Sie sind auf Seite 1von 6

Vol. 4, No.

3 2007
Drug Discovery Today: Therapeutic Strategies

Editors-in-Chief
Raymond Baker – formerly University of Southampton, UK and Merck Sharp & Dohme, UK
Eliot Ohlstein – GlaxoSmithKline, USA
DRUG DISCOVERY
TODAY
THERAPEUTIC
STRATEGIES Gastrointestinal diseases

The gut microbiota and disease – an


inner repository for drug discovery
Fergus Shanahan*, Barry Kiely
Alimentary Pharmabiotic Centre and Department Medicine, University College Cork, National University of Ireland and Alimentary Health Ltd., Cork, Ireland

The gut micobiota is tantamount to a hidden inner


Section Editor:
organ, with a gene content (microbiome) exceeding Gareth J. Sanger – Immuno-Inflammation Centre of
that of the human genome, a versatile metabolic capa- Excellence for Drug Discovery, GlaxoSmithKline, Stevenage,
Hertfordshire, UK
city rivalling that of the liver and a source of signals
required for optimal structural and functional devel-
Another advance that accelerated research into host–
opment of the host. Although some disorders, such as microbe interactions in the gut was the first identification
Crohn’s disease, might result from abnormal host– of a susceptibility gene (NOD 2/CARD 15) for Crohn’s disease
microbe interactions, and others, such as obesity, [3,4]. This illustrated the molecular nexus of the three main
interacting factors involved in the pathogenesis of Crohn’s
might be influenced by bacterial-derived metabolic
disease: genetic risk factors, environmental (microbial) mod-
signalling from the gut, the microbiota is primarily a
ifying factors and immune-mediated tissue damage. It was
health asset in defence against infectious, inflamma- the first time that a mutant gene coding for an intracellular
tory and probably neoplastic disorders. This inner bio- sensor of microbes was directly linked with risk of chronic
mass is a relatively untapped repository for ‘mining’ inflammatory disease, and it prompted a search for other
genetic risk factors for Crohn’s disease that involve the
bioactives for novel drug discovery.
interpretation or handling of the microbial environment
[5,6].
Additional drivers of the resurgence of interest in the gut
Introduction
microbiota include the desire to manipulate the gastrointest-
The gastrointestinal ecosystem is attracting scientific and
inal microenvironment with probiotics or prebiotics, the
commercial interest, both as a target for drug therapy and
recognition of the relevance of the commensal microbiota
as an inner resource for drug discovery [1,2]. A landmark
to the emergence of the global problem of antibiotic resis-
event, recently acknowledged with the award of the Nobel
tance and the current epidemic of Clostridium difficile-asso-
Prize to its discoverers, was the role of Helicobacter pylori in the
ciated enteric disease. Finally, deterrents to the study of the
pathogenesis of peptic ulcer disease. This opened minds to
microbiota, such as a dependency on traditional culture-
the possibility of a microbial contribution to other chronic
based techniques that are limited by the inability to grow
diseases. It also served a lesson that the solution to some
most intestinal bacteria on available culture media, have
diseases might never be found if research is centred exclu-
been circumvented with the advent of molecular strategies
sively on mammalian pathophysiology, without due atten-
[7].
tion to the interface between the host and its microbial
The purpose of this article is to provide a perspective on
environment.
opportunities for drug discovery by exploitation of microbe–
*Corresponding author: F. Shanahan (f.shanahan@ucc.ie) microbe and microbe–host interactions within the gut. To

1740-6773/$ ß 2008 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ddstr.2008.02.006 195
Drug Discovery Today: Therapeutic Strategies | Gastrointestinal diseases Vol. 4, No. 3 2007

that end, some of the pivotal concepts and gaps in knowledge lymphoid tissue with sparse lamina propria cellularity, low
in relation to the microbiota will be addressed in overview epithelial cell turnover and digestive enzyme activity,
but, for more detailed reviews and specific aspects of the gut reduced muscle wall thickness with impaired peristalsis, in
microbiota, the reader is referred elsewhere [7–10]. comparison with colonised littermates. By contrast, life
without bacteria (germ-free) is associated with increased
Scale and complexity of the microbial inner organ enterochromaffin cell area and increased dietary caloric
The human gut is the most densely populated ecosystems on requirements. This suggests that the microbiota is a source
the planet. It is a hidden biomass, tantamount to an inner of both positive and negative regulatory signals acting a
organ with a metabolic capacity on the scale of that of the multiple levels within the host, the identities of which are
liver. The bacterial load and diversity (>1000 species) being pursued with techniques such as laser microdissection
increases distally with the largest gradient being across the and gene array analysis [16].
ileocaecal valve, where there are approximately 108 bacteria/g Germ-free animals are more susceptible to infection, have
of ileal content rising to 1012/g of colonic content. Cross- defective immune function and disturbed stress responses
sectional variation is also evident with the ratio of anaerobes [10,17]. However restoration to normality occurs with resti-
to aerobes being lower for mucosa-adherent bacteria versus tution of the intestinal microbiota; colonisation with the
luminal populations. single species, Bacteroides thetaiotaomicron, is sufficient to
Collectively, the microbiota is a repository of genetic influence the expression of host genes regulating nutrient
information (microbiome) exceeding that of the host gen- uptake, metabolism, angiogenesis, barrier function and the
ome. Indeed, the host genome encodes insufficient informa- development of the enteric nervous system [18].
tion for full development of the gut. Sterile before birth, Host–microbial dialogue occurs over a huge surface area
optimal development of the gut is dependent upon signals within the gut (400 m2), with only a single layer of epithe-
from the microbiota colonising the lumen immediately after lium separating the lumen from the internal milieu.
birth. The composition of these pioneering commensals is Although this adaptation of structure to function is optimal
determined by several factors that include the mode of birth for nutrient absorption, it becomes a potential vulnerability
delivery, maternal microbiota, diet and environmental in the setting of enteric infection. Hence, there is a require-
hygiene. In different individuals, the composition of the ment for continual maintenance of mucosal homoeostasis;
microbiota appears to be distinct, and perhaps unique, by multiple mechanisms are required and involve contributions
molecular fingerprinting. A variety of molecular techniques, from both the host and the resident microbiota (reviewed in
mainly nucleic acid-based strategies, has revealed a scale and references [9,10]). Breakdown in homoestasis through defec-
diversity of the human microbiota greater than previously tive host–microbe exchange is thought to underlie the patho-
contemplated (reviewed in references [7,11]). For example, genesis of some chronic inflammatory disorders, such as
the acidity of the human stomach was thought by many to Crohn’s disease [5].
create an almost sterile microenvironment, once cleared of The challenge for the host is the requirement for exquisite
H. pylori, but a metagenomic approach has revealed an precision in distinguishing harmless commensal microbes
extraordinary range of bacterial residents, some of which from episodic challenge with pathogens. This is achieved
were not previously thought to be present in the human by continual sampling of the microenvironment and distinct
body [12]. cellular and molecular immunosensory processes. Immuno-
The application of culture-independent metagenomics to logical sampling of the lumen takes place across the epithe-
the gut microbiota is escalating and has already provided new lium at several sites. M cells overlying lymphoid follicles
insights into the metabolic capacity of the normal microbiota transport microbial antigens to subjacent antigen-presenting
in diseases, such as obesity, and is being applied to the cells, whereas surface enterocytes sample soluble antigens
apparent compositional changes in the microbiota in disor- and elaborate a range of chemokines, thereby alerting the
ders such as inflammatory bowel disease and irritable bowel host and directing the innate and acquired immune systems
syndrome [13–15]. Major consortia on both sides of the to the site of any breach in the mucosal barrier. Dendritic cells
Atlantic are underway to apply metagenomics to enhancing might directly sample the contents of the lumen by extend-
the understanding of the normal microbiota in health and ing dendrites between the surface enterocytes; they can
disease at various ages and different dietary conditions [11]. ingest and retain live bacteria and migrate to the mesenteric
lymph node where an immune response is induced. In this
Host–microbe dialogue way, the mesenteric lymph node serves as the gatekeeper,
Comparative studies of germ-free versus conventionally colo- limiting access by luminal bacteria to the systemic immune
nised animals first performed several decades ago showed the compartment [19].
conditioning influence of the microbiota on intestinal struc- At a molecular level, discrimination of pathogenic
ture and function [10]. Germ-free animals have rudimentary and commensal bacteria by dendritic and other immunosen-

196 www.drugdiscoverytoday.com
Vol. 4, No. 3 2007 Drug Discovery Today: Therapeutic Strategies | Gastrointestinal diseases

sory cells is mediated, partly, by two major host pattern this disease and the widespread resistance of the organism to
recognition receptor (PRR) systems – the family of surface currently available drugs.
Toll-like receptors (TLRs) and the intracellular nucleotide-
binding oligomerization domain/caspase recruitment Commensal-derived immunomodulatory drugs
domain isoforms (NOD/CARD) [9,10]. These systems are Given that germ-free mice have defective immune develop-
under precise regulation, the details of which are not fully ment that is restored upon colonisation with conventional
understood. The complexity of the process is shown by the microbiota, it follows that the gut microbiota must be a source
fact that commensals and pathogens share many PRR ligands, of immunomodulatory molecules influencing the structural
but the normal mucosal immune response to commensals is and functional development of the immune system. This
restrained. In addition, continual signalling by ligands from prediction was fulfilled by the demonstration that colonisa-
the resident microbiota using host TLRs is essential for main- tion with Bacteroides fragilis produces a zwitterionic polysac-
tenance of intestinal homeostasis and barrier function [20]. charide that directs and corrects maturational deficits in the
Furthermore, some commensal bacteria attenuate the pro- developing immune system [25,26]. Particularly noteworthy
inflammatory effects of pathogens using various mechanisms was the observation that this microbial-derived polysaccharide
[10,21]. not only influenced development of mucosal lymphoid tissue
but also corrected structural and functional systemic immune
Therapeutic strategies deficits after colonisation of germ-free animals [25].
Humankind has exploited microbes for many purposes ran- Bacterial products or constituents, other than polysacchar-
ging from producing insulin to cleaning up oil slicks. Envir- ides, might also modulate immune function. For example,
onmental microbes also continue to be used for isolation of bacterial DNA and oligonucleotides containing hypomethy-
novel antibiotics, immunomodulatory drugs and other lated CpG dinucleotides, in contrast to eukaryotic DNA
bioactives, but the inner gut ecosystem is a relatively and methylated oligonucleotides, have immunomodulatory
untapped frontier. However, a ‘bugs to drugs’ discovery strat- properties that are mediated by ligation of TLR9 on host cells
egy can be designed from current understanding of the [10]. Indeed, the beneficial effects of probiotics in murine
normal microbiota, particularly the lessons derived from models of inflammatory bowel disease have been attributed
studying life without bacteria, and as summarised below, to the actions of probiotic DNA on TLR9 [27]. However, bacteria
some predictions are already fulfilled. vary in their immunomodulatory DNA content, and CpG DNA
motifs might have unexpected or undesirable effects in experi-
Commensal-derived novel antimicrobial drugs – bacteriocins mental models of colitis depending on the timing of admin-
Because bacterial numbers within different segments of the istration. Thus, in contrast to the prophylactic effect of CpG
gut are constrained not only by the host and by mutual DNA before the onset of inflammation, exposure during acute
competitive interactions but also by microbe–microbe signal- inflammation has been reported to aggravate the inflammatory
ling, the potential to mine novel antimicrobial factors from disease in a murine model of inflammatory bowel disease [28].
the microbiota arises. A highly conserved antimicrobial sys-
tem in living organisms is the production of antimicrobial Microbial-derived anti-inflammatory inflammatory activity
peptides, such as bacteriocins and defensins. Bacteriocins are In addition to immunomodulatory and immunostimulatory
ribosomally synthesised antimicrobial peptides produced by polysaccharides and nucleotides, bacteria in the gut have
one bacterium that are active against other bacteria but to been shown to produce a variety of proteins and peptides
which the producer organism has a specific resistance with actions on the host that are broadly cytoprotective or
mechanism. They might exhibit either narrow- or broad- anti-inflammatory [29,30].
spectrum antibacterial activity and have been used for With intact whole bacteria, the impact on host cytokine
decades by the food industry where bacteriocin-producing production is highly variable even for different strains within
food-grade bacteria are deployed for food preservation a given species. However, in the case of lactobacilli, it has
(reviewed in reference [22]). been shown that a major determinant of anti-inflammatory
More recently, a bacteriocin from a specific probiotic lac- activity is the composition of the cell envelope constituent,
tobacillus has been shown to account for the protective lipotechoic acid, specifically its D-alanine content [31]. Lipo-
mechanism of probiotic action against Listeria infection techoic acids are docked in the bacterial cell membrane by a
[23]. Bacteriocins have also been shown to have clinical glycolipid anchor and represent amphiphilic polymers of
potential in animal husbandry and in human conditions repeating units of glycerophosphate speckled with D-alanine
such as Clostridium difficile [24]. The pursuit of human com- and glycosyl residues. D-alanine content might become a
mensal-derived bacteriocins with potency and a narrow spec- selection criterion for probiotics for some disease indications
trum of activity against C. difficile is a particularly appealing and has implications for future mining of microbial-derived
strategy in view of the emergence of a highly virulent form of anti-inflammatory compounds.

www.drugdiscoverytoday.com 197
Drug Discovery Today: Therapeutic Strategies | Gastrointestinal diseases Vol. 4, No. 3 2007

Nutrient and bioactive metabolite production storage has been shown by Gordon and colleagues [34]. Two
Although the enteric microbiota play a central role in diges- mechanisms are involved: the first being the induction of
tion and nutrient provision and this is of commercial rele- hepatic lipogenesis following enhanced absorption of mono-
vance particularly to the food industry, it is beyond the scope saccharides released by microbial metabolism of non-digesti-
of the present overview. However, some bacterial metabolites ble polysaccharides and the second being the enhanced
in the gut serve not only a nutrient function but also have deposition of lipid in adipocytes promoted by microbial-
intrinsic bioactive properties that can be mined for use either induced suppression of fasting-induced adipocyte factor (Fiaf,
as a functional food ingredient or perhaps as a drug. An also known as ANGPTL4), which is a member of the angio-
example is the generation of bioactive isomers of conjugated poietin-like family of proteins and an inhibitor of lipoprotein
linoleic acid (CLA) by the commensal microbiota that exhibit lipase. In a series of elegant studies, the same group of inves-
peroxisome proliferator-activated receptor (PPAR) agonist tigators showed that the microbiota of obese leptin-deficient
activity [32,33]. mice and of obese humans extract and make available more
energy to the host from a given diet than do the microbiota of
Microbial-derived metabolic signals for host metabolism and obesity their lean counterparts, and this was associated with reversible
Germ-free animals must consume more calories than colo- compositional differences in the microbiota [7].
nised animals to maintain body weight. It follows that manip- This body of work shows that the impact of the gut
ulation of the composition or metabolic activity of the microbiota in health and disease extends well beyond the
microbiota might have a bearing on the approach to obesity. gut. To underscore this, others have used metabolic profiling
Convincing evidence that the microbiota is a regulator of fat to show a contribution of the gut microbiota to fatty liver

Table 1. Representative examples of bacterial actions in the gut that might be suited to a ‘bugs to drugs’ programme of
discovery for development of bioactive ingredients in functional foodsa
Bacterial action Opportunity Advantages Potential limitations Anyone interested? Refs
1 Microbe–microbe Isolation of novel Naturally occurring Sensitive to digestive The food industry [22]
communication antibiotics Low level of resistance enzymatic degradation Alimentary
Examples: quorum (e.g. bacteriocins) Dual mechanism of action Topical action only unless Health Ltd.
sensing and Example: lacticin enteric-coated (targeting C. difficile)
antimicrobial
production
2 Bacterial stimulation Bacterial-derived Potential applications for Might involve complex [26]
or modulation of immunoregulatory immunodeficiency disorders and expensive carbohydrate
host immunity polysaccharides chemistry
Macromolecules might be
required as end products
Bacterial Applications to inflammatory Might be proinflammatory Coley [28]
immunostimulatory and neoplastic disorders in certain settings Pharmaceuticals
(CpG) DNA Pfizer Corp.
3 Commensal (and probiotic) Anti-inflammatory drugs Relevant bacteria are readily Evidence base to date [31]
anti-inflammatory effects on from bacterial components available and culturable in vitro is still very preliminary
host tissues (e.g. lipotechoic acid)
4 Nutrient production Bacterial-derived vitamins Sound evidence base The food industry [33]
& fatty acid nutrients Gut microbiota might be
Example: bioactive exploited to favourably
isomers of CLA influence the fatty acid
composition of host tissues
5 Metabolic signals Obesity prevention by Seemingly a potentially safe Will probably require The food industry [34]
reducing dietary caloric but long-term strategy if live microbes
bioavailability developed
6 Neuroactive signals Analgesic effects for Exploitation of endogenous Needs independent [36]
irritable bowel syn. analgesic mechanism confirmation
7 Genetically engineered enteric Bioactive of choice Site-specific delivery Public health and Actigenix [38]
bacteria to produce bioactives is probably unlimited Inexpensive environmental concerns
regarding containment
a
Note this table is intended to be representative not comprehensive. The well-established potential of microbial ecosystems outside the gut microbiota is acknowledged but is beyond the
scope of this overview.

198 www.drugdiscoverytoday.com
Vol. 4, No. 3 2007 Drug Discovery Today: Therapeutic Strategies | Gastrointestinal diseases

phenotype in insulin-resistant mice [35]. Unravelling the tract and has a bearing on common disorders in developed
mechanisms at the host–microbe interface in these disorders societies, such as obesity, metabolic syndrome and their
is likely to reveal new therapeutic targets, some of which complications. Although technical hurdles continue to fall,
might be suitable for novel drug strategies. the greatest challenge is still the requirement for enhanced
understanding of the normal gut microbiota and its interac-
Neuroactive signals tion with the host in health and disease.
Of the numerous examples exhibiting the diversity of bacterial
actions within the gut that might be suited to bioprospecting Conflict of interest statement
for novel drugs, the modulation of visceral pain by a subset of The authors are affiliated with a multi-departmental university
commensals (L. acidophilus) is particularly intriguing in the campus company, Alimentary Health Ltd. that investigates
context of managing patients with irritable bowel syndrome inter alia host–microbe interactions. The content of this paper
[36]. Oral administration of some, but not all, lactobacillus was neither influenced nor constrained by that fact.
strains to rodents was found to induce the expression of mu-
opioid and cannabinoid receptors on intestinal epithelia, and
References
in a rectal distension model of visceral pain an analgesic effect 1 Jia, W. et al. (2008) Gut microbiota: a potential new territory for drug
was noted that was equivalent to 1 mg/kg morphine. Because targeting. Nat. Rev. Drug Discov. 7, 123–129
there is increasing scientific interest in host–microbe interac- 2 O’Hara, A.M. and Shanahan, F. (2007) Gut microbiota: mining for
therapeutic potential. Clin. Gastroenterol. Hepatol. 5, 274–284
tions in irritable bowel syndrome [14], it might be that both the
3 Hugot, J.P. et al. (2001) Association of NOD2 leucine-rich repeat variants
cause and the solution can be mined from the enteric micro- with susceptibility to Crohn’s disease. Nature 411, 599–603
biota in this condition (Table 1). 4 Ogura, Y. et al. (2001) A frameshift mutation in NOD2 associated with
susceptibility to Crohn’s disease. Nature 411, 603–606
5 Strober, W. et al. (2007) The fundamental basis of inflammatory bowel
Genetically engineered enteric bacteria for production of bioactives disease. J. Clin. Invest. 117, 514–521
The use of commensal or food-grade bacteria engineered for 6 Baumgart, D.C. and Carding, S.R. (2007) Inflammatory bowel disease:
delivery of therapeutically relevant molecules to the gut cause and immunobiology. Lancet 369, 1627–1640
7 Marchesi, J. and Shanahan, F. (2007) The normal intestinal microbiota.
mucosa is an appealing prospect with many advantages. Curr. Opin. Infect. Dis. 20, 508–513
Proof of principle was established in murine models of 8 Ley, R.E. et al. (2006) Ecological and evolutionary forces shaping microbial
inflammatory bowel disease using a transgenic Lactococcus diversity in the human intestine. Cell 124, 837–848
9 Medzhitov, R. (2007) Recognition of microorganisms and activation of the
lactis to deliver interleukin-10 to the gut, and preliminary
immune response. Nature 449, 819–826
results in humans with Crohn’s disease have been encoura- 10 O’Hara, A.M. and Shanahan, F. (2006) The gut as a forgotten organ. EMBO
ging [37,38]. The strategy is suited to a diversity of applica- Rep. 7, 688–693
11 Turnbaugh, P.J. et al. (2007) The human microbiome project. Nature 449,
tions, such as delivery of vaccines, trefoil factors or other
804–810
bioactive molecules. Containment of the transgenic organ- 12 Bik, E.M. et al. (2006) Molecular analysis of the bacterial microbiota in the
ism has been achieved by deletion and replacement of the human stomach. Proc. Natl. Acad. Sci. U. S. A. 103, 732–737
thymidylate synthase gene with the chosen transgene, 13 Kassinen, A. et al. (2007) The fecal microbiota of irritable bowel syndrome
patients differs significantly from that of healthy subjects. Gastroenterology
thereby rendering the organism dependent on a local supply 133, 24–33
of thymidine or thymine in the gut and unable to survive 14 Shanahan, F (2007) Irritable bowel syndrome: shifting the focus toward
without these in the external environment. the gut microbiota. Gastroenterology 133, 340–342
15 Frank, D.N. et al. (2007) Molecular-phylogenetic characterization of
microbial community imbalances in human inflammatory bowel diseases.
Conclusions Proc. Natl. Acad. Sci. U. S. A. 104, 13780–13785
There is a long-standing tradition in drug discovery from 16 Hooper, L.V. et al. (2001) Molecular analysis of commensal host–microbial
relationships in the intestine. Science 291, 881–884
mining microbes and other biologic material. The gut eco-
17 Sudo, N. et al. (2004) Postnatal microbial colonization programs the
system, long neglected or considered to be impenetrable hypothalamic-pituitary-adrenal system for stress response in mice. J.
owing to technical obstacles, is now well suited to such Physiol. 558 (Pt 1), 263–275
bio-prospecting. Predictions of areas that are ripe for explora- 18 Xu, J. and Gordon, J.I. (2003) Inaugural article: honor thy symbionts. Proc.
Natl. Acad. Sci. U. S. A. 100, 10452–10459
tion and exploitation, based on comparative studies of germ- 19 Macpherson, A.J. and Smith, K. (2006) Mesenteric lymph nodes at the
free and conventionally colonised animals, are already being center of immune anatomy. J. Exp. Med. 203, 497–500
fulfilled. This occurs on a background of an increasing list of 20 Rakoff-Nahoum, S. et al. (2004) Recognition of commensal microflora by
toll-like receptors is required for intestinal homeostasis. Cell 118, 229–241
chronic gastrointestinal disorders, for which some distur-
21 Kumar, A. et al. (2007) Commensal bacteria modulate cullin-dependent
bance of host–microbe interactions appears to be contribu- signaling via generation of reactive oxygen species. EMBO J. 26, 4457–4466
tory. The importance of these conditions is reflected in 22 Cotter, P.D. et al. (2005) Bacteriocins: developing innate immunity for
food. Nat. Rev. Microbiol. 3, 777–788
reviews of the substantial global burden of gastrointestinal
23 Corr, S.C. et al. (2007) Bacteriocin production as a mechanism for the
disease [39,40]. Furthermore, the influence of the gut micro- antiinfective activity of Lactobacillus salivarius UCC118. Proc. Natl. Acad.
biota extends well beyond the confines of the gastrointestinal Sci. U. S. A. 104, 7617–7621

www.drugdiscoverytoday.com 199
Drug Discovery Today: Therapeutic Strategies | Gastrointestinal diseases Vol. 4, No. 3 2007

24 Rea, M.C. et al. (2007) Antimicrobial activity of lacticin 3,147 against 32 Coakley, M. et al. (2003) Conjugated linoleic acid biosynthesis by human-
clinical Clostridium difficile strains. J. Med. Microbiol. 56 (Pt 7), 940–946 derived Bifidobacterium species. J. Appl. Microbiol. 94, 138–145
25 Mazmanian, S.K. et al. (2005) An immunomodulatory molecule of symbiotic 33 Rosberg-Cody, E. et al. (2004) Mining the microbiota of the neonatal
bacteria directs maturation of the host immune system. Cell 122, 107–118 gastrointestinal tract for conjugated linoleic acid-producing
26 Mazmanian, S.K. and Kasper, D.L. (2006) The love-hate relationship bifidobacteria. Appl. Environ. Microbiol. 70, 4635–4641
between bacterial polysaccharides and the host immune system. Nat. Rev. 34 Bäckhed, F. et al. (2004) The gut microbiota as an environmental
Immunol. 8, 777–788 factor that regulates fat storage. Proc. Natl. Acad. Sci. U. S. A. 101, 15718–
27 Shanahan, F. (2005) Physiological basis for novel drug therapies used to 15723
treat the inflammatory bowel diseases I. Pathophysiological basis and 35 Dumas, M.E. et al. (2006) Metabolic profiling reveals a contribution of gut
prospects for probiotic therapy in inflammatory bowel disease. Am. J. microbiota to fatty liver phenotype in insulin-resistant mice. Proc. Natl.
Physiol. Gastrointest. Liver Physiol. 288, G417–G421 Acad.Sci. U. S. A. 103, 12511–12516
28 Obermeier, F. et al. (2003) Contrasting activity of cytosin-guanosin 36 Rousseaux, C. et al. (2007) Lactobacillus acidophilus modulates
dinucleotide oligonucleotides in mice with experimental colitis. Clin. Exp. intestinal pain and induces opioid and cannabinoid receptors. Nat. Med.
Immunol. 134, 217–224 13, 35–37
29 Yan, F. et al. (2007) Soluble proteins produced by probiotic bacteria 37 Shanahan, F. (2004) Making microbes work for mankind—clever trick
regulate intestinal epithelial cell survival and growth. Gastroenterology 132, or a glimpse of the future for IBD treatment? Gastroenterology 127,
562–575 667–668
30 Fujiya, M. et al. (2007) The Bacillus subtilis quorum-sensing molecule CSF 38 Braat, H. et al. (2006) A phase I trial with transgenic bacteria expressing
contributes to intestinal homeostasis via OCTN2, a host cell membrane interleukin-10 in Crohn’s disease. Clin. Gastroenterol. Hepatol. 4, 754–759
transporter. Cell Host Microbe 1, 299–308 39 Sandler, R.S. et al. (2002) The burden of selected digestive diseases in the
31 Grangette, C. et al. (2005) Enhanced antiinflammatory capacity of a United States. Gastroenterology 122, 1500–1511
Lactobacillus plantarum mutant synthesizing modified teichoic acids. Proc. 40 Hellier, M.D. and Williams, J.G. (2007) The burden of gastrointestinal
Natl. Acad. Sci. U. S. A. 102, 10321–10326 disease: implications for the provision of care in the UK. Gut 56, 165–166

200 www.drugdiscoverytoday.com

Das könnte Ihnen auch gefallen