You are on page 1of 10

Available online at www.sciencedirect.

com

Mechanisms controlling pathogen colonization of the gut


Bärbel Stecher1 and Wolf-Dietrich Hardt2

The intestinal microbiota can protect efficiently against interest in reproducible animal models and comprehen-
colonization by many enteric pathogens (‘colonization sive analytical tools.
resistance’, CR). This phenomenon has been known for
decades, but the mechanistic basis of CR is incompletely The composition of the gastrointestinal
defined. At least three mechanisms seem to contribute, that is microbiota
direct inhibition of pathogen growth by microbiota-derived The composition of the microbiota and its collective
substances, nutrient depletion by microbiota growth and genome, the microbiome, has been intensely studied.
microbiota-induced stimulation of innate and adaptive immune Early cultivation based studies suggested that the human
responses. In spite of CR, intestinal infections are well known to intestinal microbiota harbors at least 400 different, mostly
occur. In these cases, the multi-faceted interactions between the obligate anaerobic bacterial species [3,4]. This was con-
microbiota, the host and the pathogen are shifted in favor of the firmed by modern, culture-independent approaches esti-
pathogen. We are discussing recent progress in deciphering the mating that an individual’s microbiota comprises not
underlying molecular mechanisms in health and disease. more than 500 species [5]. Two predominant phyla were
Addresses observed, the Firmicutes and the Bacteroidetes. Other
1
Max von Pettenkofer Institut, Pettenkoferstrasse 9a, 80336 München, phyla such as as Proteobacteria, Actinobacteria, Fusobac-
Germany teria, Verrucomicrobia, and Cyanobacteria are minor con-
2
Institute of Microbiology, ETH Zürich, Switzerland
stituents. Of note, the murine and the human microbiota
Corresponding authors: Stecher, Bärbel are remarkably similar [6], suggesting that mouse models
(Stecher@mvp.uni-muenchen.de) and Hardt, Wolf-Dietrich can be used to study basic functional principles of
(hardt@micro.biol.ethz.ch) relevance for human health.

Current Opinion in Microbiology 2011, 14:82–91


Nevertheless, data generated by novel culture-indepen-
dent approaches (i.e. 16S rRNA gene and metagenomic
This review comes from a themed issue on sequence analyses) have to be interpreted with care as
Host–microbe interactions: bacteria
certain species may be missed or underestimated in terms
Edited by Brett Finlay and Ulla Bonas
of their abundance due to differential bacterial lysis
Available online 28th October 2010 efficiency, primer bias and variable 16S rRNA gene copy
numbers [7,8]. Moreover, PCR on mixed templates can
1369-5274/$ – see front matter
# 2010 Elsevier Ltd. All rights reserved.
give rise to significant ‘noise’ which leads to overestima-
tion of ecosystem complexity [5,9].
DOI 10.1016/j.mib.2010.10.003
Deep microbiome sequencing analyses have revealed a
wide array of shared genes (‘core’ microbiome) but also
demonstrated a considerable degree of species diversity
To understand the whole it is necessary to understand the
among the microbiota, even between closely related hosts
parts. To understand the parts, it is necessary to under-
[5]. Interestingly, despite this species diversity, different
stand the whole. Such is the circle of understanding [1].
microbiomes are functionally highly conserved as shown
by the abundance of COG categories [10]. Therefore,
A word of caution besides all variability, some general ordering principles do
The mammalian gut is a highly complex ecosystem exist and lead to functionally widely conserved micro-
shaped by the host, a complex microbial community biota properties. This explains why key functional prop-
called microbiota and profoundly affected by interactions erties of the microbiome are consistently observed in
with the outside environment, for example, the intake of different laboratories worldwide, in spite of significant
food or infection by pathogens [2]. This complexity has lab-to-lab variations in the species composition of the
been an immense obstacle for mechanistic studies. Sim- mouse colony microbiota. However, effects of the species
plified model systems and recent advances in analytic composition should be carefully addressed in each case.
methodology have fuelled significant progress. However,
it should be kept in mind that no single study has so far General functions of the microbiota
been able to monitor all relevant parameters (some of In most cases the beneficial functions of the microbiota
which might still be unknown) of the complex mamma- outweigh potentially harmful side effects. The micro-
lian gut ecosystem in parallel. This may leave room for biota provides digestive functions, modulates host metab-
alternative interpretations and explains the field’s keen olism and stimulates development of lymphatic tissue

Current Opinion in Microbiology 2011, 14:82–91 www.sciencedirect.com


Mechanisms controlling pathogen colonization Stecher and Hardt 83

Glossary Figure 1
COG: cluster of orthologous groups of proteins used for phylogenetic
classification of proteins encoded in complete genomes
Colonization resistance (CR): characteristic of the intestinal Microbiota
microbiota to block colonization of pathogens composition -
Defensin: peptide with antimicrobial activity
Gnotobiotic: colonized with bacteria of known identity
IBD: inflammatory bowel disease (e.g. Crohn’s disease and
10
Ulcerative Colitis)
Microbiome: collective genome of all present bacteria
Metagenome: collective genome of all organisms present in a given
8
ecosystem

Log10 cfu/g Salmonella


16S rRNA gene sequencing: The 16SrRNA gene is commonly used
for phylogenetic studies as it is highly conserved between different
species of bacteria and archaea 6
SPF: specified pathogen free Bacteroidetes
Firmicutes
4
Deferribacteres
and the mucosal immune system. Moreover, it can effi- Verrucomicrobia
ciently limit infection of the gut by pathogenic bacteria. 2 Proteobacteria
In fact, during pathogen infection, the microbiota may
have at least three cardinal functions: (i) it may block
growth of the pathogen and thus interfere with the in- 0

CON

Antibiotic-

Germfree
(Schaedler)
treated
fection right from the beginning. This is termed as

LCM
colonization resistance (CR; Box 1) and will be the focus
of this review. (ii) The microbiota may prime the host’s
innate and adaptive immune defenses and prevent path-
Current Opinion in Microbiology
ology which would otherwise be caused by the pathogen’s
virulence factors. This has been reviewed recently [11].
Colonization resistance against oral Salmonella infection in mice with
(iii) It can help to eliminate the pathogen from the gut
different types of microbiota. The figure summarizes published and
lumen at the end of an infection. This has recently been unpublished data from Salmonella-infected mice. Conventional mice
demonstrated [12] and will be discussed elsewhere. from different sources (n = 58), streptomycin-pretreated mice (n = 12)
and low-complexity microbiota (LCM) mice (n = 10; all from [29] and
Even in the normal, healthy host, microbial products Stecher and Hardt, unpublished) and germfree mice (n = 7; [45]) were
orally infected with Salmonella serovar Typhimurium or serovar
released from the intestinal microbiota such as peptido- Enteritidis. Intestinal loads in cfu/g cecal content are depicted. Pie plots
glycan disseminate to the mesenteric lymph nodes and illustrate microbiota composition in the respective groups
even to systemic sites and stimulate immune cell matu- (Bacteroidetes: yellow; Firmicutes: green; Deferribacteres: purple;
ration [13]. In some cases, the presence of an intestinal Verrucomicrobia: blue; Proteobacteria: red; other phyla with abundance
<1%: not depicted).
microbiota contributes to the etiology and progression of
diseases in humans, namely chronic inflammatory bowel
diseases (IBD) [14]. IBD is thought to result if barrier
functions, antimicrobial killing or dampening signaling strated recently that the microbiota can in fact be ‘trans-
pathways of the innate and/or adaptive immune system planted’ between two hosts [17].
fail [15]. In addition, some bacterial species are associated
with the aggravation of autoimmune diseases and the This review will summarize the recent literature pertain-
incidence of colorectal cancer [16]. Thus, microbiota ing to the three-way interaction between the microbiota,
manipulation by the introduction or selective elimination the host and enteric pathogens and discuss novel tech-
of relevant bacteria might represent future avenues for nologies which may aid analysis of CR in the future.
cure or prevention. A ‘proof of principle’ study demon-
Mechanisms of CR
What do we know about the mechanisms underlying CR?
Box 1 Colonization resistance (CR) has been known for more than Innate immunity, adaptive immunity and bacterial inter-
50 years. Anecdotal observations in human patients and animal actions are probably involved in modulating both the
studies had shown that disruption of the microbiota by antibiotics
dramatically increases the susceptibility towards enteric infections
composition of the microbiota and the outcome of infec-
[94,95]. This has been confirmed in numerous recent studies tions. To establish successful infection, all pathogens
[43,96,97]. Experiments in germ-free mice and ex-germfree mice need to replicate in the gut lumen in order to reach a
colonized with selected bacterial species lent further support to this sufficient population density for causing disease. In the
[29,45,98]. The mechanisms underlying this natural protection are
case of S. Typhimurium enterocolitis, this was demon-
still unresolved and a matter of debate.
strated by infecting mice with defined mixtures of the
wild-type pathogen and avirulent mutants [18]. At least

www.sciencedirect.com Current Opinion in Microbiology 2011, 14:82–91


84 Host–microbe interactions: bacteria

Figure 2 The sheer complexity of the normal microbiota should


guarantee depletion of most nutrients which might other-
wise sustain growth of an incoming pathogen. The theory,
also known as ‘Freter’s nutrient-niche hypothesis’ claims
that intestinal colonization by a given bacterial species
requires that its growth rate is faster than its individual
washout rate and that it must consume one or a small
number of growth-limiting nutrients (i.e. mucus-derived
carbohydrates) better than all other competitors in the
ecosystem [23]. This theory was followed up experimen-
tally by P. Cohen and T. Conway using E. coli mutants in
defined metabolic pathways.

These studies beautifully demonstrate that E. coli strains


can grow on mucus and consume multiple mucin-derived
sugars in parallel. Conversely, a defect in consuming one
of these sugars (i.e. arabinose, fucose, and N-acetyl-glu-
cosamine) led to a competitive disadvantage against the
respective parental strain. Interestingly, different E. coli
strains showed distinct sugar utilization preferences [24].
A pathogenic E. coli was only excluded by strains with a
similar nutrient utilization spectrum (i.e. an isogen) or a
mixture of several commensal E. coli strains [25]. Along
this line, studies in mice and humans have shown that CR
Three mechanisms of intestinal microbiota-mediated CR. There are at correlates inversely with the number of different Enter-
least three mechanistic ways how the intestinal microbiota can confer obacteriaceae as well as different E. coli biotypes [26,27].
protection against enteric pathogens. (1) Direct inhibition of the pathogen Accordingly, the density of commensal intestinal E. coli
by antimicrobial effector molecules (bacteriocins, metabolic by-products).
was inversely correlated with the degree of CR against E.
(2) Efficient competition for nutrients establishes CR by shutting down all
potentially available nutrient niches for the pathogen. (3) Indirect inhibition coli and Salmonella spp. [28,29]. This might be explained
by stimulating the host’s antimicrobial defense system. The microbiota by the fact that the same global selective pressure (i.e.
releases microbial patterns (LPS, peptidoglycan) which are sensed by the presence/absence of a common inhibitor/nutrient) acts on
host’s epithelial cells thus triggering antibacterial defenses such as release both species.
of epithelial-derived defensins, mucin secreted by goblet cells, and
secretory IgA produced by lamina propria plasma B-cells.
Therefore, different combinations of microbiota species
could confer CR against a given pathogen, as long as key
nutrients are depleted. This would be in line with the
three possible mechanisms are conceivable how the high level of host-to-host COG-conservation discussed,
microbiota may convey CR (Figure 2): above. Formation of an ecological ‘food-web’ consisting
of several primary and secondary fermenters may be
(1) Direct inhibition: Bacterial species can inhibit the required for the efficient degradation of all potential
growth of other bacteria, including pathogens, in differ- nutrient sources available to pathogens. Recently, func-
ent ways, for example, by releasing inhibitory metab- tional principles of such food-webs have been studied
olites (i.e. acetate and butyrate) and bacteriocins. more in detail. Association experiments with Bacteroides
Besides, oxygen depletion slows down the growth rates thetaiotaomicron (B. t.) in gnotobiotic mice demonstrated
of facultative anaerobic bacteria (such as most pathogens) that co-colonization with different probiotic strains can
and competition for binding sites on epithelia or mucus- affect the efficiency of B. t. metabolism [30,31]. Con-
derived receptor sites [19,20]. Direct inhibition in the versely, B. t. opens up new nutrient sources for the co-
absence of the host has been demonstrated in vitro using colonizing strains, as they themselves do not produce a
continuous colonic fermentation models [21]. Similarly, wide spectrum of secreted polysaccharide lyases and
the lag phase of pathogenic shiga-toxin producing Escher- glycoside hydrolases. Co-association also increases the
ichia coli (STEC) grown in fecal suspensions was corre- total microbiota density and boosts the yield of fermenta-
lated to the CR of the donor, suggesting that the tion products, such as short chain fatty acids (i.e. butyrate)
inhibitory mechanisms are operational when uncoupled which in turn serve as nutrient source for colonic epi-
from the intestine [22]. thelial cells and modulate host gene expression [32].

(2) Nutrient depletion: The microbiota is known to utilize (3) Stimulation of immune defenses: This mechanism is quite
numerous nutrients not absorbed by the host’s intestine. complex and difficult to analyze: On the one hand,

Current Opinion in Microbiology 2011, 14:82–91 www.sciencedirect.com


Mechanisms controlling pathogen colonization Stecher and Hardt 85

microbiota-derived stimuli are not easily provided in the Studies on the long-term effects of the microbiota on the
absence of life bacterial colonization. On the other hand, host are still at the beginning and we urgently need to
the presence of a complex bacterial community in the gut investigate the effects in more detail to avoid misinter-
does not allow disentangling inhibitory effects mediated pretation of experimental data. Most likely, one needs to
by a particular bacterial product or a certain bacterial revisit the current SPF concept: animal breeders should
species. change from a definition ‘free of pathogens’ to rather
declare a ‘positive list’ of microorganisms that are present
Defensins produced by the host’s specialized epithelium in a certain mouse flora [47].
are secreted into the gut lumen in order to increase mucosal
barrier function against luminal bacteria. Mice lacking Environmental factors affecting the
matrix metalloproteinase 7 (MMP7), and hence devoid microbiota
of mature paneth-cell a-defensins, show an altered micro- The determinants influencing stability and composition
biota composition [33]. Conversely, transgenic mice over- of the microbiota are not completely understood. Besides
expressing a human beta-defensin were protected against antibiotics other factors are known that lead to alterations
lethal S. Typhimurium challenge [34]. So far, it is unclear if of intestinal bacterial communities and may, as a con-
defensins merely protect by killing pathogens approaching sequence, affect CR. The diet has a profound effect on
the mucosal surface or indirectly, by inducing microbiota the microbiota composition [48] as shown by next-gener-
shifts and modulating CR. Besides antimicrobial peptides, ation deep sequencing analysis of the human microbiota
IgA is also secreted in the gut lumen in high quantities and [49,50]. Both studies found that the microbiota can
can protect against insult from pathogens, steady state respond rapidly to changes in diet in as little as one
penetration of bacterial products and accidental entry of day. Thus, one day it might become feasible to design
microbiota [12,35,36]. diets or nutrient additives which foster ‘favorable’ or
‘unfavorable’ microbiota compositions thus enhancing
On the other hand, the capsule polysaccharide (PSA) of CR. However, today we are clearly far away from such
Bacteroides fragilis was found to stimulate T-cell and ‘rational microbiota management’.
lymphoid organ development and protects mice from
experimental Helicobacter hepaticus colitis, presumably CR and intestinal inflammation
by stimulating Foxp-3(+) IL10 producing T-cells and Mucosal inflammation has a profound effect on the host’s
inhibiting pro-inflammatory TH17 responses [37,38]. mucosa, on the nutrient availability in the gut and on the
Conversely, antibiotic-mediated microbiota depletion microbiota composition. For practical reasons, we dis-
reduced the secretion of the antimicrobial lectin RegIIIg tinguish between chronic gut inflammation caused by
and resulted in increased susceptibility to oral Enterococ- effects of the commensal microbiota (IBD) and acute
cus infection [39]. Also, several studies linked particular inflammation caused by enteric pathogens. Nevertheless,
members of the microbiota (i.e. segmented filamentous some functional principles might be common to both
bacteria, SFB) to intestinal TH17 responses [40–42]. types of disease.
Confirmation and detailed mechanistic analysis of these
observations would require culturing of the SFB and Work on acute Salmonella and Citrobacter infection models
studies in defined model systems. Anyhow, these data has revealed that enteropathogenic bacteria can subvert
suggest that antibiotics and facility-specific microbiota the inflammation in order to out-compete the microbiota
differences may modify susceptibility to enteric infec- [51–54]. Similarly, inflammation seems to enhance gut
tions and autoimmune diseases [29,43,44]. colonization by other pathogens like Vibrio cholerae or
Clostridium difficile [55,56]. This growth advantage
Of note, the degree of CR can vary dramatically between might be attributable in part to the high levels of anti-
individual mice ranging from no detectable colonization microbial peptides/defensins secreted by the inflamed
(below 103 cfu/g) to levels reached in GF mice (>109 cfu/ mucosa [57–61] and the relatively high resistance which
g) [29,45] (Figure 1). Substantial differences, for example, pathogens like S. Typhimurium have against these com-
the presence/absence of SFB, can be observed between pounds [57,62,63]. On the other hand, inflammation alters
mice from different sources and the microbiota compo- the mix of electron donors and acceptors available in the
sition can have significant effects on the immune gut (Figure 3). The inflamed mucosa secretes large
responses of the host (e.g. TH17 responses [44]). These amounts of high energy glycosylated proteins in the
differences can be minimized by breeding littermate gut lumen, which are utilized by S. Typhimurium [64].
controls or by swapping litters (Stecher and Hardt, Furthermore, it secretes lipocalin which interferes with
unpublished observation). In conclusion, extreme care iron uptake by commensal bacteria, but not by S. Typhi-
should be taken with respect to the appropriate design of murium [65]. Finally, it was found recently that the
such animal experiments. Not only the genetic back- inflamed mucosa can convert H2S, a typical metabolic
ground of the mice must be matched (i.e. wild type product released by commensals, into tetrathionate and
and knock-out) but also the respective microbiota [46]. that the latter can serve as a primary electron acceptor for

www.sciencedirect.com Current Opinion in Microbiology 2011, 14:82–91


86 Host–microbe interactions: bacteria

Figure 3 invasive E. coli (AIEC) are thought to lead to aggravation of


disease symptoms. In patients affected by Crohn’s disease,
AIEC were found in close contact with the inflamed ileal
mucosa and even invaded intestinal epithelial cells [72,73].
These data suggest that there might be a link between
AIEC overgrowth in the inflamed intestine and aggravation
of disease symptoms in IBD.

In conclusion, chronic and acute inflammation profoundly


affects CR, but the underlying mechanisms are still not
entirely understood. Elucidating how different commen-
sal bacteria adapt to growth in the inflamed and non-
inflamed gut and how this affects their interaction with
the host will be of high relevance.

Future directions
In the future, two developments will have a great impact
on analyzing CR, simplified model systems and improved
systems biology techniques (Box 2).
Inflammation-induced Salmonella overgrowth in the gut. Salmonella may
exploit intestinal inflammation to out-compete the microbiota using Gnotobiology has already contributed significantly to our
different strategies: (1) Salmonella uses mucin-derived carbohydrates current understanding of the microbiota–host cross-talk
which are released in the gut in the course of inflammation [64]. (2)
Salmonella can use tetrathionate as an electron acceptor which is more
Box 2 Mouse models for studying CR. Conventionally raised (CON)
abundant in the inflamed gut [66]. (3) Salmonella is resistant to a variety
mice harbor a complex gut flora consisting of >500 different
of host-derived defensins and antimicrobial peptides and avoids
species conferring CR against enteropathogens including
inhibition of the host-derived antimicrobial peptide lipocalin-2 that
Salmonella enterica serovar Typhimurium (S. Typhimurium; Figure
sequesters the siderophore enterochelin by the production of
1). Upon disruption of the gut microbiota, for example, by antibiotic
salmochelin [65]. Inflamed mucosa depicted in red. PMN: transmigrated
treatment or in germfree mice (GF), susceptibility to enteric
polymorphonuclear leucocyte.
infections is greatly increased [45,96,97]. Re-association of
microbiota-depleted mice with a CON gut flora re-establishes CR
[99–101]. This enabled pioneering studies of the bacterial species
conferring CR, that is the ground-breaking work by Rolf Freter and
S. Typhimurium [66]. Respiration would allow S. colleagues in the 1960s. GF mice have an enlarged cecum (>5% of
Typhimurium to grow faster than the microbiota, most total body weight versus 1% in conventional mice; attributed to
of which are not able to respire. It will be interesting to non-digested complex carbohydrates). Association of GF mice with
see whether the tetrathionate-mediated growth species of a CON microbiota can ‘normalize’ the cecal size and
advantage might be affected by facility-specific differ- physiology [102,103]. E. coli served as an indicator strain for CR and
relative cecal size or the degree of normalization is directly linked to
ences in microbiota composition. intestinal E. coli titers [104]. Thus, CR can be quantified by orally
contaminating animals with E. coli and measuring fecal colonization
Microbial community imbalances are also observed in levels [27,95,105,106]. Analyzing CR upon re-introducing
human inflammatory bowel disease (IBD) patients as well combinations of microbiota strains R. Freter demonstrated that CR
requires obligate anaerobes [107], for example, a combination of
as experimental mouse colitis models [51,67]. However,
Clostridium spp. and three Lactobacillus strains established CR
the events initiating CR are still controversial. Reduction against E. coli in GF mice [79,80]. Recent work assigned these
of antimicrobial peptide and mucin production [68], epi- species to the Clostridium clusters XIVa and IV [81]. The
thelial cell dysfunction (ER stress), [69] and lack of mechanisms how these bacteria induce cecal normalization and CR
microbial pattern recognition receptors [70] are prime are yet to be elucidated. In order to investigate microbiota functions
including CR, efforts towards a controlled colonization of mice with
candidates that may lead to a generally weakened mucosal a defined microflora have been made and served as model in
barrier. Secondary to this, a pathologically altered micro- biological research since the 1960 s [108,109]. Interestingly, mice
biota may trigger or aggravate chronic gut inflammation. It associated with a low-complexity type of microbiota (LCM) derived
was demonstrated that the microbiota derived from from the altered Schaedler flora (ASF) [110] are highly susceptible to
enteric Salmonellosis in a similar way as GF mice [29]. Again,
animals suffering from colitis was a potent inducer of colitis
transfer of a complex flora derived from CON mice induces
after transfer and members of the Enterobacteriaceae protection against S. Typhimurium colonization and gut
family contributed to this effect [71]. Conversely, an acute inflammation [12,29,89]. The colonization status of ASF mice can be
and chronically inflamed intestine presents a considerably stably maintained in individually ventilated cages [111] making this
altered environment for the intestinal microbiota and mouse model an attractive tool in combination with a deep
16SrRNA gene pyrosequencing strategy and phylogenetic
fosters growth of pathogens and facultative anaerobic sequence analysis to identify bacteria contributing to CR.
commensals [52]. E. coli overgrowth is commonly observed
in human IBD patients. Here, the so-called adherent and

Current Opinion in Microbiology 2011, 14:82–91 www.sciencedirect.com


Mechanisms controlling pathogen colonization Stecher and Hardt 87

Table 1

Overview on host–microbiota interaction studies in gnotobiotic mice and mice colonized with undefined microbial consortia

Bacteria used in gnotobiotic model Phenotype observed Effect on CR versus Citation


germfree/conventional
situation
Segmented filamentous SFB induces IgA-producing cells, ND [78]
bacteria (SFB), MHCII expression on epithelial cells
Candidatus arthromitus Induction of TH17 responses [40,44]
Induction of EAE in mouse model [41,42]
(immunization with MOG in CFA)
Induction of rheumatoid arthritis
16 Bacteroides strains or Significantly reduced induction of IL-17 ND [44]
46 Clostridia strains production in CD4-T-cells compared to
mice associated with SFB; Increase in
IFNg producing CD4-T-cells.
Bacteroides fragilis Induction of Foxp3(+) Treg cells ND [38]
Spore-forming bacteria Induction of TH17 responses similar to ND [40]
colonization with SFB. This was not as
pronounced in mice associated with the
culturable fraction of
conventional microbiota
Human baby flora derived Protection against E. coli infection Increased CR against [22]
bacterial mixture E. coli
Human fecal flora ND [49]
Mouse flora
(1) E. coli Positive effect on cecal ‘normalization’ Increased CR from (1) to [79–81]
(2) E. coli + Lactobacillus and colonization resistance (7) against E. coli
(3) (2) + Lb + Candida
(4) (3) + 3 anaerobic fusiform
bacteria
(5) (4) + 30 anaerobes
(6) 50 strict anaerobes + 80
facultative anaerobes
(7) Chloroform-resistant
spore-forming Bacteria
(Clostridia)
Escherichia coli K-12, Exclusion of B. longum but not L. paracasei Increased CR against [82]
Lactobacillus johnsonii NCC533, from the gut; E. coli Nissle led to displacement B. longum
and Bifidobacterium of Escherichia coli K-12, Lactobacillus johnsonii
longum NCC2705 NCC533;
Altered Schaedler flora No protection against S. Typhimurium infection Decreased CR against S. [12,29]
and induction of intestinal inflammation Tm compared to CON mice
Bifidobacterium animalis Reduced fecal S. Typhimurium excretion Increased CR against S. Tm [83]
Lactobacillus delbrueckii Protection against systemic L. monocytogenes ND [84]
UFV-H2b20 infection
Bacteroides thetaiotaomicron Induction of mucin fucosylation by fucose ND [85–87]
consumption
Induction of angiogenins and RegIIIg [30,88,89]
Flexible foraging behavior (host-derived mucus
and food particles)
+Eubacterium rectale ND [90]
Ruminococcus gnavus E1, Elimination of C. perfringens from the intestine Increased CR against [91]
Bacteroides thetaiotaomicron C. perfringens
LEMF4, Clostridium hathewayi
LEMC7, and Clostridium
orbiscindens LEMH9
+Methanobrevibacter smithii Increased energy harvest from diet; higher ND [92]
yields of acetate and formate produced by
B. t.; Expansion of B. t. sugar substrate range
+Desulfovibrio piger No effect ND [92]
+Lactobacillus casei Expansion of B. t. sugar substrate range ND [89]
+Bifidobacterium longum
Acetogenic bacteria Expansion of B. t. sugar substrate range ND [31]
Lactobacillus plantarum Expansion of B. t. sugar substrate range ND [93]

www.sciencedirect.com Current Opinion in Microbiology 2011, 14:82–91


88 Host–microbe interactions: bacteria

(Table 1). We will need to invest into improved GF and This study uses a new algorithm for pyrosequencing-noise reduction in
phylogenetic data to determine the actual diversity of the gut microbiota
gnotobiotic animal models for analyzing particular strains by a deep sequencing approach.
(or combinations thereof). An increasing catalogue of 6. Dethlefsen L, McFall-Ngai M, Relman DA: An ecological and
sequenced microbial genomes and development of evolutionary perspective on human–microbe mutualism and
disease. Nature 2007, 449:811-818.
genetic tools for molecular manipulation of the
sequenced strains will allow deciphering molecular mech- 7. Daims H, Bruhl A, Amann R, Schleifer KH, Wagner M: The
domain-specific probe EUB338 is insufficient for the
anisms of CR [74,75]. Also, GF mice allow re-association detection of all bacteria: development and evaluation of a
with ‘human’ microbiota [49]. And the generation of GF more comprehensive probe set. Syst Appl Microbiol 1999,
22:434-444.
KO and transgenic mice will be important for analyzing
the host’s contributions to CR [76]. It should be men- 8. Salonen A, Nikkila J, Jalanka-Tuovinen J, Immonen O, Rajilic-
Stojanovic M, Kekkonen RA, Palva A, de Vos WM: Comparative
tioned that the methods for generating and breeding GF analysis of fecal DNA extraction methods with phylogenetic
mice have been significantly improved since their inven- microarray: effective recovery of bacterial and archaeal DNA
using mechanical cell lysis. J Microbiol Methods 2010,
tion and that key gnotobiotic techniques (embryo transfer 81:127-134.
and isolators) have become available in most animal
9. Kunin V, Engelbrektson A, Ochman H, Hugenholtz P: Wrinkles in
facilities worldwide. the rare biosphere: pyrosequencing errors lead to artificial
inflation of diversity estimates. Environ Microbiol 2009.
On the other hand, high throughput methods for systems 10. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A,
wide analysis will have a great impact. Technological Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP et al.: A core gut
microbiome in obese and lean twins. Nature 2009, 457:480-484.
quantum leaps like extremely deep sequencing and
11. Sekirov I, Russell SL, Antunes LC, Finlay BB: Gut microbiota in
RNAseq will enable a second generation of system-wide health and disease. Physiol Rev 2010, 90:859-904.
microbiome, system-transcriptome analysis [31]. These
12. Endt K, Stecher B, Chaffron S, Slack E, Tchitchek N, Benecke A,
kinds of data will allow the discovery of novel bacterial Van Maele L, Sirard JC, Mueller AJ, Heikenwalder M et al.: The
functions as recently exemplified by an algae degrading microbiota mediates pathogen clearance from the gut lumen
after non-typhoidal Salmonella diarrhea. PLoS Pathog 2010:6.
enzyme that was potentially transferred from a marine
bacterium, Zobellia galactanivorans into the genome of 13. Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN:
 Recognition of peptidoglycan from the microbiota by Nod1
Bacteroides plebeius, a commensal isolated from Japanese enhances systemic innate immunity. Nat Med 2010, 16:228-
individuals [77]. A combination of genomic approaches, 231.
This study demonstrates for the first time that microbial products can
functional screens, directed mutagenesis and gnotobiotic disseminate from the gut to systemic sites to influence host immunity.
models systems will enable us to disentangle the complex
14. Nell S, Suerbaum S, Josenhans C: The impact of the microbiota
microbiota–host–pathogen interaction-network explain- on the pathogenesis of IBD: lessons from mouse infection
ing CR and enteric disease. models. Nat Rev Microbiol 2010, 8:564-577.
15. Strober W, Fuss I, Mannon P: The fundamental basis of
Acknowledgements inflammatory bowel disease. J Clin Invest 2007, 117:514-521.
We are grateful to numerous colleagues for stimulating discussions and to 16. Arthur JC, Jobin C: The struggle within: microbial influences on
Yvonne Lötscher for assisting with figure design. Work was supported by colorectal cancer. Inflamm Bowel Dis 2010.
the Swiss National Science Foundation (310030-113623, 310030-132997 to
WDH), the UBS foundation (1004/A, to WDH) and the BMBF 17. Manichanh C, Reeder J, Gibert P, Varela E, Llopis M, Antolin M,
Infektionsgenomik (to BS). Guigo R, Knight R, Guarner F: Reshaping the gut microbiome
with bacterial transplantation and antibiotic intake. Genome
Res 2010.
References and recommended reading
Papers of particular interest, published within the period of review, 18. Ackermann M, Stecher B, Freed NE, Songhet P, Hardt WD,
have been highlighted as: Doebeli M: Self-destructive cooperation mediated by
phenotypic noise. Nature 2008, 454:987-990.
 of special interest 19. Vollaard EJ, Clasener HA: Colonization resistance. Antimicrob
 of outstanding interest Agents Chemother 1994, 38:409-414.
20. Stecher B, Hardt WD: The role of microbiota in infectious
1. Wilber K: The Eye of Spirit, An Integral Vision for a World Gone disease. Trends Microbiol 2008, 16:107-114.
Slightly Mad (1997). 21. Le Blay G, Rytka J, Zihler A, Lacroix C: New in vitro colonic
2. Foxman B, Goldberg D, Murdock C, Xi C, Gilsdorf JR: fermentation model for Salmonella infection in the child gut.
Conceptualizing human microbiota: from multicelled organ to FEMS Microbiol Ecol 2009, 67:198-207.
ecological community. Interdiscip Perspect Infect Dis 2008, 22. Momose Y, Hirayama K, Itoh K: Effect of organic acids on
2008:613979. inhibition of Escherichia coli O157:H7 colonization in
3. Moore WE, Holdeman LV: Human fecal flora: the normal flora of gnotobiotic mice associated with infant intestinal microbiota.
20 Japanese-Hawaiians. Appl Microbiol 1974, 27:961-979. Antonie Van Leeuwenhoek 2008, 93:141-149.

4. Lee A, Gordon J, Dubos R: Enumeration of the oxygen sensitive 23. Brogden KA, Cornick N, Stanton TB, Zhang Q, Nolan LK,
bacteria usually present in the intestine of healthy mice. Nature Wannemuehler MJ (Eds): Virulence Mechanisms of Bacterial
1968, 220:1137-1139. Pathogens. Washington, DC: ASM Press; 1997.

5. Turnbaugh PJ, Quince C, Faith JJ, McHardy AC, Yatsunenko T, 24. Fabich AJ, Jones SA, Chowdhury FZ, Cernosek A, Anderson A,
 Niazi F, Affourtit J, Egholm M, Henrissat B, Knight R et al.: Smalley D, McHargue JW, Hightower GA, Smith JT, Autieri SM
Organismal, genetic, and transcriptional variation in the et al.: Comparison of carbon nutrition for pathogenic and
deeply sequenced gut microbiomes of identical twins. Proc commensal Escherichia coli strains in the mouse intestine.
Natl Acad Sci U S A 2010, 107:7503-7508. Infect Immun 2008, 76:1143-1152.

Current Opinion in Microbiology 2011, 14:82–91 www.sciencedirect.com


Mechanisms controlling pathogen colonization Stecher and Hardt 89

25. Leatham MP, Banerjee S, Autieri SM, Mercado-Lubo R, Conway T, 42. Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y,
Cohen PS: Precolonized human commensal Escherichia coli Littman DR, Benoist C, Mathis D: Gut-residing segmented
strains serve as a barrier to E. coli O157:H7 growth in the filamentous bacteria drive autoimmune arthritis via T helper 17
streptomycin-treated mouse intestine. Infect Immun 2009, cells. Immunity 2010, 32:815-827.
77:2876-2886.
43. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH:
26. Apperloo-Renkema HZ, Van der Waaij BD, Van der Waaij D: Prolonged impact of antibiotics on intestinal microbial
Determination of colonization resistance of the digestive tract ecology and susceptibility to enteric Salmonella infection.
by biotyping of Enterobacteriaceae. Epidemiol Infect 1990, Infect Immun 2009, 77:2741-2753.
105:355-361.
44. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U,
27. Apperloo-Renkema HZ, van der Waaij D: Study of colonization Wei D, Goldfarb KC, Santee CA, Lynch SV et al.: Induction of
resistance for Enterobacteriaceae in man by experimental intestinal Th17 cells by segmented filamentous bacteria. Cell
contamination and biotyping as well as the possible role of 2009, 139:485-498.
antibodies in the clearance of these bacteria from the
intestines. Epidemiol Infect 1991, 107:619-626. 45. Stecher B, Macpherson AJ, Hapfelmeier S, Kremer M, Stallmach T,
Hardt WD: Comparison of Salmonella enterica serovar
28. van der Waaij D, Berghuis JM: Determination of the colonization Typhimurium colitis in germfree mice and mice pretreated with
resistance of the digestive tract of individual mice. J Hyg (Lond) streptomycin. Infect Immun 2005, 73:3228-3241.
1974, 72:379-387.
46. Muller AJ, Hoffmann C, Galle M, Van Den Broeke A,
29. Stecher B, Chaffron S, Kappeli R, Hapfelmeier S, Freedrich S, Heikenwalder M, Falter L, Misselwitz B, Kremer M, Beyaert R,
Weber TC, Kirundi J, Suar M, McCoy KD, von Mering C et al.: Like Hardt WD:: The S. Typhimurium effector SopE induces
will to like: abundances of closely related species can predict caspase-1 activation in stromal cells to initiate gut
susceptibility to intestinal colonization by pathogenic and inflammation. Cell Host Microbe 2009, 6:125-136.
commensal bacteria. PLoS Pathog 2010, 6:e1000711.
47. Norin E, Midtvedt T: Intestinal microflora functions in laboratory
30. Sonnenburg JL, Xu J, Leip DD, Chen CH, Westover BP,  mice claimed to harbor a ‘normal’ intestinal microflora is the
Weatherford J, Buhler JD, Gordon JI: Glycan foraging in vivo by SPF concept running out of date? Anaerobe 2010, 16:311-313.
an intestine-adapted bacterial symbiont. Science 2005, This review brings up an important issue regarding the standardisation of
307:1955-1959. the microbial flora of laboratory mice to increase experimental reprodu-
cibility in between different laboratories.
31. Rey FE, Faith JJ, Bain J, Muehlbauer MJ, Stevens RD,
 Newgard CB, Gordon JI: Dissecting the in vivo metabolic 48. Ley RE, Turnbaugh PJ, Klein S, Gordon JI: Microbial ecology:
potential of two human gut acetogens. J Biol Chem 2010, human gut microbes associated with obesity. Nature 2006,
285:22082-22090. 444:1022-1023.
A new method (RNAseq) is used to determine the microbial Metatran-
scriptome of acetogenic commensal bacteria in vivo 49. Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI:
The effect of diet on the human gut microbiome: a
32. Rawls JF, Mahowald MA, Ley RE, Gordon JI: Reciprocal gut metagenomic analysis in humanized gnotobiotic mice. Sci
microbiota transplants from zebrafish and mice to germ-free Transl Med 2009, 1(6):ra14.
recipients reveal host habitat selection. Cell 2006, 127:423-433.
50. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X,
33. Salzman NH, Hung K, Haribhai D, Chu H, Karlsson-Sjoberg J, Brown D, Stares MD, Scott P, Bergerat A et al.: Dominant and
Amir E, Teggatz P, Barman M, Hayward M, Eastwood D et al.: diet-responsive groups of bacteria within the human colonic
Enteric defensins are essential regulators of intestinal microbiota. ISME J 2010.
microbial ecology. Nat Immunol 2010, 11:76-83.
51. Lupp C, Robertson ML, Wickham ME, Sekirov I, Champion OL,
34. Salzman NH, Ghosh D, Huttner KM, Paterson Y, Bevins CL: Gaynor EC, Finlay BB: Host-mediated inflammation disrupts
Protection against enteric salmonellosis in transgenic mice the intestinal microbiota and promotes the overgrowth of
expressing a human intestinal defensin. Nature 2003, 422:522- enterobacteriaceae. Cell Host Microbe 2007, 2:119-129.
526.
52. Stecher B, Robbiani R, Walker AW, Westendorf AM, Barthel M,
35. Slack E, Hapfelmeier S, Stecher B, Velykoredko Y, Stoel M, Kremer M, Chaffron S, Macpherson AJ, Buer J, Parkhill J et al.:
Lawson MA, Geuking MB, Beutler B, Tedder TF, Hardt WD et al.: Salmonella enterica serovar Typhimurium exploits
Innate and adaptive immunity cooperate flexibly to maintain inflammation to compete with the intestinal microbiota. PLoS
host–microbiota mutualism. Science 2009, 325:617-620. Biol 2007, 5:e244.

36. Macpherson AJ, Uhr T: Induction of protective IgA by intestinal 53. Lawley TD, Bouley DM, Hoy YE, Gerke C, Relman DA,
dendritic cells carrying commensal bacteria. Science 2004, Monack DM: Host transmission of Salmonella enterica serovar
303:1662-1665. Typhimurium is controlled by virulence factors and indigenous
intestinal microbiota. Infect Immun 2008, 76:403-416.
37. Mazmanian SK, Round JL, Kasper DL: A microbial symbiosis
factor prevents intestinal inflammatory disease. Nature 2008, 54. Barman M, Unold D, Shifley K, Amir E, Hung K, Bos N, Salzman N:
453:620-625. Enteric salmonellosis disrupts the microbial ecology of the
murine gastrointestinal tract. Infect Immun 2008, 76:907-915.
38. Round JL, Mazmanian SK: Inducible Foxp3 + regulatory T-cell
development by a commensal bacterium of the intestinal 55. Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA,
microbiota. Proc Natl Acad Sci U S A 2010, 107:12204-12209.  Croucher N, Mastroeni P, Scott P, Raisen C, Mottram L et al.:
Antibiotic treatment of Clostridium difficile carrier mice
39. Brandl K, Plitas G, Mihu CN, Ubeda C, Jia T, Fleisher M, Schnabl B, triggers a supershedder state, spore-mediated transmission,
DeMatteo RP, Pamer EG: Vancomycin-resistant enterococci and severe disease in immunocompromised hosts. Infect
exploit antibiotic-induced innate immune deficits. Nature 2008, Immun 2009.
455:804-807. In this study a nice mouse model for C. difficile mediated antibiotic-
associated diarrhea is described which will be highly important for study-
40. Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, ing this still poorly understood disease in the future.
Bridonneau C, Rochet V, Pisi A, De Paepe M, Brandi G et al.: The
key role of segmented filamentous bacteria in the coordinated 56. Ma AT, Mekalanos JJ: In vivo actin cross-linking induced by
maturation of gut helper T cell responses. Immunity 2009, Vibrio cholerae type VI secretion system is associated with
31:677-689. intestinal inflammation. Proc Natl Acad Sci U S A 2010,
107:4365-4370.
41. Lee YK, Menezes JS, Umesaki Y, Mazmanian SK: Microbes and
Health Sackler Colloquium: proinflammatory T-cell responses 57. Cash HL, Whitham CV, Behrendt CL, Hooper LV: Symbiotic
to gut microbiota promote experimental autoimmune bacteria direct expression of an intestinal bactericidal lectin.
encephalomyelitis. Proc Natl Acad Sci U S A 2010. Science 2006, 313:1126-1130.

www.sciencedirect.com Current Opinion in Microbiology 2011, 14:82–91


90 Host–microbe interactions: bacteria

58. Brandl K, Plitas G, Schnabl B, DeMatteo RP, Pamer EG: MyD88- et al.: A catalog of reference genomes from the human
mediated signals induce the bactericidal lectin RegIII gamma microbiome. Science 2010, 328:994-999.
and protect mice against intestinal Listeria monocytogenes
infection. J Exp Med 2007, 204:1891-1900. 75. Goodman AL, McNulty NP, Zhao Y, Leip D, Mitra RD,
Lozupone CA, Knight R, Gordon JI: Identifying genetic
59. Dessein R, Gironella M, Vignal C, Peyrin-Biroulet L, Sokol H, determinants needed to establish a human gut symbiont in its
Secher T, Lacas-Gervais S, Gratadoux JJ, Lafont F, Dagorn JC habitat. Cell Host Microbe 2009, 6:279-289.
et al.: Toll-like receptor 2 is critical for induction of Reg3 beta
expression and intestinal clearance of Yersinia 76. Faith JJ, Rey FE, O’Donnell D, Karlsson M, McNulty NP,
pseudotuberculosis. Gut 2009, 58:771-776. Kallstrom G, Goodman AL, Gordon JI: Creating and
characterizing communities of human gut microbes in
60. Rodenburg W, Bovee-Oudenhoven IM, Kramer E, van der Meer R, gnotobiotic mice. ISME J 2010, 4:1094-1098.
Keijer J: Gene expression response of the rat small intestine
following oral Salmonella infection. Physiol Genomics 2007, 77. Hehemann JH, Correc G, Barbeyron T, Helbert W, Czjzek M,
30:123-133. Michel G: Transfer of carbohydrate-active enzymes from
marine bacteria to Japanese gut microbiota. Nature 2010,
61. Rodenburg W, Keijer J, Kramer E, Roosing S, Vink C, Katan MB, 464:908-912.
van der Meer R, Bovee-Oudenhoven IM: Salmonella induces
prominent gene expression in the rat colon. BMC Microbiol 78. Umesaki Y, Okada Y, Matsumoto S, Imaoka A, Setoyama H:
2007, 7:84. Segmented filamentous bacteria are indigenous intestinal
bacteria that activate intraepithelial lymphocytes and induce
62. Miller SI, Ernst RK, Bader MW: LPS: TLR4 and infectious disease MHC class II molecules and fucosyl asialo GM1 glycolipids on
diversity. Nat Rev Microbiol 2005, 3:36-46. the small intestinal epithelial cells in the ex-germ-free mouse.
Microbiol Immunol 1995, 39:555-562.
63. Peschel A: How do bacteria resist human antimicrobial
peptides? Trends Microbiol2002, 10:179-186. 79. Syed SA, Abrams GD, Freter R: Efficiency of various intestinal
bacteria in assuming normal functions of enteric flora after
64. Stecher B, Barthel M, Schlumberger MC, Haberli L, Rabsch W, association with germ-free mice. Infect Immun 1970, 2:376-386.
Kremer M, Hardt WD: Motility allows S. Typhimurium to benefit
from the mucosal defence. Cell Microbiol 2008, 10:1166-1180. 80. Itoh K, Freter R: Control of Escherichia coli populations by a
combination of indigenous clostridia and lactobacilli in
65. Raffatellu M, George MD, Akiyama Y, Hornsby MJ, Nuccio SP, gnotobiotic mice and continuous-flow cultures. Infect Immun
Paixao TA, Butler BP, Chu H, Santos RL, Berger T et al.: Lipocalin- 1989, 57:559-565.
2 resistance confers an advantage to Salmonella enterica
serotype Typhimurium for growth and survival in the inflamed 81. Momose Y, Maruyama A, Iwasaki T, Miyamoto Y, Itoh K: 16S rRNA
intestine. Cell Host Microbe 2009, 5:476-486. gene sequence-based analysis of clostridia related to
conversion of germfree mice to the normal state. J Appl
66. Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL, Microbiol 2009.
 Crawford RW, Russell JM, Bevins CL, Adams LG, Tsolis RM et al.:
Gut inflammation provides a respiratory electron acceptor for 82. Denou E, Rezzonico E, Panoff JM, Arigoni F, Brussow H: A
Salmonella. Nature 2010, 467:426-429. Mesocosm of Lactobacillus johnsonii, Bifidobacterium
The authors discovered a new mechanism how pathogens can exploit longum, and Escherichia coli in the mouse gut. DNA Cell Biol
host inflammatory responses in the gut to outcompete the commensal 2009, 28:413-422.
microflora.
83. Martins AK, Martins FS, Gomes DA, Elian SD, Vieira AT, Teixeira MM,
67. Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Cara DC, Nardi RM, Nicoli JR: Evaluation of in vitro antagonism
Pace NR: Molecular-phylogenetic characterization of microbial and of in vivo immune modulation and protection against
community imbalances in human inflammatory bowel diseases. pathogenic experimental challenge of two probiotic strains of
Proc Natl Acad Sci U S A 2007, 104:13780-13785. Bifidobacterium animalis var. lactis. Arch Microbiol 2010.

68. Van der Sluis M, De Koning BA, De Bruijn AC, Velcich A, 84. Dos Santos LM, Santos MM, de Souza Silva HP, Arantes RM,
Meijerink JP, Van Goudoever JB, Buller HA, Dekker J, Van Nicoli JR, Vieira LQ: Monoassociation with probiotic
Seuningen I, Renes IB et al.: Muc2-deficient mice Lactobacillus delbrueckii UFV-H2b20 stimulates the immune
spontaneously develop colitis, indicating that MUC2 is system and protects germfree mice against Listeria
critical for colonic protection. Gastroenterology 2006, monocytogenes infection. Med Microbiol Immunol 2010.
131:117-129.
85. Stappenbeck TS, Hooper LV, Gordon JI: Developmental
69. Hormannsperger G, Haller D: Molecular crosstalk of probiotic regulation of intestinal angiogenesis by indigenous microbes
bacteria with the intestinal immune system: clinical relevance via Paneth cells. Proc Natl Acad Sci U S A 2002, 99:15451-15455.
in the context of inflammatory bowel disease. Int J Med
Microbiol 2010, 300:63-73. 86. Hooper LV, Gordon JI: Commensal host–bacterial relationships
in the gut. Science 2001, 292:1115-1118.
70. Strober W, Kitani A, Fuss I, Asano N, Watanabe T: The molecular
basis of NOD2 susceptibility mutations in Crohn’s disease. 87. Hooper LV, Xu J, Falk PG, Midtvedt T, Gordon JI: A molecular
Mucosal Immunol 2008, 1(Suppl. 1):S5-9. sensor that allows a gut commensal to control its nutrient
foundation in a competitive ecosystem. Proc Natl Acad Sci U S
71. Garrett WS, Gallini CA, Yatsunenko T, Michaud M, DuBois A, A 1999, 96:9833-9838.
Delaney ML, Punit S, Karlsson M, Bry L, Glickman JN et al.:
Enterobacteriaceae act in concert with the gut microbiota to 88. Bry L, Falk PG, Midtvedt T, Gordon JI: A model of host–microbial
induce spontaneous and maternally transmitted colitis. Cell interactions in an open mammalian ecosystem. Science 1996,
Host Microbe 2010, 8:292-300. 273:1380-1383.

72. Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, 89. Sonnenburg JL, Chen CT, Gordon JI: Genomic metabolic
Barnich N, Bringer MA, Swidsinski A, Beaugerie L, Colombel JF: studies of the impact of probiotics on a model gut symbiont
High prevalence of adherent-invasive Escherichia coli and host. PLoS Biol 2006, 4:e413.
associated with ileal mucosa in Crohn’s disease.
Gastroenterology 2004, 127:412-421. 90. Mahowald MA, Rey FE, Seedorf H, Turnbaugh PJ, Fulton RS,
Wollam A, Shah N, Wang C, Magrini V, Wilson RK et al.:
73. Barnich N, Carvalho FA, Glasser AL, Darcha C, Jantscheff P, Characterizing a model human gut microbiota composed of
Allez M, Peeters H, Bommelaer G, Desreumaux P, Colombel JF members of its two dominant bacterial phyla. Proc Natl Acad
et al.: CEACAM6 acts as a receptor for adherent-invasive E. Sci U S A 2009, 106:5859-5864.
coli, supporting ileal mucosa colonization in Crohn disease. J
Clin Invest 2007, 117:1566-1574. 91. Crost EH, Pujol A, Ladire M, Dabard J, Raibaud P, Carlier JP,
Fons M: Production of an antibacterial substance in the
74. Nelson KE, Weinstock GM, Highlander SK, Worley KC, Creasy HH, digestive tract involved in colonization-resistance against
Wortman JR, Rusch DB, Mitreva M, Sodergren E, Chinwalla AT Clostridium perfringens. Anaerobe 2010.

Current Opinion in Microbiology 2011, 14:82–91 www.sciencedirect.com


Mechanisms controlling pathogen colonization Stecher and Hardt 91

92. Samuel BS, Gordon JI: A humanized gnotobiotic mouse model characteristics of the intestines of ex-germfree mice. Infect
of host–archaeal–bacterial mutualism. Proc Natl Acad Sci U S A Immun 1994, 62:5442-5446.
2006, 103:10011-10016.
102. Hudson JA, Luckey TD: Bacteria induced morphologic
93. Marco ML, Peters TH, Bongers RS, Molenaar D, van Hemert S, changes. Proc Soc Exp Biol Med 1964, 116:628-631.
Sonnenburg JL, Gordon JI, Kleerebezem M: Lifestyle of
Lactobacillus plantarum in the mouse caecum. Environ 103. Skelly BJ, Trexler PC, Tanami J: Effect of a Clostridium species
Microbiol 2009, 11:2747-2757. upon cecal size of gnotobiotic mice. Proc Soc Exp Biol Med
1962, 100:455-458.
94. Bohnhoff M, Drake BL, Miller CP: Effect of streptomycin on
susceptibility of intestinal tract to experimental Salmonella 104. Dubos R, Schaedler RW, Costello R, Hoet P: Indigenous normal
infection. Proc Soc Exp Biol, NY 1954, 86:132-137. and autochthonous flora of the gastrointestinal tract. J Exp
Med 1965, 122:67-76.
95. van der Waaij D, Berghuis-de Vries JM, Lekkerkerk Lv:
Colonization resistance of the digestive tract in conventional 105. Freter R, Abrams GD: Function of various intestinal bacteria in
and antibiotic-treated mice. J Hyg (Lond) 1971, 69:405-411. converting germfree mice to the normal state. Infect Immun
1972, 6:119-126.
96. Barthel M, Hapfelmeier S, Quintanilla-Martinez L, Kremer M,
Rohde M, Hogardt M, Pfeffer K, Russmann H, Hardt WD: 106. Koopman JP, Kennis HM, Mullink JW, Prins RA, Stadhouders AM,
Pretreatment of mice with streptomycin provides a Salmonella De Boer H, Hectors MP: ‘Normalization’ of germfree mice with
enterica serovar Typhimurium colitis model that allows anaerobically cultured caecal flora of ‘normal’ mice. Lab Anim
analysis of both pathogen and host. Infect Immun 2003, 1984, 18:188-194.
71:2839-2858.
107. Arank A, Syed SA, Kenney EB, Freter R: Isolation of anaerobic
97. Sekirov I, Tam NM, Jogova M, Robertson ML, Li Y, Lupp C,
bacteria from human gingiva and mouse cecum by means of
Finlay BB: Antibiotic-induced perturbations of the intestinal
a simplified glove box procedure. Appl Microbiol 1969,
microbiota alter host susceptibility to enteric infection. Infect
17:568-576.
Immun 2008, 76:4726-4736.
98. Nardi RM, Silva ME, Vieira EC, Bambirra EA, Nicoli JR: 108. Schaedler RW, Dubos R, Costello R: The development of the
Intragastric infection of germfree and conventional mice bacterial flora in the gastrointestinal tract of mice. J Exp Med
with Salmonella typhimurium. Braz J Med Biol Res 1989, 1965, 122:59-66.
22:1389-1392.
109. Wilson KH, Sheagren JN, Freter R, Weatherbee L, Lyerly D:
99. Itoh K, Mitsuoka T: Production of gnotobiotic mice with normal Gnotobiotic models for study of the microbial ecology of
physiological functions I. Selection of useful bacteria from Clostridium difficile and Escherichia coli. J Infect Dis 1986,
feces of conventional mice. Z Versuchstierkd 1980, 22:173-178. 153:547-551.

100. Koopman JP, Kennis HM, Lankhorst A, Welling GW, Hectors MP, 110. Dewhirst FE, Chien CC, Paster BJ, Ericson RL, Orcutt RP,
Nagengast F: ‘Normalization’ of germfree mice after direct and Schauer DB, Fox JG: Phylogeny of the defined murine
indirect contact with mice having a ‘normal’ intestinal microbiota: altered Schaedler flora. Appl Environ Microbiol
microflora. Lab Anim 1986, 20:286-290. 1999, 65:3287-3292.
101. Okada Y, Setoyama H, Matsumoto S, Imaoka A, Nanno M, 111. Stehr M, Greweling MC, Tischer S, Singh M, Blocker H,
Kawaguchi M, Umesaki Y: Effects of fecal microorganisms and Monner DA, Charles WC: River altered Schaedler flora (CRASF)
their chloroform-resistant variants derived from mice, rats, remained stable for four years in a mouse colony housed in
and humans on immunological and physiological individually ventilated cages. Lab Anim 2009, 43:362-370.

www.sciencedirect.com Current Opinion in Microbiology 2011, 14:82–91

You might also like