Vous êtes sur la page 1sur 15

Current Pharmaceutical Design, 2009, 15, 2051-2065 2051

The Intestinal Environment in Health and Disease – Recent Insights on the


Potential of Intestinal Bacteria to Influence Human Health

Sam Possemiers1,#,*, Charlotte Grootaert1,#, Joan Vermeiren1,#, Gabriele Gross1,2,#,


Massimo Marzorati1, Willy Verstraete1 and Tom Van de Wiele1,#

1
Laboratory of Microbial Ecology and Technology (LabMET), Ghent University, Belgium and 2Advanced Food
Microbiology, Unilever R&D Vlaardingen, The Netherlands

Abstract: The human intestine is colonized by a complex microbial ecosystem, which could be considered as a separate
organ within the human host, having a coding capacity which exceeds the liver by a factor 100. On the one hand, this ex-
tensive microbiome is closely involved in the first-pass metabolism and bioavailability of food and drug compounds. Un-
derstanding to which extent each individual’s gut microbiota affects the bioavailability and response to orally adminis-
tered drugs is therefore a first important challenge towards novel drug development strategies. On the other hand, as our
microbiota is directly or indirectly involved in the onset of a number of disease states, a new generation of therapeutics
may be developed that affect the structure and functioning of the intestinal microbiota and interfere with their specific
cross-talk with the human host. Ultimately, the intestinal microbiota may even be used as a biomarker for impending dis-
eases inside or outside the gastrointestinal tract and for the evaluation of responses to specific therapeutic interventions.
This review will therefore highlight the importance of the indigenous microbial community and its enormous metabolic
potential, microbe-microbe interactions, mechanisms of host-bacterium cross-talk and will discuss the onset of obesity, a
specific disease state in which the role of intestinal bacteria becomes more and more apparent. Understanding the impor-
tance of the intestinal ecosystem in these phenomena may open the door for new strategies which target the management
of the intestinal microbiome into the desired direction and therefore to a completely new type of nutrition research and
pharmaceutical design.

1. INTRODUCTION Driven by selection forces at both microbial and host


level, the microbial community has coevolved in a mutualis-
For many years, it was believed that the main function of
tic relation with the human host with the capacity to commu-
the large intestine was the resorption of water and salt by the
nicate with other species and with the host. Due to this sig-
body and the facilitated disposal of waste materials. How-
naling system, the microbiome is closely involved in the
ever, this task definition proved to be far from complete, as it
host’s health, for instance by the extraction of energy from
did not consider the activity of the microbial content of the
large intestine. Indeed, the human colon harbors a highly otherwise indigestible compounds [4], the stimulation of the
gut immune system [5], the regulation of cell proliferation
complex microbial ecosystem of about 200 g living cells, at
[6], the synthesis of essential vitamins K and B [7] and by
concentrations of 1012 microorganisms per gram gut content,
enhancing pathogen resistance [8]. On the other hand, spe-
the highest recorded for any microbial habitat [1]. As this
cific community assemblage may also be seen as a risk fac-
number is approximately 10 times greater than the total
tor contributing to a disease state. This is shown by recent
number of our somatic and germ cells, it seems appropriate
to view ourselves as a composite of human cells and bacteria reports linking intestinal bacteria with diseases ranging from
allergies [9] to bowel inflammation [10] and obesity [11].
and our genetic landscape as a ‘metagenome’, an amalgam
of genes embedded in our genome and in the genomes of all When we consider our body therefore as the host of a
our microbial partners [2]. It is estimated that the collection complex and prominent microbiome, this opens up a totally
of all microbial genomes in the gut comprises between 2 new dimension of potential therapeutic strategies. On the one
million and 4 million genes, which is 70-140 times more hand, the extensive gene pool present in the intestinal micro-
than that of their host. This ‘microbiome’ encompasses all biome is closely involved in the first-pass metabolism of
genes that are responsible for numerous processes such as compounds that are poorly or incompletely absorbed by the
substrate breakdown, protein synthesis, biomass production, gut. Understanding to which extent each individual’s gut
production of signaling molecules, antimicrobial compounds microbiota affects the bioavailability and response to orally
and encodes biochemical pathways that humans have not administered drugs is therefore a first important challenge
evolved [3]. Thus, the intestinal microbiota can be regarded towards new drug development strategies. On the other hand,
as a separate organ within the human host, which is capable as our microbiota is directly or indirectly involved in the
of even more conversions than the human liver. onset of a number of disease states, a new generation of
therapeutics may be developed that affects the structure and
*Address correspondence to this author at the LabMET – UGent. Coupure functioning of the intestinal microbiota and interferes with
Links 653, B-9000 Gent, Belgium; Tel: +32 9 264 59 76; Fax: + 32 9 264 their specific cross-talk with the human host. Ultimately, the
62 48; E-mail: sam.possemiers@ugent.be intestinal microbiota may even be used as a biomarker for
#
These authors equally contributed to the manuscript.

1381-6128/09 $55.00+.00 © 2009 Bentham Science Publishers Ltd.


2052 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

impending diseases inside or outside the gastrointestinal tract ria are thus allowed to have a direct contact with the host
and for the evaluation of responses to specific therapeutic (adhesion), thereby positively activating innate and adaptive
interventions. This review will therefore highlight the impor- immunity [18]. The diversity found in the gastrointestinal
tance of the indigenous microbial community and its enor- tract, namely a few divisions represented by very tight clus-
mous metabolic potential, the mechanisms of bacterium-host ters of related bacteria, may therefore reflect strong host se-
interaction and a specific disease state in which the role of lection for specific bacteria whose emergent collective be-
intestinal bacteria becomes more and more apparent. havior is beneficial for the host [14].

2. THE INTESTINAL MICROBIOTA: A WORLD IN- 2.2. Mechanisms of Cross-Talk between the Colonic Mi-
SIDE US crobiota and Its Host
2.1. Structure and Functional Organization The gastrointestinal tract is the site in the body where the
divergent needs of nutrient absorption on the one hand and
One of the most important accomplishments of Life Sci-
host defense and host-bacterium cross-talk on the other hand
ences in the last decades was the completion of the Human
collide. Whereas nutrient absorption requires a thin epithe-
Genome Project, which characterized over 25 000 genes in
lium and large surface area, optimal communication with
the human cell. Five years ago, the US National Institutes for
commensal bacteria and simultaneous protection against
Health started another ambitious project which has the aim pathogen invasion requires an extensive and selective de-
of exploring the vast microbial community that interacts with
fense system. The investment by the gut in protecting itself
the human host: the Human Microbiome Project [2]. The use
is evident in the abundant lymphoid tissue and immune cells
of advanced culture-independent approaches [12, 13] and of
it harbors. At the same time, multiple mechanisms exist for
new ecological theories about evolutionary forces shaping
host-bacterium cross-talk, ultimately influencing human
the microbial community in the intestine [6, 14, 15] have
health. It is therefore important to understand how these ef-
already provided new insights into the structure of our mi- fects are mediated, and how bacteria communicate with
crobial ecosystem. The adult human gastrointestinal tract
epithelial cells in the intestine.
contains all three domains of life, bacteria, archaea and eu-
karya, with the largest community residing in our colon. Al- In general, gut bacteria can interact with the host by
though 55 and 13 divisions of respectively bacteria and ar- common mechanisms like attachment to the mucosal surface
chaea have been described, only 8 bacterial divisions have or by excretion of soluble factors, but many species have
been identified so far in the gastrointestinal tract and the gut developed unique approaches of bacterial-epithelial cros-
microbiota is dominated by only two bacterial divisions, the stalk, thereby inducing specific immune responses [19, 20].
Cytophaga-Flavobacterium-Bacteroides (bacteroides) and It has been proposed that this cross-kingdom cell-to-cell sig-
Firmicutes (clostridia, eubacteria, …), with lower levels of naling could also involve small molecules, such as hormones
Actinobacteria (bifidobacteria), and by mainly one member that are produced by the eukaryotes (adrenaline, noradrena-
of the archaea, Methanobrevibacter smithii. At this level, the line,…) and hormone-like chemicals (autoinducer signal 3 -
intestinal communities of all humans therefore appear quite AI-3 and acyl homoserine lactones) that are produced by
similar. However, within these divisions, a limited number bacteria for quorum sensing purposes [21]. In many cases
of lineages terminate in broad, shallow radiations comprising however, the varying underlying molecular mechanisms are
hundreds of species and thousands of strains, making the not yet completely understood. Importantly, innate immune
microbiota of an individual as personalized as a fingerprint recognition of endogenous bacteria plays a crucial role in
[14, 15]. maintaining intestinal homeostasis under healthy conditions
[22, 23]. To sense bacteria that are present in the intestinal
The unique shape of this microbial ecosystem is related
tract, the host innate immune system is able to recognize
to a specific functional organization. The structure and com-
specific molecular structures that are unique to pathogenic as
position of this ecosystem reflects a natural selection at both
well as non-pathogenic microorganisms, referred to as Mi-
microbial and host levels, which promote a mutual coopera-
crobe-Associated Molecular Patterns [24]. The best-charac-
tion in the search of a functional stability [16]. Indeed, a re-
terized class of pattern recognition receptors are the Toll-like
cent paper from Turnbaugh et al. [17] showed that, although receptors (TLR) that elicit adequate inflammatory and antim-
the community composition may be as individual as a fin-
icrobial responses after activation by their microbial ligands
gerprint, a ‘core’ gut microbiome would exist at the level of
[24, 25]. For example, lipoteichoic acids and bacterial DNA
shared functional genes. It has been proposed that such equi-
can exert immunomodulatory properties towards the host
librium requires a series of evolved, nested equilibria to
mediated by respectively TLR2 and TLR9 [26-28]. Currently
achieve the overall homeostasis, in analogy with the ‘Evolu-
known molecular modes of action of probiotic bacteria -
tionary Stable Strategy’ theory. In other words, microbial added to functional food products to improve host health and
success in a host requires the ability to grow and overcome
well-being - have been reviewed elsewhere [29]. For ins-
the host’s defenses, yet cannot affect the fitness of the host,
tance, interactions of lactobacilli with the pattern-recogni-
as this would ultimately reduce the number of hosts and
tion receptor DC-SIGN (dendritic cell-specific intercellular
thereby lead to loss of the bacteria’s own habitat [16]. Such a
adhesion molecule 3-grabbing non-integrin) and with man-
selection primarily occurs along mucosal surfaces via a host-
nose-residues on the epithelial surface have been described
microbiota cross-talk that leads to the modulation of host [30, 31]. Although recently various studies have been pub-
immunity. The gastrointestinal system has to constantly deal
lished on cross-talk of intestinal bacteria with the host im-
with a large number of bacteria and only those recognized as
mune system, the complex connections between the diversity
“friendly” are considered to be tolerated. Commensal bacte-
of bacterial triggers and the various components of possible
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2053

host responses are not yet fully understood (reviewed in Due to the use of germ-free animals in the majority of
Corthesy et al. [18] and Medhzitov et al. [24]). studies, evidence on the impact of similar mechanisms in the
presence of an endogenous microbiota in conventional ani-
After the initial contact between microbes and epithelial
mals is limited. However, modulation of host gene expres-
cells, the host organism may react to these bacterial signals.
Different bacterial triggers in the intestine can induce a vari- sion by commensal species in germ-free animals demon-
strates the importance of the composition of the intestinal
ety of host responses, which are reflected in changes in gene
microbiota for physiological functions of the host [35, 37].
expression and can be detected simultaneously by for exam-
Mucosal immune responses are highly adapted to the pres-
ple transcriptome analyses. Several studies using this ap-
ence of large numbers of endogenous bacteria in the lower
proach have been performed with pathogenic, commensal
intestine, constantly present in close proximity to host tissues
and probiotic bacterial species, employing different host
models [30, 32-34]. Colonization of germ-free mice with [41]. This is indicated by the fact that germ-free mice pos-
sess small and underdeveloped Peyer’s patches and that the
commensal and probiotic bacteria such as B. thetaiotaomi-
restitution of these mice with a microbial microbiota is suffi-
cron, Bifidobacterium longum and some Lactobacillus spp.
cient to restore the mucosal immune system [9]. One of the
has been shown to cause substantial changes in gene expres-
future challenges of gastrointestinal microbiology research
sion of the host, including the induction of host genes in-
will be to study the regulatory mechanisms within this con-
volved in innate immunity, mucosal barrier function, nutrient
absorption, and intestinal maturation [35-37]. stant co-existence and its implications for health in more
detail.
One interesting example of a gene whose expression has
been shown to be differently affected by interactions with 2.3. Towards an Integrative Approach to Study the Intes-
various bacterial species in different animal models is tinal Microbiota and Host-Microbe Interactions
RegIII (human counterpart: pancreatitis-associated protein,
PAP). RegIII/PAP has been proposed to be directly antimi- As many specific interactions of intestinal bacteria with
the host have not been elucidated yet, molecular approaches
crobial and could therefore play a role during increased bac-
are needed to identify mechanisms at both bacterial and host
terial-epithelial contact to limit potential microbial penetra-
level (Fig. 1) by which specific bacteria may confer defined
tion and maintain mucosal integrity [38]. RegIII/PAP
health benefits [29]. Genome sequence analyses of an in-
(mRNA) expression has been shown to be up- or downregu-
creasing number of bacterial strains provide the tools to
lated, respectively, by B. thetaiotaomicron, E. coli and Sal-
monella, segmented filamentous bacteria, Schaedler’s E. study genetic parameters that determine their functions re-
lated to host health [57-59]. Currently, specific bacterial
coli, Listeria innocua, Lactobacillus plantarum, and B.
genes can be characterized by the construction of (multiple)
longum [30, 32, 37-40]. Interestingly, the ability of bacteria
gene-specific mutant strains in an isogenic background [60].
to repress host expression of this bactericidal protein may
This offers much more elegant and targeted research strate-
possibly promote their own survival in the gut and influence
gies than the former use of spontaneous mutants with unde-
the microbial ecology [37].

Fig. (1). Schematic overview of host-microbe interactions in the intestine and currently available (molecular) methods to investigate various
factors involved. References indicate studies for which a similar approach has been applied.
2054 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

fined genetic alterations [53, 61]. Furthermore, it has become bacterium interactions was developed to study the joint me-
possible to assess responses of particular strains to gastroin- tabolism between liver and gut microbiota. In an elegant
testinal conditions in terms of bacterial gene expression dur- two-compartment reactor, Laube et al. succeeded in the co-
ing passage through the gastrointestinal tract [43, 62]. culture of hepatocytes and fecal microbiota under aerobic
With regard to gut microbial ecology, several molecular and anaerobic conditions, respectively [75]. This way, the
sequential metabolism of 7-ethoxycoumarin and a thiadiazi-
high-throughput techniques are currently available to moni-
non derivative by hepatocytes and fecal bacteria was investi-
tor intestinal bacterial population dynamics such as 16S ribo-
gated in vitro.
somal DNA terminal restriction fragment pattern analysis
(T-RFLP), denaturing gradient gel electrophoresis (DGGE), Strategies which target the intestinal microbiota may
phylogenetic microarrays as well as metagenomics and consist of affecting or introducing specific bacterial species
metaproteomics approaches [12, 47, 63-65]. Most of these or strains, or may lead to modulation of the composition or
culture-independent methods are based on phylogenetic activity of the general microbial ecosystem. A typical exam-
analysis of rRNA genes and can provide important insights ple of the introduction of specific bacterial strains is the use
into the types of bacteria present in the gastrointestinal tract, of engineered lactic acid bacteria as mucosal-delivery vehi-
their distribution, functionality and development under cer- cles and vaccine carriers, expressing immunomodulatory
tain conditions [66]. At present, comparative metagenomics proteins [76]. Several applications of lactic acid bacteria
have been used e.g. in the Human Microbiome Project to delivery have already been tested in animal models, targeting
find out whether all humans have an identifiable ‘core’ mi- the possible prevention and treatment of Inflammatory
crobiome (see above) and to compare intestinal microbial Bowel Disease (IBD), allergies and different infections (re-
community composition among different populations [2]. viewed in [77]). In many cases however, the final health ef-
Linking gut microbial community composition profiles to fect cannot be attributed to one specific species, but is the
e.g. host metabolomics data can indicate key functional mi- result of a complex interplay between various bacteria,
crobiome members that are particularly important for host which interact through various mechanisms with the host. In
metabolism and health [67]. analogy with the recently developed ecological concept of
Microbial Resource Management [78, 79], which provides a
Concerning the host, the rapidly progressing develop-
number of tools that can help in the interpretation of ecologi-
ment of whole genome sequences and commercially avail-
cal surveys, we therefore introduce the concept of Gastroin-
able microarrays for increasing numbers of species improves
the possibilities to study transcriptomes which illustrate host testinal Resource Management (GRM), i.e. the management
of the complex gut microbiota and its metabolism with the
responses upon environmental and e.g. bacterial stimuli [32,
aim of improving the health of the host (Fig. 2).
68, 69]. However, truly integrated approaches, which focus
on both specific bacterial genes potentially involved in a The rationale behind this is that microorganisms acting as
particular cross-talk mechanism as well as on their molecular consortia are comparable with human beings working to-
effects on the host, are still scarce [30, 50]. gether in organizations and firms, requiring human resource
management. To put the basics of GRM in perspective one
In order to gain more insight in this complex host-
needs to know:
bacterium interplay and to understand how this can affect
human health, the development and application of appropri- a) Who is there? The genomic methods currently allow the
ate model systems will be very important. Several in vitro listing of the various organisms present. Besides the mo-
model systems have been developed to study the intestinal lecular analysis of microbial communities, one should
microbiota as a complex ecosystem and its metabolic poten- also explore new ways to isolate new species by new cul-
tial. Examples of well-designed models are the Simulator of tivation approaches.
the Human Intestinal Microbial Ecosystem (SHIME) [70,
b) Who is doing what? There are a variety of new ap-
71], a similar three-step colon model [72] and the EnteroMix
proaches to identify the actuators [43-47]. All these
model [73]. Such systems enable fast reproducible experi-
methods will allow making up the “catalogues” of the
ments under controlled conditions and can be used to study species involved in the different processes. Yet, they will
the metabolic potential of intestinal bacteria and microbe-
only give a limited picture of the overall ”happiness” and
microbe interactions such as horizontal gene transfer. Vali-
”functioning” of the microbial consortia involved.
dation of the results may be obtained by using well-
controlled in vivo models for the human gut, such as germ- c) Who is doing what with whom? To deal with complex
free and human microbiota-associated animal models [74]. groups of living entities, it is required to know the inter-
These results should help to formulate and direct hypothesis- relationships. We need to learn about the standard type
based research of the microbiota’s metabolome in humans. exchanges between different groups of organisms, par-
ticularly about types of “trading” of electron donors
To study specific host-bacterium cross-talk, optimal ap-
and/or acceptors.
proaches should include in vitro as well as in vivo experi-
ments to investigate possible health effects and to integrate If we can link the answer to those three questions to-
mechanistic exploratory indications with the complex multi- gether, this will allow to understand, and therefore also to
factorial interactions that occur in vivo. To do this, a number manage, what makes the intestinal community to act in mu-
of in vivo models have been developed in which specific tualism with the host and which factors trigger the intestinal
genes are switched off (e.g. leptin-deficient ob/ob mice) or microbiome to become a pathogenic one. Understanding the
which are colonized with specific microbiota. One particu- structure and function of the intestinal ecosystem as a whole,
larly interesting example of an in vitro model to study host- will therefore be an important step in finding new drug tar-
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2055

Fig. (2). Introducing the ecological concept of Gastrointestinal Resource Management, the management of the complex gut microbiome with
the aim to improve host health. Central questions are who is there, who is doing what with whom and how can one adjust, control and/or
steer these mixed cultures and communities.

gets which help to manage and steer the intestinal micro- human health. A selected class of food compounds that will
biome into the desired direction. shortly be described here as a good example of biologically
active compounds, is the group of (poly-)phenols. The daily
3. METABOLIC POTENTIAL OF INTESTINAL MI- intake ranges from 100 mg to over 2 g and primarily origi-
CROBIOTA AND ITS IMPLICATIONS FOR HUMAN nates from the consumption of coffee, black tea, fruits and
HEALTH vegetables. From the ingested polyphenols, only 5% is ab-
3.1. Microbial Metabolism in the Intestine sorbed in the duodenum and only 5% of the absorbed frac-
tion, mainly flavanols, reaches the blood circulation intact.
As the colon microbial community receives a large diver- The rest of the absorbed fraction is conjugated through the
sity of non- or partly digested food components and host liver biotransformation process. The 90 to 95% of the in-
excretion products as nutrient and energy source, the set of gested compounds together with the biliary secreted conju-
bacterial genes that code for metabolic enzymes is highly gated compounds reach the colon where they become avail-
diverse and redundant to ensure the metabolism of many able to the resident microbiota (reviewed by Clifford [83]).
different substrates under the fluctuating nutritional condi- The extensive transformations taking place include the re-
tions. Carbohydrates, resistant to digestion, drive colonic moval of sugars, removal of phenolic hydroxyl groups, fis-
bacterial fermentation and the resulting end products (SCFA) sion of aromatic rings, and further degradation [84]. Many
are considered beneficial for the host. In contrast, when pro- microbial metabolites have been found and include phenols
teins are fermented, the end products include toxic com- and aromatic/phenolic acids/lactones possessing 0, 1 or 2
pounds, such as amines and phenols [80]. Besides carbohy- phenolic hydroxyl groups and up to five carbons in the side
drates and proteins, many other components, such as plant chain (reviewed by [83]).
chemicals, food or environmental contaminants and xenobi-
otics in general also become available to the gut microbiota. Flavonoids are the best studied polyphenols and can be
It has been shown that the gut microbial community has the divided into several groups, of which flavanols and proan-
ability to metabolize xenobiotics far more extensively than thocyanidins are generally unconjugated, whereas the other
any other part of the body (reviewed by [81]). This microbial substances normally occur as glycosides [83]. Amongst
factor in the metabolism of ingested chemicals has often these, isoflavones, some flavanones and stilbenes, some lig-
been overlooked, yet the last decade of scientific research nans and some coumarins are characterized as phytoestro-
has brought new insights and surprising findings. Ilett dem- gens for their putative estrogenic and/or anti-androgenic
onstrated that gut bacterial metabolism has a strong potential properties. These phytoestrogens (isoflavones from soy,
for both bioactivation and detoxification of xenobiotics [82]. prenylflavonoids from hop, coumestans and lignans from
Table 1 presents the different types of microbial enzymes in fruits and cereals) are typically considered as a good exam-
the gut. In contrast to the oxidative and conjugative nature of ple of the importance of intestinal metabolism of food com-
liver metabolism, which generates hydrophilic high molecu- pounds. This is related to the fact that the potency of phy-
lar weight biotransformation products, the metabolic nature toestrogens to prevent or modulate hormone-dependent dis-
of the gut microbial community in an anaerobic environment eases such as breast- and prostate cancer, or osteoporosis and
is mainly reductive and hydrolytic, generating non-polar low menopausal complaints, is largely governed by the presence
molecular weight byproducts [81]. Yet, oxidative reactions of specific intestinal microorganisms. Lignans only show
can also occur. estrogenic properties after metabolism by colon microorgan-
isms. Hop contains the most powerful phytoestrogen known
3.2. Implications for the Activity of Dietary Constituents thus far, namely 8-prenylnaringenin (8-PN). Rowland et al.
discussed the role of the gut microbial community in the
Foodstuffs represent a significant source of components
bioavailability and metabolism of estrogens in general and
that have the potential to positively or negatively influence
2056 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

Table 1. Metabolic Potency of Human Gastrointestinal Microbiota (After: Ilett et al. [82])

Reactions Enzyme Bacterial Species / Origin of Sample

Hydrolysis

Glucuronides -Glucuronidase Escherichia coli

Glycosides -Glucosidase Enterococcus faecalis, Eubacterium rectale, Clostridium sphenoides

Amides Amide hydrolase E. coli, Bacillus subtilis

Esters Deacetylase E. faecalis

Sulfamates Arylsulfotransferase Clostridia, enterobacteria, enterococci

Reduction

Azo-compounds Azoreductase Clostridia, lactobacilli

Unsaturated lacton Unsat. glycoside hydrogenase Eubacterium lentum

Aliphatic double bounds Unsat. fatty acid hydrogenase E. faecalis

Nitro-compounds Nitroreductase E. coli, bacteroides

N-oxides N-oxide reductase Human colon

S-oxides Sulfoxide reductase E. coli

Ketones Hydrogenase Rat caecum

Hydroxylamines Nitroreductase Rat GIT

Dehydroxylation

Demethylaton Demethylase Enterococci, lactobacilli, clostridia

Deamination Deaminase Bacteroides, clostridia

Decarboxylation Decarboxylase E. faecalis

Dehydrogenation Dehydrogenase Clostridium welchii

Dehalogenation Dehalogenase E. coli, Aerobacter aerogenes

Synthetic Reactions

Esterification Acetyltransferase E. coli

N-nitrosation E. faecalis, E. coli

Other Reactions

Oxidation Oxidase E. coli, E. faecalis

Isomerization Isomerase E. rectale, C. sphenoides

Fission aliphatic Tryptophanase E. coli, Bacillus alvei

Activity towards cystein or GSH conjugates C-S lyase Pig GIT, Eubacterium aerofaciens

phytoestrogens more in particular [85]. Intestinal bacteria logical activity (Fig. 3) [86]. For example, equol, 8-PN and
contribute to the bioavailability and biological activity of enterodiol and enterolacton are bacterial metabolites from
phytoestrogens in a number of ways. Firstly, intestinal bacte- daidzein (soy) [87], isoxanthohumol (hop) [88] and secoiso-
ria produce ß-glucosidases thereby cleaving the naturally lariciresinol diglucoside (fruit/cereal lignans) [89, 90], re-
occurring glycosylated phytoestrogens into an aglycon. Sec- spectively and have a much higher biological activity than
ondly, the gut bacteria elicit glucuronidase and sulfatase ac- their precursor product. A remarkable aspect of the microbial
tivity, thereby deconjugating phase II metabolites from the conversion of daidzein into equol and of isoxanthohumol
liver and releasing aglycons again. Thirdly and most impor- into 8-PN is the large interindividual variability in the con-
tantly, intestinal bacteria are capable of transforming the version efficiency [52]. This was nicely illustrated for equol
original components to metabolites that have a higher bio- [91] and 8-PN production [92].
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2057

Fig. (3). The most relevant groups of phytoestrogens, the isoflavones, lignans and prenylflavonoids, are present in food as less active precur-
sors. Inside the human body, they can be activated into the respective active metabolites equol, enterodiol + enterolacton and 8-
prenylnaringenin. Although some transformations are of chemical origin (C) or catalyzed by human enzymes (H), microbial enzymes (M)
have a crucial role in the generation of the final metabolites for each group of phytoestrogens (after Possemiers, et al. [86]).

Other examples of gut microbial polyphenol metabolism cooking. Additionally, guinea pig experiments showed that
include for instance proanthocyanidins, which are complex the hydrolysis of benzo(a)pyrene conjugates by the intestinal
flavonoid polymers naturally present in fruits, legume seeds, microbiota resulted in de novo production of toxic
cereal grains, and different beverages (wine, tea, cocoa/ benzo(a)pyrene intermediates which caused the formation of
chocolate, cider) (reviewed by Santos-Buelga et al. [93]), DNA adducts in the colon [96].
and are, therefore, an integral part of the human diet. The
The ability of gut bacteria to metabolize drugs came un-
most common proanthocyanidins are procyanidins, which
der the attention of pharmaceutical companies since an acci-
consist of flavan-3-ol (catechin, epicatechin) elementary
dent in 1993 with the antiviral drug orivudine revealed that
units with the typical C6-C3-C6 flavonoid skeleton. Procya-
its gut microbial metabolite (E)-5-(2-bromovinyl)uracil in-
nidins are not/poorly absorbed through the intestinal barrier, terfered with the clearance of a co-administered anti-cancer
but they are degraded by the colonic microbiota into low
drug 5-fluoro-uracil. This resulted in the death of 18 patients
molecular weight phenolic acids [94], which are more read-
[97]. Recently, Sousa et al. reviewed the conversion of over
ily absorbed.
30 drug compounds by gut microorganisms and the related
consequences for their biological effect in the human body
3.3. Implications for the Activity of Xenobiotics
[81]. Examples are the reduction of omeprazole and digoxin,
Besides the interest in the microbial conversion of health- hydrolysis of lactulose and sorivudine, acetylation of 5-
promoting components from ingested foodstuffs, much at- aminosalicylic acid, proteolysis of insulin and calcitonin and
tention is also given to the role of intestinal bacteria in the N-demethylation of methamphetamine. Additionally, de-
conversion of ingested drugs, environmental or food con- methylation, deamination, decarboxylation and dehalogena-
taminants or xenobiotics in general. Firstly, the intestinal tion reactions have also been described, next to other reac-
microbiota may interfere with the clearance of ingested tions such as aromatization, esterification and N-nitrosation
xenobiotics through deconjugation reactions of phase II [82].
biotransformation products from enterocytes and primarily
Quite remarkably, gut bacteria can also perform oxida-
hepatocytes. This results in the enterohepatic circulation of
tive reactions despite a reducing intestinal environment. For
xenobiotic compounds. Compounds absorbed in the intestine
instance, Enterococcus faecalis can use micromolar amounts
and subsequently detoxified are usually conjugated with po-
of oxygen, diffusing across the epithelium from the serosal
lar groups (glucuronic acid, glycine, sulfate, glutathion and to the luminal side, to form hydroxylated radicals from D-
taurine) in the liver prior to secretion with the bile [82]. Once
phenylalanine and N-tert-butylnitrone. This may increase the
released in the intestinal lumen, bacterial enzymes such as -
oxidative stress on the intestinal epithelium [98]. Further-
glucuronidase, sulfatase and other glycosidases may hydro-
more, IQ is converted to the direct acting mutagen 7-
lyze these conjugates again. This would negate the detoxifi-
hydroxy-IQ [99] by bacteria belonging to predominant intes-
cation cycle and delay the excretion of many exogenous
tinal populations, such as B. thetaiotaomicron, Clostridium
compounds since the original compounds or phase I metabo- clostridiiforme and E. coli [100], although a large interindi-
lites are more prone to intestinal absorption than their phase
vidual variability exists. As a third example, oxidative reac-
II conjugates. In one particular study, it was shown that gut
tions were also described with the hydroxylation of poly-
microbial -glucuronidase activity increased the genotoxicity
cyclic aromatic hydrocarbons (PAH) by in vitro cultured
of 2-amino-3-methylimidazo[4,5-f]quinoline (IQ) [95], a
intestinal microbiota [101]. Hydroxylation of PAH gives
food-borne carcinogen that is formed in meat and fish during
these compounds an affinity for the human estrogen recep-
2058 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

tor, so that they may interfere with normal hormone-driven food constituents may modulate the gut microbial commu-
processes in the body. Therefore, microbial oxidation prod- nity composition, thereby affecting the metabolic potency of
ucts of PAH may actively interfere with the host’s physiol- the microbiota as such, but also influencing the host
ogy. biotransformation capacity.
Finally, also the microbial modifications of food con-
3.5. Perspectives
taminants may interfere with intestinal health. For example,
2-Amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), a In summary, the true scale of the metabolic potential of
carcinogenic heterocyclic aromatic amine formed in meat the gut microorganisms is only recentlty becoming evident.
products during cooking, is converted to PhIP-M1 by En- Not only is this important for e.g. the biological activity of
terococcus faecium strains and Lactobacillus reuteri [102]. many functional food components, but it also has toxicologi-
Whether it is a detoxification is still a matter of debate, since cal consequences when assessing the risks from ingested
PhIP-M1 has been shown to be cytotoxic toward Caco-2 contaminants or the pharmacological effects of administerd
cells but is not mutagenic in the Ames assay [103]. As a sec- drugs. Mikov pointed out the importance of gut microbiota
ond example, incubation of arsenobetaine, an important or- stating that the gut microbial metabolism must be considered
ganoarsenical compound in fish and shellfish, with intestinal an integral part of drug/xenobiotic metabolism and toxicity
microorganisms results in dimethylarsinic acid, dimethylars- studies [109]. New developments in metabonomics covering
inoylacetic acid and trimethylarsine oxide production. The the metabolic potency of the entire gut microbiome will re-
toxicological consequences of these metabolites in the colon veal new metabolites and pathways of biotransformation and
environment are not known. In addition, metal(loid)s such as biodegradation. Additionally, emphasis needs to be put on
As, Se, Te, Bi have been shown to be volatilized through the interindividual variability in terms of microbial commu-
methylation (e.g. As(CH3)3) and hydride formation (AsH3 ) nity composition and hence, also in its metabolic capabili-
(Diaz and Van de Wiele, submitted). Methanoarcheae such ties. Li et al. concluded from their work that interindividual
as M. smithii are now proposed as an important player in this differences in xenobiotic metabolism are associated to a
process [104]. This is of important toxicological significance large extent with the gut microbiota composition, which pre-
as volatile metal(loid)s easily diffuse across the gut epithe- disposes a different pathophysiological outcome upon die-
lium and thus become bioavailable to the host. tary factors or chemical stimuli [110]. Clearly, unraveling
the metabolic pathways of the gut microbiota but also ex-
3.4. Indirect Effects of the Intestinal Microbiota on Host ploring the interindividual differences presents an enormous
Metabolism challenge for the scientific community.
During their research of microbe-host interactions,
4. HOST-BACTERIUM CROSS-TALK: GUT BACTE-
Hooper et al. made an important observation concerning the
RIA AND OBESITY
effect from gut bacteria towards the host [105]. Colonization
of germ-free rats by B. thetaiotaomicron resulted in modula- 4.1. Obesity and the Intestinal Microbial Community
tion of epithelial cell gene expression involved in several
Obesity has been recognized as one of the most important
important functions. This not only involved mucosal barrier
growing problems in our current society leading towards
formation, angiogenesis and nutrient absorption but, impor-
health problems such as diabetes type II, certain types of
tantly, also xenobiotic metabolism. In addition, Li et al. de-
cancer and cardiovascular diseases [111]. Besides an im-
monstrated a "transgenomic" approach to link gut micro-
paired balance between energy uptake and physical activity,
biome and metabolic phenotype (metabotype) variation.
Combining spectroscopic, microbiomic, and multivariate the composition of the intestinal microbiota has recently
been presented as a factor playing a substantial role in the
statistical tools, they analyzed fecal and urinary samples
development of obesity. Ley et al. showed that the state of
from different individuals and modeled the microbe-host
obesity determines the composition of the intestinal micro-
metabolic connectivity. They introduced the concept of func-
bial community [112]. Both mice and human trials revealed
tional metagenomics by which they characterized key func-
that obese individuals had a decrease of up to 50% in the
tional members of the microbiome, such as Faecalibacte-
rium prausnitzii, that most influence host metabolism and abundance of bacteria belonging to the division of the Bac-
teroidetes, and a proportional increase in Firmicutes, com-
hence health [48]. Other examples of bacterial modulation of
pared to lean individuals. Moreover, this proportion of Bac-
host metabolism include the studies performed by Villarini et
teroidetes to Firmicutes increased with weight loss on low-
al. who recently showed that Lactobacillus casei decreased
calorie diets [113]. A corresponding decrease in the propoer-
the production of liver xenobiotic metabolizing enzymes
tion of Bacteroidetes was also observed in a recent study on
thereby lowering the genotoxicity of 1,2-dimethylhydrazine
[106]. Additionally, lactulose fermentation products influ- 154 individuals with, in this case, also a significant increase
in Actinobacteria [17]. It has also been demonstrated that the
ences the activities of liver and intestinal xenobiotic metabo-
composition of the microbial community may also determine
lizing enzymes in rats inoculated with Clostridium parapu-
the state of obesity [114]. In this study, the intestinal content
trificum [107]. Furthermore, Humblot et al. showed that
of both lean and genetically modified ob/ob mice (with
Brussels sprouts, oligosaccharides, and fermented milk low-
leptin deficiency and thus developing obesity) was isolated,
ered the genotoxicity of 2-amino-3-methylimidazo[4,5-
f]quinoline by changing not only the composition of the in- and transplanted in normal, germ-free mice. It was observed
that mice, conventionalized with microbiota originating from
testinal microbial community, but also the xenobiotic me-
obese mice, had a higher percentage increase in body fat
tabolizing enzymes [108]. Thus, supplementation of func-
over two weeks than mice conventionalized with intestinal
tional foods under form of dietary fibers, probiotics or other
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2059

bacteria from lean mice. Furthermore, it was shown that a presence of glucose in the intestine also enhances the GLP-1
high fat diet alone is not sufficient to induce obesity [11], production in the L-cells [116]. These mechanisms may ex-
indicating that host-bacterium cross-talk may play an essen- plain why gut peptide modulation is only observed with
tial role in the host’s fat storage. highly fermentable fibers [131].
Given this potentially crucial role of intestinal bacterial
4.3. Gut Peptides Involved in fat Storage
in the process of metabolic homeostasis, i.e. the control of
energy uptake and metabolism in the body, the next para- In vivo studies with germ-free and conventionalized mice
graphs will discuss the potential mechanisms of this host- revealed that intestinal bacteria may not only impact the in-
bacterium cross-talk and which future strategies may be en- take of energy but that they may also modulate the host’s fat
visaged to prevent or treat obesity and metabolic disorders storage by repressing the intestinal production of a cytokine
by targeting the intestinal microbiota (Table 2). involved in this process, called the fasting-induced adipose
factor (FIAF) [4]. Once secreted in the blood, FIAF inhibits
4.2. Gut Peptides Involved in Appetite Control the activity of lipoprotein lipase, an enzyme responsible for
the conversion of triglycerides to monoglycerides and fatty
Appetite control is a first important factor in the concept
acids from circulating lipoproteins [118, 132]. As a conse-
of metabolic homeostasis and involves the production of a
number of gut peptides. Glucagon-like peptide 1 (GLP-1) quence, these triglycerides cannot be stored in the fat tissue,
resulting in a lower body weight [4]. The fact that germ-free
and peptide YY (PYY) are satiety stimulating hormones,
mice showed elevated serum FIAF concentration and that
released in response to nutrient ingestion by L-cells in
conventionalization of these animals with intestinal bacteria
mainly ileum and colon. GLP-1 promotes insulin secretion
decreased FIAF levels and increased fat storage and weight
and pancreatic -cell proliferation and controls glycogen
gain, led to the conclusion that intestinal bacteria may impact
synthesis in muscle cells [115], while PYY slows down gas-
tric emptying. In contrast, ghrelin stimulates appetite and is metabolic homeostasis through this process and may there-
fore be a potentially important future target in the protection
mainly produced by P/D1 cells in the stomach and -cells of
against obesity [11]. Until now, it is not known which
the pancreas [126]. Non-digestible carbohydrates, such as
mechanisms in this host-bacterium cross-talk occur during
oligofructose [127], lactitol [128] and resistant starch [117],
the repression of FIAF production by the gut epithelium. It
are effective to induce satiety by modulating the production
was observed that, when conventionalizing germ-free mice
of the gut peptides GLP-1, PYY and ghrelin through a
mechanism that also involves modulation of the intestinal with B. thetaiotaomicron and/or M. smithii, FIAF production
was more repressed by the presence of a combination of both
microbial community [129]. Rat studies demonstrated that an
micro-organisms, than by each of them separately [11].
oligofructose-enriched diet significantly increased GLP-1
However, the mechanism remains unclear. Furthermore, it is
and decreased ghrelin production, and doubled the number
not known whether different microbial species have the same
of GLP-1-expressing cells in the proximal colon [127, 130].
FIAF repressing potential.
Lactitol mainly increased PYY production in rats and hu-
mans [128], while resistant starch significantly affected both
4.4. Interference with Lipid Metabolism and Nutrient
PYY and GLP-1 production [117]. The latter authors also
Uptake
demonstrated that bacterial regulation of gut peptides is me-
diated by short chain fatty acids produced from indigestible Several pro- and prebiotics have been shown to alter se-
substrates. Physiological concentrations of acetate, propion- rum cholesterol and lipid levels, thereby not only affecting
ate and butyrate, but also a pH decrease from 7.5 to 6, sig- fat storage, but also the development of cardiovascular dis-
nificantly increased proglucagon and PYY in the entero- eases [133]. These effects are often attributed to the produc-
endocrine colon cell line STC-1 [117]. Furthermore, the tion of propionate, which inhibits hepatic cholesterol synthe-

Table 2. Examples of Mechanisms, Conventional Drugs, Potential Microbial Alternatives and Possible Advantages of a Microbial
Approach to Prevent or Treat Obesity

Example of Potential Microbial Advantages of the


Mechanism Target References
Conventional Drugs Alternative Microbial Alternative

GLP-1
GLP-1, PYY Pre/probiotics stimulating glucose No rapid degradation,
Satiety gut peptides PYY [115-117]
and analogues release and SCFA production no injection needed
Ghrelin

Fenofibrate, Only intestinal, not in


Lipase inhibitors FIAF Intestinal microbiota [4, 118, 119]
thiazolidinediones other organs

Cholesterol, Prebiotics stimulating propionate,


Liver lipogenesis Statines No muscle damage [120-123]
Fatty acids bile salt-degrading probiotics

No lactic acidosis,
Liver gluconeogenesis Glucose, Insuline Metformin Prebiotics [124, 125]
no hypoglycemia
2060 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

sis from acetate [123, 134, 135]. Furthermore, intestinal bac- high blood pressure. Furthermore, the use of gut peptides is
teria interfere with cholesterol absorption from the gut by not evident because of their rapid enzymatic degradation
deconjugating bile salts [136] or by directly assimilating after injection [115, 117]. As the previous paragraphs show
cholesterol, as reviewed by St-Onge et al. [122]. Examples that intestinal microbiota may have an important role in both
of probiotic bacteria influencing cholesterol and lipid me- the intake of energy (appetite control), uptake of sugars and
tabolism are Lactococcus lactis [137], Streptococcus ther- fat from the gut, energy metabolism in the body and fat stor-
mophilus [138, 139], Lactobacillus acidophilus [140], E. age in the adipose tissue, alternative strategies may be devel-
faecium [139], Bifidobacterium bifidum [141, 142] and B. oped in which the specific host-bacterium cross-talk in-
longum [143]. Bacteria present in fermented foods, such as volved in these processes is targeted. Into this respect, pre-
bacteroides, bifidobacteria, fusobacteria, clostridia, pep- and probiotics seem to be particularly promising as can be
tostreptococci, lactobacilli, and streptococci are known to deduced from rapidly expanding scientific evidence.
hydrolyze conjugated bile acids [144]. Several in vivo stud-
ies confirmed the cholesterol and lipid lowering effects of 5. CONCLUDING REMARKS
oligofructose [120, 145-147] and resistant starch [148, 149],
If we accept the view of our body as a functional system
representing a prebiotic approach.
composed of both human cells and bacterial species, with a
Hepatic lipogenesis is not only regulated by short chain prominent role for the intestinal microbiome, an enormous
fatty acids, but also by serum glucose and insulin levels potential for future drug development is offered. The differ-
[150]. Enhanced sugar uptake has been observed in presence ent topics described above clearly show that the intestinal
of gut bacteria compared to germ-free conditions, which can microbiota may have a profound role in health and disease
be explained by several mechanisms. First of all, the pres- and the true impact of our microbial ecosystem on human
ence of an intestinal microbial community leads towards an health is yet far from fully understood. Targeting the intesti-
increase in the amount of capillaries that underlie the small nal microbiota may therefore offer unique prevention or
intestinal villus epithelium [151]. Secondly, host monosac- treatment strategies for a variety of disease states in which
charide transporters are induced by the polysaccharide- the microbial factor is related to its metabolic potential, mi-
processing activity of the microbiota, as was demonstrated crobe-microbe interactions or host-bacterium cross-talk. This
by studies with germ-free mice colonized with B. thetaio- review aimed at describing these factors, based on specific
taomicron [35]. The monomers generated from indigestible disease states in which existing scientific evidence shows an
polysaccharides are delivered as substrates for lipid produc- important role of the intestinal microbiota.
tion in the liver. Besides, they can also activate the lipogenic
One of the important drivers for investigating and under-
enzymes in the liver by ChREBP- and SREBP-1- mediated
standing the metabolic ability of gut microbiota, is the modu-
mechanisms [4]. Hence, the polysaccharide-degrading poten-
lating effect it has towards the biological activity of food and
tial of an intestinal microbial community is considered an
pharmaceutical compounds. The examples described in this
important determinant for hepatic lipid production. In obese
review illustrate that the gut microbial metabolic potency is
ob/ob mice, the intestinal microbial community is enriched sometimes a crucial variable when assessing the benefits or
for genes that are able to harvest calories from complex
risks from ingested compounds and drug substances.
plant-derived polysaccharides compared to lean mice. These
genes code for enzymes involved in sugar degradation, sugar Horizontal gene transfer is an important mechanism by
transport and acetate and butyrate production [114]. Al- which bacteria may interact with each other and efficiently
though more and more insight is gained in the importance of exchange information (microbe-microbe interactions). This
polysaccharide degradation in the host’s fat production and does not only enable bacteria to adapt themselves to rapidly
storage, it is neither evident nor desirable to modulate the changing environmental conditions, but is also an important
composition of the microbial community to completely in- contributing factor to for instance the rapidly spreading anti-
hibit polysaccharide degradation and thus obesity. However, biotic resistance between bacteria under clinical conditions.
modulation of the sugar uptake by bacteria so that a slower Whereas antibiotic treatment is still considered a standard
glucose release and glycemic response is obtained, might be treatment strategy in case of pathogen infections, thorough
of interest. Several studies demonstrate that both oligofruc- understanding of microbe-microbe interactions such as gene
tose and resistant starch are effective against obesity-induced transfer systems may offer important alternative strategies to
diabetes type II, because they reduce glycemia [152], im- counteract infectious diseases [155]. Originally susceptible
prove glucose tolerance and insulin sensitivity [125, 149], pathogens may acquire resistance factors during antibiotic
and lower endotoxaemia caused by lipopolysaccharides from therapy, thereby reducing the treatment’s efficiency. Simul-
Gram-negative intestinal bacteria, all factors which may lead taneous administration of antibiotics with inhibitors of hori-
towards obesity and type II diabetes [129]. zontal gene transfer may prevent the acquisition of these
resistance factors, making that such inhibitors could offer
4.5. Perspectives interesting future applications that selectively target bacteria
capable of transferring antibiotic resistance and generating
Conventional anti-obesity drugs induce weight losses by
multidrug resistant strains [156]. This may be an important
several mechanisms, such as suppressing appetite (gut pep-
step in the prevention of resistance propagation during clini-
tides and analogues, anorectics), interference with nutrient cal treatment of bacterial infections and in extending the
absorption (pancreatic lipase inhibitors) and increasing the
lifetime of our antibiotic arsenal.
host’s metabolism (cannaboid receptor antagonists) [153,
154]. Although effective, some of these drugs may cause Whereas modern medicine has managed, at least in the
undesired side effects, such as oily bowel movements and a westernized world, to take control over most infectious dis-
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2061

ease of the gut, gastrointestinal food allergies and different SHIME = Simulator of the Human Intestinal Microbial
intestinal inflammatory conditions have dramatically in- Ecosystem
creased. Although the reason for this remains unknown, a
TLR = Toll-like receptor
prevailing notion is that we now face inflammation without
infection. This may even be related to the lack of sufficient T-RFLP = Terminal restriction fragment pattern analysis
gut infection and stimulation of the immune system (the so-
called ‘hygiene hypothesis’), thereby disturbing the balance REFERENCES
and the cross-talk between the normal bacteria which colo- References 158-160 are related articles recently published.
nize the gut and the host immune system [9]. However, more
and more evidences arise that disturbance in the microbial [1] Whitman WB, Coleman DC, Wiebe WJ. Prokaryotes: The unseen
majority. Proc Natl Acad Sci USA 1998; 95: 6578-83.
ecosystem and the host-bacterium cross-talk would not only [2] Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R,
be involved in disease states of the gut itself but also in the Gordon JI. The human microbiome project. Nature 2007; 449: 804-
rest of our body. Typical examples are the findings that in- 10.
testinal microbiota may play a crucial role in the onset and [3] Egert M, de Graaf AA, Smidt H, de Vos WM, Venema K. Beyond
development of obesity [114, 157]. diversity: functional microbiomics of the human colon. Trends Mi-
crobiol 2006; 14: 86-91.
Recent findings on - for instance - the role of intestinal [4] Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al.
bacteria in the latter disease states have shown that changes The gut microbiota as an environmental factor that regulates fat
occur in the general ecosystem structure rather than the storage. Proc Natl Acad Sci USA 2004; 101: 15718-23.
[5] Salminen S, Bouley C, Boutron-Ruault MC, Cummings JH, Franck
(dis)appearance of one particular species. For this reason, A, Gibson GR, et al. Functional food science and gastrointestinal
treatment strategies should also target the intestinal microbi- physiology and function. Br J Nutr 1998; 80: S147-71.
ota as a multi-cellular structure. A deeper insight in its popu- [6] Dethlefsen L, Eckburg PB, Bik EM, Relman DA. Assembly of the
lation dynamics, variability and structure-function relation- human intestinal microbiota. Trends Ecol Evol 2006; 21: 517-23.
ships will lead to a better understanding of the extent to [7] Conly JM, Stein K. Quantitative and qualitative measurements of
which each individual’s gut microbiota affects the bioavail- vitamin K in human intestinal contents. Am J Gastroenterol 1992;
87: 311-6.
ability and response to administered drugs and the impact of [8] Hopkins MJ, Macfarlane GT. Nondigestible oligosaccharides en-
interventions that alter our microbial ecology. hance bacterial colonization resistance against Clostridium difficile
in vitro. Appl Environ Microbiol 2003; 69: 1920-7.
ACKNOWLEDGEMENTS [9] MacDonald TT, Monteleone G. Immunity, inflammation, and
allergy in the gut. Science 2005; 307: 1920-5.
Sam Possemiers and Tom Van de Wiele benefit from a [10] Elson CO, Cong Y, Qi F, Hershberg RM, Targan SR. Molecular
post-doctoral grant from the Research foundation – Flanders Approaches to the role of the microbiota in inflammatory bowel
(FWO Vlaanderen). Joan Vermeiren is supported by a Con- disease. Ann NY Acad Sci 2006; 1072: 39-51.
certed Research Action of the Flemish Community (GOA) [11] Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mecha-
nisms underlying the resistance to diet-induced obesity in germ-
(GOA/24J/BAS/DOC/DOS/PDO/ZAP). Charlotte Grootaert free mice. Proc Natl Acad Sci USA 2007; 104: 979-84.
profits from a IWT (Institute for the Promotion of Innovation [12] Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, Samuel
by Science and Technology in Flanders) scholarship. We BS, et al. Metagenomic analysis of the human distal gut micro-
acknowledge the financial support of the European Commu- biome. Science 2006; 312: 1355-9.
nity under the Marie Curie Transfer of Knowledge – Indus- [13] Zoetendal EG, Collier CT, Koike S, Mackie RI, Gaskins HR. Mo-
try-Academia Strategic Partnership Scheme, MTKI-CT- lecular ecological analysis of the gastrointestinal microbiota: a re-
view. J Nutr 2004; 134: 465-72.
2006-042786 (GUTSYSTEM), especially for the second- [14] Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI.
ment of Gabriele Gross to Gent University. Host-bacterial mutualism in the human intestine. Science 2005;
307: 1915-20.
ABBREVIATIONS [15] Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary
forces shaping microbial diversity in the human intestine. Cell
DGGE = Denaturing gradient gel electrophoresis 2006; 124: 837-48.
[16] O'Hara AM, Shanahan F. The gut flora as a forgotten organ.
FIAF = fasting-induced adipose factor EMBO Rep 2006; 7: 688-93.
GLP-1 = Glucagon-like peptide 1 [17] Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A,
Ley RE, et al. A core gut microbiome in obese and lean twins. Na-
GRM = Gastrointestinal resource management ture 2009; 457: 480-4.
[18] Corthesy B, Gaskins HR, Mercenier A. Cross-talk between probi-
IBD = Inflammatory Bowel Disease otic bacteria and the host immune system. J Nutr 2007; 137: 781S-
IQ = 2-Amino-3-methylimidazo[4,5-f]quinoline 90.
[19] Dogi CA, Galdeano CM, Perdigon G. Gut immune stimulation by
PAH = Polycyclic aromatic hydrocarbons non pathogenic Gram(+) and Gram(-) bacteria. Comparison with a
probiotic strain. Cytokine 2008; 41: 223-31.
PAP = Pancreatitis-associated protein [20] Lu L, Walker WA. Pathologic and physiologic interactions of
bacteria with the gastrointestinal epithelium. Am J Clin Nutr 2001;
PhIP = 2-Amino-1-methyl-6-phenylimidazo[4,5- 73: 1124S-30.
b]pyridine [21] Hughes DT, Sperandio V. Inter-kingdom signalling: communica-
8-PN = 8-Prenylnaringenin tion between bacteria and their hosts. Nat Rev Microbiol 2008; 6:
111-20.
PYY = Peptide YY [22] Kelly D, Conway S, Aminov R. Commensal gut bacteria: mecha-
nisms of immune modulation. Trends Immunol 2005; 26: 326-33.
SCFA = Short-chain fatty acids
2062 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

[23] Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, [44] Goldenberg O, Herrmann S, Marjoram G, Noyer-Weidner M, Hong
Medzhitov R. Recognition of commensal microflora by toll-like re- G, Bereswill S, et al. Molecular monitoring of the intestinal flora
ceptors is required for intestinal homeostasis. Cell 2004; 118: 229- by denaturing high performance liquid chromatography. J Micro-
41. biol Methods 2007; 68: 94-105.
[24] Medzhitov R. Recognition of microorganisms and activation of the [45] Gross G, Wildner J, Schonewille A, Rademaker JL, van der Meer
immune response. Nature 2007; 449: 819-26. R, Snel J. Probiotic Lactobacillus plantarum 299v does not coun-
[25] Takeda K, Akira S. TLR signaling pathways. Semin Immunol teract unfavorable phytohemagglutinin-induced changes in the rat
2004; 16: 3-9. intestinal microbiota. Appl Environ Microbiol 2008; 74: 5244-9.
[26] Frances R, Munoz C, Zapater P, Uceda F, Gascon I, Pascual S, et [46] Jacobs DM, Deltimple N, van Velzen E, van Dorsten FA, Bingham
al. Bacterial DNA activates cell mediated immune response and ni- M, Vaughan EE, et al. 1H NMR metabolite profiling of feces as a
tric oxide overproduction in peritoneal macrophages from patients tool to assess the impact of nutrition on the human microbiome.
with cirrhosis and ascites. Gut 2004; 53: 860-4. NMR Biomed 2007; 21: 615-26.
[27] Grangette C, Nutten S, Palumbo E, Morath S, Hermann C, Dewulf [47] Klaassens ES, de Vos WM, Vaughan EE. Metaproteomics ap-
J, et al. Enhanced antiinflammatory capacity of a Lactobacillus proach to study the functionality of the microbiota in the human in-
plantarum mutant synthesizing modified teichoic acids. Proc Natl fant gastrointestinal tract. Appl Environ Microbiol 2007; 73: 1388-
Acad Sci USA 2005; 102: 10321-6. 92.
[28] Rachmilewitz D, Katakura K, Karmeli F, Hayashi T, Reinus C, [48] Li M, Wang BH, Zhang MH, Rantalainen M, Wang SY, Zhou HK,
Rudensky B, et al. Toll-like receptor 9 signaling mediates the anti- et al. Symbiotic gut microbes modulate human metabolic pheno-
inflammatory effects of probiotics in murine experimental colitis. types. Proc Natl Acad Sci USA 2008; 105: 2117-22.
Gastroenterology 2004; 126: 520-8. [49] Macpherson AJ, Uhr T. Induction of protective IgA by intestinal
[29] Marco ML, Pavan S, Kleerebezem M. Towards understanding dendritic cells carrying commensal bacteria. Science 2004; 303:
molecular modes of probiotic action. Curr Opin Biotechnol 2006; 1662-5.
17: 204-10. [50] Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis
[30] Gross G, van der Meulen J, Snel J, van der Meer R, Kleerebezem factor prevents intestinal inflammatory disease. Nature 2008; 453:
M, Niewold TA, et al. Mannose-specific interaction of Lactobacil- 620-5.
lus plantarum with porcine jejunal epithelium. FEMS Immunol [51] Palmer C, Bik EM, Digiulio DB, Relman DA, Brown PO. Devel-
Med Microbiol 2008; 54: 215-23. opment of the human infant intestinal microbiota. PLoS Biol 2007;
[31] Smits HH, Engering A, van der Kleij D, de Jong EC, Schipper K, 5: e177.
van Capel TM, et al. Selective probiotic bacteria induce IL-10- [52] Possemiers S, Bolca S, Eeckhaut E, Depypere H, Verstraete W.
producing regulatory T cells in vitro by modulating dendritic cell Metabolism of isoflavones, lignans and prenylflavonoids by intes-
function through dendritic cell-specific intercellular adhesion tinal bacteria: producer phenotyping and relation with intestinal
molecule 3-grabbing nonintegrin. J Allergy Clin Immunol 2005; community. FEMS Microbiol Ecol 2007; 61: 372-83.
115: 1260-7. [53] Pretzer G, Snel J, Molenaar D, Wiersma A, Bron PA, Lambert J, et
[32] Niewold TA, Kerstens HH, van der Meulen J, Smits MA, Hulst al. Biodiversity-based identification and functional characterization
MM. Development of a porcine small intestinal cDNA micro-array: of the mannose-specific adhesin of Lactobacillus plantarum. J Bac-
characterization and functional analysis of the response to entero- teriol 2005; 187: 6128-36.
toxigenic E. coli. Vet Immunol Immunopathol 2005; 105: 317-29. [54] Ten Bruggencate SJ, Bovee-Oudenhoven IM, Lettink-Wissink ML,
[33] Rodenburg W, Bovee-Oudenhoven IM, Kramer E, van der Meer R, Van der Meer R. Dietary fructooligosaccharides increase intestinal
Keijer J. Gene expression response of the rat small intestine follow- permeability in rats. J Nutr 2005; 135: 837-42.
ing oral Salmonella infection. Physiol Genomics 2007; 30: 123-33. [55] Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV.
[34] Troost FJ, van Baarlen P, Lindsey P, Kodde A, de Vos WM, Paneth cells directly sense gut commensals and maintain homeosta-
Kleerebezem M, et al. Identification of the transcriptional response sis at the intestinal host-microbial interface. Proc Natl Acad Sci
of human intestinal mucosa to Lactobacillus plantarum WCFS1 in USA 2008; 105: 20858-63.
vivo. BMC Genomics 2008; 9: 374. [56] van de Wiele T, Boon N, Possemiers S, Jabobs H, Verstraete W.
[35] Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon JI. Inulin-type fructans of longer degree of polymerization exert more
Molecular analysis of commensal host-microbial relationships in pronounced in vitro prebiotic effects. J Appl Microbiol 2007; 102:
the intestine. Science 2001; 291: 881-4. 452-60.
[36] Lebeer S, Vanderleyden J, De Keersmaecker SCJ. Genes and [57] Altermann E, Russell WM, Azcarate-Peril MA, Barrangou R, Buck
molecules of lactobacilli supporting probiotic action. Microbiol BL, McAuliffe O, et al. Complete genome sequence of the probi-
Mol Biol Rev 2008; 72: 728-+. otic lactic acid bacterium Lactobacillus acidophilus NCFM. Proc
[37] Sonnenburg JL, Chen CT, Gordon JI. Genomic and metabolic Natl Acad Sci USA 2005; 102: 3906-12.
studies of the impact of probiotics on a model gut symbiont and [58] Klaenhammer TR, Barrangou R, Buck BL, Azcarate-Peril MA,
host. PLoS Biol 2006; 4: 2213-26. Altermann E. Genomic features of lactic acid bacteria effecting
[38] Cash HL, Whitham CV, Behrendt CL, Hooper LV. Symbiotic bioprocessing and health. FEMS Microbiol Rev 2005; 29: 393-409.
bacteria direct expression of an intestinal bactericidal lectin. [59] Kleerebezem M, Boekhorst J, van Kranenburg R, Molenaar D,
Science 2006; 313: 1126-30. Kuipers OP, Leer R, et al. Complete genome sequence of Lactoba-
[39] Keilbaugh SA, Shin ME, Banchereau RF, McVay LD, Boyko N, cillus plantarum WCFS1. Proc Natl Acad Sci USA 2003; 100:
Artis D, et al. Activation of RegIIIbeta/gamma and interferon 1990-5.
gamma expression in the intestinal tract of SCID mice: an innate [60] Lambert JM, Bongers RS, Kleerebezem M. Cre-lox-based system
response to bacterial colonisation of the gut. Gut 2005; 54: 623-9. for multiple gene deletions and selectable-marker removal in Lac-
[40] Rodenburg W, Keijer J, Kramer E, Roosing S, Vink C, Katan MB, tobacillus plantarum. Appl Environ Microbiol 2007; 73: 1126-35.
et al. Salmonella induces prominent gene expression in the rat co- [61] Mack DR, Ahrne S, Hyde L, Wei S, Hollingsworth MA. Extracel-
lon. BMC Microbiol 2007; 7: 84. lular MUC3 mucin secretion follows adherence of Lactobacillus
[41] Macpherson AJ, Geuking MB, McCoy KD. Immune responses that strains to intestinal epithelial cells in vitro. Gut 2003; 52: 827-33.
adapt the intestinal mucosa to commensal intestinal bacteria. [62] Marco ML, Bongers RS, de Vos WM, Kleerebezem M. Spatial and
Immunology 2005; 115: 153-62. temporal expression of Lactobacillus plantarum genes in the gas-
[42] Armougom F, Raoult D. Use of pyrosequencing and DNA bar- trointestinal tracts of mice. Appl Environ Microbiol 2007; 73: 124-
codes to monitor variations in Firmicutes and Bacteroidetes com- 32.
munities in the gut microbiota of obese humans. BMC Genom [63] Kaplan CW, Astaire JC, Sanders ME, Reddy BS, Kitts CL. 16S
2008; 9: 576. ribosomal DNA terminal restriction fragment pattern analysis of
[43] Bron PA, Grangette C, Mercenier A, de Vos WM, Kleerebezem M. bacterial communities in feces of rats fed Lactobacillus acidophilus
Identification of Lactobacillus plantarum genes that are induced in NCFM. Appl Environ Microbiol 2001; 67: 1935-9.
the gastrointestinal tract of mice. J Bacteriol 2004; 186: 5721-9.
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2063

[64] Wang RF, Beggs ML, Erickson BD, Cerniglia CE. DNA microar- of the beer flavonoid isoxanthohumol. Oxford: Elsevier 2009; pp.
ray analysis of predominant human intestinal bacteria in fecal sam- 523-39.
ples. Mol Cell Probes 2004; 18: 223-34. [87] Decroos K, Vanhemmens S, Cattoir S, Boon N, Verstraete W.
[65] Zoetendal EG, Rajilic-Stojanovic M, de Vos WM. High-throughput Isolation and characterisation of an equol-producing mixed micro-
diversity and functionality analysis of the gastrointestinal tract mi- bial culture from a human fecal sample and its activity under gas-
crobiota. Gut 2008; 57: 1605-15. trointestinal conditions. Arch Microbiol 2005; 183: 45-55.
[66] Frank DN, Pace NR. Gastrointestinal microbiology enters the me- [88] Possemiers S, Heyerick A, Robbens V, De Keukeleire D, Ver-
tagenomics era. Curr Opin Gastroenterol 2008; 24: 4-10. straete W. Activation of proestrogens from hops (Humulus lupulus
[67] Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, et al. L.) by intestinal microbiota; Conversion of isoxanthohumol into 8-
Symbiotic gut microbes modulate human metabolic phenotypes. prenylnaringenin. J Agric Food Chem 2005; 53: 6281-8.
Proc Natl Acad Sci USA 2008; 105: 2117-22. [89] Clavel T, Henderson G, Alpert CA, Philippe C, Rigottier-Gois L,
[68] Dalma-Weiszhausz DD, Warrington J, Tanimoto EY, Miyada CG. Dore J, et al. Intestinal bacterial communities that produce active
The affymetrix GeneChip platform: an overview. Methods Enzy- estrogen-like compounds enterodiol and enterolactone in humans.
mol 2006; 410: 3-28. Appl Environ Microbiol 2005; 71: 6077-85.
[69] van Hemert S, Ebbelaar BH, Smits MA, Rebel JM. Generation of [90] Eeckhaut E, Struijs K, Possemiers S, Vincken JP, De Keukeleire D,
EST and microarray resources for functional genomic studies on Verstraete W. Metabolism of the lignan macromolecule into enter-
chicken intestinal health. Anim Biotechnol 2003; 14: 133-43. olignans in the gastrointestinal lumen as determined in the simula-
[70] Molly K, Woestyne MV, Verstraete W. Development of a 5-step tor of the human intestinal microbial ecosystem. J Agric Food
multichamber reactor as a simulation of the human intestinal mi- Chem 2008; 56: 4806-12.
crobial ecosystem. Appl Microbiol Biotechnol 1993; 39: 254-8. [91] Bolca S, Possemiers S, Herregat A, Huybrechts I, Heyerick A, De
[71] Possemiers S, Verthe K, Uyttendaele S, Verstraete W. PCR- Vriese S, et al. Microbial and dietary factors are associated with the
DGGE-based quantification of stability of the microbial commu- equol producer phenotype in healthy postmenopausal women. J
nity in a simulator of the human intestinal microbial ecosystem. Nutr 2007; 137: 2242-6.
FEMS Microbiol Ecol 2004; 49: 495-507. [92] Bolca S, Possemiers S, Maervoet V, Huybrechts I, Heyerick A,
[72] Macfarlane GT, Macfarlane S, Gibson GR. Validation of a three- Vervarcke S, et al. Microbial and dietary factors associated with
stage compound continuous culture system for investigating the ef- the 8-prenylnaringenin producer phenotype: a dietary intervention
fect of retention time on the ecology and metabolism of bacteria in trial with fifty healthy post-menopausal Caucasian women. Br J
the human colon. Microb Ecol 1998; 35: 180-7. Nutr. 2007; 98: 950-9.
[73] Makivuokko HA, Saarinen MT, Ouwehand AC, Rautonen NE. [93] Santos-Buelga C, Scalbert A. Proanthocyanidins and tannin-like
Effects of lactose on colon microbial community structure and compounds - nature, occurrence, dietary intake and effects on nutri-
function in a four-stage semi-continuous culture system. Biosci tion and health. J Sci Food Agric 2000; 80: 1094-117.
Biotechnol Biochem 2006; 70: 2056-63. [94] Gonthier MP, Verny MA, Besson C, Remesy C, Scalbert A.
[74] Possemiers S, Rabot S, Espin JC, Bruneau A, Philippe C, Gon- Chlorogenic acid bioavailability largely depends on its metabolism
zalez-Sarrias A, et al. Eubacterium limosum activates isoxantho- by the gut microflora in rats. J Nutr 2003; 133: 1853-9.
humol from hops (Humulus lupulus L.) into the potent phytoestro- [95] Humblot C, Murkovic M, Rigottier-Gois L, Bensaada M, Bouclet
gen 8-prenylnaringenin in vitro and in the rat intestine. J Nutr 2008; A, Andrieux C, et al. beta-Glucuronidase in human intestinal mi-
138: 1310-6. crobiota is necessary for the colonic genotoxicity of the food-borne
[75] Laube B, Winkler S, Ladstetter B, Scheller T, Schwarz LR. Estab- carcinogen 2-amino-3-methylimidazo[4,5-f]quinoline in rats. Car-
lishment of a novel in vitro system for studying the interaction of cinogenesis 2007; 28: 2419-25.
xenobiotic metabolism of liver and intestinal microflora. Arch [96] Bowes SG, Renwick AG. The intestinal metabolism and DNA-
Toxicol 2000; 74: 379-87. binding of benzo[a]pyrene in guinea pigs fed normal, high-fat and
[76] Braat H, Rottiers P, Hommes DW, Huyghebaert N, Remaut E, high-cholesterol diets. Xenobiotica 1986; 16: 543-53.
Remon JP, et al. A phase I trial with transgenic bacteria expressing [97] Okuda H, Ogura K, Kato A, Takubo H, Watabe T. A possible
interleukin-10 in Crohn's disease. Clin Gastroenterol Hepatol 2006; mechanism of eighteen patient deaths caused by interactions of
4: 754-9. sorivudine, a new antiviral drug, with oral 5-fluorouracil prodrugs.
[77] Wells JM, Mercenier A. Mucosal delivery of therapeutic and pro- J Pharmacol Exp Ther 1998; 287: 791-9.
phylactic molecules using lactic acid bacteria. Nat Rev 2008; 6: [98] Huycke MM, Moore DR. In vivo production of hydroxyl radical by
349-62. Enterococcus faecalis colonizing the intestinal tract using aromatic
[78] Marzorati M, Wittebolle L, Boon N, Daffonchio D, Verstraete W. hydroxylation. Free Rad Biol Med 2002; 33: 818-26.
How to get more out of molecular fingerprints: practical tools for [99] Van Tassell RL, Carman RJ, Kingston DG, Wilkins TD. Bacterial
microbial ecology. Environ Microbiol 2008; 10: 1571-81. metabolism in humans of the carcinogen IQ to the direct acting
[79] Verstraete W. Microbial ecology and environmental biotechnology. mutagen hydroxy-IQ. Microb Ecol Health Dis 1989; 2: 123-9.
ISME J 2007; 1: 4-8. [100] Humblot C, Combourieu B, Vaisanen ML, Furet JP, Delort AM,
[80] Tuohy KM, Hinton DJS, Davies SJ, Crabbe MJC, Gibson GR, Rabot S. 1H nuclear magnetic resonance spectroscopy-based stud-
Ames JM. Metabolism of Maillard reaction products by the human ies of the metabolism of food-borne carcinogen 2-amino-3-
gut microbiota - implications for health. Mol Nutr Food Res 2006; methylimidazo[4,5-f]quinoline by human intestinal microbiota.
50: 847-57. Appl Environ Microbiol 2005; 71: 5116-23.
[81] Sousa T, Paterson R, Moore V, Carlsson A, Abrahamsson B, Basit [101] Van de Wiele T, Vanhaecke L, Boeckaert C, Peru K, Headley J,
AW. The gastrointestinal microbiota as a site for the biotransforma- Verstraete W, et al. Human colon microbiota transform polycyclic
tion of drugs. Int J Pharm 2008; 363: 1-25. aromatic hydrocarbons to estrogenic metabolites. Environ Health
[82] Ilett KF, Tee LBG, Reeves PT, Minchin RF. Metabolism of drugs Perspect 2005; 113: 6-10.
and other xenobiotics in the gut lumen and wall. Pharmacol Thera- [102] Vanhaecke L, Vercruysse F, Boon N, Verstraete W, Cleenwerck I,
peut 1990; 46: 67-93. De Wachter M, et al. Isolation and characterization of human intes-
[83] Clifford MN. Diet-derived Phenols in plasma and tissues and their tinal bacteria capable of transforming the dietary carcinogen 2-
implications for health. Planta Med 2004; 70: 1103-14. amino-1-methyl-6-phenylimidazo[4,5-b]pyridine. Appl Environ
Microbiol 2008; 74: 1469-77.
[84] Walle T, Walle UK, Halushka PV. Carbon dioxide is the major
metabolite of quercetin in humans. J Nutr 2001; 131: 2648-52. [103] Vanhaecke L, Derycke L, Le Curieux F, Lust S, Marzin D, Ver-
[85] Rowland I, Faughnan M, Hoey L, Wahala K, Williamson G, Cas- straete W, et al. The microbial PhIP metabolite 7-hydroxy-5-
methyl-3-phenyl-6,7,8,9-tetrahydropyrido[3 ',2 ': 4,5]imidazo[1,2-
sidy A. Bioavailability of phytoestrogens. Br J Nutr 2003; 89: S45-
58. a]pyrimidin-5-ium chloride (PhIP-M1) induces DNA damage,
apoptosis and cell cycle arrest towards Caco-2 cells. Toxicol Lett
[86] Possemiers S, Verstraete W, Van de Wiele T. In: Preedy VR, Ed.
2008; 178: 61-9.
Estrogenicity of beer: the role of intestinal bacteria in the activation
2064 Current Pharmaceutical Design, 2009, Vol. 15, No. 18 Possemiers et al.

[104] Meyer J, Michalke K, Kouril T, Hensel R. Volatilisation of metals sitivity by oligofructose requires a functional glucagon-like peptide
and metalloids: An inherent feature of methanoarchaea? Syst Appl 1 receptor. Diabetes 2006; 55: 1484-90.
Microbiol 2008; 31: 81-7. [126] Inui A, Asakawa A, Bowers CY, Mantovani G, Laviano A, Meguid
[105] Hooper LV, Gordon JI. Commensal host-bacterial relationships in MM, et al. Ghrelin, appetite, and gastric motility: the emerging role
the gut. Science 2001; 292: 1115-8. of the stomach as an endocrine organ. FASEB J 2004; 18: 439-56.
[106] Villarini M, Caldini G, Moretti M, Trotta F, Pasquini R, Cenci G. [127] Cani PD, Hoste S, Guiot Y, Delzenne NM. Dietary non-digestible
Modulatory activity of a lactobacillus casei strain on 1.2- carbohydrates promote L-cell differentiation in the proximal colon
dimethylhydrazine-induced genotoxicity in rats. Environ Mol of rats. Br J Nutr 2007; 98: 32-7.
Mutagen 2008; 49: 192-9. [128] Gee JM, Johnson IT. Dietary lactitol fermentation increases circu-
[107] Lhoste EF, Nugon-Baudon L, Lory S, Meslin JC, Andrieux C. The lating peptide YY and glucagon-like peptide-1 in rats and humans.
fermentation of lactulose in rats inoculated with Clostridium Nutrition 2005; 21: 1036-43.
paraputrificum influences the activities of liver and intestinal [129] Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy
xenobiotic-metabolising enzymes. J Sci Food Agric 2001; 81: KM, et al. Selective increases of bifidobacteria in gut microflora
1397-404. improve high-fat-diet-induced diabetes in mice through a mecha-
[108] Humblot C, Lhoste E, Knasmuller S, Gloux K, Bruneau A, Ben- nism associated with endotoxaemia. Diabetologia 2007; 50: 2374-
saada M, et al. Protective effects of Brussels sprouts, oligosaccha- 83.
rides and fermented milk towards 2-amino-3-methylimidazo[4,5- [130] Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate
f]quinoline (IQ)-induced genotoxicity in the human flora associated gastrointestinal peptides involved in appetite regulation (glucagon-
F344 rat: role of xenobiotic metabolising enzymes and intestinal like peptide-1 and ghrelin) in rats. Br J Nutr 2004; 92: 521-6.
microflora. J Chromatogr B 2004; 802: 231-7. [131] Massimino SP, McBurney MI, Field CJ, Thomson ABR, Keelan
[109] Mikov M. The metabolism of drugs by the gut flora. Eur J Drug M, Hayek MG, et al. Fermentable dietary fiber increases GLP-1
Metab Pharmacokinet 1994; 19: 201-7. secretion and improves glucose homeostasis despite increased in-
[110] Li HK, Ni Y, Su MM, Qiu YP, Zhou MM, Qiu MF, et al. Phar- testinal glucose transport capacity in healthy dogs. J Nutr 1998;
macometabonomic phenotyping reveals different responses to 128: 1786-93.
xenobiotic intervention in rats. J Proteome Res 2007; 6: 1364-70. [132] Yoshida K, Shimizugawa T, Ono M, Furukawa H. Angiopoietin-
[111] Wyatt SB, Winters KP, Dubbert PM. Overweight and obesity: like protein 4 is a potent hyperlipidemia-inducing factor in mice
prevalence, consequences, and causes of a growing public health and inhibitor of lipoprotein lipase. J Lipid Res 2002; 43: 1770-2.
problem. Am J Med Sci 2006; 331: 166-74. [133] Williams CM. Postprandial lipid metabolism: effects of dietary
[112] Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, fatty acids. Proc Nutr Soc 1997; 56: 679-92.
Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad [134] Berggren A, Nyman EMGL, Lundquist I, Björck IME. Influence of
Sci USA 2005; 102: 11070-5. orally and rectally administered propionate on cholesterol and glu-
[113] Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: cose metabolism in obese rats. Br J Nutr 1996; 76: 287-94.
Human gut microbes associated with obesity. Nature 2006; 444: [135] Lin Y, Vonk RJ, Slooff MJ, Kuipers F, Smit MJ. Differences in
1022-3. propionate-induced inhibition of cholesterol and triacylglycerol
[114] Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, synthesis between human and rat hepatocytes in primary culture. Br
Gordon JI. An obesity-associated gut microbiome with increased J Nutr 1995; 74: 197-207.
capacity for energy harvest. Nature 2006; 444: 1027-131. [136] Bongaerts GP, Severijnen RS, Tangerman A, Verrips A, Tolboom
[115] Delzenne NM, Cani PD, Neyrinck AM. Modulation of glucagon- JJ. Bile acid deconjugation by Lactobacilli and its effects in pa-
like peptide 1 and energy metabolism by inulin and oligofructose: tients with a short small bowel. J Gastroenterol 2000; 35: 801-4.
Experimental data. J Nutr 2007; 137: 2547S-51. [137] Nakajima H, Suzuki Y, Kaizu H, Hirota T. Cholesterol lowering
[116] Egan JM, Margolskee RF. Taste cells of the gut and gastro- activity of ropy fermented milk. J Food Sci 1992; 57: 1327-9.
intestinal chemosensation. Mol Interv 2008; 8: 78-81. [138] Pulusani SR, Rao DR. Whole body, liver and plasma cholesterol
[117] Zhou J, Martin RJ, Tulley RT, Raggio AM, McCutcheon KL, Shen levels in rats fed Thermophilus bulgaricus and acidophilus milks. J
L, et al. Dietary resistant starch upregulates total GLP-1 and PYY Food Sci 1983; 48: 280-1.
in a sustained day-long manner through fermentation in rodents. [139] Richelsen B, Kristensen K, Pedersen SB. Long-term (6 months)
Am J Physiol Endocrinol Metab 2008; 295: E1160-6. effect of a new fermented milk product on the level of plasma lipo-
[118] Mandard S, Zandbergen F, van Straten E, Wahli W, Kuipers F, proteins--a placebo-controlled and double blind study. Eur J Clin
Muller M, et al. The fasting-induced adipose factor/angiopoietin- Nutr 1996; 50: 811-5.
like protein 4 is physically associated with lipoproteins and governs [140] Gilliland SE, Nelson CR, Maxwell C. Assimilation of cholesterol
plasma lipid levels and adiposity. J Biol Chem 2006; 281: 934-44. by Lactobacillus acidophilus. Appl Environ Microbiol 1985; 49:
[119] Yoon JC, Chickering TW, Rosen ED, Dussault B, Qin YB, Soukas 377-81.
A, et al. Peroxisome proliferator-activated receptor gamma target [141] Beena A, Prasad V. Effect of yogurt and bifidus yogurt fortified
gene encoding a novel angiopoietin-related protein associated with with skim milk powder, condensed whey and lactose-hydrolysed
adipose differentiation. Mol Cell Biol 2000; 20: 5343-9. condensed whey on serum cholesterol and triacylglycerol levels in
[120] Delzenne NM, Kok N, Fiordaliso M, Deboyser D, Goethals F, rats. J Dairy Res 1997; 64: 453-7.
Roberfroid MB. Dietary fructo-oligosaecharides modifies lipid me- [142] Mohan B, Kadirvel R, Natarajan A, Bhaskaran M. Effect of probi-
tabolism in rats. Am J Clin Nutr 1993; 57: 8205. otic supplementation on growth, nitrogen utilization and serum
[121] Hanai JI, Cao P, Tanksale P, Imamura S, Koshimizu E, Zhao JH, et cholesterol in broilers. Br Poult Sci 1996; 37: 395-401.
al. The muscle-specific ubiquitin ligase atrogin-1/MAFbx mediates [143] Xiao JZ, Kondo S, Takahashi N, Miyaji K, Oshida K, Hiramatsu A,
statin-induced muscle toxicity. J Clin Invest 2007; 117: 3940-51. et al. Effects of milk products fermented by Bifidobacterium
[122] St-Onge M-P, Farnworth ER, Jones PJH. Consumption of fer- longum on blood lipids in rats and healthy adult male volunteers. J
mented and nonfermented dairy products: effects on cholesterol Dairy Sci 2003; 86: 2452-61.
concentrations and metabolism. Am J Clin Nutr 2000; 71: 674-81. [144] Hentges DJ. Biotransformation of bile acids and cholesterol by the
[123] Todesco T, Rao AV, Bosello O, Jenkins DJA. Propionate lowers intestinal microflora. New York: Academic Press 1983.
blood glucose and alters lipid-metabolism in healthy subjects. Am J [145] Fiordaliso MF, Kok N, Desager JP, Goethals F, Deboyser D,
Clin Nutr 1991; 54: 860-5. Roberfroid MB, et al. Dietary oligofructose lowers triacylglycerols,
[124] Bodmer M, Meier C, Krahenbuhl S, Jick SS, Meier CR. Met- phospholipids and cholesterol in serum and very low density lipo-
formin, sulfonylureas, or other antidiabetes drugs and the risk of proteins of rats. Lipids 1995; 30: 163-7.
lactic acidosis or hypoglycemia. A nested case-control analysis. [146] Trautwein EA, Rieckhoff D, Erbersdobler HF. Dietary inulin low-
Diabetes Care 2008; 31: 2086-91. ers plasma cholesterol and triacylglycerol and alters biliary bile
[125] Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM, Bur- acid profile in hamsters. J Nutr 1998; 128: 1937-43.
celin R. Improvement of glucose tolerance and hepatic insulin sen- [147] Williams CM, Jackson KG. Inulin and oligofructose: effects on
lipid metabolism from human studies. Br J Nutr 2002; 87: S261-4.
Intestinal Bacteria and Human Health Current Pharmaceutical Design, 2009, Vol. 15, No. 18 2065

[148] Cheng H-H, Lai M-H. Fermentation of resistant rice starch pro- peptide-1 by anorexigenic cannabinoid compounds, SR141716A
duces propionate reducing serum and hepatic cholesterol in rats. J (rimonabant) and oleoylethanolamide. Br J Nutr 2004; 92: 757-61.
Nutr 2000; 130: 1991-5. [155] Zaneveld J, Turnbaugh PJ, Lozupone C, Ley RE, Hamady M,
[149] Venter CS, Vorster HH, Cummings JH. Effects of dietary propion- Gordon JI, et al. Host-bacterial coevolution and the search for new
ate on carbohydrate and lipid metabolism in healthy volunteers. drug targets. Curr Opin Chem Biol 2008; 12: 109-14.
Am J Gastroenterol 1990; 85: 549-53. [156] Lujan SA, Guogas LM, Ragonese H, Matson SW, Redinbo MR.
[150] Towle HC. Glucose and cAMP: Adversaries in the regulation of Disrupting antibiotic resistance propagation by inhibiting the con-
hepatic gene expression. Proc Natl Acad Sci USA 2001; 98: 13476- jugative DNA relaxase. Proc Natl Acad Sci USA 2007; 104:
8. 12282-7.
[151] Hooper LV, Midtveld T, Gordon JI. How host-microbial interac- [157] Cani PD, Delzenne NM. Gut microflora as a target for energy and
tions shape the nutrient environment of the mammalian intestine. metabolic homeostasis. Curr Opin Clin Nutr Metab Care 2007; 10:
Ann Rev Nutr 2002; 22: 283-307. 729-34.
[152] Cani PD, Daubioul CA, Reusens B, Remacle C, Catillon G, Del- [158] Corcoran BM, Stanton C, Fitzgerald G, Ross R P. Life under stress:
zenne NM. Involvement of endogenous glucagon-like peptide-1(7- the probiotic stress response and how it may be manipulated. Curr
36) amide on glycaemia-lowering effect of oligofructose in strepto- Pharm Des 2008; 14(14): 1382-99.
zotocin-treated rats. J Endocrinol 2005; 185: 457-65. [159] O'Sullivan DJ. Genomics can advance the potential for probiotic
[153] Bray GA, Greenway FL. Current and potential drugs for treatment cultures to improve liver and overall health. Curr Pharm Des 2008;
of obesity. Endocr Rev 1999; 20: 805-75. 14(14): 1376-81.
[154] Cani PD, Montoya ML, Neyrinck AM, Delzenne NM, Lambert [160] Singh SB. Pelaez F. Biodiversity, chemical diversity and drug
DM. Potential modulation of plasma ghrelin and glucagon-like discovery. Prog Drug Res 2008; 65: 141, 143-74.

Received: March 4, 2009 Accepted: March 11, 2009

Vous aimerez peut-être aussi