F prausnitzii is reduced in abundance inT2DM.98 In terms of gene function, there is an enrichment ofgenes involved in membrane transport,99 sulfate reduction,and oxidative stress resistance functions and a decrease infunctions involving cofactor and vitamin metabolism andbutyrate production.100 Therefore, many of the same shiftsin function of the gut microbiota, and even specific taxa, areseen across these diseases, suggesting the existence ofgeneralized features that relate these diseases and the se-lection for an auxotrophic microbiota that can thrive in aninflammatory environment.

IBD Treatments Affecting theMicrobiome

An array of antibiotics have been shown to lead to abloom of E coli.101 Because increased Enterobacteriaceae isa distinctive feature of intestinal inflammation and oxidativestress, the relationship between microbial composition,inflammation, and antibiotic use forms an important topicfor future research. In contrast, some promising data showthat antibiotic therapy specifically in IBD does induceremission or prevent relapse, but this topic will requirefurther controlled trials.102 To better understand the con-sequences of perturbing the gut microbiota of patients andthe role of the microbiota in treatment outcome, studies thatmonitor the temporal response at the levels of microbialecology and functional composition will be required.103

Thus far, several studies of healthy humans brieflyexposed to some antibiotics show the substantial pertur-bation, and the level of resilience, of the gut microbiota.104

Repeated exposures to a single antibiotic in healthy sub-jects result in cumulative and persistent changes to the gutmicrobial composition.105 Microbial homeostasis is typicallydisrupted by the loss of species complexity, particularly ofprotective microbes and thereby potentially resulting in anincreased risk of infections,106 or dysbiosis.45 Anothermechanism by which antibiotics lead to increased gut in-fections is by causing a thinning of the mucus layer, therebyweakening its barrier function.107

Instead of perturbing the existing microbiome byremoving diversity through antibiotics, repopulating the guthabitat with a healthy community has gained popularity inthe past few years. This will be an exciting new direction forthe pharmaceutical industry, expanding the focus beyondtraditional small molecules and biologics.108 The complexity

Page 6: Microbiota & ibd gastroenterology 2014

1494 Kostic et al Gastroenterology Vol. 146, No. 6

THEGUT

MICROBIOM

EAND

DISEASE

and composition that will be used to repopulate the gutcommunity will be very important. The success of probioticsin the management of IBD ranges from mixed results toconsiderable potential109 and is dependent on the strainsused and disease subtype targeted. In contrast, the evidencethat fecal microbiota transplantation (FMT) can be highlyeffective in replenishing our complex microbiota hasreceived considerable attention because of a convincingclinical trial of treatment of relapsing Clostridium difficileinfection.110 Related studies have shown that the use of awell-selected community subset rather than whole fecalcommunities can be sufficient for recovery.111 The highsuccess rate reported for relapsing C difficile infection haselevated FMT as an emerging treatment for several gastro-intestinal and metabolic disorders112 and is actively beingconsidered for IBD.113 So far, the sparse results reported forcases of IBD have been variable with regard to the successrate for inducing remission, and well-designed randomizedcontrolled trials are currently still lacking.114 Althoughchanges in the composition of the intestinal microbiota weresignificant, and reductions in Proteobacteria as well as anincrease in Bacteroides after FMT were observed, reachingor maintaining remission has been less frequent.115

Changing protocols toward repeated FMT procedures for

multiple days in a row seems to increase the chances ofachieving clinical remission.116

Future DirectionsStudies thus far have been able to address many aspects

of IBD, including genetics, immune responses, microbialdysbiosis, and microbial functional activity. However,because of the complexity of the human microbiome as adynamically interacting system, only limited data have beenproduced to bridge the gap between pathogenesis in a hu-man host, individual microbes, and alterations in microbialmetabolism and function. This suggests the need for a moremultifaceted approach to the microbiome in IBD (Figure 2).Because the gaps in IBD are arguably the narrowest amongdiseases in the microbiome field, as indicated by the prog-ress reviewed in the preceding text, it can be considered amodel for systems-level investigations of human-associatedmicrobial communities and their interactions with the host’simmune system. Increasing our mechanistic understandingof host-microbe interactions through such a systems-levelapproach will provide new opportunities to develop di-agnostics and treatments117 but is certainly not withoutchallenges.118 Recent technological advances, including

Figure 2. A multifacetedapproach to study the roleof the microbiome in IBD.Future microbiome studiesin the context of diseasewill shift toward multi-omics approaches tostudy host-microbe re-lations more comprehen-sively. Optimized samplecollection, detailed clinicalannotation, and sampleprocessing will be key toexpanding data generationfar beyond the typicalmarker gene and shotgunsequencing approaches. Anumber of assays on thehost side (red) and micro-bial end (blue) will gainincreasing attention goingforward.

Page 7: Microbiota & ibd gastroenterology 2014

May 2014 The Microbiome in Inflammatory Bowel Disease 1495

THEGU

TMICRO

BIOM

EAN

DDISE

ASE

improvements in sequencing and computational biology, arecontributing novel methods to study human-associated mi-crobial communities,119 with an increasing focus on func-tional follow-up using cultured microbes and germ-freeanimal models.

Essential to further explorations, more longitudinal sur-veys of patients before, during, and after treatment as well aslarge-scale clinical trials that take into account bothmicrobialand genetic heterogeneity will need to be performed andshould take a multifaceted approach. One approach to in-crease sample size is to combine different cohort studies.However, sample type,120 collection,121 and extraction122 canintroduce artifactual differences in microbial composition,which unfortunately makes it more challenging to combinedata sets produced under different protocols.123 Stream-lining experimental protocols across cohorts will be essentialto preserve statistical power for identifying true biologicaleffects in microbiome data sets. This will require growingefforts for standardizing the collection of patient samples andtheir clinical information. The discussion around the value ofuniform data collection has resulted in solutions already,124

but these need further adaptation for clinical information.Pursuing a true systems biology approach will require asolution for simultaneous measurement of the host state, themicrobiome, and the multidirectional signaling betweenthem. Although solutions for sequential isolation of metabo-lites, RNA, DNA, and proteins from the same unique samplehave been described,125 engineering a solution that can easilybe deployed as a self-sampling kit for patients will requirefurther exploration. Such challenges have been addressedwith other technologies, for example, in the use of micro-arrays as a tool for biomarker detection in clinical applica-tions, which led to the establishment of a consortium with amandate to set up standards and quality measures.126 Suchefforts are now also initiated in the microbiome space withboth environmental and clinical relevance (see www.hmpdacc.org, http://www.mbqc.org, and http://www.earthmicrobiome.org) and could eventually allow us tocombine several largewell-characterized cohorts without thechallenge of study-introduced biases.123

Despite promising correlations between shifts in micro-bial composition and disease phenotypes, to date no causa-tive role for the microbiome has been established, and ourunderstanding of the dynamic role of the human microbiomein IBD remains incomplete. The biological questions of in-terest enabled by prospective longitudinal studies would beto (1) identify the potential role of the intestinal microbiomein triggering disease, (2) determine if microbial compositionpredicts subsequent risk of activity flares, and (3) examinewhether the luminal flora predicts response to therapy. Theidentification of a correlative microbial pattern in humansthat induces antimicrobial defense and ameliorates inflam-mation would have considerable promise as a novel diag-nostic and therapeutic approach for themanagement of thesecomplex diseases. Existing correlative genetic-microbialstudies have helped to motivate this area but cannot speakto causality, response to treatment, or risk prediction in theabsence of multifaceted longitudinal measurements.

References

1.Sartor RB. Microbial influences in inflammatory boweldiseases. Gastroenterology 2008;134:577–594.

2.Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal florain inflammatory bowel disease. Gastroenterology 2002;122:44–54.

3. Loftus EV Jr. Clinical epidemiology of inflammatorybowel disease: incidence, prevalence, and environmentalinfluences. Gastroenterology 2004;126:1504–1517.

4. Jostins L, Ripke S, Weersma RK, et al. Host-microbeinteractions have shaped the genetic architecture of in-flammatory bowel disease. Nature 2012;491:119–124.

5.KhorB,Gardet A, Xavier RJ.Genetics andpathogenesis ofinflammatory bowel disease. Nature 2011;474:307–317.

6.Molodecky NA, Soon IS, Rabi DM, et al. Increasingincidence and prevalence of the inflammatory boweldiseases with time, based on systematic review.Gastroenterology 2012;142:46–54.e42; quiz e30.

7.Halme L, Paavola-Sakki P, Turunen U, et al. Family andtwin studies in inflammatory bowel disease. World JGastroenterol 2006;12:3668–3672.

8.Pillai S. Rethinking mechanisms of autoimmune patho-genesis. J Autoimmun 2013;45:97–103.

9. Flint HJ, Scott KP, Louis P, et al. The role of the gutmicrobiota in nutrition and health. Nat Rev GastroenterolHepatol 2012;9:577–589.

10.Dave M, Higgins PD, Middha S, et al. The human gutmicrobiome: current knowledge, challenges, and futuredirections. Transl Res 2012;160:246–257.

11.O’Hara AM, Shanahan F. The gut flora as a forgottenorgan. EMBO Rep 2006;7:688–693.

12.Qin J, Li R, Raes J, et al. A human gut microbial genecatalogue established by metagenomic sequencing.Nature 2010;464:59–65.

13.Human Microbiome Project Consortium. Structure,function and diversity of the healthy human microbiome.Nature 2012;486:207–214.

14.Blaser MJ, Falkow S. What are the consequences of thedisappearing human microbiota? Nat Rev Microbiol2009;7:887–894.

15.Ley RE, Hamady M, Lozupone C, et al. Evolution ofmammals and their gut microbes. Science 2008;320:1647–1651.

16.Wu GD, Chen J, Hoffmann C, et al. Linking long-termdietary patterns with gut microbial enterotypes. Science2011;334:105–108.

17.Zimmer J, Lange B, Frick JS, et al. A vegan or vegetariandiet substantially alters the human colonic faecal micro-biota. Eur J Clin Nutr 2012;66:53–60.

18.Moschen AR, Wieser V, Tilg H. Dietary factors: major reg-ulators of the gut’s microbiota. Gut Liver 2012;6:411–416.

19.Martin-de-Carpi J, Rodriguez A, RamosE, et al. Increasingincidence of pediatric inflammatory bowel disease inSpain (1996-2009): the SPIRIT Registry. Inflamm BowelDis 2013;19:73–80.

20.Spor A, Koren O, Ley R. Unravelling the effects of theenvironment and host genotype on the gut microbiome.Nat Rev Microbiol 2011;9:279–290.

Page 8: Microbiota & ibd gastroenterology 2014

1496 Kostic et al Gastroenterology Vol. 146, No. 6

THEGUT

MICROBIOM

EAND

DISEASE

21.Dominguez-Bello MG, Blaser MJ, Ley RE, et al. Devel-opment of the human gastrointestinal microbiota andinsights from high-throughput sequencing. Gastroenter-ology 2011;140:1713–1719.

22.Lozupone CA, Stombaugh JI, Gordon JI, et al. Diversity,stability and resilience of the human gut microbiota.Nature 2012;489:220–230.

23.Claesson MJ, Cusack S, O’Sullivan O, et al. Composi-tion, variability, and temporal stability of the intestinalmicrobiota of the elderly. Proc Natl Acad Sci U S A 2011;108(suppl 1):4586–4591.

24.Ogura Y, Bonen DK, Inohara N, et al. A frameshift mu-tation in NOD2 associated with susceptibility to Crohn’sdisease. Nature 2001;411:603–606.

25.Stappenbeck TS, Hooper LV, Gordon JI. Developmentalregulation of intestinal angiogenesis by indigenous mi-crobes via Paneth cells. Proc Natl Acad Sci U S A 2002;99:15451–15455.

26.Philpott DJ, Girardin SE. Crohn’s disease-associatedNod2 mutants reduce IL10 transcription. Nat Immunol2009;10:455–457.

27.Frank DN, Robertson CE, Hamm CM, et al. Diseasephenotype and genotype are associated with shifts inintestinal-associated microbiota in inflammatory boweldiseases. Inflamm Bowel Dis 2011;17:179–184.

28.McGovern DP, Jones MR, Taylor KD, et al. Fucosyl-transferase 2 (FUT2) non-secretor status is associatedwith Crohn’s disease. Hum Mol Genet 2010;19:3468–3476.

29.Rausch P, Rehman A, Kunzel S, et al. Colonic mucosa-associated microbiota is influenced by an interaction ofCrohn disease and FUT2 (Secretor) genotype. Proc NatlAcad Sci U S A 2011;108:19030–19035.

30.Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A coregut microbiome in obese and lean twins. Nature 2009;457:480–484.

31.Smeekens SP, Huttenhower C, Riza A, et al. Skinmicrobiome imbalance in patients with STAT1/STAT3defects impairs innate host defense responses. J InnateImmun 2013 Jun 22 [Epub ahead of print].

32.Strober W, Fuss I, Mannon P. The fundamental basis ofinflammatory bowel disease. J Clin Invest 2007;117:514–521.

33.Casellas F, Borruel N, Papo M, et al. Antiinflammatoryeffects of enterically coated amoxicillin-clavulanic acid inactive ulcerative colitis. Inflamm Bowel Dis 1998;4:1–5.

34.Sartor RB. Therapeutic manipulation of the entericmicroflora in inflammatory bowel diseases: antibiotics,probiotics, and prebiotics. Gastroenterology 2004;126:1620–1633.

35.Rietdijk ST, D’Haens GR. Recent developments in thetreatment of inflammatory bowel disease. J Dig Dis 2013;14:282–287.

36.Macpherson A, Khoo UY, Forgacs I, et al. Mucosal an-tibodies in inflammatory bowel disease are directedagainst intestinal bacteria. Gut 1996;38:365–375.

37.Pirzer U, Schonhaar A, Fleischer B, et al. Reactivity ofinfiltrating T lymphocytes with microbial antigens inCrohn’s disease. Lancet 1991;338:1238–1239.

38.Frank DN, St Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial communityimbalances in human inflammatory bowel diseases. ProcNatl Acad Sci U S A 2007;104:13780–13785.

39.Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of theintestinal microbiome in inflammatory bowel disease andtreatment. Genome Biol 2012;13:R79.

40.Manichanh C, Rigottier-Gois L, Bonnaud E, et al.Reduced diversity of faecal microbiota in Crohn’s dis-ease revealed by a metagenomic approach. Gut 2006;55:205–211.

41.Dicksved J, Halfvarson J, Rosenquist M, et al. Molecularanalysis of the gut microbiota of identical twins withCrohn’s disease. ISME J 2008;2:716–727.

42.Kang S, Denman SE, Morrison M, et al. Dysbiosis of fecalmicrobiota in Crohn’s disease patients as revealed by acustom phylogenetic microarray. Inflamm Bowel Dis2010;16:2034–2042.

43.Martinez C, Antolin M, Santos J, et al. Unstablecomposition of the fecal microbiota in ulcerative colitisduring clinical remission. Am J Gastroenterol 2008;103:643–648.

44.Sepehri S, Kotlowski R, Bernstein CN, et al. Microbialdiversity of inflamed and noninflamed gut biopsy tissuesin inflammatory bowel disease. Inflamm Bowel Dis 2007;13:675–683.

45.Gevers D, Kugathasan S, Denson LA, et al. The treat-ment-naïve microbiome in new-onset Crohn’s disease.Cell Host Microbe 2014;15:382–392.

46.Hunter P. The secret garden’s gardeners. Researchincreasingly appreciates the crucial role of gut virusesfor human health and disease. EMBO Rep 2013;14:683–685.

47.Cadwell K, Patel KK, Maloney NS, et al. Virus-plus-sus-ceptibility gene interaction determines Crohn’s diseasegene Atg16L1 phenotypes in intestine. Cell 2010;141:1135–1145.

48.Trojanowska D, Zwolinska-Wcislo M, Tokarczyk M, et al.The role of Candida in inflammatory bowel disease.Estimation of transmission of C. albicans fungi ingastrointestinal tract based on genetic affinity betweenstrains. Med Sci Monit 2010;16:CR451–CR457.

49.Ott SJ, Kuhbacher T, Musfeldt M, et al. Fungi and in-flammatory bowel diseases: alterations of compositionand diversity. Scand J Gastroenterol 2008;43:831–841.

50.Lupp C, Robertson ML, Wickham ME, et al. Host-medi-ated inflammation disrupts the intestinal microbiota andpromotes the overgrowth of Enterobacteriaceae. CellHost Microbe 2007;2:119–129.

51.Darfeuille-Michaud A, Boudeau J, Bulois P, et al. Highprevalence of adherent-invasive Escherichia coli asso-ciated with ileal mucosa in Crohn’s disease. Gastroen-terology 2004;127:412–421.

52.Sokol H, Lepage P, Seksik P, et al. Temperature gradientgel electrophoresis of fecal 16S rRNA reveals activeEscherichia coli in the microbiota of patients with ulcer-ative colitis. J Clin Microbiol 2006;44:3172–3177.

53.Chassaing B, Darfeuille-Michaud A. The commensalmicrobiota and enteropathogens in the pathogenesis of

Page 9: Microbiota & ibd gastroenterology 2014

May 2014 The Microbiome in Inflammatory Bowel Disease 1497

THEGU

TMICRO

BIOM

EAN

DDISE

ASE

inflammatory bowel diseases. Gastroenterology 2011;140:1720–1728.

54.Benjamin JL, Hedin CR, Koutsoumpas A, et al. Smokerswith active Crohn’s disease have a clinically relevantdysbiosis of the gastrointestinal microbiota. InflammBowel Dis 2012;18:1092–1100.

55.Johansson ME, Larsson JM, Hansson GC. The twomucus layers of colon are organized by the MUC2 mucin,whereas the outer layer is a legislator of host-microbialinteractions. Proc Natl Acad Sci U S A 2011;108(suppl 1):4659–6465.

56.Cario E. Microbiota and innate immunity in intestinalinflammation and neoplasia. Curr Opin Gastroenterol2013;29:85–91.

57.Shanahan F. The colonic microbiota in health and dis-ease. Curr Opin Gastroenterol 2013;29:49–54.

58.Vaishnava S, Yamamoto M, Severson KM, et al. Theantibacterial lectin RegIIIgamma promotes the spatialsegregation of microbiota and host in the intestine.Science 2011;334:255–258.

59.Hooper LV, Littman DR, Macpherson AJ. Interactionsbetween the microbiota and the immune system.Science 2012;336:1268–1273.

60.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 BowelDis 2009;15:872–882.

61.Glasser AL, Boudeau J, Barnich N, et al. Adherent inva-sive Escherichia coli strains from patients with Crohn’sdisease survive and replicate within macrophageswithout inducing host cell death. Infect Immun 2001;69:5529–5537.

62.Meconi S, Vercellone A, Levillain F, et al. Adherent-invasive Escherichia coli isolated from Crohn’s diseasepatients induce granulomas in vitro. Cell Microbiol 2007;9:1252–1261.

63.Ryan P, Kelly RG, Lee G, et al. Bacterial DNA withingranulomas of patients with Crohn’s disease—detectionby laser capture microdissection and PCR. Am J Gas-troenterol 2004;99:1539–1543.

64.Ohkusa T, Sato N, Ogihara T, et al. Fusobacterium var-ium localized in the colonic mucosa of patients with ul-cerative colitis stimulates species-specific antibody.J Gastroenterol Hepatol 2002;17:849–853.

65.OhkusaT,YoshidaT,SatoN, et al.Commensalbacteria canenter colonic epithelial cells and induce proinflammatorycytokine secretion: a possible pathogenic mechanism ofulcerative colitis. J Med Microbiol 2009;58:535–545.

66.Ohkusa T, Okayasu I, Ogihara T, et al. Induction ofexperimental ulcerative colitis by Fusobacterium variumisolated from colonic mucosa of patients with ulcerativecolitis. Gut 2003;52:79–83.

67.Strauss J, Kaplan GG, Beck PL, et al. Invasive potentialof gut mucosa-derived Fusobacterium nucleatum posi-tively correlates with IBD status of the host. InflammBowel Dis 2011;17:1971–1978.

68.Kostic AD, Gevers D, Pedamallu CS, et al. Genomicanalysis identifies association of Fusobacterium withcolorectal carcinoma. Genome Res 2012;22:292–298.

69.Castellarin M, Warren RL, Freeman DJ, et al. Fuso-bacterium nucleatum infection is prevalent in humancolorectal carcinoma. Genome Res 2012;22:299–306.

70.Rubinstein MR, Wang X, Liu W, et al. Fusobacteriumnucleatum promotes colorectal carcinogenesis bymodulating E-cadherin/beta-catenin signaling via itsFadA adhesin. Cell Host Microbe 2013;14:195–206.

71.Kostic AD, Chun E, Robertson L, et al. Fusobacteriumnucleatum potentiates intestinal tumorigenesis andmodulates the tumor-immune microenvironment. CellHost Microbe 2013;14:207–215.

72.Kitajima S, Morimoto M, Sagara E, et al. Dextran sodiumsulfate-induced colitis in germ-free IQI/Jic mice. ExpAnim 2001;50:387–395.

73.Callaway TR, Edrington TS, Anderson RC, et al. Pro-biotics, prebiotics and competitive exclusion for pro-phylaxis against bacterial disease. Anim Health Res Rev2008;9:217–225.

74.Kamada N, Chen G, Nunez G. A complex microworld inthe gut: harnessing pathogen-commensal relations. NatMed 2012;18:1190–1191.

75.Kane M, Case LK, Kopaskie K, et al. Successful trans-mission of a retrovirus depends on the commensalmicrobiota. Science 2011;334:245–249.

76.Medellin-Pena MJ, Wang H, Johnson R, et al. Probioticsaffect virulence-related gene expression in Escherichiacoli O157:H7. Appl EnvironMicrobiol 2007;73:4259–4267.

77.Atarashi K, Tanoue T, Oshima K, et al. Treg induction bya rationally selected mixture of Clostridia strains from thehuman microbiota. Nature 2013;500:232–236.

78.Kelly D, Campbell JI, King TP, et al. Commensal anaer-obic gut bacteria attenuate inflammation by regulatingnuclear-cytoplasmic shuttling of PPAR-gamma andRelA. Nat Immunol 2004;5:104–112.

79.Llopis M, Antolin M, Carol M, et al. Lactobacillus caseidownregulates commensals’ inflammatory signals inCrohn’s disease mucosa. Inflamm Bowel Dis 2009;15:275–283.

80.Sokol H, Pigneur B, Watterlot L, et al. Faecalibacteriumprausnitzii is an anti-inflammatory commensal bacteriumidentified by gut microbiota analysis of Crohn diseasepatients. Proc Natl Acad Sci U S A 2008;105:16731–16736.

81.Sokol H, Seksik P, Furet JP, et al. Low counts of Fae-calibacterium prausnitzii in colitis microbiota. InflammBowel Dis 2009;15:1183–1189.

82.Willing B, Halfvarson J, Dicksved J, et al. Twin studiesreveal specific imbalances in the mucosa-associatedmicrobiota of patients with ileal Crohn’s disease.Inflamm Bowel Dis 2009;15:653–660.

83.Varela E, Manichanh C, Gallart M, et al. Colonisation byFaecalibacterium prausnitzii and maintenance of clinicalremission in patients with ulcerative colitis. AlimentPharmacol Ther 2013;38:151–161.

84.Ahmad MS, Krishnan S, Ramakrishna BS, et al. Butyrateand glucose metabolism by colonocytes in experimentalcolitis in mice. Gut 2000;46:493–499.

85.Smith PM, Howitt MR, Panikov N, et al. The microbialmetabolites, short-chain fatty acids, regulate colonicTreg cell homeostasis. Science 2013;341:569–573.

Page 10: Microbiota & ibd gastroenterology 2014

1498 Kostic et al Gastroenterology Vol. 146, No. 6

THEGUT

MICROBIOM

EAND

DISEASE

86.Duncan SH, Hold GL, Barcenilla A, et al. Roseburiaintestinalis sp. nov., a novel saccharolytic, butyrate-producing bacterium from human faeces. Int J SystEvol Microbiol 2002;52:1615–1620.

87.Meyer F, Trimble WL, Chang EB, et al. Functional pre-dictions from inference and observation in sequence-based inflammatory bowel disease research. GenomeBiol 2012;13:169.

88.Presley LL, Ye J, Li X, et al. Host-microbe relationships ininflammatory bowel disease detected by bacterial andmetaproteomic analysis of the mucosal-luminal interface.Inflamm Bowel Dis 2012;18:409–417.

89.Erickson AR, Cantarel BL, Lamendella R, et al. Integratedmetagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS One2012;7:e49138.

90.Rowan F, Docherty NG, Murphy M, et al. Desulfovibriobacterial species are increased in ulcerative colitis. DisColon Rectum 2010;53:1530–1536.

91.Pitcher MC, Beatty ER, Cummings JH. The contributionof sulphate reducing bacteria and 5-aminosalicylic acidto faecal sulphide in patients with ulcerative colitis. Gut2000;46:64–72.

92.Devkota S, Wang Y, Musch MW, et al. Dietary-fat-induced taurocholic acid promotes pathobiont expan-sion and colitis in Il10-/- mice. Nature 2012;487:104–108.

93.Le Chatelier E, Nielsen T, Qin J, et al. Richness of humangut microbiome correlates with metabolic markers.Nature 2013;500:541–546.

94.Larsen N, Vogensen FK, van den Berg FW, et al. Gutmicrobiota in human adults with type 2 diabetes differsfrom non-diabetic adults. PLoS One 2010;5:e9085.

95.Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity forenergy harvest. Nature 2006;444:1027–1031.

96.Schwiertz A, Taras D, Schafer K, et al. Microbiota andSCFA in lean and overweight healthy subjects. Obesity(Silver Spring) 2010;18:190–195.

97.Kalliomaki M, Collado MC, Salminen S, et al. Early dif-ferences in fecal microbiota composition in children maypredict overweight. Am J Clin Nutr 2008;87:534–538.

98.Furet JP, Kong LC, Tap J, et al. Differential adaptation ofhuman gut microbiota to bariatric surgery-inducedweight loss: links with metabolic and low-grade inflam-mation markers. Diabetes 2010;59:3049–3057.

99.Greenblum S, Turnbaugh PJ, Borenstein E. Meta-genomic systems biology of the human gut microbiomereveals topological shifts associated with obesity andinflammatory bowel disease. Proc Natl Acad Sci U S A2012;109:594–599.

100.Qin J, Li Y, Cai Z, et al. A metagenome-wide associationstudy of gut microbiota in type 2 diabetes. Nature 2012;490:55–60.

101.Looft T, Allen HK. Collateral effects of antibiotics onmammalian gut microbiomes. Gut Microbes 2012;3:463–467.

102.Khan KJ, Ullman TA, Ford AC, et al. Antibiotic therapy ininflammatory bowel disease: a systematic review andmeta-analysis. Am J Gastroenterol 2011;106:661–673.

103.Lemon KP, Armitage GC, Relman DA, et al. Microbiota-targeted therapies: an ecological perspective. Sci TranslMed 2012;4:137rv5.

104.Relman DA. The human microbiome: ecosystem resil-ience and health. Nutr Rev 2012;70(suppl 1):S2–S9.

105.Dethlefsen L, Relman DA. Incomplete recovery andindividualized responses of the human distal gut micro-biota to repeated antibiotic perturbation. Proc Natl AcadSci U S A 2011;108(suppl 1):4554–4561.

106.Khosravi A, Mazmanian SK. Disruption of the gutmicrobiome as a risk factor for microbial infections. CurrOpin Microbiol 2013;16:221–227.

107.Wlodarska M, Willing B, Keeney KM, et al. Antibiotictreatment alters the colonic mucus layer and predisposesthe host to exacerbated Citrobacter rodentium-inducedcolitis. Infect Immun 2011;79:1536–1545.

108.Fischbach MA, Bluestone JA, Lim WA. Cell-based ther-apeutics: the next pillar of medicine. Sci Transl Med2013;5:179ps7.

109.Whelan K, Quigley EM. Probiotics in the management ofirritable bowel syndrome and inflammatory bowel dis-ease. Curr Opin Gastroenterol 2013;29:184–189.

110.van Nood E, Vrieze A, Nieuwdorp M, et al. Duodenalinfusion of donor feces for recurrent Clostridium difficile.N Engl J Med 2013;368:407–415.

111.Shahinas D, Silverman M, Sittler T, et al. Toward an un-derstanding of changes in diversity associated with fecalmicrobiome transplantation based on 16S rRNA genedeep sequencing. MBio 2012;3.

112.Smits LP, Bouter KE, de Vos WM, et al. Therapeuticpotential of fecal microbiota transplantation. Gastroen-terology 2013;145:946–953.

113.Damman CJ, Miller SI, Surawicz CM, et al. The micro-biome and inflammatory bowel disease: is there a ther-apeutic role for fecal microbiota transplantation? Am JGastroenterol 2012;107:1452–1459.

114.Anderson JL, Edney RJ, Whelan K. Systematic review:faecal microbiota transplantation in the management ofinflammatory bowel disease. Aliment Pharmacol Ther2012;36:503–516.

115.Kump PK, Grochenig HP, Lackner S, et al. Alteration ofintestinal dysbiosis by fecal microbiota transplantationdoes not induce remission in patients with chronicactive ulcerative colitis. Inflamm Bowel Dis 2013;19:2155–2165.

116.Kunde S, Pham A, Bonczyk S, et al. Safety, tolerability,and clinical response after fecal transplantation in chil-dren and young adults with ulcerative colitis. J PediatrGastroenterol Nutr 2013;56:597–601.

117.Olle B. Medicines from microbiota. Nat Biotechnol 2013;31:309–315.

118.Brown J, de Vos WM, DiStefano PS, et al. Translating thehuman microbiome. Nat Biotechnol 2013;31:304–308.

119.Gevers D, Pop M, Schloss PD, et al. Bioinformatics forthe Human Microbiome Project. PLoS Comput Biol 2012;8:e1002779.

120.Stearns JC, Lynch MD, Senadheera DB, et al. Bacterialbiogeography of the human digestive tract. Sci Rep2011;1:170.

Page 11: Microbiota & ibd gastroenterology 2014

May 2014 The Microbiome in Inflammatory Bowel Disease 1499

THEGU

TMICRO

BIOM

EAN

DDISE

ASE

121.Rubin BE, Gibbons SM, Kennedy S, et al. Investigatingthe impact of storage conditions on microbial commu-nity composition in soil samples. PLoS One 2013;8:e70460.

122.Momozawa Y, Deffontaine V, Louis E, et al. Charac-terization of bacteria in biopsies of colon and stools byhigh throughput sequencing of the V2 region of bac-terial 16S rRNA gene in human. PLoS One 2011;6:e16952.

123.Lozupone CA, Stombaugh J, Gonzalez A, et al. Meta-analyses of studies of the human microbiota. GenomeRes 2013;23:1704–1714.

124.Yilmaz P, Kottmann R, Field D, et al. Minimum informa-tion about a marker gene sequence (MIMARKS) andminimum information about any (x) sequence (MIxS)specifications. Nat Biotechnol 2011;29:415–420.

125.Roume H, Heintz-Buschart A, Muller EE, et al. Sequentialisolation of metabolites, RNA, DNA, and proteins from

the same unique sample. Methods Enzymol 2013;531:219–236.

126.Tillinghast GW. Microarrays in the clinic. Nat Biotechnol2010;28:810–812.

Received October 29, 2013. Accepted February 17, 2014.

Reprint requestsAddress requests for reprints to: Ramnik J. Xavier, MD, PhD, Center forComputational and Integrative Biology, Richard B. Simches ResearchCenter, Massachusetts General Hospital, 185 Cambridge Street, Boston,Massachusetts 02114. e-mail: [email protected]; fax: (617) 7246832.

Conflicts of interestThe authors disclose no conflicts.

FundingSupported by grants from the Crohn’s and Colitis Foundation of America (toR.J.X.) and National Institutes of Health grants U54 DE023798 and R01DK092405 (to R.J.X.).