You are on page 1of 14

ARTICLE IN PRESS

Clinical Nutrition (2005) 24, 339–352

http://intl.elsevierhealth.com/journals/clnu

REVIEW

Aetiology of inflammatory bowel disease (IBD):


Role of intestinal microbiota and gut-associated
lymphoid tissue immune response
Oscar C. Thompson-Chagoyána, José Maldonadob, Angel Gilc,
’’
a
Department of Paediatrics, Los Venados’’ General Hospital, Mexican Institute of Social Security,
México City, México
b
Department of Paediatrics, Faculty of Medicine, University of Granada, Granada, Spain
c
Department of Biochemistry and Molecular Biology, School of Pharmacy, Faculty of Pharmacy,
University of Granada, Campus Cartuja, 18071 Granada, Spain

Received 18 February 2005; accepted 21 February 2005

KEYWORDS Summary The aetiology of inflammatory bowel disease (IBD) probably involves a
Inflammatory bowel combination of genetic predisposition and environmental factors that may be
disease; channelled through an abnormality in gut-barrier function, with a loss of antigen
Intestinal tolerance. Some genetic markers that predispose to inflammatory disease have been
microbiota; identified (alleles DR2, DRB1*0103, DRB1*12 and mutations in the NOD2/CARD15
Intestinal tolerance; gene on chromosome 16). Alterations in the pattern of cytokine production by T cell
Probiotics subclasses leading to loss of tolerance to oral antigens have been documented.
Moreover, a number of environmental factors (cigarette smoking, use of non-steroid
anti-inflammatory drugs, psychological stress and the presence of the caecal
appendix) have been postulated as a trigger of IBD. It has also been suggested that
the gut microbiota plays a major role in the development and persistence of IBD, and
numerous modifications of intestinal microbiota composition have been identified.
As a result, manipulation of the microbiota with antibiotics is a current therapeutic
strategy; more recently, however, a number of studies have reported promising
results when using probiotic organisms to manipulate gut microbiota composition in
order to restore tolerance to microbial antigens of the host’s own microbiota.
& 2005 Elsevier Ltd. All rights reserved.

Corresponding author. Tel.: +34 958 246139; fax: +34 958 248960.
E-mail address: agil@ugr.es (A. Gil).

0261-5614/$ - see front matter & 2005 Elsevier Ltd. All rights reserved.
doi:10.1016/j.clnu.2005.02.009
ARTICLE IN PRESS
340 O.C. Thompson-Chagoyán et al.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 340

Aetiological theories of IBD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 341


Inappropriate reaction to a specific persistent pathogenic infection of the intestine . . . . . . . . . . . . . . . . 341
Subtle alterations in bacterial composition and function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342
Overexposure to normal resident luminal bacterial products that induce an impairment of the mucosal
barrier . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342
Aberrant host immune response including loss of tolerance to normal ubiquitous luminal antigens . . . . . . . 342

Intestinal microbiota in healthy and IBD subjects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342


Functions of gut microbiota . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
Metabolic functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
Trophic functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
Protective functions: the barrier effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 343
Intestinal microbiota in human IBD patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 344
Immune response to antigens in the gut . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 344

Immune response and oral tolerance in IBD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 344


Mechanisms of oral tolerance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345
Cellular mechanisms of oral tolerance. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 346
Active suppression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 346
Clonal anergy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 346
Clonal deletion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 347

Potential roles of probiotics in the treatment of IBD: A way of inducing oral tolerance . . . . . . . . . . . . . . . . . 347
Probiotics in experimental models of colitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348
Evidence supporting the successful response to probiotics for the prevention or treatment of IBD in
animals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348
Probiotics in human inflammatory bowel disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 348
Possible mechanisms of probiotic action in IBD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349

Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350

Introduction respond in vitro to the antigens of their own flora


with a Th1 polarised response, indicating a loss of
Inflammatory bowel disease (IBD) is a chronic tolerance to their intestinal microbiota.5 This Th1
inflammatory condition of the gastrointestinal tract response induces an increment in the production of
that manifests as ulcerative colitis (UC) or Crohn’s INF-g by CD4 and activated macrophages that
disease (CD). Notwithstanding intensive research, secrete TNFa and IL-126 (Fig. 1).
the aetiology of this condition remains unknown; In UC a positive association has also been
however, it is thought to result from a combination reported with DR2, DRB1*0103 and DRB1*12 al-
of genetic predisposition and environmental factors leles,7,8 and genome-wide scanning studies have
that may be channelled through an abnormality in shown linkage between UC and regions of chromo-
gut-barrier function.1 somes 3, 7, and 12.7 An increasing number of single
Between 17% and 25% of patients with CD have nucleotide polymorphisms and alleles associated
mutations in the NOD2/CARD15 gene on chromo- with IBD have been reported [186 for 1997, 261 for
some 16 that predispose to CD.2,3 The NOD2/ 1999 and 590 for 2003 for B HLA-1 polymorphism;
CARD15 gene codes for a protein that activates 4235 and 70 for C HLA 1 polymorphism, and 83,125
monocyte NFkB in response to bacterial lipopoly- and 282 for A HLA-1 polymorphism, respectively].
saccharides, and it has been suggested that the Similar increases have also been reported for HLA-
NOD2/CARD15 gene product confers susceptibility A*02 alleles in the same years (10, 22 and 65,
to CD by altering the recognition of bacterial respectively). Moreover, it has very recently been
components and/or altering the activation of host proposed that the differential expression of HLA-G
response to bacteria.4 It has been demonstrated in UC and CD provides a potential way to distinguish
that T cells from the lamina propria in CD patients between these two entities.9
ARTICLE IN PRESS
Aetiology of IBD 341

Tolerance Inflammation

sIgA Antigens

sIgA

TGF-β
INF-γ, IL-2
Apoptosis TNF-α
Th3
APC
IL-10
IL-4 Peyer’s patches

Th2 Th1
CD-4

IL-10 IL-12
TNF-α
Thr Systemic recirculation

Figure 1 Tolerance vs. inflammation in gut-associated lymphoid tissue (GALT). APC: antigen-presenting cell; IL:
Interleukin; INF g: Interferon g; sIgA: secretory IgA; Th1, Th2, Th3 and Thr: T helper lymphocyte subclasses; TGF-b:
transforming growth factor b; and TNF-a: tumour necrosis factor a.

Several environmental factors associated with could have a beneficial impact on IBD, influencing
IBD have been reported, and many researchers both microbiota composition and GALT immune
believe that environmental factors are more responses. Current data on probiotics and IBD are
important than genetic factors, at least in UC; reviewed here.
the main argument adduced in support of this claim
is the low rate of concordance between mono-
zygotic twins with UC (6–14%) compared with twins
Aetiological theories of IBD
affected by CD (44–50%).8
Other factors frequently associated with IBD3,8
IBD is thought to be due to an impairment of
include cigarette smoking (protective to UC and
functional immune responses. The increase in
detrimental to CD), use of non-steroid anti-inflam-
mucosal B cell and plasma cell populations in IBD
matory drugs, psychological stress and the pre-
suggests that the disease is antibody-mediated and
sence of caecal appendix (appendectomy at a
complement-dependent, but the main abnormality
young age has a protective effect on development
driving the inflammation is an exaggerated T cell
of UC), but the microbial factor is the strongest
response causing mucosal hyper-responsiveness to
environmental factor studied until now. Although a
commensal bacteria.11
long search has so far failed to confirm a direct
pathogenic role for a specific infectious agent,
ample evidence suggests that commensal enteric
’’ Inappropriate reaction to a specific
bacteria and their products are the triggers’’ that persistent pathogenic infection of the
initiate and perpetuate IBD.10 intestine
The aim of this review is to examine some of the
main factors influencing the onset and develop- It has long been proposed that resident bacterial
ment of IBD focusing on the alteration of intestinal flora play an essential role in the pathogenesis of
microbiota and gut-associated lymphoid tissue human IBD. Indeed, direct interaction of commen-
(GALT) immune responses leading to loss of oral sal microbiota with the intestinal mucosa is
tolerance to antigens. Since IBD may result from believed to stimulate inflammatory activity in gut
alterations in intestinal bacterial composition and lesions. Because CD and UC closely mimic defined
overexposure to luminal bacterial products, the intestinal infections, and occur in areas with the
review will focus on how faecal microbial colonisa- highest luminal bacterial concentrations, many
tion and intestinal microbiota composition may microbial pathogens have been suggested as
affect the course of the disease. By contrast, some causes of IBD. These include: Mycobacterium
microorganisms, mainly lactic acid bacteria (LAB), paratuberculosis and M. kansaii, Escherichia coli,
ARTICLE IN PRESS
342 O.C. Thompson-Chagoyán et al.

Diplostreptococcus sp, Virus (measles, RNA virus), bacterial products, toxins, and food antigens.
Lysteria monocytogenes, Fusobacterium necro- Normal luminal bacteria may induce and perpe-
phorum, Chlamydia sp., Listeria monocytogenes, tuate intestinal inflammation in IBD patients by
Pseudomona maltophila and Helicobacter hepati- initiating an aberrant host immune response,
cus.10,12 Moreover, faecal stream diversion has although in most cases no specific pathogen can
been shown to prevent recurrence of CD, whereas be identified.
infusion of intestinal contents to the excluded ileal Progression or resolution of acute inflammation
segments reactivates mucosal lesions.12 after bacterial stimulation depends on the delicate
balance between proinflammatory and immunosup-
Subtle alterations in bacterial composition pressive forces.10,13 It has been shown that both
and function mice and humans are normally tolerant to auto-
logous microbiota, and that breakdown of this
The gut microbiota represents an extremely com- tolerance is associated with the development of
plex ecosystem. Because the microbiota in healthy chronic intestinal inflammation.5 It may be that
humans is relatively stable when diet remains potentially pathological responses to components
unchanged, subtle alterations in bacterial composi- of the intestinal flora do occur under physiological
tion and function may have profound implications conditions, but these are suppressed by immunor-
for mucosal barrier function and immune response. egulatory mechanisms.15
Some modified E. coli strains have been reported in There is increasing evidence that the antigens
patients with UC and CD; similarly, patients with driving the tissue immune response are derived
IBD have larger amounts of bacteria attached to from normal bacterial flora. Proof of this concept
their epithelial surfaces than do healthy people. has come from studies of at least 11 models of IBD,
Isolated bacteria belong to a wide range of genera; in which inflammation is found to be dependent on
some are identified within the epithelial layer, the presence of normal flora; the absence of
while others are found in intracellular locations. normal flora is associated with non-appearance of
Nevertheless, the role of these altered bacteria in the illness.13,16–18 This phenomenon is seen in
IBD pathogenesis remains unclear.13,14 different species (mice, rats and guinea pigs), and
occurs in manipulated organisms such as transgenic
mice with targeted deletion of the T cell receptor
Overexposure to normal resident luminal
(TCRa), that spontaneously develop colitis in
bacterial products that induce an response to the gut microbiota.19 It also has been
impairment of the mucosal barrier observed in rats and mice with IBD treated with
broad-spectrum antibiotics, which have been
An alternative mechanism by which enteric micro- shown to mitigate mucosal inflammation.20 More-
bial agents could stimulate chronic enterocolitis over; in some models, colonisation with normal
and systemic inflammation is by means of an flora rapidly results in T-cell-mediated gut inflam-
impairment of the intestinal mucosal barrier, mation, and disease can be transferred to other
allowing increased uptake of antigens and proin- animals using activated T cells against enteric
flammatory molecules, including endotoxin and bacteria.21 Nevertheless, it appears that a constant
other bacterial products. It has been reported that bacterial stimulus is needed for the induction and
mucosal permeability is increased in most intestinal continuation of inflammatory process21 and that
inflammatory conditions. In such cases, bacterial not all commensal bacteria have an equivalent
components can activate intestinal immune cells, ability to induce mucosal inflammation, but instead
endothelial cells and epithelial cells to secrete are influenced by the host genetic background.22
many substances (cytokines, eicosanoids, oxygen
metabolites, nitrogen oxide and proteases), indu-
cing local damage and inflammation that contri-
butes to disrupting mucosal barrier function.10 Intestinal microbiota in healthy and IBD
subjects
Aberrant host immune response including
loss of tolerance to normal ubiquitous The alimentary tract is a specialised organ that
luminal antigens extends from the lips to the anus, and represents
the largest contact area between the body and the
The inflammatory bowel syndrome may be the external environment.23 It has been estimated that
result of an abnormal host response to ubiquitous the adult human body contains 1014 cells, of which
antigens, including the normal resident microbiota, only 10% constitute the body proper; the remaining
ARTICLE IN PRESS
Aetiology of IBD 343

90% are microbiota cells, mostly living in the lumen and proliferation. Fermentation of carbohydrates is
of the large intestine.13,23–28 The fact that we are a major source of energy in the colon. Non-
90% bacteria is frequently overlooked, and the digestible carbohydrates include large polysacchar-
significance of the relationship between prokar- ides (resistant starches, cellulose, hemicellulose,
yotes and eukaryotes is only just beginning to be pectins, and gums), some oligosaccharides that
appreciated. escape digestion, and unabsorbed sugars and
The mammalian intestinal tract hosts a complex, alcohols.32 The metabolic endpoint is the genera-
dynamic, and diverse society of bacteria. Although tion of the short-chain fatty acids (SCFA) acetate,
the precise quantitative and qualitative composi- propionate and butyrate. Anaerobic metabolism of
tion of the indigenous digestive microbiota is not peptides and proteins (putrefaction) by the micro-
yet known, more than 100 different bacterial biota also produces SCFA but, at the same time, it
species have been identified in faecal specimens generates a series of potentially toxic substances
from normal individuals, and it is thought that at included ammonia, amines, phenols, thiols, and
least 500 species are present in the digestive indols.29
tract.24,25,29 On a quantitative basis, approximately Intestinal microorganisms are also involved in
10–20 genera probably predominate,25 but the real vitamin synthesis29 and in the absorption of
magnitude of the intestinal microbiota is under- calcium, magnesium and iron.33 Absorption of ions
lined by the fact that the number of genes it in the caecum is improved by carbohydrate
contains is about 50–100 times that of our own fermentation and SCFA production. All of these
genome.28 fatty acids have important functions in host
The gastrointestinal microbiota can be consid- physiology.29
ered a functionally and metabolically adaptable
and rapidly renewable organ of the body.4,13,30 Its Trophic functions
relationship with the host is so specific that any Possibly the most important role of the intestinal
alteration in the balance of organisms might result microbiota in colon physiology is its trophic effect
in illness; some of its members are considered on the intestinal epithelium.29 Epithelial cell
potentially harmful in view of their involvement in differentiation is considerably influenced by inter-
toxin production, mucosal invasion, or activation of action with resident microorganisms and their
carcinogens and inflammatory responses. However, metabolic products, mainly short chain fatty acids;
more recently attention has turned to indigenous all of these stimulate epithelial cell proliferation
non-pathogenic microorganisms and the ways in and differentiation of the small and large bowel.34
which they benefit the host,13,25,31 since there is
evidence that they play a major role in host Protective functions: the barrier effect
nutrition, physiology, and control of the immune Intestinal bacteria form a barrier against non-
system.23 indigenous microorganisms, including pathogens.35
All mechanisms that do not allow the installation of
pathogenic bacteria in the human gut are said to
Functions of gut microbiota
exert a mucosal barrier effect. Intact indigenous
flora are known to represent a formidable barrier to
Evidence obtained with animals feed under germ-
the establishment of pathogenic populations on
’’
free conditions suggests that gut microbiota have
host surfaces; this phenomenon is called bacterial
’’
important and highly specific metabolic, trophic,
antagonism’’, bacterial interference’’ or more
and protective functions.29
’’
commonly colonisation resistance’’. This barrier
effect includes many activities such as production
Metabolic functions of bacteriocins by indigenous flora components,
One major metabolic function of the intestinal production of metabolic products which will be
microbiota is the fermentation of non-digestible toxic for pathogens, conditions inhibitory to patho-
dietary residue and endogenous mucus produced by genic organisms, such as low pH and depletion of
the epithelia.29 Gut-bacteria metabolism is respon- nutrients required for their multiplication.36 Ad-
sible for the conversion of many substances into herent non-pathogenic bacteria can prevent at-
metabolites that can be absorbed and used by the tachment and subsequent entry of pathogenic
host (60–70% of their energy requirements).29 The enteroinvasive bacteria into epithelial cells.37
overall outcomes of this complex metabolic activity Furthermore, bacteria compete for nutrient avail-
are the recovery of metabolic energy and absorb- ability in ecological niches, and maintain their
able substrates for the host, and the supply of collective habitat by administering and consuming
energy and nutritive products for bacterial growth all resources. The host actively provides a nutrient
ARTICLE IN PRESS
344 O.C. Thompson-Chagoyán et al.

that the bacterium needs, and the bacterium regulatory cells (Tr) are the main source of IL-
actively indicates to the host how much it needs. 10.40,41 An altered pattern of cytokines is observed
This symbiotic relationship prevents unwanted in IBD compared with tolerant subjects (Fig. 1).
overproduction of the nutrient, which would favour Increased release of IL-2, INF-g TNF-a, which
intrusion of microbial competitors with potential mediate cellular immune responses, and decreased
pathogenicity for the host.29 Finally, bacteria can release of IL-4, IL-5, IL-6, IL-10, which are
also inhibit the growth of their competitors by pH implicated in humoral immune responses, in con-
modification, control of motility, nutrient depletion junction with decreased secretion of TGF-b by Th3
and production of anti-microbial substances called cells and IL-10 by Thr, are well known to occur in
bacteriocins.30 IBD (Table 1).
Lack of peripheral T cell response may be
Intestinal microbiota in human IBD patients achieved in three different ways: (1) the T cells
encounter the antigen in an irrelevant immunolo-
Major modifications of the intestinal microbiota gical form and are thus unaware of it; (2) the T cells
reported in patients with IBD include: high con- may encounter the antigen in circumstances that
centrations of mucosal bacteria in both inflamed result in the subsequent functional or actual
and non-inflamed bowel tissue14; increased num- elimination of the cell (anergy or deletion); (3)
bers of coliforms and bacteroids, and a decrease in regulatory cells or mediators can modify the
LAB38; and increased E. coli numbers in the colon.39 immune response.
Whether antigens induce peripheral immunity or
tolerance may be dependent on costimulatory
Immune response to antigens in the gut signals like CD40, CD44v7, CD80 and CD86, all of
them provided to the T cells by APC.15,40
After reaching the lamina propria, the antigens
encounter various host defence mechanisms and
are internalised and processed by antigen-present-
ing cells (APC) for presentation to T cells. Recogni- Immune response and oral tolerance in
tion of antigen by T cells requires interaction IBD
between the antigen, the major histocompatibility
antigen complex on the surface of the APC and the The gastrointestinal tract provides a protective
T cell antigen–receptor complex (TCR) on the T interface between the internal environment and
helper cell. An appropriate interaction leads to the constant challenge from food-derived antigens
activation of the T cell. T cells are classified as CD4+ and from microorganisms in the external environ-
and CD8+ T cells; CD8+ cells (suppressor cells) have ment. A mutual interdependence exists between
a downregulatory influence on the immune system the gut microbiota and the host immune system.
and can kill cells infected with cytosolic pathogens. Successful coexistence with a complex microbiota
CD4+ cells (helper cells) are currently divided into presents a particular challenge to the immune
four types—Th1, Th2, Th3 and Tr—distinguishable system of the host. On the one hand, the host needs
by their differential pattern of cytokine secretion: to avoid an overly aggressive response to this
Th1 cells release interleukin (IL)-2, interferon (INF) microbial population that would lead to the
g, and tumour necrosis factor (TNF)-a; Th2 cells elimination of beneficial organisms and would
secrete IL-4, IL-5, IL-6 and IL-10; Th3 cells secrete almost certainly result in inflammation and exten-
mainly transforming growth factor (TGF)-b; and T sive tissue damage. On the other, the ability to

Table 1 Alteration in cytokine production by CD4+ Th cells in IBD.

CD4+ T cell (Helper T cell)

mTh1 CD4+ kTh2 CD4+ kTh3 kThr


mRelease kRelease kRelease kRelease
IL2 IL-4 TGF-b IL10
IFN-g IL-5
TNF-a IL-6
IL-10
Mediate cellular immune responses Mediate humoral immune responses
ARTICLE IN PRESS
Aetiology of IBD 345

limit the spread of bacteria from the lumen into Mechanisms of oral tolerance
underlying tissues and to mount an effective
response to intestinal pathogens needs to be The presence of large numbers of immunogenic and
maintained.4 Mucosal immune response and non- pro-inflammatory molecules in the intestinal lu-
response are marked by their ability to distinguish men, together with evidence for the interaction of
between the antigenic challenge posed by patho- the host’s immune system with this material, has
genic invaders, and not dangerous resident organ- led to the concept that the gut mucosa is in a state
isms or proteins in the food stream. of restrained immune reactivity. This state has
’’ ’’
The first line of host defence is directed towards been termed controlled’’ or physiological’’
the exclusion of antigens, the elimination of foreign inflammation4; when the body comes into contact
antigens that have penetrated the mucosa, and the with a specific antigen, it should be able to identify
regulation of the ensuing antigen-specific immune whether this molecule is dangerous, whether it
response. As a result, the gastrointestinal barrier must be eliminated or if it is possible to tolerate its
controls antigen transport and the generation of presence.
immunological phenomena in the gut; but even in The best-understood mechanism of immune
physiological conditions, an insignificant but im- tolerance is the central tolerance established in
munologically important fraction of antigens bypass thymus and bone marrow. However, not all antigens
the defence barrier4 and the intestinal immune are present in the lymphoid organs; therefore,
system is faced with the dilemma of discriminating central tolerance must be complemented by addi-
between antigens with no pathogenic potential and tional mechanisms of peripheral tolerance. Figure 2
antigens and microorganisms that are potentially depicts some of the most important mechanisms
harmful. It has long been recognised that the involved in central and peripheral tolerance. Most
encounter of dietary antigens by the immune data concerning this selective immune response
system normally leads to abrogation of the immune ’’ have been obtained from experimental animals
response. This phenomenon has been termed oral (ovalbumin-TCR transgenic mice). When an antigen
tolerance’’41 and is defined as the induction of a is tolerated, the site of tolerance induction and the
state of systemic immune non-responsiveness to type of APC generating the tolerogenic immune
orally administered antigen upon subsequent anti- response are not clear, although T cells appear to
gen challenge. This mechanism presumably pre- be the major target of tolerance, and the reduction
vents the development of an immune reaction or in antibody responses after antigen feeding is due
allergy against intestinal intraluminal antigens. to the reduction in T helper activity.42

CD4+ CD8+
Thymus

CD4+ CD8+ CD4+ CD8+

Peripheria

CD4+ +
CD8+

CD8+ CD45hi CD4+ CD4+


CD45RBhi CD45RBlow
Th2
- +

Autoimmunity and inflammation TGF-β


IBD, experimental granulomatose IL- 4
colitis, autoimmune thyroiditis IL-10 ?

Figure 2 Mechanisms of central and peripheral tolerance. IBD: inflammatory bowel disease; IL: Interleukin; and TGF-b:
transforming growth factor b.
ARTICLE IN PRESS
346 O.C. Thompson-Chagoyán et al.

Cellular mechanisms of oral tolerance of antigen-specific effector cells. These suppressor


It is accepted that the major mechanisms of cells can cause cytokine suppression in other
tolerance induction are the development of sup- lymphoid cells in the surrounding area; this
’’
pressor T cells and the induction of clonal deletion phenomenon is known as bystander suppression’’
and/or anergy clonal deletion, and it is believed and a specific antigen is required for the activation
that different mechanisms are favoured at differ- of suppressor cells. When these activated cells
ent antigen doses40: low doses of antigen favour reencounter the same antigen, they release TGF-b
active suppression, whereas high doses are that inhibits the response of other neighbouring T
mediated principally by anergy or clonal dele- cells that are sensitive to antigens other than those
tion28,40 (Fig. 3). initiating an antigen-mediated response.44,45

Active suppression Clonal anergy


Active suppression occurs when T cells obtained The administration of high doses of antigen
from an antigen-fed donor animal and transferred produces clonal anergy. Clonal anergy exists when
to a recipient animal induce unresponsiveness to T cells are unable to secret IL-2 or to proliferate;
the administration of any antigen. Without prior this process is not passive, and seems to be
cell transfer, the administration of the same preceded by activation and proliferation of anti-
antigen induces a strong immune response.43 The gen-specific T cells in lymph nodes.46 The tolerance
transferred cells are CD4+ T cells originating in induced after administration of high doses of
Peyer’s patches,15,43 and the primary mechanism of antigen resembles the tolerance induced by admin-
active cellular suppression is via the secretion of istering antigen intravenously, but it is now
suppressive cytokines. The crucial contributor to accepted that high-dose oral tolerance occurs due
oral tolerance appears to be TGF-b, and the to the absence of adequate costimulation, since T-
response could be summarised as follows: after cell activation requires two signals, one via TCR
the generation of antigen-specific regulatory cells interaction with the antigen–MHC complex and a
in GALT, these cells migrate to peripheral lymphoid second via the B7–CD28 interaction. If the costi-
organs, and suppress the immune response by mulatory signal is missing, the T cell will undergo
secreting TGF-b, thereby inhibiting the generation deletion or become anergic. Likewise, B7 interacts

Oral administration of antigens

Macrophages, dendritic cells,


APC B cells, enterocytes

APC-T cell Interaction

Low doses High doses

T cell
T cell

Th2 Th3 Th1 Th2

IL-4, IL-10 TGFβ Anergy, deletion

Inhibition of autoimmunity Oral tolerance


and inflammation

Figure 3 Mechanisms of dose-dependent oral tolerance in the gut-associated lymphoid tissue (GALT). APC: antigen-
presenting cell; IL: Interleukin; TGF-b: transforming growth factor b; and Th2: T helper lymphocyte subclass 2.
ARTICLE IN PRESS
Aetiology of IBD 347

APC

B7s B7s

TGF-β
MHC II Th3

TCR
CD3 CD28 -
CTLA-4
T cell

Th1
IL-12
T cell
IL-2
TGF-β Activation
IL-4
- Th2

Figure 4 Function of TGF-b in the suppression of T cell response mediated by CTLA-4 receptor. APC: antigen-presenting
cell; MHCII: major hystocompatibility complex II; TGF-b: transforming growth factor b; and Th1, Th2, Th3 and Thr: T
helper lymphocyte subclasses.

with CTLA-4 leading to TGF-b secretion and im- immunological strategies, including administration
portant molecule in the induction of oral tolerance of neutralising monoclonal antibodies or anti-
since it inhibits T cell activation41 (Fig. 4). inflammatory cytokines with the intention of
modulating the immune alterations causing IBD.47
Clonal deletion Nevertheless, for various reasons long-term
This mechanism has been observed under non- therapy is often the fate of patients suffering from
physiological conditions in transgenic animals, and IBD; unfortunately, however, unpleasant side ef-
occurs following the activation of T cells by strange fects may result from drug use. Accordingly,
proteins.46 researchers have sought to develop methods which
allow for a more localised delivery of therapeutics.
The most widely used techniques are: colonic
Potential roles of probiotics in the instillation of conventional chemical therapy or
treatment of IBD: A way of inducing oral interleukins (IL-4 and IL-10), administration of SCFA
tolerance enemas,48 intranasal instillation of DNA delivery
systems,47 and injection of IL-4 using human
In the absence of an aetiological therapy, the adenovirus as a vector.49 Finally, due to the possible
inflammatory process itself has become the current role of indigenous microbiota in the pathogenesis of
target for the treatment of IBD. A large number of IBD,11 the external manipulation of its composition
chemical compounds have been used in an attempt using probiotic organisms seems to be a promising
to revert to the non-inflammatory state, as well as therapeutic form.
for the maintenance of the remission state. The It appears that normal humans develop tolerance
manipulation of intestinal microbiota has received to their own microbiota and do not mount an
little attention, and the few studies addressing this immune response to these bacteria. However, IBD
option have been carried out with antibiotics. patients seem to lose this tolerance and their
Although corticoid drugs remain the most effective immune response is upregulated50; for this reason,
treatment for active IBD, other therapeutic agents and with the support of findings in experimental
such as mesalazine, azathioprine, 6-mercaptopur- models, it has been postulated that IBD is the result
ine, methotrexate and cyclosporine have also been of genetically determined defective immunosup-
tested.47 With the knowledge that activated T cells pression resulting in an overly aggressive response
can produce pro-inflammatory and anti-inflamma- to ubiquitous luminal bacteria constituents.14,17,38
tory cytokines, physicians have begun to use It has been suggested that manipulation of
ARTICLE IN PRESS
348 O.C. Thompson-Chagoyán et al.

Table 2 Criteria for considering bacteria as develop disease when they are reared in a germ-
probiotics. free environment. However, once the normal
intestinal microbiota is restored, inflammatory
Human origin disease occurs.56
Non pathogenic for the host It has likewise been demonstrated that among
Resistant to destruction by gastric acid, bile or the commensal gut microbes colonising both IBD
technical processing patients and experimental animals, anaerobic
Be able to adhere to gut epithelial tissue
bacteria are thought to be responsible for the
Capacity to colonize the gastrointestinal tract
Produce antimicrobial substances development of immune response and inflamma-
Modulate immune responses tion.57,58
Influence host metabolic activities Finally, in mice with IBD, the tolerance to
commensal microorganisms is lost while colitis is
Source: Annu. Rev. Nutr. 2002;22:107–38. active.5

intestinal bacteria may be a useful approach in the


Evidence supporting the successful response to
treatment of these patients, and that probiotics
probiotics for the prevention or treatment of IBD
offer a promising option.51 in animals
Probiotics are live microbial food supplements
Several studies in animal IBD models have yielded
that beneficially affect the host by improving its
encouraging results, although they have also shown
intestinal microbial balance; when administered in
that a single microorganism may not be ideally
adequate amounts, they confer a health benefit.52
suited to all patients; use of probiotic cocktails has
Recent research has expanded the definition of
been tested with promising results in patients with
probiotics, as it has been shown that genetically
UC,59 but more rigorous scientific study is required
engineered microbes and non-viable microbes may
before they can be recommended for routine use.
equally possess such potential.53 Current criteria The most promising results obtained to date are as
for defining probiotics are shown in Table 2.
follows:
There is some evidence that administration of
probiotics could be effective in the prevention and
treatment of infant acute infectious diarrhoea, 1. Administration of Lactobacillus reuteri prevents
mainly of viral origin, and in the prevention of acetic acid colitis in a rat model.60
antibiotic-associated diarrhoea, as well as in the 2. Ingestion of Lactobacillus, and especially of L.
treatment of Helicobacter pylori infection and in plantarum, improves colitis induced by intraper-
the prevention and management of atopic diseases. itoneal methotrexate administration in rats.61
One of the mechanisms of action is the modification 3. In the neonatal period, IL-10 gene-deficient
of the host gut microbiota.54 mice have decreased levels of Lactobacillus sp.
In view of these preliminary successes, attempts in the colon. Daily anal administration of
have been made to modify the microbiota in lactobacilli reduces colonic mucosal adherent
patients with IBD: The scientific rationale for and translocated bacteria and prevents colitis.18
therapeutic manipulation of enteric microbiota in In the same model, Lactobacillus salivarius
IBD can be divided into experimental and human inhibits the development of both colitis and
evidence. colon cancer.62
4. Administration of a non-pathogenic E. coli
(serotype O6:K5:H1) prevents relapses of UC.63
Probiotics in experimental models of colitis 5. Genetically engineered Lactobacillus lactis se-
creting active IL-10 has been used successfully in
The findings of several studies suggest that some the treatment and prevention of IBD in murine
bacterial products influence epithelial proinflam- models.64
matory responses and exchange regulatory signals
with subepithelial cells and the mucosal immune Probiotics in human inflammatory bowel
system51; as a result, it is difficult to induce disease
intestinal inflammation in germ-free animals.55
Similarly, while transgenic rats with normal intest- The main evidence supporting the involvement of
inal flora spontaneously develop colitis, in those the gut microbiota in IBD is as follows:
bred in germ-free conditions, the inflammation is
either attenuated or non-existent.16 Moreover, IL- 1. High mucosal bacteria counts are reported in
2, IL-10, and T-cell receptor knockout mice17 do not patients with bowel inflammation, and counts
ARTICLE IN PRESS
Aetiology of IBD 349

increase progressively with the severity of 3. Short-term administration of Lactobacillus GG in


disease, in both inflamed and non-inflamed children with CD increases the intestinal IgA
colon tissue.14 immune response71; long-term administration of
2. It has been demonstrated that T cells from the this probiotic reduces intestinal permeability
lamina propria of CD patients respond in vitro and improves clinical status.72
to antigens of their own microbiota with a Th1 4. Saccharomyces boulardii, in association with
polarised response, indicating a loss of toler- mesalazine, increases the time without relapses
ance of their normal microbiota.4 in patients with stable CD.73
3. Among the commensal intestinal microbes 5. In 120 patients with UC, 12-week administration
colonising both IBD patients and experimental of a non-pathogenic E. coli strain was compared
animals, anaerobic Gram positive bacteria, with mesalazine; no differences were found in
particularly those of the Bacteroides genus, the proportion of remission at the end of the
are thought to be responsible for the develop- study63; when the E. coli strain was admini-
ment of inflammation and immune response,65 strated over 1 year, it prevented relapses.68
and evidence suggests that bifidobacteria 6. In patients with pouchitis, treatment with a
might be used as a probiotic to ameliorate combined preparation of probiotics (four strains
IBD through the suppression of Bacteroides in of Lactobacilli, three strains of Bifidobacteria
the intestinal microbiota.58 and Streptococcus salivarius) containing 5  1011
4. Coliforms and Bacteroides are potentially bacteria per gram dramatically reduced relapses
pathogenic to the gut mucosa, and an increase (15% in patients with probiotics vs. 100% of the
in both bacteria, together with a decrease in placebo group); 4 months after the end of study,
LAB, has been reported in patients with IBD.38 all patients with bacteriotherapy were in remis-
5. Normal gut microorganisms have a considerable sion.74 The same mixture was successful as a
influence on the integrity and development of maintenance treatment of UC in an open trial.75
the intestinal mucosa.36,66 7. Oral administration of Lactobacillus casei strain
6. UC shares certain histological features with GG leads to a significant increase in mucosa IgA
infective colitis.67 levels, suggesting an improvement of the im-
7. Lymphocytes in contact with an extract of E. munological barrier in patients with CD.71
coli lipopolysaccharides exhibit cytotoxicity to 8. Treatment with E. coli strain Nissle 1917 reduces
colon cells; patients with UC are more likely to the risk of relapse and decreases corticoid
carry E. coli strains with these lipopolysacchar- requirements in patients with CD.76
ides.68
8. Tobramycin, an antibiotic with activity against
Gram negative bacteria, when administrated Possible mechanisms of probiotic action in
orally together with corticosteroids, prompts IBD
an improvement in patients with active UC.69
9. An exaggerated antibody response against the There are a number of possible mechanisms by
intestinal microbiota has been reported in IBD which probiotics produce beneficial effects in IBD.
patients.57 These mechanisms, which influence most of the
10. Surgical bypass leads to remission in patients interactions between the host and his/her intest-
with IBD and its reconnection produces a inal microbiota, include:
relapse.39 Production of inhibitory substances: including
11. High serum E. coli antibody levels are reported the modification of pH and the production of
in IBD patients; this species has been isolated in organic acids, hydrogen peroxide and bacteriocins.
65% of patients with CD.39 These compounds reduce the number of viable cells
and affect bacterial metabolism or toxin produc-
A number of trials in patients with IBD report tion.70
successful use of probiotics: Blocking of adhesion sites: displacement of
harmful microorganisms may be a consequence of
1. The administration of Lactobacillus GG may be physical competition. The ability of probiotics to
an effective therapeutic agent for pouchitis, a adhere to epithelial cells produces competitive
complication of ileal reservoir surgery occurring inhibition of bacterial adhesion sites, thus impeding
in 10–20% of patients undergoing surgical treat- invasion by foreign organisms.29,47,77 Also, some
ment for chronic UC.70 bacteria are able to block the intestinal mucosal
2. Bifidobacteria ameliorates IBD through suppres- receptor, thus preventing the establishment of
sion of Bacteroides in the gut.58 adhesive pathogenic bacteria.68
ARTICLE IN PRESS
350 O.C. Thompson-Chagoyán et al.

Competition for essential nutrients: probiotics luminal bacterial products. Moreover, it has been
utilise nutrients that would otherwise be consumed observed that patients with UC and CD have
by potentially harmful microorganisms, thus inhi- abnormal intestinal microbiota and this alteration
biting their growth.70 induces the release of pro-inflammatory mediators.
Degradation of toxin receptor: this mechanism Loss of tolerance to indigenous bacteria could be
protects against Clostridium difficile intestinal implicated in the appearance of IBD; mechanisms
disease.78 involved include the breakdown of active suppres-
Stimulation of immunity: there is strong evi- sion, clonal anergy, clonal deletion and the release
dence to suggest that probiotics modulate both of pro-inflammatory cytokines.
innate and acquired immunity. Probiotics could Treatment with probiotics represents an alter-
interact with indigenous bacteria and/or host native to current drug treatment in IBD patients. In
mucosal cells to induce or modulate the immune these cases, previous intestinal ecology can be
response, modifying the number of CD4 T cells; it is restored by manipulating the microbiota through
well known that probiotics can actively interfere administration of probiotic bacteria. Probiotics
with anti-inflammatory and proinflammatory sig- could also be genetically modified to release
nalling pathways, inducing production of IL-10 and specific anti-inflammatory cytokines.
reducing INF and TNF release.39 Finally, the It must be remembered that, despite the
increase in local IgA levels and IgA-secreting plasma potential for application of probiotics in the
cell numbers found in IBD patients results from the treatment and prevention of IBD, more controlled
ingestion of probiotics.47,71 studies are required before their routine use can be
Promotion of gut integrity: Nitric oxide (NO) at recommended.
high concentrations is a very potent pro-inflamma-
tory stimulator. At low levels however, NO is a
promoter of gut integrity. It has been proven that References
Lactobacillus plantarum has the ability to produce
NO in low doses with beneficial effects on the 1. Blumberg RS, Strober W. Prospects for research in inflam-
intestinal mucosa.47 It has also been observed that matory bowel disease. JAMA 2001;285:643–7.
disruption of the epithelial barrier leads to a large 2. Hugot JP, Chamaillard M, Zouali H, et al. Association of NOD2
leucine-rich repeat variants with susceptibility to Crohn’s
influx of antigen into the mucosa and therefore to disease. Nature 2001;411:599–603.
an enhanced intestinal immune response. In addi- 3. Gil A. Interacciones medio ambiente genes. Enfermedad
tion, the administration of L. casei or Clostridium Inflamatoria Intestinal al dı́a 2004;3:38–44.
butyricum markedly enhances gut epithelial cell 4. Hart AL, Stagg AJ, Frame M, et al. Review article: the role of
the gut flora in health and disease, and its modification as
proliferation (up to 200% in rat colon), thus
therapy. Aliment Pharmacol Ther 2002;16:1383–93.
enhancing tissue repair.79 5. Duchmann R, Kaiser I, Hermann E, Mayet W, Ewe K, Meyer
It has been suggested that SCFA (acetate, zum Büschenfelde KH. Tolerance exists towards resident
propionate and butyrate) deficiency promotes intestinal flora but is broken in active inflammatory bowel
colitis,80 and that probiotic bacteria have the disease (IBD). Clin Exp Immunol 1995;102:448–55.
capacity to produce SCFA,77 which in turn promote 6. Stallmach A, Marth T, Adrian N, et al. Increased expression
of interleukine-12 receptor beta (2) on lamina propria
mucosal intestinal restitution.47 mononuclear cells of patients with active Crohn’s disease.
Finally, probiotic bacteria can increase the gut Int J Colorectal Dis 2002;17:303–10.
epithelial mucus that provides a barrier shielding 7. Satsangi J, Welsh KI, Bunce M, et al. Contribution of genes of
intestinal immune cells from the antigen-rich the major histocompatibility complex to susceptibility and
luminal matrix by inducing genetic expression.81 disease phenotype in inflammatory bowel disease. Lancet
1996;347:1212–7.
8. Farrell RJ, Peppercorn MA. Ulcerative colitis. Lancet
2002;359:331–40.
9. Torres MI, Le Discorde M, Lorite P, et al. Expression of HLA-G
Conclusions in inflammatory bowel disease provides a potential way to
distinguish between ulcerative colitis and Crohn’s disease.
Int Immunol 2004;16:579–83.
Both genetic and environmental factors are in- 10. Farrell RJ, La Mont JT. Microbial factors in inflammatory
volved in the pathogenesis of IBD. bowel disease. Gastroenterol Clin North Am 2002;31(1):
Most of the proposed aetiological theories for IBD 41–62.
include a pathogenic role for the normal intestinal 11. Sartor RB. Pathogenesis and immune mechanisms of chronic
resident bacteria. These theories include: an inflammatory bowel disease. Am J Gastroenterol 1997;
2(suppl):5–11.
inappropriate reaction to a specific infection of 12. Casellas F, Borruel N, Papo M, et al. Anti-inflammatory
the intestine, subtle alterations of bacterial com- effects of enterically coated amoxicilin-clavulanic acid in
position and function, and overexposure to resident ulcerative colitis. Inflamm Bowel Dis 1998;4:1–5.
ARTICLE IN PRESS
Aetiology of IBD 351

13. Teitelbaum JE, Walker WA. Nutritional impact of pre- and 34. Gordon JI, Hoper LV, McNevin MS, Wong M, Bry L. Epigthelial
probiotics as protective gastrointestinal organisms. Annu cell growth and differentiation. III. Promoting diversity in
Rev Nutr 2002;22:107–38. the intestine: conversations between the microbiota,
14. Swidsinski A, Ladhoff A, Pernthaler A, et al. Mucosal flora epithelium, and diffuse GALT. Am J Physiol 1997;273:
in inflammatory bowel disease. Gastroenterology 2002; 565–70.
122:44–54. 35. Guérin-Danan C, Popot AF, Vaissade CP, et al. Pattern of
15. Garside P, McI Mowat A, Khoruts A. Oral tolerance in disease. metabolism and composition of the fecal microbiota in
Gut 1999;44:137–42. infants 10 to 18 months old from day care centers. J Pediatr
16. Taurog JD, Richardson JA, Croft JT, et al. The germfree state Gastroenterol Nutr 1997;25:281–9.
prevents development of gut and joint inflammatory disease 36. Bezirtzoglou E. The intestinal microbiota during the first
in HLA-B27 transgenic rats. J Exp Med 1994;180:2359–64. weeks of life. Anaerobe 1997;3:173–7.
17. Sellon RK, Tonkonogy S, Schultz M, et al. Resident enteric 37. Bernet MF, Brassart D, Neeser JR, Servin AL. Lactobacillus
bacteria are necessary for development of spontaneous acidophilus LA 1 binds to cultured human cell lines and
colitis and immune system activation in interleukin 10- inhibits cell attachment and cell invasion by enterovirulent
deficient mice. Infect Immun 1998;66:5224–31. bacteria. Gut 1994;35:483–9.
18. Madsen KL, Doyle JS, Jewell LD, Tavernini MM, Fedorak RN. 38. Kennedy RJ, Kirk SJ, Gardiner KR. Promotion for a favorable
Lactobacillus species prevents colitis interleukin 10 gene- gut flora in inflammatory bowel disease. JPEN 2000;
deficient mice. Gastroenterology 1999;116:1107–14. 24:189–95.
19. Mombaerts P, Mizoguchi E, Grusby MJ, et al. Spontaneous 39. Borruel N, Carol M, Casellas F, et al. Increased mucosal TNFa
development of inflammatory bowel disease in T cell production in Crohn’s disease can be modulated in vitro by
receptor mutant mice. Cell 1993;75:275–82. probiotic bacteria. Gut 2002;5:659–64.
20. Videla S, Vilaseca J, Guarner F, et al. Role of intestinal 40. Gil A. +Por qué la tolerancia oral? Enfermedad Inflamatoria
microbiota in chronic inflammation and ulceration of the rat Intestinal al dı́a 2004;3:66–70.
colon. Gut 1994;35:1090–7. 41. Spiekermann GM, Walker WA. Oral tolerance and its role in
21. Aranda R, Sydora BC, McAllister PL, et al. Analysis of clinical disease. J Pediatr Gastroenterol Nutr 2001;
intestinal lymphocytes in mouse colitis mediated by transfer 32:237–55.
of CD4+, CD45RBhigh T cells to SCID recipients. J Immunol 42. Brandtzaeg PE. Current understanding of gastrointestinal
1997;158:3464–73. immunoregulation and its relation to food allergy. Ann NY
22. Veltkamp C, Tonkonogy SL, De Jong YP, et al. Continuous Acad Sci 2002;964:13–45.
stimulation by normal luminal bacteria is essential for the 43. Mowat AM. The role of antigen recognition and suppressor
development and perpetuation of colitis in Tg(epsilon26) cells in mice with oral tolerance to ovoalbumin. Immunology
mice. Gastroenterology 2001;120:900–13. 1995;56:253–60.
23. Mackie RI, Sghir A, Gaskins HR. Developmental microbial 44. Miller A, Lider O, Weiner HL. Antigen driven bystander
ecology of neonatal gastrointestinal tract. Am J Clin Nutr suppression after oral administration of antigens. J Exp Med
1999;69(suppl):1035–45. 1991;174:791–8.
24. Kleesen B, Bezirtzoglou E, Matto J. Cultured-based knowl- 45. Sun J, Dirden-Kramer B, Ito K, Ernst PB, Van Houten N.
edge on biodiversity, development and stability of human Antigen specific T cell activation and proliferation during
gastrointestinal microbiota. Microb Ecol Health Dis oral tolerance induction. J Immunol 1999;162:5868–75.
2000;2(suppl):53–63. 46. Chen Y, Inobe J, Marks R, Gonella P, Kuchroo VK, Weiner HL.
25. Salminen S, Bouley C, Boutron-Ruault MC, et al. Functional Peripheral deletion of antigen reactive T cells in oral
food science and gastrointestinal physiology and fuction. Br tolerance. Nature 1995;376:177–80.
J Nutr 1998;80(suppl):147–71. 47. Steidler L. Microbial and immunological strategies for
26. Saavedra JM. Microbes to fight microbes: a not so novel treatment of inflammatory bowel disease. Microb Infect
approach to controlling diarrheal disease. J Pediatr Gastro- 2001;3:1157–66.
enterol Nutr 1995;21:125–9. 48. Steinhart AH, Brzezinski A, Baker JP. Treatment of refractory
27. Bengmark S. Colonic food: pre- and probiotics. Am J ulcerative proctosigmoiditis with butyrate enemas. Am J
Gastroenterol 2000;95(suppl):5–7. Gastroenterol 1994;89:179–83.
28. Hooper LV, Bry L, Falk PG, Gordon JI. Host-microbial 49. Hogaboam CM, Vallance BA, Kumar A, Addison CL, Graham
symbiosis in the mammalian intestine: exploring as internal FL. Gauldie J. Therapeutic effects of interleukin 4 gene
ecosystem. Bioessays 1998;20:336–43. transfer in experimental inflammatory bowel disease. J Clin
29. Guarner F, Melagelada JR. Gut flora in health and disease. Invest 1997;100:2766–76.
Lancet 2003;361:512–9. 50. McDonald TT. Breakdown of tolerance to the intestinal
30. Falk PG, Hooper LV, Midtvedt T, Gordon JI. Creating and bacterial flora in inflammatory bowel disease (IBD). Clin Exp
maintaining the gastrointestinal ecosystem: what we know Immunol 1995;102:445–7.
and need to know from gnotobiology. Microbiol Mol Biol Rev 51. Shanahan F. Therapeutic manipulation of gut flora. Science
1998;62:1157–70. 2000;289:1311–2.
31. Gismondo MR, Drago L, Lombardi A. Review of probiotics 52. Gill HS, Guarner F. Probiotics and human health: a clinical
available to modify gastrointestinal flora. Int J Antimicrob perspective. Postgrad Med J 2004;80:516–26.
Agents 1999;12:287–92. 53. Isolauri E, Kirjavainen PV, Salminen S. Probiotics: a role in
32. Cummings JH, Beatty ER, Kingman SM, Binghman SA, Englyst the treatment of intestinal infection and inflammation? Gut
HN. Digestion and physiological properties of resistant 2002;50(suppl):54–9.
starch in the human large bowel. Br J Nutr 1996;75:733–47. 54. Gil A, Rueda R. Interaction of early diet and the develop-
33. Miyazawa E, Iwabuchi A, Yoshida T. Phytate breakdown and ment of the immune system. Nutr Res Rev 2002;15:263–92.
apparent absorption of phosphorus, calcium and magnesium 55. Sartor RB, Rath HC, Sellon RK. Microbial factors in chronic
in germfree and conventional rats. Nutr Res 1996;16: intestinal inflammation. Curr Opin Gastroenterol 1996;12:
603–13. 327–33.
ARTICLE IN PRESS
352 O.C. Thompson-Chagoyán et al.

56. Ehrhardt RO, Ludviksson BR, Gray B, Naurath M, Strober W. treatment of ulcerative colitis: a randomised trial. Lancet
induction and prevention of colonic inflammation in IL-2 1999;354:635–9.
deficient mice. J Immunol 1997;158:566–73. 69. Burke DA, Axon AT, Clayden SA, Dixon MF, Johnston D, Lacey
57. Bamba T, Matsuda H, Endo M, Fujiyama Y. The pathogenic RW. The efficacy of tobramycin in the treatment of
role of Bacteroides vulgatus in patients with ulcerative ulcerative colitis. Amilent Pharmacol Ther 1990;4:123–9.
colitis. J Gastroenterol 1995;30:45–7. 70. Rolfe RD. The role of probiotic cultures in the control of
58. Shiba T, Aiba Y, Ashikawa H, et al. The suppressive effect of gastrointestinal health. J Nutr 2000;130(suppl):396–402.
Bifidobacteria on Bacteroides vulgatus, a putative pathio- 71. Malin M, Soumalainen H, Saxelin M, Isolauri E. Promotion of
genic microbe in inflammatory bowel disease. Microbiol IgA immune response in patients with Crohn’s disease by oral
Immunol 2003;47:371–8. bacteriotherapy with Lactobacillus GG. Ann Nutr Metab
59. Ventury A, Gionchetti P, Rizzello F, et al. Impact of the 1996;40:137–45.
composition of the faecal flora by a new probiotic prepara- 72. Gupta P, Andrew H, Kirschner BS, Guandalini S. Is Lactoba-
tion: preliminary data on maintenance treatment of cillus GG helpful in children with Crohn’s disease. Results of
patients with ulcerative colitis. Aliment Pharmacol Ther a preliminary, open-label study. J Pediatr Gastroenterol
1999;13:1103–8. Nutr 2000;31:453–7.
60. Kasper H. Protection against gastrointestinal diseases- 73. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces
Present facts and future developments. Int J Food Microbiol boulardii in maintenance treatment of Crohn’s disease. Dig
1998;41:127–31. Dis Sci 2000;45:1462–4.
74. Gionchetti P, Rizzello F, Venturi A, et al. Oral bacteriother-
61. Mao Y, Nobaeck S, Kasravi B, et al. The effects of
apy as maintenance treatment in patients with chronic
Lactobacillus strains and oat fiber on methotrexate-induced
pouchitis: a double blind, placebo-controlled trial. Gastro-
enterocolitis in rats. Gastroenterology 1996;111:334–44.
enterology 2000;119:305–9.
62. O’Mahony L, Feeney M, O’Halloran S, et al. Probiotic impact
75. Venturi A, Gionchetti P, Rizzello F, et al. Impact of fecal flora
on microbial flora, inflammation and tumour development in
by a new probiotic preparation: preliminary data on
IL-10 knockout mice. Aliment Pharmacol Ther 2001;15:
maintenance treatment of patients with ulcerative colitis.
1219–25.
Aliment Pharmacol Ther 1999;13:1103–8.
63. Kruis W, Schültz E, Fric P, Fixa B, Judmaier G, Stolte M.
76. Malchow HA. Crohn’s disease and E. coli. A new approach in
Double blind comparison of an oral E. coli preparation and therapy to maintain remission of colonic Crohn’s disease. J
mesalazine in maintaining remission of ulcerative colitis. Clin Gastroenterol 1997;25:653–8.
Aliment Pharmacol Ther 1997;11:853–8. 77. Thompson-Chagoyán OC, Maldonado J, Gil A. La microbiota
64. Steidler L, Hans W, Schotte L, et al. Treatment of murine intestinal en el niño y la influencia de la dieta sobre su
colitis by Lactobacillus lactis secreting interleukin-10. composición. Alim Nutr Salud 2004;11:37–48.
Science 2000;289:1352–5. 78. Castagliuolo I, Riegler MF, Valenick L, Lamont JT, Pothoulakis
65. Tvede M, Bondesen S, Nielson OH, Rasmussen SN. Serum C. Saccharomyces bourlardii protease inhibits the effects of
antibodies to Bacteroides species in chronic inflammatory Clostridium difficile toxins A and B in human colonic mucosa.
bowel disease. Scand J Gastroenterol 1983;18:783–9. Infect Immun 1999;67:302–7.
66. Hooper LV, Wong MH, Thelin A, Hansson L, Falk PG, Gordon 79. Ichikawa H, Kuroiwa T, Inagaki A, et al. Probiotic bacteria
JI. Molecular analysis of commensal host-microbial relation- stimulate gut epithelial cell proliferation in rat. Dig Dis Sci
ships in the intestine. Science 2001;291:881–4. 1999;44:2119–23.
67. Tedesco FJ, Hardin RD, Harper RN, Edwards BH. Infectious 80. Roediger WE. The colonic epithelium in ulcerative colitis: an
colitis endoscopically simulating inflammatory bowel dis- energy-deficient disease? Lancet 1980;2:712.5.
ease: a prospective evaluation. Gastrointest Endosc 1983; 81. Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA.
29:195–7. Probiotics inhibit enteropathogenic E coli adherence in vitro
68. Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon by inducing intestinal mucin gene expression. Am J Physiol
ATR. Non-pathogenic E. coli versus mesalazine for the 1999;276:G941–G50.

You might also like