Sunteți pe pagina 1din 11

CLINICAL GASTROENTEROLOGY AND HEPATOLOGY 2007;5:274 284

REVIEWS
Gut Microbiota: Mining for Therapeutic Potential

ANN M. OHARA* and FERGUS SHANAHAN*,


*Alimentary Pharmabiotic Centre, and Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland

The resident microbiota of the human intestine exerts a The 2005 Nobel prize awarded to Barry Marshall and Robin
conditioning effect on intestinal homeostasis, delivering Warren is a sobering reminder that the solution to some chronic
regulatory signals to the epithelium and instructing muco- disorders cannot be unraveled by exclusive investigation of the
sal immune responses. Pattern recognition receptors are host response. The host response must be considered in terms of
key mediators of innate host defense, and in healthy indi- bacterial residents and prokaryotic-eukaryotic interactions. Mod-
viduals, the mucosal immune system exhibits an exquisitely ern molecular techniques are facilitating our understanding of
regulated restrained response to the resident microbiota. microbe-microbe and host-microbe communications at mucosal
However, in genetically susceptible hosts, unrestrained mu- interfaces and are providing glimpses of tangible therapeutic strat-
cosal immune activation in response to local bacterial sig- egies. Therefore, the intent here is to present an overview of the
nals can contribute to the pathogenesis of inflammatory commensal microbiota, their interactions with the intestinal mu-
bowel disease. Manipulation of the microbiota to enhance cosa, and clinical relevance to IBD.
its beneficial components thus represents a potential ther-
apeutic strategy for inflammatory bowel disease. Moreover, Indigenous Gut Microbiota
the microbiota might be a rich repository of metabolites
that can be exploited for therapeutic benefit. Modern mo- Composition of the Normal Gut Microbiota
lecular techniques are facilitating improved understanding Dialogue between commensal bacteria and the host
of host-microbe dialogue in health and in several disease occurs primarily along mucosal surfaces, and the largest inter-
processes, including inflammatory bowel disease. It follows face is the human gastrointestinal mucosa. The intestine is
that elucidating the molecular mechanisms of host-micro- habitat to a dynamic and diverse bacterial community that is
bial interactions is now a prerequisite for a bugs to drugs separated from the internal milieu by only a single layer of
program of discovery. epithelial cells. Intestinal bacteria outnumber human somatic
and germ cells 10-fold12 and represent a combined microbiome
well in excess of the human genome. Co-evolution of the host

T he chronic inflammatory bowel diseases (IBDs), Crohns


disease and ulcerative colitis, are characterized by bouts of
uncontrolled, chronic mucosal inflammation, followed by re-
and indigenous microbes has fostered mutually beneficial and
cooperative interactions mediated by bidirectional host-micro-
biota exchange.
modeling processes that occur during periods of remission. Traditionally, studies of indigenous bacteria focused on the
Together, these result in tissue-damaging inflammatory re- characterization of fecal diversity. Detailed analysis was hin-
sponses in the intestine that might result in much personal dered by conventional microbiology techniques, and most bac-
suffering and impaired quality of life. Crohns disease and terial species still cannot be cultured. Modern molecular tech-
ulcerative colitis have a combined prevalence of 250/100,000 in niques such as broad-range sequencing of 16S ribosomal RNA
the Western world, and they represent a substantial economic (rRNA) from amplified bacterial nucleic acid extracted from
burden on health care resources. Although the precise etiology feces or biopsies indicate evolutionary divergence and can be
of IBD remains to be elucidated, a complex interaction of used to identify and classify bacteria. The availability of bacte-
environmental, genetic, and immunoregulatory factors contrib- rial sequence data has facilitated the development of molecular
utes to the initiation and perpetuation of the disease.1 probes for DNA microarrays, fluorescent in situ hybridization,
Under normal circumstances, the mucosal immune system is and gene chips that identify and enumerate specific microbial
exquisitely regulated and exhibits a restrained response to the species. These molecular approaches are being used to examine
resident microbiota while retaining an ability to mount appro- the individuality and temporal stability of the microbiota and
priate immune responses to pathogenic bacteria. However, sev-
eral lines of evidence indicate that genetically influenced dys-
regulation of the mucosal immune response to antigens of the Abbreviations used in this paper: AIEC, adherent-invasive Esche-
indigenous microbiota can contribute to the pathogenesis of richia coli; CARD, caspase recruitment domain; DC, dendritic cell; IBD,
IBD.2,3 In susceptible individuals, environmental triggers can inammatory bowel disease; IL, interleukin; IFN, interferon; MAP,
impact on the initiation or reactivation of disease, and tissue Mycobacterium avium subspecies paratuberculosis; MDP, muramyl
dipeptide; NF, nuclear factor; NOD, nucleotide-binding oligomerization
damage might result from immune cell misperception of dan-
domain; PPAR, peroxisome proliferator activated receptor; PRR, pat-
ger within the indigenous microbiota or from failure of normal
tern recognition receptor; rRNA, ribosomal RNA; Th1/Th2, T-helper cell
tolerance to enteric bacteria.2,4 Within the gastrointestinal tract, type 1/2; TLR, toll-like receptor; TNF, tumor necrosis factor.
the inflammatory capacity of commensal bacteria is varied. 2007 by the AGA Institute
Some resident bacteria are proinflammatory, whereas others 1542-3565/07/$32.00
attenuate inflammatory responses.511 doi:10.1016/j.cgh.2006.12.009
March 2007 THERAPEUTIC POTENTIAL OF THE GUT MICROBIOTA 275

Figure 1. Changes in intestinal structure and function in germ-free animals compared with colonized animals raised under conventional conditions.
Reconstitution of germ-free mice with a microbiota restores the mucosal immune system, and commensal bacteria exert numerous protective,
structural, and metabolic effects on the intestinal epithelium.

the impact of weaning, antibiotics, or dietary changes on its between breast-fed and formula-fed infants.21 These pioneer
composition. It is now realized that the mucosa-associated bacteria can modulate gene expression in the host to create a
community is significantly different from the luminal and fecal suitable environment for themselves and can prevent growth of
communities.13 Although every adult intestine harbors a par- other bacteria introduced later in the ecosystem.23,24 Increased
ticular combination of predominant species, the composition credence is being given to the hypothesis that the modern
of the intestinal microbiota might alter with lifestyle, diet, and sanitized environment of developed societies has altered the
age.14,15 Nonetheless, a comparative study of the microbiota of normal pattern of gut colonization during infancy. This might
human adults with varying degrees of genetic relatedness, in- result in a lack of tolerance to otherwise harmless food proteins
cluding monozygotic twins, emphasized the prevailing influ- and other antigens, including those of the intestinal micro-
ence of host genotype over diet in determining the microbial biota.25
composition of the gut.16
Acid, bile, and pancreatic secretions hinder the colonization Functions of the Normal Gut Microbiota
of the stomach and proximal small intestine by most bacteria. Enteric bacteria confer many benefits to intestinal phys-
However, bacterial density dramatically increases in the distal iology including protective, structural, and metabolic effects.
small intestine and in the large intestine increases to approxi- Their influence on intestinal structure and function has been
mately 10111012 bacteria per gram of colonic content, which demonstrated in comparative studies of germ-free and colo-
contribute to 60% of fecal mass.17,18 Although archaea and nized animals. Some of the differences between animals raised
eukarya are also represented,19 bacteria predominate, and co- under germ-free and conventional conditions are listed in Fig-
lon-residing bacteria achieve the highest cell densities recorded ure 1. Such comparisons, together with studies indicating that
for any ecosystem.20 The most common anaerobic genera in- reconstitution of gnotobiotic mice with a microbiota is suffi-
clude Bifidobacterium, Clostridium, Bacteroides, and Eubacterium, cient to restore the mucosal immune system,26 illustrate that
and aerobic Escherichia, Enterococcus, Streptococcus, and Klebsiella the microbiota provide regulatory signals that instruct intesti-
are also found. However, sequencing of 16S rRNA gene clone nal development and function. Indeed, colonization of germ-
libraries has indicated that a significant fraction of the bacteria free mice with a single species, Bacteroides thetaiotaomicron, af-
represent uncultivated species and novel microorganisms.13 fects the expression of a variety of host genes influencing
Mammalian fetuses are born germ-free, but immediately nutrient uptake, metabolism, angiogenesis, mucosal barrier
after birth, establishment of the resident microbiota is driven by function, and the development of the enteric nervous system.23
environmental factors such as mode of delivery, type of infant Furthermore, ligands from resident bacteria and commensal-
diet, hygiene levels, and medication.21,22 Enterobacteria and derived symbiosis factors influence the normal development
bifidobacteria species represent early colonizers, but differences and function of mucosal immunity.27,28 Indigenous bacteria
in gut microbial composition and incidence of infection occur educate the mucosal immune system and modulate the fine
276 OHARA AND SHANAHAN CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 5, No. 3

tuning of T-cell repertoires and T-helper cell type 1 (Th1)/Th2 antigen-presenting cells. Intestinal DCs themselves participate
cytokine profiles.29 Taken together, the composition of the in immune surveillance and can directly sample gut contents by
colonizing microbiota might influence individual variations in either entering or extending dendrites between surface entero-
immunity. cytes without disrupting tight junctions.46 DCs can ingest and
Along the epithelium, enteric bacteria form a natural defense retain live commensal bacteria and transit to the mesenteric
barrier against exogenous microbes. Colonization resistance lymph node where immune responses to these bacteria are
involves several mechanisms including displacement, competi- induced locally. This prevents access of commensal bacteria to
tion for nutrients and epithelial binding sites by the resident the internal milieu.47 In addition, enterocyte-derived mediators
microbiota, and production of antimicrobial factors such as such as thymic stromal lymphopoietin can induce noninflam-
lactic acids and bacteriocins.30 Commensal bacteria also help matory DCs, suggesting that physiologic interactions between
fortify the epithelial barrier by various mechanisms that include DCs and surface enterocytes contribute to intestinal homeosta-
the induction of the complement inhibitor decay-accelerating sis.48
factor or increasing barrier function.31,32 Exposure of colonic
epithelial cell lines to bacterial ligands also results in apical Mucosal Recognition of Bacteria
tightening and sealing of the tight junctional protein ZO-1 and
The ability of immunosensory cells to discriminate
increased transepithelial resistance.33
pathogen from commensal is mediated, in part, by 2 major host
The metabolic activity of the microbiota is equivalent to that pattern recognition receptor (PRR) systems. These are the fam-
of an organ within an organ.34 Through the production of short ily of toll-like receptors (TLRs) and the nucleotide-binding
chain fatty acids, resident microorganisms positively influence oligomerization domain/caspase recruitment domain (NOD/
the differentiation and proliferation of intestinal enterocytes.35 CARD) molecules.49 Several of the signal transduction path-
Resident bacteria can break down dietary carcinogens; synthe- ways induced by these receptors have been elucidated and their
size biotin, folate, and vitamin K; ferment nondigestible dietary effector responses characterized.49 53 Both TLRs and NOD pro-
residue, particularly carbohydrates and endogenous epithelial-
teins trigger innate and adaptive immune responses, including
derived mucus; and assist in the absorption of calcium, mag-
the synthesis of proinflammatory cytokines and chemokines,
nesium, and iron.36 38 Together, this complex metabolic activity
and TLR signaling also impacts on subsequent T-cell responses
recovers valuable energy and absorbable substrates for the host by activating DCs that overcome the suppressive effects of T
and provides energy and nutrients for bacterial growth and regulatory cells.54 Together, these PRRs play a fundamental role
proliferation. Colonization increases glucose uptake in the gut,
in immune cell activation in response to specific microbial-
and compared with colonized mice, germ-free mice require a
associated molecular patterns (Table 1).
greater caloric intake to sustain a normal body weight.39 This
TLRs and NOD proteins are expressed by surface enterocytes
implicates intestinal bacteria as modulators of fat deposition in and DCs,51 and in the gut, TLRs and NOD proteins appear to
the host. It has been proposed that an individuals gut micro-
be crucial for bacterial-host communication. Decreased entero-
biota has a specific metabolic efficiency, and differences in gut
cyte proliferation and levels of cytoprotective factors have been
microbial composition between individuals might regulate en-
observed in TLR-defective mice compared with wild-type
ergy storage and predispose to obesity.19 Therefore, exploration
mice.27 TLR signals mediated by commensal bacteria or their
of the metabolic activity of the microbiota offers strong poten- ligands are essential for intestinal barrier function and repair of
tial for exploitation of the microbiota as biomarkers for im- the gut.55 Many PRR ligands are expressed by commensal bac-
pending disease and the development of novel therapies.
teria, yet the healthy gut does not evoke inflammatory re-
sponses to these bacteria.11 PRRs participate in several recently
Host-Microbiota Dialogue at the Mucosal described host molecular immune mechanisms that mediate
Surface intestinal homeostasis (Table 2), and these are reviewed else-
Host defense demands precise interpretation of the where.56 In the healthy gut, regulatory T cells and tolerance-
microenvironment to distinguish commensal from pathogenic inducing DCs contribute to the control of excessive Th1 re-
microorganism and must entail exquisite regulation of re- sponses to the indigenous microbiota also.57,58 Phenotypic
sponses. Disruption of these processes might lead to inappro- alterations in regulatory T-cell populations have been observed
priate responses. Thus, although the microbiota are generally a in IBD patients,59 and several lines of evidence indicate that
health asset, they might become a liability in certain circum- peripheral tolerance, as well as local tolerance to commensal
stances. These include syndromes of bacterial overgrowth bacteria, is mediated by the suppressive effects of regulatory T
and/or translocation, metabolite-mediated conversion of pro- cells.59 61
carcinogens to carcinogens, and IBD.29,40,41 Commensal bacteria have been shown to suppress inflam-
Along the epithelium, active sampling of commensal bacte- matory responses and inhibit specific intracellular signal trans-
ria, pathogens, and other antigens is mediated by various types duction pathways, thereby contributing to the maintenance of
of immunosensory cells. These include surface enterocytes, M mucosal homeostasis. The transcription factor, nuclear factor
cells, and dendritic cells (DCs). Surface enterocytes are inter- (NF)-B, is a master coordinator of immune responses to
connected with tight junctions and overlain with mucus. They pathogenic bacteria and other stress signals. However, most
serve as afferent sensors of danger signals within the luminal commensal bacteria do not activate NF-B8,11; instead, some
microenvironment by secreting defensins, immunoglobulin A, commensal strains antagonize NF-B within epithelial cells by
chemokines, and cytokines that alert and direct innate and a variety of mechanisms. These include inhibition of epithelial
adaptive immune responses.42 45 M cells, specialized epithelial proteasome function or degradation of the NF-B counter-
cells, that overlie lymphoid follicles sample the environment regulatory factor IB- or by the nuclear export of the p65
and transport luminal antigens to subadjacent DCs and other subunit of NF-B in a peroxisome proliferator activated recep-
March 2007 THERAPEUTIC POTENTIAL OF THE GUT MICROBIOTA 277

Table 1. List of PRRs and Their Ligands


TLR Ligand Source

TLR1 Triacyl lipopeptides Bacteria & mycobacteria


TLR2 Lipoprotein Most bacteria
Peptidoglycan Gram-positive bacteria
Lipoteichoic acid Gram-negative bacteria
Lipoarabinomannan Mycobacteria
TLR3 Double-stranded RNA Viruses
TLR4 Lipopolysaccharide Gram-negative bacteria
TLR5 Flagellin Flagellated bacteria
TLR6 Diacyl lipopeptides Mycoplasma
Lipoteichoic acids Gram-positive bacteria
Zymosan Fungi
TLR7 Single-stranded RNA Viruses
TLR8 Single-stranded RNA Viruses
TLR9 Unmethylated CpG-containing DNA Bacteria & viruses
TLR10 Undetermined Undetermined
TLR11 Profilin-like ligands Protozoa
NOD
NOD1/CARD4 -D-glutamyl-meso-diamino-pimelic acid of peptidoglycan Gram-negative bacteria
NOD2/CARD15 MDPs of peptidoglycan Most bacteria

tor (PPAR)-dependent manner.7,8,62 Some commensal bacte- factors. The normal physiologic response to the indigenous
ria can attenuate colonic inflammation through TLR4 by ele- microbiota is one of immunologic quiescence. Deviations from
vating PPAR- expression in intestinal enterocytes and this and, in particular, genetically influenced aberrant immune
uncoupling NF-B dependent target genes in a negative feed- responses to luminal antigens can lead to the development of
back loop.8,63 It is clear that many of these mechanisms are IBD. Crohns disease bears the immunologic signature of an
strain-specific, and not all commensal bacteria with immuno- exaggerated Th1 response, with excess interleukin (IL)-12, IL-
modulatory effects use these mechanisms; other signal trans- 18, interferon (IFN)-, and tumor necrosis factor (TNF).64
duction pathways are likely to account for their anti-inflamma- Conversely, ulcerative colitis is associated with a dominant
tory effects. atypical Th2 response that is probably driven by the production
of IL-13.65
Pathogenesis of Inflammatory Bowel An environmental contribution to the pathogenesis of IBD
Disease is clear from studies of monozygotic twins in which an incom-
Both forms of IBD represent the clinical outcome of a plete accordance rate has been observed for both Crohns dis-
complex interaction of environmental, genetic, and immune ease (50%) and ulcerative colitis (10%).66 Smoking, dietary

Table 2. Host Systems That Limit PRR Signaling and Inflammation


Mechanism Example Reference

Decreased ligand recognition due to TLR Low expression of functional TLRs (eg, TLR2 and TLR4) 107, 130, 131
expression profiles TLR localization (surface vs cytoplasmic expression) 108, 132, 133
State of cell differentiation (apical vs basolateral 132, 134
expression)
Limited repertoire of TLR co-receptor Low expression of TLR co-receptors (eg, MD-2) 130
expression Lack of expression of TLR co-receptors (eg, CD14) 108
Inhibitors of TLR signaling High expression of TLR signaling suppressors (eg, toll- 133, 135137
interacting protein/Tollip, toll IL-1 receptor 8/Tir8)
Sequestration of the TLR signaling adaptor proteins 138
MyD88 and Mal by ST-2 (toll IL-1 receptor family
member)a
Enzyme-mediated inhibition of TLR Proteolytic degradation of TLR4 and TLR9 by TRIAD3A, 139
signaling a ubiquitin ligasea
Deubiquitination of TLR adaptor proteins by the zinc 140, 141
finger protein A20a
Cross-regulation by other PRRs Negative regulation of TLR signaling by NOD2 103, 104, 142, 143
Ligand neutralization Secretion of immunoglobulin A by intestinal 47
enterocytes
aThe regulatory effects of ST2, TRIAD3A, and A20 on TLR signals have been identified in other systems, but it remains to be determined whether
these suppressors mediate TLR signaling in the gut.
278 OHARA AND SHANAHAN CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 5, No. 3

factors, nonsteroidal anti-inflammatory drugs, socioeconomic Microbial Contribution to Inflammatory


conditions, and the more hygienic lifestyles of developed coun- Bowel Disease
tries have all been associated with IBD.67 Contributing environ- Although a specific infectious cause for IBDs has not
mental factors are probably multiple and complex. Numerous been established, current clinical and experimental data incrim-
studies have pursued a specific infectious cause for IBD. The inate a loss of tolerance to commensal bacteria in the develop-
most notable inconclusively incriminate either Mycobacterium ment of chronic intestinal inflammation.67 Animal models of
avium subspecies paratuberculosis (MAP) or adherent-invasive IBD provide persuasive evidence implicating the resident mi-
Escherichia coli (AIEC) in the development of Crohns disease.68,69 crobiota as a causative factor of IBD. Irrespective of the under-
Viable MAP have occasionally been detected in peripheral lying genetic defect(s) in various animal models of the disease,
blood and intestinal tissue in a higher proportion of individuals colonization with commensal bacteria is required for an inflam-
with Crohns disease than in controls, and antibodies to the matory phenotype, and germ-free animals do not develop IBD-
organism have been disease-associated.68,70,71 A recent study like lesions.29,79 82 Of note, antibiotics are beneficial in a num-
identified 2 sets of cross-reactive MAP and human peptides that ber of animal models. It has been shown in some models that
are specific for Crohns disease.72 Although these peptide pairs the disease can be adoptively transferred to immune-deficient
might represent structural mimics whereby an anti-MAP im- recipients when reconstituted with T cells from a diseased
mune response could trigger a host autoimmune response, donor that was sensitized to enteric bacteria.83 Moreover, mod-
definite proof for a role of molecular mimicry in Crohns ification of the microbiota by the administration of probiotic
diseasespecific autoimmunity has not been established. This bacteria, which are commensal organisms that can be harnessed
study did not test the existence of cross-reactive autoantibodies for therapeutic benefits, has been shown to delay the onset of
or T-cell immunity with pathogenic activity. Nonetheless, there disease and attenuate the inflammatory process.84 86 Although
is limited evidence for T-cell immunity to MAP in association these animal models do not reveal whether commensal bacteria
with Crohns disease, and a cause and effect relationship re- passively or directly elicit the inflammatory response, they
mains unproved.73 Among several arguments against persistent clearly demonstrate that exposure to the microbiota is essential
MAP infection as a cause of Crohns disease, the most persua- for expression of the disease.
sive is the clinical experience with anti-TNF therapy. Therapeu- In patient-related studies, increased numbers of bacteria
tic blockade of TNF- greatly impairs host resistance to myco- within the mucosa of IBD patients compared with that of
noninflamed and inflammatory disease controls have been re-
bacterial infection, but this is not associated with disseminated
ported.87,88 Furthermore, the distribution of lesions and inci-
MAP in patients with IBD.73 Furthermore, controlled trials have
dence of inflammation in either Crohns disease or ulcerative
failed to show a therapeutic effect of anti-tuberculosis therapy
colitis is greatest in the area with the highest concentrations of
in Crohns disease.74
luminal bacteria.29 Diversion of the fecal stream is associated
A number of studies have reported abnormally high num-
with an improvement in disease severity in the distal bowel, and
bers of virulent AIEC and hemagglutinin-expressing Esche-
cellular and humoral immune reactivity against components of
richia coli adjacent to the ileal or colonic mucosa in Crohns the bacterial microbiota is evident in patients with IBD.2,29,89
disease.69,75 AIEC can adhere to epithelial cells, invade mac- Flagellin derived from commensal bacteria has been identified
rophages without inducing apoptosis, and persist and repli- as a dominant antigen in Crohns disease, suggesting that
cate within phagocytes.76 Nonetheless, these reports do not TLR5-dependent recognition of flagellin might play a role in
fulfil Kochs postulates, and several lines of available evi- the immune responses to the commensal bacteria observed in
dence that are reviewed elsewhere stand against an etiologic IBD.90 In addition, therapeutic efficacy from the administration of
role for any single pathogenic microorganism in the patho- antibiotics and probiotics in IBD patients has been reported.3,9193
genesis of IBD.67 Moreover, it is difficult to reconcile the Together, these and other circumstantial observations in humans
concept of an infectious etiology with the beneficial effects of that are reviewed in more detail elsewhere29,94 substantiate the
host immunosuppression; any infection putatively stimulat- current dogma that IBD is a heterogeneous syndrome caused by
ing such an intense inflammatory reaction as seen in Crohns dysregulated immunity to normal microbiota. However, the model
disease is unlikely to respond to long-term suppression of must allow for heterogeneity within both Crohns disease and
host immune defenses. However, after the lesson of Helico- ulcerative colitis. It remains unclear whether the inflammatory
bacter pylori and peptic ulcer disease, it would be unwise to responses in IBD, both within the gut and at extraintestinal sites,
completely dismiss an infectious contribution to the patho- are elicited in response to a specific subset of intestinal microbes,
genesis of IBD. It is interesting to note that both MAP DNA or whether tolerance to commensal bacteria in general is affected.
and E coli DNA have been detected in the granulomas of
resected tissues from some Crohns disease patients.77,78 Genetic Influences on Microbial Perception in
However, other forms of bacterial DNA were also detected,67 Inflammatory Bowel Disease
and rather than reflect a specific causal relationship, it might The pathophysiologic impact of bacterial-mucosal in-
reflect impaired innate handling of luminal bacteria in teractions has been emphasized by an increased awareness of
Crohns disease. It is possible that in a subset of genetically the association of genetic aberrations with increased suscepti-
susceptible Crohns disease patients, particularly those with bility to IBD. In particular, mutations of CARD15, which en-
a defect in their ability to sense bacterial ligands or clear codes NOD2, and certain TLR polymorphisms have been
intracellular infections, persistent infection with MAP, AIEC, strongly associated with Crohns disease. This implicates a role
or an as yet unidentified pathogen might underlie their of PRR dysfunction and impaired bacterial sensing in the
disease. pathogenesis of IBD.
March 2007 THERAPEUTIC POTENTIAL OF THE GUT MICROBIOTA 279

Three major polymorphisms in the CARD15 gene on chro- Therefore, it is probable that additional IBD susceptibility genes
mosome 16q12 have been specifically associated with 15% of that affect the interpretation of the microbial microenvironment
Crohns disease patients, and individuals who carry 2 copies of and/or regulate the host response to it remain to be identified.
the risk alleles have a 20-fold to 40-fold increase in their risk of
developing Crohns disease.9597 However, this risk factor is Modifying the Ecosystem as
neither sufficient nor necessary for development of the dis- Therapeutic Strategy
ease.98 Moreover, NOD2-deficient mice do not develop sponta- Traditional drug therapies for IBD address only one
neous gut inflammation, and NOD2 knockout mice that lack aspect of the pathogenesis by targeting the mucosal inflamma-
full-length NOD2 protein do not demonstrate significantly tory response, but this strategy remains suboptimal in terms of
enhanced susceptibility to experiment-induced colitis.99,100 disease management and efficacy. Optimal disease management
The ligands for NOD2 are muramyl dipeptides (MDP) of might require the microbial contribution to be addressed. Ma-
bacterial peptidoglycan (Table 1). All 3 Crohns diseaseassoci- nipulating the microbiota and exploiting host-microbial signal-
ated mutations occur in the ligand-binding domain of NOD2. ing pathways could provide benefit for the treatment of both
Intracellular triggering of NOD2 leads to NF-B activation. acute and chronic intestinal diseases. This underpins the ratio-
Thus, mutations in the NOD2 protein compromise MDP-in- nale for therapeutic manipulation of the microbiota with pro-
duced signaling and result in aberrant or defective NF-B re- biotic bacteria or other pharmabiotics.
sponses to gut bacteria.101,102 However, stimulation of NOD2 Lactobacilli and bifidobacteria have traditionally been the most
with MDP has been shown to inhibit TLR2-driven NF-B acti- common probiotic candidates, but multi-strain cocktails (eg,
vation and Th1 cytokine responses. This suggests that in the VSL#3), nonpathogenic E coli, and nonbacterial organisms such as
absence of NOD2, stimulation with MDP results in NF-B Saccharomyces boulardii and nematode parasites have been used for
dysregulation and imbalanced TLR2-mediated cytokine pro- probiotic effect.113,114 Pharmabiotic is an umbrella term to encom-
duction (elevated IL-12, decreased IL-10).103,104 Together these pass any form of therapeutic exploitation of the gut microbiota. In
might contribute to the pathogenesis of Crohns disease (Figure addition to live probiotic bacteria, pharmabiotics might include
2A). On the other hand, in a study of knock-in mice expressing bacterial constituents such as DNA, probiotic-derived biologically
the most common Crohns diseaseassociated NOD2/CARD15 active metabolites, food ingredients that modulate the composi-
mutation, it was shown that MDP triggers increased NF-B tion of the microbiota (prebiotics), probiotic/prebiotic combina-
activation, IL-1 secretion, and apoptosis105 (Figure 2B). More- tions (synbiotics), or genetically modified commensal bacteria.
over, NOD2-deficient mice are unusually susceptible to infec- Recently, in the first human trial with genetically engineered ther-
tion with Listeria monocytogenes and have reduced production of apeutic bacteria, modified Lactococcus lactis was used to deliver IL-10
defensin-like antimicrobial peptides termed cryptidins.100 Con- locally to the gut in 10 Crohns disease patients. The treatment was
sidering that reduced expression of -defensin has been ob- safe, disease activity was reduced, and the modified bacteria were
served in Crohns disease patients with NOD2 mutations,106 biologically contained.115 Therefore, bacterial-based topical deliv-
this has led to the proposal that NOD2 mutations might ery of biologically active proteins represents a highly promising
predispose to Crohns disease by reducing -defensinmediated and safe therapeutic strategy for combating mucosal diseases.
innate antimicrobial activity100 (Figure 2C). To date, these dif- Increasing evidence supports a therapeutic role for probiotic
ferent experimental models have implicated conflicting loss of strategies for treating enteric infections, post-antibiotic syn-
function, loss of regulation, or gain of function phenotypes for dromes, necrotizing enterocolitis, and irritable bowel syn-
NOD2 mutations, and the precise biologic role of NOD2 in drome.116 119 In murine models of colitis, probiotics have also
normal intestinal physiology and in the pathogenesis of demonstrated prophylactic effects that are associated with a
Crohns disease remains elusive. reduction in proinflammatory cytokines and an induction of
Expression of some TLRs, including TLR4, is differentially regulatory cytokines.84,120 However, the role of probiotics in
altered in Crohns disease and ulcerative colitis.107,108 It has been human IBD is more complex. The best evidence for probiotic
proposed that increased TLR4 expression in patients with IBD efficacy in IBD is in the prevention of pouchitis,121 but results
confers lipopolysaccharide hyperresponsiveness.107 Alterna- in clinical practice appear to be inconsistent. This might relate
tively, it might reflect a loss of response. The TLR4 gene is to variability in patient populations or the quality or choice of
localized on chromosome 9q32-33, a region harboring a probiotic preparation. In ulcerative colitis, E coli Nissle 1917,
Crohns disease susceptibility gene,109 and in selective popula- Lactobacillus rhamnosus GG, and VSL#3 have shown efficacy sim-
tions, a TLR4 polymorphism has been associated with IBD.49,110 ilar to the drug mesalazine in maintaining remission.93,122,123
It is plausible that variant alleles in the TLR4 gene could induce Probiotics have induced remission of acute ulcerative colitis
functional dysregulation in response to lipopolysaccharide. also.93,124 However, in a randomized, double-blind, placebo-
However, the functional phenotypic consequences of TLR4 controlled trial involving 157 patients, neither Bifidobacterium
polymorphisms in IBD remain unresolved. A preliminary report infantis 35624 nor Lactobacillus salivarius subspecies salivarius
from a single German cohort study has suggested that Crohns UCC118 demonstrated a significant benefit over placebo in the
disease might also be associated with TLR9 promoter polymor- maintenance of steroid-induced remission of ulcerative coli-
phisms.111 Recent reports demonstrating a hyperreactivity to tis.125 These strains have attenuated disease severity in animal
flagellins in sera from Crohns disease patients90,112 suggest that models of IBD.85,126 It might be that the differences in efficacy
these TLR or other PRR polymorphisms could lead to impaired between animal and human IBD might reflect the timing of
bacterial clearance and thus increased load of bacterial antigens, administration, differences in disease severity, or effective pro-
including flagellin, in the lumen. This would be consistent with biotic dose/body weight.
evidence for defective handling of bacteria and increased bacterial The evidence for probiotics in Crohns disease is even weaker.
numbers in the mucosa of patients with Crohns disease.90,112 Controlled studies have not established efficacy for L rhamnosus
280 OHARA AND SHANAHAN CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 5, No. 3

Figure 2. Possible mechanisms contributing to the association of mutations in the CARD15 gene with Crohns disease. CARD15 encodes the NOD
protein, NOD2, which is expressed in the cytosol of cells of the macrophage/monocyte lineage, DCs, and enterocytes. Stimulation of NOD2 by MDP
of bacterial peptidoglycan leads to the activation of the transcription factor NF-B, and peptidoglycan can signal also to antigen-presenting cells
through TLR2. (A) In normal mucosa, TLR2-driven activation of NF-B is negatively regulated by non-mutated NOD2, thereby preventing excessive
production of the proinflammatory Th1 cytokines IL-12 and IFN-. Conversely, in NOD2-deficient cells, IL-10 secretion is decreased, and Th1-cell
mediated inflammation occurs through IL-12 as a result of NOD2 to negatively regulate TLR2 signaling. (B) Alternatively, it has been postulated that
in antigen-presenting cells expressing mutated NOD2, MDP elevates NF-B activation and results in excessive IL-1 production and uncontrolled
apoptosis, which together might mediate the pathogenesis of Crohns disease. (C) Deficient expression of NOD2 by intestinal enterocytes,
particularly by Paneths cells at the base of the small intestinal crypts, results in decreased secretion of antimicrobial peptides known as -defensins.
Impaired NOD-2 dependent -defensin production might lead to increased bacterial colonization and a type of bacterial overgrowth that triggers an
immunologic response to the microbiota.

GG or Lactobacillus johnsonii LA1 as maintenance therapies for pharmabiotic therapy. It might be that multi-strain probiotic com-
Crohns disease.127,128 Crohns disease is a complex condition, vari- binations are required, and strain selection, dosing, and coloniza-
able in its location as well as its manifestation. Variability in the tion issues need more intensive investigation. Moreover, there
composition and diversity of the microbiota along and over the might be a role for probiotics in the enhancement of epithelial
cross-sectional axis of the gastrointestinal tract suggests that de- barrier function in the very early stages of Crohns disease.
pending on the topographic distribution of lesions in Crohns Several unresolved issues impede the clinical evaluation of
disease, a single probiotic might not be equally suited to different probiotics.67,129 These include determination of optimal dose
subsets of patients. Colonic location of disease seems to respond and vehicle of delivery, development of reliable predictors of in
better to antibiotics and might, as a result, be more susceptible to vivo performance, regulation and verification of product stabil-
March 2007 THERAPEUTIC POTENTIAL OF THE GUT MICROBIOTA 281

ity, determination of which combinations of probiotics or other wards resident intestinal flora but is broken in active
pharmabiotics are synergistic or antagonistic, and strain-strain inflammatory bowel disease (IBD). Clin Exp Immunol 1995;
comparisons of probiotic performance in different indications. 102:448 455.
Furthermore, the microbial, immunologic, and functional char- 3. Rutgeerts P, Hiele M, Geboes K, et al. Controlled trial of metro-
acteristics of individual probiotic strains and their mechanisms nidazole treatment for prevention of Crohns recurrence after
ileal resection. Gastroenterology 1995;108:16171621.
of action in different clinical settings require clarification. In-
4. Shanahan F. Nutrient tasting and signaling mechanisms in the
dividual variability in composition of the enteric microbiota gut V: mechanisms of immunologic sensation of intestinal con-
might also be a determining factor for optimal strain selection. tents. Am J Physiol Gastrointest Liver Physiol 2000;278:G191
To protect consumers, there is also a pressing need for more G196.
stringent regulation of unsubstantiated health claims. 5. Haller D, Russo MP, Sartor RB, et al. IKK beta and phosphati-
Bacterial-mediated host immune responses involve several dylinositol 3-kinase/Akt participate in non-pathogenic Gram-neg-
pathways and signal transduction systems, and several modes ative enteric bacteria-induced RelA phosphorylation and NF-
of action by which commensal and probiotic bacteria dampen kappa B activation in both primary and intestinal epithelial cell
NF-B signaling have already been elucidated. More detailed lines. J Biol Chem 2002;277:38168 38178.
clarification of probiotic mechanisms will be facilitated by the 6. Kagnoff MF, Eckmann L. Epithelial cells as sensors for microbial
identification of the molecular basis of prokaryotic-eukaryotic infection. J Clin Invest 1997;100:6 10.
7. Neish AS, Gewirtz AT, Zeng H, et al. Prokaryotic regulation of
dialogue. Furthermore, the increasing availability of commen-
epithelial responses by inhibition of IkappaB-alpha ubiquitina-
sal/probiotic genomes should facilitate the identification of tion. Science 2000;289:1560 1563.
commensal effector molecules or other components with 8. Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut
pharmabiotic potential. The possibility of using these mole- bacteria attenuate inflammation by regulating nuclear-cytoplas-
cules to specifically target distinct points of intracellular signal- mic shuttling of PPAR-gamma and RelA. Nat Immunol 2004;5:
ing cascades might alleviate inflammation in a target area and 104 112.
overcome the global immunosuppressive effects associated with 9. Otte JM, Podolsky DK. Functional modulation of enterocytes by
current therapies. In patients with severe IBD, the use of genet- gram-positive and gram-negative microorganisms. Am J Physiol
ically engineered or designer probiotics might offer a new Gastrointest Liver Physiol 2004;286:G613G626.
strategy for more targeted delivery of anti-inflammatory mole- 10. Ma D, Forsythe P, Bienenstock J. Live Lactobacillus reuteri is
essential for the inhibitory effect on tumor necrosis factor alpha-
cules to the inflamed mucosa.
induced interleukin-8 expression. Infect Immun 2004;72:
5308 5314.
Conclusions and Future Perspectives 11. OHara AM, ORegan P, Fanning A, et al. Functional modulation
of human intestinal epithelial cell responses by Bifidobacterium
The rationale of pharmabiotic therapies appears to be infantis and Lactobacillus salivarius. Immunology 2006;118:
justified, and gastroenterologists need to be aware of an ex- 202215.
panding scope for the microbiota in clinical practice. The ap- 12. Savage DC. Microbial ecology of the gastrointestinal tract. Annu
peal of probiotic strategies in IBD is their safety in comparison Rev Microbiol 1977;31:107133.
with immunosuppressive drugs and corticosteroids. However, 13. Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human
safety concerns surrounding other pharmabiotic strategies in- intestinal microbial flora. Science 2005;308:16351638.
cluding the modification of food-grade bacteria to achieve spe- 14. Moore WE, Moore LH. Intestinal floras of populations that have
cific functional activity in vivo need to be resolved. Clinical a high risk of colon cancer. Appl Environ Microbiol 1995;61:
32023207.
evidence of efficacy also requires substantiation, and scientifi-
15. Hopkins MJ, Sharp R, Macfarlane GT. Age and disease related
cally accredited medical evidence should underlie a clinicians changes in intestinal bacterial populations assessed by cell
choice of pharmabiotic strategy. Moreover, mining the meta- culture, 16S rRNA abundance, and community cellular fatty acid
bolic activity of the indigenous microbiota might yield novel profiles. Gut 2001;48:198 205.
therapeutic agents. Nevertheless, therapeutic manipulation of 16. Zoetendal EG, Akkermans ADL, Akkermans-van Vilet WM, et al.
the indigenous microbiota with any form of pharmabiotic re- The host genotype affects the bacterial community in the human
mains suboptimal as a result of incomplete understanding of gastrointestinl tract. Microb Ecol Health Dis 2001;13:129
commensal bacteria, their anti-inflammatory properties, and 134.
host-microbial interactions. The importance of PRRs in the 17. Berg RD. The indigenous gastrointestinal microflora. Trends
regulation of intestinal innate immunity and homeostatic in- Microbiol 1996;4:430 435.
teractions with the commensal microbiota has only begun to be 18. Stephen AM, Cummings JH. The microbial contribution to hu-
man faecal mass. J Med Microbiol 1980;13:4556.
appreciated, but it is clear that engagement with host immune
19. Backhed F, Ley RE, Sonnenburg JL, et al. Host-bacterial mutu-
cells is central to pharmabiotic action. Further studies of phys- alism in the human intestine. Science 2005;307:19151920.
iologic interactions within the complex network of host cell 20. Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: the unseen
commensalPRRligand signaling in gut health and disease majority. Proc Natl Acad Sci U S A 1998;95:6578 6583.
should lead to the optimal exploitation of pharmabiotic ap- 21. Mountzouris KC, McCartney AL, Gibson GR. Intestinal microflora
proaches to alleviate mucosal inflammation in IBD and possi- of human infants and current trends for its nutritional modula-
bly other intestinal diseases. tion. Br J Nutr 2002;87:405 420.
22. Gronlund MM, Lehtonen OP, Eerola E, et al. Fecal microflora in
healthy infants born by different methods of delivery: permanent
References changes in intestinal flora after cesarean delivery. J Pediatr
1. Elson CO. Genes, microbes, and T cells: new therapeutic targets Gastroenterol Nutr 1999;28:19 25.
in Crohns disease. N Engl J Med 2002;346:614 616. 23. Hooper LV, Gordon JI. Commensal host-bacterial relationships
2. Duchmann R, Kaiser I, Hermann E, et al. Tolerance exists to- in the gut. Science 2001;292:11151118.
282 OHARA AND SHANAHAN CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 5, No. 3

24. Xu J, Gordon JI. Inaugural article: honor thy symbionts. Proc Natl basis and prospects for probiotic therapy in inflammatory bowel
Acad Sci U S A 2003;100:1045210459. disease. Am J Physiol Gastrointest Liver Physiol 2005;288:
25. Rook GA, Brunet LR. Microbes, immunoregulation, and the gut. G417G421.
Gut 2005;54:317320. 46. Rescigno M, Urbano M, Valzasina B, et al. Dendritic cells ex-
26. Umesaki Y, Okada Y, Matsumoto S, et al. Segmented filamen- press tight junction proteins and penetrate gut epithelial mono-
tous bacteria are indigenous intestinal bacteria that activate layers to sample bacteria. Nat Immunol 2001;2:361367.
intraepithelial lymphocytes and induce MHC class II molecules 47. Macpherson AJ, Uhr T. Induction of protective IgA by intestinal
and fucosyl asialo GM1 glycolipids on the small intestinal epi- dendritic cells carrying commensal bacteria. Science 2004;
thelial cells in the ex-germ-free mouse. Microbiol Immunol 303:16621665.
1995;39:555562. 48. Rimoldi M, Chieppa M, Salucci V, et al. Intestinal immune
27. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recogni- homeostasis is regulated by the crosstalk between epithelial
tion of commensal microflora by toll-like receptors is required for cells and dendritic cells. Nat Immunol 2005;6:507514.
intestinal homeostasis. Cell 2004;118:229 241. 49. Cario E. Bacterial interactions with cells of the intestinal mu-
28. Mazmanian SK, Liu CH, Tzianabos AO, et al. An immunomodu- cosa: toll-like receptors and NOD2. Gut 2005;54:11821193.
latory molecule of symbiotic bacteria directs maturation of the 50. Takeda K, Akira S. Toll-like receptors in innate immunity. Int
host immune system. Cell 2005;122:107118. Immunol 2005;17:114.
29. Shanahan F. The host-microbe interface within the gut. Best 51. Abreu MT, Fukata M, Arditi M. TLR signaling in the gut in health
Pract Res Clin Gastroenterol 2002;16:915931. and disease. J Immunol 2005;174:4453 4460.
30. Falk PG, Hooper LV, Midtvedt T, et al. Creating and maintaining 52. Takeda K, Akira S. TLR signaling pathways. Semin Immunol
the gastrointestinal ecosystem: what we know and need to 2004;16:39.
know from gnotobiology. Microbiol Mol Biol Rev 1998;62:1157 53. Inohara N, Chamaillard M, McDonald C, et al. NOD-LRR pro-
1170. teins: role in host-microbial interactions and inflammatory dis-
31. Madsen K, Cornish A, Soper P, et al. Probiotic bacteria enhance ease. Annu Rev Biochem 2005;74:355383.
murine and human intestinal epithelial barrier function. Gastro- 54. Pasare C, Medzhitov R. Toll pathway-dependent blockade of
enterology 2001;121:580 591. CD4CD25 T cell-mediated suppression by dendritic cells.
32. Hooper LV, Wong MH, Thelin A, et al. Molecular analysis of Science 2003;299:10331036.
commensal host-microbial relationships in the intestine. Sci- 55. Fukata M, Michelsen KS, Eri R, et al. Toll-like receptor-4 is
ence 2001;291:881 884. required for intestinal response to epithelial injury and limiting
33. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 enhances bacterial translocation in a murine model of acute colitis. Am J
ZO-1-associated intestinal epithelial barrier integrity via protein Physiol Gastrointest Liver Physiol 2005;288:G1055G1065.
kinase C. Gastroenterology 2004;127:224 238. 56. Cario E, Podolsky DK. Intestinal epithelial TOLLerance versus
34. Bocci V. The neglected organ: bacterial flora has a crucial im- inTOLLerance of commensals. Mol Immunol 2005;42:887
munostimulatory role. Perspect Biol Med 1992;35:251260. 893.
35. Gordon JI, Hooper LV, McNevin MS, et al. Epithelial cell growth 57. Coombes JL, Robinson NJ, Maloy KJ, et al. Regulatory T cells
and differentiation. III. Promoting diversity in the intestine: con- and intestinal homeostasis. Immunol Rev 2005;204:184 194.
versations between the microflora, epithelium, and diffuse 58. Hart AL, Lammers K, Brigidi P, et al. Modulation of human
GALT. Am J Physiol 1997;273:G565G570. dendritic cell phenotype and function by probiotic bacteria. Gut
36. Conly JM, Stein K, Worobetz L, et al. The contribution of vitamin 2004;53:16021609.
K2 (menaquinones) produced by the intestinal microflora to 59. Hart AL, Kamm MA, Knight SC, et al. Quantitative and functional
human nutritional requirements for vitamin K. Am J Gastroen- characteristics of intestinal-homing memory T cells: analysis of
terol 1994;89:915923. Crohns disease patients and healthy controls. Clin Exp Immu-
37. Hill MJ. Intestinal flora and endogenous vitamin synthesis. Eur J nol 2004;135:137145.
Cancer Prev 1997;6(Suppl 1):S43S45. 60. Groux H, OGarra A, Bigler M, et al. A CD4 T-cell subset inhibits
38. Roberfroid MB, Bornet F, Bouley C, et al. Colonic microflora: antigen-specific T-cell responses and prevents colitis. Nature
nutrition and health: summary and conclusions of an Interna- 1997;389:737742.
tional Life Sciences Institute (ILSI) [Europe] workshop held in 61. Duchmann R, May E, Heike M, et al. T cell specificity and cross
Barcelona, Spain. Nutr Rev 1995;53:127130. reactivity towards enterobacteria, bacteroides, bifidobacterium,
39. Backhed F, Ding H, Wang T, et al. The gut microbiota as an and antigens from resident intestinal flora in humans. Gut
environmental factor that regulates fat storage. Proc Natl Acad 1999;44:812 818.
Sci U S A 2004;101:15718 15723. 62. Petrof EO, Kojima K, Ropeleski MJ, et al. Probiotics inhibit
40. Berg RD. Bacterial translocation from the gastrointestinal tract. nuclear factor-kappaB and induce heat shock proteins in colonic
Adv Exp Med Biol 1999;473:1130. epithelial cells through proteasome inhibition. Gastroenterology
41. Hope ME, Hold GL, Kain R, et al. Sporadic colorectal cancer: 2004;127:1474 1487.
role of the commensal microbiota. FEMS Microbiol Lett 2005; 63. Dubuquoy L, Jansson EA, Deeb S, et al. Impaired expression of
244:17. peroxisome proliferator-activated receptor gamma in ulcerative
42. Takahashi A, Wada A, Ogushi K, et al. Production of beta- colitis. Gastroenterology 2003;124:12651276.
defensin-2 by human colonic epithelial cells induced by Salmo- 64. Cobrin GM, Abreu MT. Defects in mucosal immunity leading to
nella enteritidis flagella filament structural protein. FEBS Lett Crohns disease. Immunol Rev 2005;206:277295.
2001;508:484 488. 65. Targan SR, Karp LC. Defects in mucosal immunity leading to
43. Suzuki K, Meek B, Doi Y, et al. Aberrant expansion of seg- ulcerative colitis. Immunol Rev 2005;206:296 305.
mented filamentous bacteria in IgA-deficient gut. Proc Natl Acad 66. Tysk C, Lindberg E, Jarnerot G, et al. Ulcerative colitis and
Sci U S A 2004;101:19811986. Crohns disease in an unselected population of monozygotic
44. Stadnyk AW. Intestinal epithelial cells as a source of inflamma- and dizygotic twins: a study of heritability and the influence of
tory cytokines and chemokines. Can J Gastroenterol 2002;16: smoking. Gut 1988;29:990 996.
241246. 67. Shanahan F. Probiotics in inflammatory bowel disease: thera-
45. Shanahan F. Physiological basis for novel drug therapies used peutic rationale and role. Adv Drug Deliv Rev 2004;56:
to treat the inflammatory bowel diseases I. Pathophysiological 809 818.
March 2007 THERAPEUTIC POTENTIAL OF THE GUT MICROBIOTA 283

68. Naser SA, Ghobrial G, Romero C, et al. Culture of Mycobacte- 88. Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora in
rium avium subspecies paratuberculosis from the blood of pa- inflammatory bowel disease. Gastroenterology 2002;122:44
tients with Crohns disease. Lancet 2004;364:1039 1044. 54.
69. Darfeuille-Michaud A, Boudeau J, Bulois P, et al. High preva- 89. Macpherson A, Khoo UY, Forgacs I, et al. Mucosal antibodies in
lence of adherent-invasive Escherichia coli associated with ileal inflammatory bowel disease are directed against intestinal bac-
mucosa in Crohns disease. Gastroenterology 2004;127:412 teria. Gut 1996;38:365375.
421. 90. Lodes MJ, Cong Y, Elson CO, et al. Bacterial flagellin is a
70. Suenaga K, Yokoyama Y, Nishimori I, et al. Serum antibodies to dominant antigen in Crohn disease. J Clin Invest 2004;113:
Mycobacterium paratuberculosis in patients with Crohns dis- 1296 1306.
ease. Dig Dis Sci 1999;44:12021207. 91. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriotherapy as
71. Olsen I, Wiker HG, Johnson E, et al. Elevated antibody re- maintenance treatment in patients with chronic pouchitis: a
sponses in patients with Crohns disease against a 14-kDa double-blind, placebo-controlled trial. Gastroenterology 2000;
secreted protein purified from Mycobacterium avium subsp. 119:305309.
paratuberculosis. Scand J Immunol 2001;53:198 203. 92. Lammers KM, Vergopoulos A, Babel N, et al. Probiotic therapy in
72. Polymeros D, Bogdanos DP, Day R, et al. Does cross-reactivity the prevention of pouchitis onset: decreased interleukin-1beta,
between mycobacterium avium paratuberculosis and human intes- interleukin-8, and interferon-gamma gene expression. Inflamm
tinal antigens characterize Crohns disease? Gastroenterology Bowel Dis 2005;11:447 454.
2006;131:8596. 93. Bibiloni R, Fedorak RN, Tannock GW, et al. VSL#3 Probiotic-
73. Shanahan F, OMahony J. The mycobacteria story in Crohns mixture induces remission in patients with active ulcerative
disease. Am J Gastroenterol 2005;100:15371538. colitis. Am J Gastroenterol 2005;100:1539 1546.
74. Thomas GA, Swift GL, Green JT, et al. Controlled trial of antitu- 94. Mahida YR, Rolfe VE. Host-bacterial interactions in inflammatory
berculous chemotherapy in Crohns disease: a five year follow bowel disease. Clin Sci (Lond) 2004;107:331341.
up study. Gut 1998;42:497500. 95. Macdonald TT, Monteleone G. Immunity, inflammation, and al-
75. Martin HM, Campbell BJ, Hart CA, et al. Enhanced Escherichia lergy in the gut. Science 2005;307:1920 1925.
coli adherence and invasion in Crohns disease and colon can- 96. Bairead E, Harmon DL, Curtis AM, et al. Association of NOD2
cer. Gastroenterology 2004;127:80 93. with Crohns disease in a homogenous Irish population. Eur J
76. Bringer MA, Glasser AL, Tung CH, et al. The Crohns disease- Hum Genet 2003;11:237244.
associated adherent-invasive Escherichia coli strain LF82 repli- 97. Ogura Y, Bonen DK, Inohara N, et al. A frameshift mutation in
cates in mature phagolysosomes within J774 macrophages. NOD2 associated with susceptibility to Crohns disease. Nature
Cell Microbiol 2006;8:471 484. 2001;411:603 606.
77. Ryan P, Bennett MW, Aarons S, et al. PCR detection of Myco- 98. Inoue N, Tamura K, Kinouchi Y, et al. Lack of common NOD2
bacterium paratuberculosis in Crohns disease granulomas iso- variants in Japanese patients with Crohns disease. Gastroen-
lated by laser capture microdissection. Gut 2002;51:665 670. terology 2002;123:86 91.
78. Ryan P, Kelly RG, Lee G, et al. Bacterial DNA within granulomas 99. Pauleau AL, Murray PJ. Role of nod2 in the response of macro-
of patients with Crohns disease: detection by laser capture phages to toll-like receptor agonists. Mol Cell Biol 2003;23:
microdissection and PCR. Am J Gastroenterol 2004;99:1539 75317539.
1543. 100. Kobayashi KS, Chamaillard M, Ogura Y, et al. Nod2-dependent
79. Blumberg RS, Saubermann LJ, Strober W. Animal models of regulation of innate and adaptive immunity in the intestinal
mucosal inflammation and their relation to human inflammatory tract. Science 2005;307:731734.
bowel disease. Curr Opin Immunol 1999;11:648 656. 101. Li J, Moran T, Swanson E, et al. Regulation of IL-8 and IL-1beta
80. Kosiewicz MM, Nast CC, Krishnan A, et al. Th1-type responses expression in Crohns disease associated NOD2/CARD15 mu-
mediate spontaneous ileitis in a novel murine model of Crohns tations. Hum Mol Genet 2004;13:17151725.
disease. J Clin Invest 2001;107:695702. 102. Hisamatsu T, Suzuki M, Reinecker HC, et al. CARD15/NOD2
81. Onderdonk AB, Franklin ML, Cisneros RL. Production of experi- functions as an antibacterial factor in human intestinal epithe-
mental ulcerative colitis in gnotobiotic guinea pigs with simpli- lial cells. Gastroenterology 2003;124:9931000.
fied microflora. Infect Immun 1981;32:225231. 103. Netea MG, Kullberg BJ, de Jong DJ, et al. NOD2 mediates
82. Strober W, Fuss IJ, Blumberg RS. The immunology of mucosal anti-inflammatory signals induced by TLR2 ligands: implications
models of inflammation. Annu Rev Immunol 2002;20:495549. for Crohns disease. Eur J Immunol 2004;34:20522059.
83. Cong Y, Brandwein SL, McCabe RP, et al. CD4 T cells reactive 104. Watanabe T, Kitani A, Murray PJ, et al. NOD2 is a negative
to enteric bacterial antigens in spontaneously colitic C3H/He- regulator of Toll-like receptor 2-mediated T helper type 1 re-
JBir mice: increased T helper cell type 1 response and ability to sponses. Nat Immunol 2004;5:800 808.
transfer disease. J Exp Med 1998;187:855 864. 105. Maeda S, Hsu LC, Liu H, et al. Nod2 mutation in Crohns
84. McCarthy J, OMahony L, OCallaghan L, et al. Double blind, disease potentiates NF-kappaB activity and IL-1beta process-
placebo controlled trial of two probiotic strains in interleukin 10 ing. Science 2005;307:734 738.
knockout mice and mechanistic link with cytokine balance. Gut 106. Wehkamp J, Harder J, Weichenthal M, et al. NOD2 (CARD15)
2003;52:975980. mutations in Crohns disease are associated with diminished
85. Sheil B, McCarthy J, OMahony L, et al. Is the mucosal route of mucosal alpha-defensin expression. Gut 2004;53:1658
administration essential for probiotic function? subcutaneous 1664.
administration is associated with attenuation of murine colitis 107. Cario E, Podolsky DK. Differential alteration in intestinal epithe-
and arthritis. Gut 2004;53:694 700. lial cell expression of toll-like receptor 3 (TLR3) and TLR4 in
86. Madsen KL, Doyle JS, Jewell LD, et al. Lactobacillus species inflammatory bowel disease. Infect Immun 2000;68:7010
prevents colitis in interleukin 10 gene-deficient mice. Gastroen- 7017.
terology 1999;116:11071114. 108. Cario E, Rosenberg IM, Brandwein SL, et al. Lipopolysaccharide
87. Schultsz C, Van Den Berg FM, Ten Kate FW, et al. The intestinal activates distinct signaling pathways in intestinal epithelial cell
mucus layer from patients with inflammatory bowel disease lines expressing Toll-like receptors. J Immunol 2000;164:
harbors high numbers of bacteria compared with controls. Gas- 966 972.
troenterology 1999;117:1089 1097. 109. Cho JH, Nicolae DL, Gold LH, et al. Identification of novel
284 OHARA AND SHANAHAN CLINICAL GASTROENTEROLOGY AND HEPATOLOGY Vol. 5, No. 3

susceptibility loci for inflammatory bowel disease on 128. Marteau P, Lemann M, Seksik P, et al. Ineffectiveness of Lac-
chromosomes 1p, 3q, and 4q: evidence for epistasis between tobacillus johnsonii LA1 for prophylaxis of postoperative recur-
1p and IBD1. Proc Natl Acad Sci U S A 1998;95:75027507. rence in Crohns disease: a randomised, double blind, placebo
110. Franchimont D, Vermeire S, El Housni H, et al. Deficient host- controlled GETAID trial. Gut 2006;55:842 847.
bacteria interactions in inflammatory bowel disease? the toll- 129. Shanahan F. Probiotics: a perspective on problems and pitfalls.
like receptor (TLR)-4 Asp299gly polymorphism is associated Scand J Gastroenterol Suppl 2003:34 36.
with Crohns disease and ulcerative colitis. Gut 2004;53:987 130. Abreu MT, Vora P, Faure E, et al. Decreased expression of
992. Toll-like receptor-4 and MD-2 correlates with intestinal epithelial
111. Torok HP, Glas J, Tonenchi L, et al. Crohns disease is associ- cell protection against dysregulated proinflammatory gene ex-
ated with a toll-like receptor-9 polymorphism. Gastroenterology pression in response to bacterial lipopolysaccharide. J Immunol
2004;127:365366. 2001;167:1609 1616.
112. Sitaraman SV, Klapproth JM, Moore DA 3rd, et al. Elevated 131. Wang JH, Doyle M, Manning BJ, et al. Induction of bacterial
flagellin-specific immunoglobulins in Crohns disease. Am J lipoprotein tolerance is associated with suppression of toll-like
Physiol Gastrointest Liver Physiol 2005;288:G403G406. receptor 2 expression. J Biol Chem 2002;277:36068 36075.
113. Shanahan F. Probiotics and inflammatory bowel disease: is there 132. Cario E, Brown D, McKee M, et al. Commensal-associated
a scientific rationale? Inflamm Bowel Dis 2000;6:107115. molecular patterns induce selective toll-like receptor-trafficking
114. Shanahan F. Host-flora interactions in inflammatory bowel dis- from apical membrane to cytoplasmic compartments in polar-
ease. Inflamm Bowel Dis 2004;10(Suppl 1):S16 S24. ized intestinal epithelium. Am J Pathol 2002;160:165173.
115. Braat H, Rottiers P, Hommes DW, et al. A phase I trial with 133. Otte JM, Cario E, Podolsky DK. Mechanisms of cross hypore-
transgenic bacteria expressing interleukin-10 in Crohns dis- sponsiveness to Toll-like receptor bacterial ligands in intestinal
ease. Clin Gastroenterol Hepatol 2006;4:754 759. epithelial cells. Gastroenterology 2004;126:1054 1070.
116. Allen SJ, Okoko B, Martinez E, et al. Probiotics for treating 134. Gewirtz AT, Navas TA, Lyons S, et al. Cutting edge: bacterial
infectious diarrhoea. Cochrane Database Syst Rev 2004: flagellin activates basolaterally expressed TLR5 to induce epi-
CD003048. thelial proinflammatory gene expression. J Immunol 2001;167:
117. DSouza AL, Rajkumar C, Cooke J, et al. Probiotics in prevention 18821885.
of antibiotic associated diarrhoea: meta-analysis. BMJ 2002; 135. Melmed G, Thomas LS, Lee N, et al. Human intestinal epithelial
324:1361. cells are broadly unresponsive to Toll-like receptor 2-dependent
118. OMahony L, McCarthy J, Kelly P, et al. Lactobacillus and bi- bacterial ligands: implications for host-microbial interactions in
fidobacterium in irritable bowel syndrome: symptom responses the gut. J Immunol 2003;170:1406 1415.
and relationship to cytokine profiles. Gastroenterology 2005; 136. Garlanda C, Riva F, Polentarutti N, et al. Intestinal inflammation
128:541551. in mice deficient in Tir8, an inhibitory member of the IL-1 recep-
119. Lin HC, Su BH, Chen AC, et al. Oral probiotics reduce the tor family. Proc Natl Acad Sci U S A 2004;101:35223526.
incidence and severity of necrotizing enterocolitis in very low 137. Polentarutti N, Rol GP, Muzio M, et al. Unique pattern of expres-
birth weight infants. Pediatrics 2005;115:1 4. sion and inhibition of IL-1 signaling by the IL-1 receptor family
120. Schultz M, Veltkamp C, Dieleman LA, et al. Lactobacillus plan- member TIR8/SIGIRR. Eur Cytokine Netw 2003;14:211218.
tarum 299V in the treatment and prevention of spontaneous 138. Brint EK, Xu D, Liu H, et al. ST2 is an inhibitor of interleukin 1
colitis in interleukin-10-deficient mice. Inflamm Bowel Dis 2002; receptor and Toll-like receptor 4 signaling and maintains endo-
8:71 80. toxin tolerance. Nat Immunol 2004;5:373379.
121. Gionchetti P, Rizzello F, Helwig U, et al. Prophylaxis of pouchitis 139. Chuang TH, Ulevitch RJ. Triad3A, an E3 ubiquitin-protein ligase
onset with probiotic therapy: a double-blind, placebo-controlled regulating Toll-like receptors. Nat Immunol 2004;5:495502.
trial. Gastroenterology 2003;124:12021209. 140. Boone DL, Turer EE, Lee EG, et al. The ubiquitin-modifying
122. Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of enzyme A20 is required for termination of Toll-like receptor
ulcerative colitis with the probiotic Escherichia coli Nissle 1917 responses. Nat Immunol 2004;5:10521060.
is as effective as with standard mesalazine. Gut 2004;53: 141. Wang YY, Li L, Han KJ, et al. A20 is a potent inhibitor of TLR3-
16171623. and Sendai virus-induced activation of NF-kappaB and ISRE and
123. Zocco MA, dal Verme LZ, Cremonini F, et al. Efficacy of Lacto- IFN-beta promoter. FEBS Lett 2004;576:86 90.
bacillus GG in maintaining remission of ulcerative colitis. Ali- 142. Netea MG, Ferwerda G, de Jong DJ, et al. Nucleotide-binding
ment Pharmacol Ther 2006;23:15671574. oligomerization domain-2 modulates specific TLR pathways for
124. Rembacken BJ, Snelling AM, Hawkey PM, et al. Non-pathogenic the induction of cytokine release. J Immunol 2005;174:6518
Escherichia coli versus mesalazine for the treatment of ulcer- 6523.
ative colitis: a randomised trial. Lancet 1999;354:635 639. 143. van Heel DA, Ghosh S, Hunt KA, et al. Synergy between TLR9
125. Shanahan F, Guarner F, Von Wright A, Vilpponen-Salmela T, et and NOD2 innate immune responses is lost in genetic Crohns
al. A one year, randomised, double-blind, placebo controlled disease. Gut 2005;54:15531557.
trial of a lactobacillus or a bifidobacterium probiotic for mainte-
nance of steroid-induced remission of ulcerative colitis. Gastro-
enterology 2006;130:A-44. Address requests for reprints to: Professor Fergus Shanahan, Ali-
126. Sheil B, MacSharry J, OCallaghan L, et al. Role of interleukin mentary Pharmabiotic Centre, Clinical Science Building, Cork Univer-
(IL-10) in probiotic-mediated immune modulation: an assess- sity Hospital, Cork, Ireland. e-mail: f.shanahan@ucc.ie; fax: 353-21-
ment in wild-type and IL-10 knock-out mice. Clin Exp Immunol 4345300.
2006;144:273280. Supported in part by Science Foundation Ireland. F.S. is also sup-
127. Bousvaros A, Guandalini S, Baldassano RN, et al. A random- ported by the Irish Health Research Board, the Higher Education
ized, double-blind trial of Lactobacillus GG versus placebo in Authority of Ireland, and the European Union (PROGID QLK-2000-
addition to standard maintenance therapy for children with 00563) and has been afliated with a campus-based company (Ali-
Crohns disease. Inflamm Bowel Dis 2005;11:833 839. mentary Health Ltd).

S-ar putea să vă placă și