Cong 2003

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Current Drug Targets - Inflammation & Allergy, 2003, 2, 145-154 145

Probiotics and Immune Regulation of Inflammatory Bowel Diseases


Yingzi Cong*, Astrid Konrad, Nuzhat Iqbal and Charles O. Elson

Division of Gastroenterology and Hepatology, The University of Alabama at Birmingham, Birmingham,


AL 35294, USA
Abstract: Intestinal microflora play an important role in the pathogenesis of inflammatory bowel diseases
(IBD). Certain probiotic bacteria provide beneficial effects for human health and intervention of IBD. Possible
mechanisms underlying these effects are diverse and include many aspects of the interaction of the host with
its commensal microflora, with immunological and non-immunological effects. This review paper will focus
on the recent progress in the use of probiotics in the treatment of IBD, and discuss the potential
immunological mechanisms underlying their effects, such as the modulation of mucosal T cell, B cell,
epithelial cell, dendrtic cell, macrophage, nature killer cell, antibody, and cytokine responses.
Keywords: Probiotics; Inflammatory Bowel Diseases; Immune Regulation; Microflora.; Mucosa.

1. INTRODUCTION The human body contains 10 times more microbes than


eukaryotic cells. These microbes have evolved in concert
The importance of commensal microflora for human
with their host to occupy specific regions and niches in the
health was first addressed by Elie Metchnikoff at the
gastrointestinal tract. A balanced, complex microflora is
beginning of the 20th century. He attributed beneficial
necessary for normal digestion and to maintain the
effects on human health to the consumption of Lactobacillus
homeostasis of the intestinal ecosystem [9]. Bacteria are
present in yogurt [1]. The concept that currently groups a
present throughout the gastrointestinal tract, but their
number of bacterial strains as “probiotics” was introduced in
distribution pattern and concentration vary greatly. The
1965 by Lilly and Stillwell [2], and is defined as living
microbial biota varies qualitatively and quantitatively along
non-pathogenic microorganisms, which upon ingestion exert
the length of the intestine with an increasing gradient of
a positive influence on the health or physiology of the host
microbes from the stomach to the colon, reaching 1011-12
beyond inherent basic nutrition [3]. Probiotics include
organisms/gram of stool in the colon. The viability of
bacteria, such as Lactobacilli sp and Bifidobacteria sp,
ingested bacteria is reduced dramatically by contact with
some Escherichia coli, Enterococci, and certain yeast,
gastric acid, and the stomach normally contains only very
mainly Saccharomyces. Their fate in the gastrointestinal tract
few bacteria (< 103 c.f.u/mL) that are predominantly Gram-
and their effects differ among strains [4]. To date, therapeutic
positive and aerobic. The human small intestine represents a
and preventive effects of probiotics on infections, intestinal
transitional zone between the sparse population of aerobic
disorders, and food allergy have been described [5]. Some
flora found in the stomach and the profuse bacterial flora of
probiotics demonstrate immunostimulatory properties by
the colon. The microflora of the proximal small bowel is
enhancing innate immune responses [6], and some others
similar to that of the stomach. In the distal ileum, the
may help eradicate pathogens in chronic carriers of
concentration of microorganisms increases, and Gram
S a l m o n e l l a and provide a beneficial effect during
negative bacteria begin to outnumber Gram-positive
Clostridium difficile infection [7, 8]. These studies suggest
organisms. A dramatic increase of bacterial concentration
that probiotics may enhance immune response and positively
occurs across the ileocecal valve; within the colon, the
affect indigenous microflora. Although many health claims
bacterial concentration is between 1011 to 1012 and anaerobes
have been made concerning the potential of probiotics to
outnumber aerobes by a factor of 102-104 [10].
prevent or treat intestinal disorders, only a few probiotic
strains have shown to be effective in randomized placebo- The enteric biota is complex with an estimate of many
controlled clinical trials. In this paper, we review some of hundreds of different cultivable organisms present [11].
recent progress in the use of probiotics in the treatment of However, not all microbes known to be present in the GI
inflammatory bowel diseases, and discuss the potential tract can or have been cultured, and the majority of bacteria
immunological mechanisms underlying their effects. remain uncultured [12]; so this is an underestimation.
Beneficial metabolic activities of microflora include
synthesis of vitamin B and K, production of epithelial
2. INTESTINAL COMMENSAL FLORA nutrients such as short-chain fatty acids, metabolism of
Gastrointestinal tract is colonized by a vast and diverse dietary carcinogens to inactive compounds and the
community of microbes that are essential to its function. conversion of pro-drugs into active drugs. Direct
involvement of the flora in host defense is demonstrated
*Address correspondence to this author at the Division of when disturbed by broad-spectrum antibiotics therapy that is
Gastroenterology and Hepatology, The University of Alabama at occasionally complicated by overgrowth of Clostridium
Birmingham, 633 Ziegler Research Building, 703 South 19th Street, difficile [13]. The indigenous flora also promotes host
Birmingham, AL 35294-0007, USA; Tel: +1-205-934-6061; Fax: +1-205- defense indirectly by influencing the development and
934-8493; E-mail: ycong@uab.edu

1568-010X/03 $41.00+.00 © 2003 Bentham Science Publishers Ltd.


146 Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 Cong et al.

function of mucosal immune response. This is shown in Bifidobacteria in the gut, whereas formula-fed infants have a
adult germ-free animals that have little or no mucosal more complex microflora. After weaning, the composition of
lymphoid tissue or S-IgA, despite exposure to a variety of the microflora resembles that of the adult flora [38].
food antigens. When germ-free animals are colonized by an
One potentially important immunoregulatory function of
intestinal biota, their mucosal lymphoid tissue greatly
the intestinal microflora is their involvement in the
expands along the length of the intestine and S-IgA is
generation of the immunocompetent cells during
produced in quantity [14]. In segmented filamentous bacteria
development and maintenance of homeostasis of the gut-
(SFB)-monoassociated mice, TCRαβ + IELs expand and
associated immune system [22, 39]. Bacterial colonization is
express cytotoxic activity from 2-3 weeks after
required for newborns to make serum antibodies to T-
administration [15]. The capacity to generate IgA-producing
dependent and type 2 T-independent antigens [40]. Neo-natal
cells progressively increases in response to intestinal
immune response is initially characterized by a Th2 cell
antigenic stimulation, particularly with the establishment of
cytokine profile, and later the bacterial stimuli emerging
the gut microflora [16, 17]. The intestinal IgA fully
from the post-natal gut seems to play a major role in driving
develops by the age of 6 weeks in conventionally raised
this Th2-skewed immune response toward a more finely
mice [18]. Upon colonization, some bacteria have been
balanced Th1 and Th2 immune responses. Although no
shown to translocate to the mesenteric lymph node (MLN),
bacterial species has been found to be uniquely involved in
but the number of translocating bacteria begins to decrease
these processes, Lactobacilli are potential candidates because
with the onset of specific IgA response, reflecting maturation
many species of Lactobacillus have been demonstrated to be
of the intestinal immunologic defense mechanisms. Among
immunomodulatory both in vitro and in vivo [41, 42].
the many bacterial strains that comprise the normal enteric
biota, not all are equally able to stimulate the mucosal
immune system [19]. When given to germ-free mice, some 3. MICROFLORA IN PATHOGENESIS OF
bacteria are more effective than others in inducing germinal INFLAMMATORY BOWEL DISEASE
center reactions in Peyer’s patches [20, 21], stimulating IgA
plasma cells in the gut lamina propria [17, 22], and specific Inflammatory bowel diseases (IBD), consisting of
antibodies in the circulation [23-25]. Bacterial colonization Crohn’s disease (CD) and ulcerative colitis (UC), are chronic
can also alter gene expression of the epithelium, such as immune-mediated diseases of the intestinal tract of unknown
production of fucosylated glycoconjugates and α , 2- etiology [43]. Similar to other chronic inflammatory and
fucosyltransferase mRNA in the small-intestinal epithelium autoimmune disorders, initiation of IBD appears to involve
[26-28], which may have secondary effects on the mucosal interactions among genetic, environmental and immune
immune system. The selective antibody response to bacterial factors. Although the pathogenesis of IBD is poorly
species found in colitic mice suggests that variability in understood, the characteristic histopathology of the lesion as
immuno-stimulatory capability of gut bacteria extends to well as an abundance of immunologic abnormalities
pathologic states [29]. Consistent with these observations, indicates that it involves a state of local immune
reconstitution of germ-free HLA-B27/ß2m transgenic rats hyperreactivity. Much of this immune reactivity appears to
and IL-10 deficient mice with certain bacterial species, but be directed at the enteric bacterial biota. The commensal
not others, result in the induction of intestinal inflammation flora conditions the level of activation of mucosal immune
[30, 31]. response and appears to be a key factor in driving mucosal
inflammation in genetically susceptible individuals.
During neo-natal colonization of mice with the enteric Increasing number of both clinical and laboratory-derived
bacterial biota, there are large swings in bacterial strains that observations implicate the normal flora in the pathogenesis
occur in a sequential manner [32-35]. This occurs in mice of IBD. It has been recognized for a long time that the
from about the 10th to the 18th day after birth. This period is anatomic sites of the highest bacterial concentration (distal
a time of profound rearrangement of bacterial populations in ileum and colon) are the sites most frequently affected by
mice, which eventually reaches the “adult” pattern by 3 to 4 inflammation in patients with IBD [44]. There is growing
weeks and then is thought to remain stable throughout adult evidence that the fecal flora, including the mucosal-
life. This period of colonization with bacterial biota associated flora, differs in patients with active IBD compared
corresponds to a time of immaturity of the systemic immune with quiescent disease or healthy volunteers [45]. Several
system and to a rapid development of the mucosal immune studies demonstrate decreased luminal concentrations of
system, and thus this encounter of the host with the rich lactobacilli and bifidobacteria concentrations in patients with
assortment of bacterial antigens could have profound active Crohn's disease or ulcerative colitis (UC) [46-48].
consequences later in life. Neo-natal antigen exposure has High concentrations of bacteria adherent to the mucosa have
been shown to alter immune responses to the same antigen been found in patients with IBD, but not in control subjects,
when re-encountered again as an adult [36]. The intestinal and the concentrations of adherent mucosal bacteria increase
flora from 1-23 days old mice is inefficient, whereas that progressively with the severity of disease [49]. There is
from 25 days old mice can induce intestinal IgA persuasive evidence for increased immune reactivity to
plasmocytes [18]. In humans, microbial colonization begins components of the commensal flora in experimental models
after birth. The maternal intestinal flora is the source of and IBD patients [50, 51, 56]. The most compelling
bacteria colonizing the newborn's intestine. Colonization is evidence for the role of normal flora in pathogenesis of IBD
also determined by contact with surroundings. Initially, comes from the experimental animal models which have
facultative anaerobic strains dominate. Later, the lactic acid significant advantages in studying interactions among
bacteria and coliforms become the predominant part of the environmental, immune, and genetic components that lead
microflora [37]. Breast-feeding encourages the growth of to chronic intestinal inflammation. While a diversity of
Probiotics and Immune Regulation of Inflammatory Bowel Diseases Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 147

sporadic and engineered genetic defects have been described plantarum decreased weight loss, intestinal permeability and
which a predisposed to chronic inflammatory bowel diseases myeloperoxidase level, re-established intestinal
in mice, with varying immunologic mechanisms mediating microecology, and reduced bacterial translocation to
inflammation, colonization with normal enteric flora is extraintestinal sites [62], with L. plantarum more effective
required for full expression of the disease. In a number of than L. reuteri. Feeding of Lactobacillus salivarius to IL-10
models, such as IL-2 deficient mice, IL-10 deficient mice and knockout mice consistently decreased mucosal inflammation
HLA-B27/ß2m transgenic rats, no disease occurs when the and significantly reduced fecal coliform and enterococci
animals are raised under germ-free conditions [53-55], or the levels [64]. At 2 weeks of age, IL-10 deficient mice showed
disease is dramatically ameliorated when the animals are no colonic injury but displayed abnormal colonic bacterial
raised under specific pathogen-free conditions [54]. Most colonization with reduced Lactobacillus sp. levels and
importantly, colitis has been induced in SCID mice by increased number of bacteria adherent to mucosa and of
adoptive transfer of CD4+ T cells reactive to enteric bacterial translocated bacteria. In association with this abnormal
antigens, providing direct evidence that bacterial-reactive T colonic bacterial flora, colitis developed by 4 weeks of age.
cells can mediate inflammatory bowel disease [56]. Daily intrarectal inoculation of Lactobacillus sp or oral
lactulose therapy which stimulates Lactobacilli resulted in
4. THERAPEUTIC APPLICATION OF PROBIOTICS Lactobacillus sp repopulation, reduced numbers of bacteria
IN IBD adherent to mucosa and of translocated bacteria and
prevented the onset of colitis [63]. Administration of
Conventional and experimental therapy of IBD mainly combinations of Lactobacillus salivarius subsp. salivarius
targets the inflammatory process by suppressing the host UCC118 and Bifidobacterium longum infantis 35624 to IL-
response. Because of the key role of the intestinal microflora 10 deficient mice also ameliorated colitis [52]. More recent
in the development of IBD, manipulating this component studies demonstrated prevention and treatment of the colitis
provides a very intriguing novel therapeutic approach. Not in IL-10 deficient mice by continuous feeding of L .
all members of the gastrointestinal flora are considered plantarum. This attenuated established colitis as manifested
proinflammatory. HLA-B27/ß2m transgenic rats mono- by decreased mucosal IL-12, IFNγ, and IgG2a levels.
associated with Bacteroides vulgatus developed colitis, Gnotobiotic IL-10 deficient mice mono-associated with L.
whereas those mono-associated with E. coli remained plantarum exhibited mild immune system activation but no
healthy [30]. Administration of probiotic bacteria to IL-10 colitis. Germ-free mice did not develop colitis. Colonization
deficient mice ameliorated their colitis [57]. Multiple reports of IL-10 deficient germ free mice with L. plantarum, and
have documented that strains of lactobacilli and subsequent exposure to a complex SPF flora and continued
bifidobacteria, which may contain strains with probiotic probiotic therapy significantly decreased the severity of
activities, are decreased in patients with active Crohn's inflammation [57].
disease or ulcerative colitis [46, 47, 58]. However, many
reports include vague or imprecise data. Only recently have
the effects of probiotics on human and animal health been 4.3 Clinical Application of Probiotics to IBD
evaluated using elegant scientific tools. Acceptance of benefit to human health from putative
probiotic bacteria requires demonstration of effect in formal
4.1 Selection of Probiotics clinical studies. Some recent clinical trials have provided
evidence of positive effects. Treatment with non-pathogenic
Probiotic strains are often selected empirically from E. coli Nissle 1917 has almost an equivalent effect of
bacterial pools, isolated from normal humans or from mesalazine in maintaining remission of ulcerative colitis
traditional fermented foods. A number of criteria have been [65, 66]. Lactobacillus GG is a safe probiotic bacterium
proposed to allow more efficient and rational selection. known to transiently colonize the human intestine. In an
Attractive properties of potentially effective probiotic open-label pilot trial of children with stable Crohn's disease,
bacteria include human origin, non-pathogenic behavior, administration of Lactobacillus GG in enteric coated tablets
resistance to technologic processing and bile toxicity, significantly improved gut barrier function and clinical
tolerance to gastric acid, adhesion to intestinal epithelial activity [67]. Saccharomyces boulardii, non-pathogenic
surfaces, ability to persist within the gastrointestinal tract, yeast with beneficial effects on the human intestine, may
production of anti-microbial substances, ability to modulate also be beneficial in the maintenance treatment of Crohn's
immune responses, and antagonism of potentially disease when combined with mesalamine [68]. Recently,
pathogenic micro-organisms [59-61]. more promising effects of probiotics on IBD have been
observed with a new combined probiotic preparation, VSL #
4.2 Therapeutic Application of Probiotics in Animal 3, which contains 5x10 1 1 cells/g of 3 strains of
Model of IBD Bifidobacteria (B. longum, B breve, and B. infantis), 4
Recent studies have suggested a potential therapeutic role strains of L a c t o b a c i l l i (L. casei, L. plantarum, L.
of probiotics in experimental inflammatory bowel disease, in acidophilus, and L. delbruekii subsp. bulgaricus), and 1
that different strains of lactobacilli have been shown to strain of Streptococcus salivarius subsp. thermssophilus. In
prevent the development or attenuate active inflammation in a small pilot trial, VSL#3 was effective in maintaining
methotrexate [62] and in acid acetic-induced colitis [58], and remission in mesalazine intolerant patients with ulcerative
to prevent colitis in IL-10 deficient mice [57, 63, 64]. colitis [69]. In a double-blind, placebo-controlled trial, oral
administration of VSL#3 was effective in the treatment of
In methotrexate-induced enterocolitis in rats, chronic pouchitis, an inflammatory complication that can
administration of Lactobacillus reuteri and Lactobacillus
148 Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 Cong et al.

arise after ileal pouch-anal anastomosis surgery for the colonic lymphoid cells was mainly on the numbers of IgA-
treatment of ulcerative colitis [70, 71]. secreting B cells and T cells, with a marked increase in T
cells. Interestingly, although the number of macrophages
was increased, there was no enhancement in their activity
4.4 Possible Mechanisms
that might have harmed the host by producing an
The possible mechanisms underlying the beneficial inflammatory response. Different probiotic strains have
effects of probiotics in IBD are rather diverse and include different effects on intestinal IgA levels [83, 84, 92].
numerous aspects of the interactions of the host with its
4.4.2 Effects of Probiotics on Circulating Antibody
commensal microflora. They can be divided into
Responses
immunological and non-immunological effects. For non-
immunological effects, most lactobacilli species are able to Kaila et al. reported that when patients with diarrhea
adhere to colonic epithelial cells, thereby interfering with the were given Lactobacillus GG, the duration of diarrhea was
adherence of other potentially pathogenic bacteria [72] and significantly shorter than in the placebo group. This effect
possibly competing for nutrients while they temporarily was associated with a significantly enhanced nonspecific
colonize the intestine. Recent in vitro data suggest that humoral response during the acute phase of the infection, in
inhibition of the adherence of attaching and effacing E. coli that total peripheral blood IgG-, IgA-, and IgM-secreting
to intestinal epithelial cells by lactobacilli is mediated cells, as well as IgA anti-rotavirus, were greatly increased
through increased expression of intestinal mucin genes [73]. [89]. Oral administration of Wistar and Brown Norway rats
Some probiotic bacterial species are able to restore damaged with viable Lactobacillus casei Shirota strain YIT9029
intestinal permeability [63, 74, 75], which is deranged in significantly enhanced the T. spiralis-specific delayed-type
IBD and experimental colitis. An additional mechanism of hypersensitivity (DTH) response together with increased
protection is the direct inhibition of the growth of potential serum T. spiralis-specific IgG2b antibody production.
pathogenic bacterial species. For example, Lactobacillus GG Importantly, no significant effects of L. casei on larval
produces a bacteriocin [76], and both lactobacilli and counts or inflammatory reactions in the tongue musculature,
bifidobacteria lower the luminal pH, which can be inhibitory body weights, or lymphoid organ weights were observed
for certain microbes [38, 69, 77]. More detailed information [93]. In another study, eight different common Lactobacilli
regarding non-immunological effects of probiotics is strains were assessed for their ability to induce pro- and anti-
described in an excellent review by Steidler [78]. inflammatory cytokines, IgA-producing plasma cells in the
gut, as well as systemic antibody responses against a
Because IBD involves abnormal immune responses to
parenterally administered antigen. Oral administration of L.
enteric flora, the modulation of the mucosal immune system
r e u t e r i and L. brevis induced expression of the
by probiotics may play a key role in their therapeutic
proinflammatory Th1 cytokines IFNγ, IL-2 and IL-1. Oral
mechanisms. Probiotic organisms can directly or indirectly
administration of these two strains and L. fermentum also
influence mucosal and systemic immune function by altering
significantly enhanced the IgG response against parenterally
cytokine gene expression, antibody production, and by
administered haptenated chicken gamma globulin. L. reuteri
modulating cellular immune responses [71, 79-81]. Some
induced relatively high levels of IgG2a against haptenated
reports have demonstrated that probiotic bacteria promote
chicken gamma globulin compared to L. murine. The five
intestinal immunologic barrier by enhancing humoral
other strains did not show any mucosal adjuvanticity [85].
immune responses [82-84], stimulate nonspecific host
resistance to microbial pathogens [85-87], and modulate the 4.4.3 Effects of Probiotics on T Cells
host immune responses to potentially harmful antigens with T cells have been shown to play a key role in immune
a potential to down-regulate hypersensitivity reactions [88]. responses against foreign pathogens and in control of
4.4.1 Effects of Probiotics on Intestinal IgA immune homeostasis. Oral administration of mice with
probiotic-supplemented yogurts containing Streptococcus
The increase in local intestinal IgA levels resulting from
thermophilus, Lactobacillus bulgaricus, Bifidobacterium,
ingestion of probiotics may contribute to enhancement of the
and Lactobacillus acidophilus significantly increased
mucosal resistance against gastrointestinal infections.
percentage of CD4+ T cells in spleen [94]. Oral ingestion of
Several reports demonstrate that probiotic strains enhance lactobacilli greatly increased T cell proliferative responses to
intestinal IgA production. An increase in systemic and bacteria in Peyer's patches, peripheral blood and spleen, but
mucosal IgA response to dietary antigens was shown after did not affect macrophage and T cell proportions and
oral administration of lactobacilli [74, 89]. When formula lymphocyte subset composition in these three sites [95],
containing viable bifidobacteria was given to children, the suggesting that live lactobacilli may interact with the
administered strain was detected in feces together with intestinal immune system and influence the systemic
higher fecal levels of total IgA and anti-poliovirus IgA [90, immune system. Different probiotic strains vary in their
91]. Oral administration of Lactobacillus casei strain GG to ability to modulate T cell and B cell responses. Some
patients with Chrohn's disease greatly increased the promote Th1 type responses, and some others enhance both
intestinal IgA response and thus promoted the gut- Th1 and Th2 type responses. In vitro stimulation with heat-
immunological barrier [82]. In a mouse model, killed Lactobacillus casei strongly enhanced murine
administration of a diet with yogurt containing various splenocyte IL-12 and IFN-γ production, but inhibited IL-4
lactobacilli, the number of IgA secreting cells in small and IL-5 secretion, and markedly suppressed total and
intestine increased, while the values for cells secreting IgM antigen-specific IgE secretion by antigen (OVA)-stimulated
and IgG and for T lymphocytes remained similar to those of splenocytes, indicating that in this situation L. casei
the controls. The effect of giving yogurt with probiotics on preferentially induce Th1 development [96]. Lactobacillus
Probiotics and Immune Regulation of Inflammatory Bowel Diseases Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 149

acidophilus treatment enhanced T cell proliferation to Con A secrete IL-10 [115]. Recently, it was shown that treatment of
and B-cell response to LPS, whereas Lactobacillus casei, human peripheral blood mononuclear cells (PBMC) with
Lactobacillus gasseri, and Lactobacillus rhamnosus Lactobacillus johnsonii selectively activated CD3-CD56 +
inhibited T cell and B cell proliferation, indicating that NK cells, detected by enhanced expression of the activating
Lactobacillus strains can have strain-specific effects on B- antigens-CD69 and CD25. Purified NK cells upregulated
and T-cell responses [97, 98]. CD25 and proliferated when stimulated with Lactobacillus
johnsonii and Lactobacillus sakei in vitro [81]. Stimulation
4.4.4 Induction of Regulatory T Cells
of PBMC with Staphylococcus aureus and Lactobacillius
The inhibitory activity of regulatory T cells is believed johnsonii resulted in the activation of CD3- CD16+ CD56+
to be central to the prevention of autoimmune diseases, human peripheral blood NK cells to express activation
allergies, transplant rejection, immune-deficiency disorders antigen CD69 and the secretion of IL-12 and IFNγ. These
and inflammation. Multiple types of regulatory T cells have effects required cell contact-dependent costimulation by
been identified, including CD4+ CD25 + T cells, Tr1 cells, autologous monocytes [116], suggesting that phagocytosis
and Th3 cells [99-105]. Recently, bacterial specific-T of such probiotic strains provided additional co-activation
regulatory cells have been identified that inhibit pathogenic signals on accessory cells that may differ from those induced
responses to enteric flora and appear to play an important by cytokines. This was not observed when PBMC were
role in the maintenance of normal intestinal immune stimulated with non-pathogenic E. coli or lipopolysaccharide
homeostasis [106]. Possibly related to these observations are (LPS) [81].
the data regarding effects of certain strains of lactic acid
4.4.6 Effects of Probiotics on Epithelial Cells
bacteria, including L. johnsonii, L. gasseri, L. paracasei,
L. acidophilus, L. casei strain Shirota, and L. casei strain Epithelial cells that line the intestinal tract participate in
GG, to modulate specific T cell responses. All strains tested the initiation and regulation of the mucosal immune
in this study induced various levels of both IL-12 and IL-10 response to bacteria by interacting with immune cells of the
production in murine splenic cells in vitro. Among these gut-associated lymphoid tissue, such as lamina propria
strains, L. paracasei induced the highest levels of IL-12 and lymphocytes (LPL) and intra-epithelial lymphocytes (IEL)
IL-10. Interestingly, when L. paracasei bacteria were added [117-121]. Intestinal epithelial cells (IEC) may change
to mixed lymphocyte cultures in which BALB/c CD4+ T phenotype as a consequence of stimulation by IEL derived
cells were stimulated weekly in the presence of irradiated soluble mediators, such as IFNγ and regulated production of
allogeneic splenocytes, the L. paracasei strongly inhibited IFN-inducible T-cell chemoattractants [120, 122, 123]. In
the proliferative response of CD4+ T cells in a dose- the case of the colonized intestinal mucosa, however,
dependent fashion. This was accompanied by a marked protection against microbial pathogens is mediated in part
decrease of both Th1 and Th2 cytokines, including IFNγ, by the microflora [124, 125]. IEC may be implicated in the
IL-4, and IL-5, but maintenance of IL-10 and induction of recognition of components of the intestinal microflora and
TGF-β in a dose-dependent manner [107]. It seems possible the transduction of bacterial-derived signals to resident
that this probiotic is stimulating regulatory T cells similar mucosal immune cells [126-129]. Changes in IEC, LPL,
to the Tr1 cells that are involved in the maintenance of and IEL have been described in response to modifications of
intestinal immune homeostasis [106, 108]. the intestinal microflora [6, 130-132]. It has been shown that
4.4.5 Effects of Probiotics on Natural Killer Cells treatment of IL-10 deficient mice with VSL #3 resulted in an
enhancement of epithelial barrier function and a reduction of
Natural killer (NK) cells have been implicated in immune TNFα production. Direct application of VSL #3 on to the
surveillance in innate immune responses [109]. Activated apical surface of gut epithelial cell T84 monolayers greatly
NK cells circulate through blood and lymph and accumulate increased their barrier function and resistance to Salmonella
at sites of injury, infections, and tumors [110], and quickly invasion [133]. L. casei or Clostridium butyricum greatly
respond to immune activation signals, in particular for enhanced gut epithelial cell proliferation in rats and thus
inducing the early production of IFNγ that is necessary to possibly promoted tissue repair [134]. In a CaCo-2 epithelial
control microbial pathogen infections [111, 112]. The rapid cell-leucocyte co-culture system, probiotic bacteria elicited a
activation of NK cells is characteristic of innate immunity differential cytokine response by human IEC in the presence
and constitutes a first line of defense against invading of human blood leucocytes. Leucocyte-sensitized CaCo-2
pathogens. Signals generated during the early host response cells in vitro distinguished between signals delivered by
have an additional role in the shaping of subsequent adaptive different probiotic bacteria and in turn transduced a
immune response. Production of IFN-γ ensures activation of discriminative signal to underlying PBMC. L. sakei
phagocytosis by monocytes and T cell differentiation to the stimulated IEC TNFα and IL-1 production, similar to non-
Th1 phenotype, thus directing the adaptive immune response pathogenic E. coli stimulation. In these co-cultures,
towards an appropriate effector pathway to clear the differences in the regulation of inflammatory immune
infection. Administration of a dietary supplement with response by IEC were observed between enteropathogenic E.
probiotic lactic acid bacteria promoted activity of circulating coli and probiotic non-pathogenic bacteria, suggesting that
NK cells in the elderly [113]. Several studies have shown IEC discriminated between enteropathogens and commensal
that commensal lactobacilli isolated from the human bacteria. L. johnsonii had a low potential to induce
gastrointestinal tract activate human mononuclear cells and proinflammatory response but rather favored induction of
are potent inducers of IFN-γ and monocyte-derived IL-12 TGFβ expression in CaCo-2 cells [80]. TGFβ is a key factor
[79, 114]. The combination of IL-12 and IL-18 is a potent implicated in the regulation of intestinal barrier function
inducer of IFN-γ in human CD56bright NK cells, whereas the [135-137], and is thought to mediate tolerance to the
combination of IL-12 and IL-15 stimulates NK cells to indigenous microflora via bystander suppression [138-142],
150 Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 Cong et al.

as well as mediate the inhibitory function of T regulatory remained unaltered. In analogous fashion, L. reuteri reduced
cells [143]. These results support the hypothesis that L. casei-induced up-regulation of CD86. These results
bacterial signaling at the mucosal surface requires a network suggest that different strains of Lactobacilli exert very
of cellular interactions. According to these results, different DC activation patterns, and some strains may be
sensitization of CaCo-2 cells by neighboring capable of inhibiting activities of other strains in the species.
immunocompetent cells constitutes a crucial step for the IL-12 and IL-10 produced by APCs exert largely opposite
recognition of non-pathogenic bacteria because CaCo-2 cells immune effects. IL-12 is critical for the induction of Th1
alone remained hyporesponsive to probiotic bacterial cells and elicits IFNγ production by T cells and by NK cells
stimuli. Interestingly, L sakei which induced biosynthesis of [167]. IL-10 is an anti-inflammatory cytokine that
TNF-α and IL-1 in CaCo-2-leucocyte co-cultures also suppresses IL-12 production, and acts on macrophages to
strongly stimulated IL-10 secretion from underlying PBMC, prevent their activation and elaboration of proinflammatory
indicating that a negative feedback loop might be activated molecules and chemokines, thus inhibiting T cell
[80]. IL-10 is crucial in the control of gut homeostasis and recruitment into the intestine [148, 149, 168]. Thus, by
immunity [144-152], and downregulates chemokine response inhibiting DC IL-12 production and promoting IL-10
in activated IEC [153]. Interestingly, Lactobacilli and production some probiotics may enhance regulatory T cell
Bifidobacteria strains from VSL#3 and Lactobacillus GG induction through modulation of DC maturation [160-163].
did not induce IL-8 of epithelial cell HT29/19A monolayers,
4.4.8 Effects of Probiotics on Macrophages and
whereas both cell debris and cell extracts from E. coli Nissle
Monocytes
1917 induced IL-8 production in a dose-dependent way,
suggesting that probiotic VSL#3 and Lactobacillus GG and Macrophages and monocytes have a central role in
E. coli Nissle 1917 may exert their beneficial effects on the initiating the innate immune response, which leads to
host by a different mechanism [154]. It is noteworthy that activation of the adaptive response. Macrophages
certain bacteria can actively interfere with the phagocytose infected cells present antigens to T and B cells,
proinflammatory I-κB/NF-κB signaling pathway in IEC by and produce cytokines and chemokines that modulate
blockade of I-κBα degradation [155]. In addition to the immune responses [169, 170]. These cytokines include
effects on IEC cytokine production, L a c t o b a c i l l u s TNFα and IL-6, which are among the first produced during
rhamnosus promoted survival of IEC by inhibition of the innate immune response toward bacteria. Some probiotic
apoptosis in TNFα stimulated mouse IEC line YAMC and bacteria have the potential to influence macrophages and
human epithelial cell line HT29. Live Lactobacillus monocytes in a very specific way. Strain-dependent
rhamnosus GG or its supernatant stimulated anti-apoptotic induction of cytokines has been described after oral
Akt activation, but did not alter ERK, NF-κ B or Lactobacillus administration and may provide a way to
SAPK/JNK activities [156]. modulate local immune responses in any desired direction
[85, 88, 114, 171]. In vitro stimulation with heat-killed
4.4.7 Effects of Probiotics on Dendritic Cells
Lactobacillus casei strongly enhanced IL-12 by antigen
Dendritic cells (DC) are highly specialized APC of the OVA-stimulated splenocytes, and such IL-12 augmentation
immune system [157]. Depending on their maturational was macrophage-dependent [96]. Lactic acid bacteria
state, they possess different functional properties. Fully stimulate human peripheral blood mononuclear cell TNFα,
mature DCs are potent activators of naïve T cells and are IL-6, and IL-10 production [79]. Lactobacillus rhamnosus
regarded as important initiators of primary T cell responses. GG induce NF-κB and STAT DNA-binding activity in
Immature DCs or “tolerant DC” are poor stimulator of T cell human primary macrophages. NF-κB activation was rapid
responses and have been shown to induce anergic or and was not inhibited by a protein synthesis inhibitor
regulatory T cells [158-163]. DCs are present throughout the cycloheximide, suggesting that these bacteria could directly
gastrointestinal tract [164, 165], and are in close proximity activate NF-κB [172]. Feeding of Lactobacillus rhamnosus,
to the intestinal microbiota. Thus, DC may be the targets for Lactobacillus acidophilus and Bifidobacterium lactis
immune modulation by probiotics. In that regard, a recent significantly enhanced natural and acquired immunity in
study has shown that species of Lactobacilli differentially healthy mice, in that the phagocytic activity of peripheral
activated DC by modulating expression of cytokines and blood leucocytes and peritoneal macrophages was greatly
surface costimulatory molecules [166]. Mouse DC IL-12 and increased compared to the control mice [173]. An interesting
TNFα production was substantially different when treated recent report showed that increased intestinal mucosal TNFα
with various strains of Lactobacillus. Similar but less production in Crohn’s disease was downregulated ex vivo by
pronounced differences were observed among lactobacilli in the probiotic bacteria L. casei or L. bulgaricus. However,
the induction of IL-6 and IL-10. The cytokine induction was these bacteria did not induce changes in non-inflamed
bacterial dose-dependent. Low concentrations of L. casei mucosa [174]. Treatment of patients with pouchitis with
yielded maximal levels of IL-6, IL-12, and TNFα but VSL#3 increased tissue levels of IL-10 and decreased TNFα
virtually no IL-10 induction. At higher concentrations of L. and IL-1α, inducible nitric oxide synthase, and matrix
casei, IL-10 production increased radically with no change in metalloproteinase [71]. These effects may favor the
the level of other cytokines. This observation indicates that generation of regulatory T cells and down-regulation of
the threshold of bacterial concentration necessary to induce inflammation.
cytokine production varies among cytokines. All strains up-
As described above, different probiotic strains exert very
regulated surface MHC class II and CD86, which is
different effects on DC and macrophage cytokine induction.
indicative of DC maturation. Interestingly, Lactobacillus
DC and macrophage recognition and response to molecular
reuteri DSM12246 inhibited IL-12, IL-6, and TNFα
structures on bacteria (pathogen-associated molecular
induction by L. casei CHCC3139, whereas IL-10 production
Probiotics and Immune Regulation of Inflammatory Bowel Diseases Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 151

patterns) occur through Toll-like receptors (TLRs). and claims made for one probiotic strain cannot readily be
Activation of these surface receptors by bacterial components applied to another even within the same species. Thus,
is a key factor for regulation of the immune response and a probiotic bacterial strains must be evaluated individually.
link between innate and adaptive immune responses [175]. Because many of the probiotic effects are mediated via
Differential activation of TLR by multi-type pathogen- immune regulation, in particular by control of the balance of
associated molecular patterns could induce different cytokine proinflammatory and anti-inflammatory cytokines, the future
patterns. This might explain the observed differences in studies should focus on defining the specific
cytokine induction found by various probiotics. TLR2 has immunomodulatory properties of probiotic bacteria.
been found to be a receptor for peptidoglycan, lipoteichoic
acid, and bacterial lipoprotein in addition to LPS [176,
177]. LPS also binds to TLR4 and CD14, thus these REFERENCES
receptors constitute alternative activation pathways for LPS [1] Metchnikoff, E. The Prolongation of Life. Optimistic
[178-180]. Recent evidence suggests that there are structural studies. In London: William Heinemann, 1907,
and functional differences in the LPS produced by different [2] Lilly, D.M.; Stillwell, R.H. Science, 1965, 47, 747.
bacteria [181, 182]. It has been proposed that the shape of [3] Fuller, R. J. Appl. Bacteriol., 1989, 66, 365.
the lipid A component determines the bioactivity of LPS [4] Marteau, P.; Pochart, P.; Bouhnik, Y.; Rambaud, J.C.
[183], with lipid A that adopts a conical conformation being World. Rev. Nutr. Diet., 1993, 74, 1.
[5] Isolauri, E.; Sutas, Y.; Kankaanpaa, P.; Arvilommi, H.;
more active than lipid A that adopts a cylindrical shape. Salminen, S. Am. J. Clin. Nutr., 2001, 73, 444S.
Some of the cylindrical LPS molecules may be antagonists. [6] Link, H.; Rochat, F.; Saudan, K.Y.; Schiffrin, E. Adv. Exp.
Variation in the molecular composition and the three- Med. Biol., 1995, 371A, 465.
dimensional conformation of the lipid A component of [7] Alm, L. Prog. Food Nutr. Sci., 1983, 7, 13.
different types of LPS may explain the activation of different [8] Biller, J.A.; Katz, A.J.; Flores, A.F.; Buie, T.M.; Gorbach,
TLR signaling pathways. Conical-shaped LPS stimulates S.L. J. Pediatr. Gastroenterol. Nutr., 1995, 21, 224.
cells through TLR4, whereas cylindrical-shaped LPS [9] Simon, G.L. and Gorbach, S.L. Dig. Dis. Sci., 1986, 31,
activates via TLR2 [184]. Therefore, it could be speculated 147S.
that the inhibitory effect of some probiotics is mediated [10] Tancrede, C. Eur. J. Clin. Microbiol. Infect. Dis., 1992,
through interference with TLRs, perhaps via alterations in 11, 1012.
[11] Macfarlane, G.T.; Macfarlane, S. Scand. J. Gastroenterol.
the shape of LPS, or other TLR ligands. The involvement of (Suppl.), 1997, 222, 3.
TLR in the differential activation capacities of species of [12] Zoetendal, E.G.; Akkermans, A.D.L.; de Vos, W.M. Appl.
probiotic bacteria on DC and macrophages provides an Environ. Microbiol., 1998, 64, 3854.
intriguing target for future mechanistic studies. [13] Shanahan, F. Br. J. Nutr., 2002, 88 Suppl 1, S5.
[14] Crabbe, P.A.; Bazin, H.; Eyssen, H.; Heremans, J.F. Int.
Arch. Allergy, 1968, 34, 362.
5. SUMMARY [15] Umesaki, Y.; Okada, Y.; Matsumoto, S.; Imaoka, A.;
The immune regulation of IBD by probiotics has been Setoyama, H. Microbiol. Immunol., 1995, 39, 555.
[16] Shroff, K.E.; Meslin, K.; Cebra, J.J. Infect. Immun., 1995,
shown to be promising and paradoxical. Proinflammatory 63, 3904.
cytokines, IL-1, TNFα, and IFNγ, play a pivotal role in [17] Moreau, M.C.; Ducluzeau, R.; Guy-Grand, D.; Muller,
inflammation. Cytokine transgenic or knockout mice have M.C. Infect. Immun., 1978, 21, 532.
been shown that a harmless immune response to commensal [18] Moreau, M.C.; Raibaud, P.; Muller, M.C. Ann. Immunol.
intestinal microflora becomes a harmful inflammatory state (Paris), 1982, 133D, 29.
in the absence of certain cytokines [30, 53-55]. These [19] Okada, Y.; Setoyama, H.; Matsumoto, S.; Imaoka, A.;
observations indicate that inflammation is induced by an Nanno, M.; Kawaguchi, M.; Umesaki, Y. Infect. Immun.,
unbalanced local or systemic cytokine milieu. As described 1994, 62, 5442.
above, treatment with some probiotic bacteria can modulate [20] Pollard, M.; Sharon, N. Infect. Immun., 1970, 2, 96.
TNFα and IFNγ production. Thus, ingestion of probiotic [21] Carter, P.B.; Pollard, M. J. Reticuloendothel. Soc., 1971,
9, 580.
bacteria may stabilize the immunologic barrier of the [22] Umesaki, Y.; Setoyama, H. Microbes Infect., 2000, 2,
intestinal mucosa by reducing the generation of local 1343.
proinflammatory cytokines. Specific probiotic strains can [23] Berg, R.D.; Savage, D.C. Am. J. Clin. Nutr., 1972, 25,
normalize aberrant antigen-induced production of the 1364.
proinflammatory cytokines [185]. These data suggest that [24] Berg, R.D. In Human intestinal microflora in health and
the immunomodulating effect of probiotic bacteria may disease, D.C. Savage, Ed.; Academic Press: New York,
depend on the immunologic state of the host, and there may 1983; pp. 101-126.
be differences between specific probiotic strains. [25] Foo, L. Infect. Immun., 1972, 6, 525.
[26] Bry, L.; Falk, P.G.; Midtvedt, T.; Gordon, J.I. Science,
1996, 273, 1380.
6. FUTURE TARGETS [27] Hooper, L.V.; Bry, L.; Falk, P.G.; Gordon, J.I. Bioessays,
1998, 20, 336.
Probiotics allow modulation of the intestinal immune [28] Gordon, J.I.; Hooper, L.V.; McNevin, M.S.; Wong, M.;
system and the commensal flora. However, the promise of Bry, L. Am. J. Physiol., 1997, 273, G565.
probiotics has often been overstated. Rigorously designed, [29] Brandwein, S.L.; McCabe, R.P.; Cong, Y.; Waites, K.B.;
controlled clinical trials of efficacy in well-categorized Ridwan, B.U.; Dean, P.A.; Ohkusa, T.; Birkenmeier, E.H.;
patients are required for future progress. It is now clear that Sundberg, J.P.; Elson, C.O. J. Immunol., 1997, 159, 44.
different probiotic bacteria have different effects on the host
152 Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 Cong et al.

[30] Rath, H.C.; Herfarth, H.H.; Ikeda, J.S.; Grenther, W.B.; [58] Fabia, R.; Ar'Rajab, A.; Johansson, M.L.; Andersson, R.;
Hamm, T.E., Jr.; Balish, E.; Taurog, J.D.; Hammer, R.E.; Willen, R.; Jeppsson, B.; Molin, G.; Bengmark, S. Digest,
Wilson, K.H.; Sartor, R.B. J. Clin. Invest., 1996, 98, 945. 1993, 54, 248.
[31] Sellon, R.K.; Tonkonogy, S.; Schultz, M.; Dieleman, L.A.; [59] Marteau, P.; Rambaud, J.C. FEMS Microbiol. Rev., 1993,
Grenther, W.; Balish, E.; Rennick, D.M.; Sartor, R.B. 12, 207.
Infect. Immun., 1998, 66, 5224. [60] Guarner, F.; Schaafsma, G.J. Int. J. Food Microbiol.,
[32] Schaedler, R.W.; Dubos, R.; Costello, R. J. Exp. Med., 1998, 39, 237.
1974, 122, 59. [61] Dunne, C.; O'Mahony, L.; Murphy, L.; Thornton, G.;
[33] Savage, D.C.; Dubos, R. J. Exp. Med., 1968, 128, 97. Morrissey, D.; O'Halloran, S.; Feeney, M.; Flynn, S.;
[34] Dubos, R. Gastroenterol., 1966, 51, 868. Fitzgerald, G.; Daly, C.; Kiely, B.; O'Sullivan, G.C.;
[35] Mushin, R.; Dubos, R. J. Exp. Med., 1965, 122, 745. Shanahan, F.; Collins, J.K. Am. J. Clin. Nutr., 2001, 73,
[36] Singh, R.R.; Hahn, B.H.; Sercarz, E.E. J. Exp. Med., 1996, 386S.
183, 1613. [62] Mao, Y.; Nobaek, S.; Kasravi, B.; Adawi, D.; Stenram, U.;
[37] Berg, D.; Davidson, N.; Kuhn, R.; Muller, W.; Menon, S.; Molin, G.; Jeppsson, B. Gastroenterol., 1996, 111, 334.
Holland, G.; Thompson-Snipes, L.; Leach, M.; Rennick, [63] Madsen, K.L.; Doyle, J.S.; Jewell, L.D.; Tavernini, M.M.;
D. J. Clin. Invest., 1996, 98, 1010. Fedorak, R.N. Gastroenterol., 1999, 116, 1107.
[38] Salminen, S.; Bouley, C.; Boutron-Ruault, M.C.; [64] O'Mahony, L.; Feeney, M.; O'Halloran, S.; Murphy, L.;
Cummings, J.H.; Franck, A.; Gibson, G.R.; Isolauri, E.; Kiely, B.; Fitzgibbon, J.; Lee, G.; O'Sullivan, G.;
Moreau, M.C.; Roberfroid, M.; Rowland, I. Br. J. Nutr., Shanahan, F.; Collins, J.K. Aliment Pharmacol. Ther.,
1998, 80 Suppl 1, S147. 2001, 15, 1219.
[39] Chin, J.; Turner, B.; Barchia, I.; Mullbacher, A. Immunol. [65] Kruis, W.; Schutz, E.; Fric, P.; Fixa, B.; Judmaier, G.;
Cell Biol., 2000, 78, 55. Stolte, M. Aliment Pharmacol. Ther., 1997, 11, 853.
[40] Butler, J.E.; Weber, P.; Sinkora, M.; Baker, D.; [66] Rembacken, B.J.; Snelling, A.M.; Hawkey, P.M.;
Schoenherr, A.; Mayer, B.; Francis, D. J. Immunol., 2002, Chalmers, D.M.; Axon, A.T. Lancet, 1999, 354, 635.
169, 6822. [67] Gupta, P.; Andrew, H.; Kirschner, B.S.; Guandalini, S. J.
[41] Ouwehand, A.C.; Salminen, S.; Isolauri, E. Antonie Van Pediatr. Gastroenterol. Nutr., 2000, 31, 453.
Leeuwenhoek, 2002, 82, 279. [68] Guslandi, M.; Mezzi, G.; Sorghi, M.; Testoni, P.A. Dig.
[42] Erickson, K.L.; Hubbard, N.E. J. Nutr., 2000, 130, 403S. Dis. Sci., 2000, 45, 1462.
[43] McCabe, R.P.; Dean, P.; Elson, C.O. Cur. Opi. [69] Venturi, A.; Gionchetti, P.; Rizzello, F.; Johansson, R.;
Gastroenterol., 1996, 12, 340. Zucconi, E.; Brigidi, P.; Matteuzzi, D.; Campieri, M.
[44] Sartor, R.B. Res. Immunol., 1997, 148, 567. Aliment Pharmacol. Ther., 1999, 13, 1103.
[45] Schultsz, C.; Van Den Berg, F.M.; Ten Kate, F.W.; Tytgat, [70] Gionchetti, P.; Rizzello, F.; Venturi, A.; Brigidi, P.;
G.N.; Dankert, J. Gastroenterol., 1999, 117, 1089. Matteuzzi, D.; Bazzocchi, G.; Poggioli, G.; Miglioli, M.;
[46] Favier, C.; Neut, C.; Mizon, C.; Cortot, A.; Colombel, Campieri, M. Gastroenterol., 2000, 119, 305.
J.F.; Mizon, J. Dig. Dis. Sci., 1997, 42, 817. [71] Ulisse, S.; Gionchetti, P.; D'Alo, S.; Russo, F.P.; Pesce, I.;
[47] Giaffer, M.H.; Holdsworth, C.D.; Duerden, B.I. J. Med. Ricci, G.; Rizzello, F.; Helwig, U.; Cifone, M.G.;
Microbiol., 1991, 35, 238. Campieri, M.; De Simone, C. Am. J. Gastroenterol., 2001,
[48] Neut, C.; Colombel, J.F.; Guillemot, F.; Cortot, A.; 96, 2691.
Gower, P.; Quandalle, P.; Ribet, M.; Romond, C.; Paris, [72] Chauviere, G.; Coconnier, M.H.; Kerneis, S.; Fourniat, J.;
J.C. Gut, 1989, 30, 1094. Servin, A.L. J. Gen. Microbiol., 1992, 138 ( Pt 8), 1689.
[49] Swidsinski, A.; Ladhoff, A.; Pernthaler, A.; Swidsinski, [73] Mack, D.R.; Michail, S.; Wei, S.; McDougall, L.;
S.; Loening-Baucke, V.; Ortner, M.; Weber, J.; Hoffmann, Hollingsworth, M.A. Am. J. Physiol., 1999, 276, G941.
U.; Schreiber, S.; Dietel, M.; Lochs, H. Gastroenterol., [74] Isolauri, E.; Majamaa, H.; Arvola, T.; Rantala, I.; Virtanen,
2002, 122, 44. E.; Arvilommi, H. Gastroenterol., 1993, 105, 1643.
[50] Duchmann, R.; Kaiser, I.; Hermann, E.; Mayet, W.; Ewe, [75] Salminen, S.; Isolauri, E.; Salminen, E. Antonie Van
K.; Meyer zum Buschenfelde, K.H. Clin. Exp. Immunol., Leeuwenhoek, 1996, 70, 347.
1995, 102, 448. [76] Silva, M.; Jacobus, N.V.; Deneke, C.; Gorbach, S.L.
[51] Duchmann, R.; Schmitt, E.; Knolle, E.; Meyer Zum Antimicrob. Agents Chemother., 1987, 31, 1231.
Buschenfelde, K.-H.; Neurath, M. Eur. J. Immunol., 1996, [77] Rowland, I.R.; Rumney, C.J.; Coutts, J.T.; Lievense, L.C.
26, 934. Carcinogenesis, 1998, 19, 281.
[52] Dunne, C.; Murphy, L.; Flynn, S.; O'Mahony, L.; [78] Steidler, L. Microbes Infect., 2001, 3, 1157.
O'Halloran, S.; Feeney, M.; Morrissey, D.; Thornton, G.; [79] Miettinen, M.; Vuopio-Varkila, J.; Varkila, K. Infect.
Fitzgerald, G.; Daly, C.; Kiely, B.; Quigley, E.M.; Immun., 1996, 64, 5403.
O'Sullivan, G.C.; Shanahan, F.; Collins, J.K. Antonie van [80] Haller, D.; Bode, C.; Hammes, W.P.; Pfeifer, A.M.;
Leeuwenhoek, 1999, 76, 279. Schiffrin, E.J.; Blum, S. Gut, 2000, 47, 79.
[53] Sadlack, B.; Merz, H.; Schorle, H.; Schimpl, A.; Feller, [81] Haller, D.; Blum, S.; Bode, C.; Hammes, W.P.; Schiffrin,
A.C.; Horak, I. Cell, 1993, 75, 253. E.J. Infect. Immun., 2000, 68, 752.
[54] Kuhn, R.; Lohler, J.; Rennick, D.; Rajewsky, K.; Muller, [82] Malin, M.; Suomalainen, H.; Saxelin, M.; Isolauri, E. Ann.
W. Cell, 1993, 75, 263. Nutr. Metab., 1996, 40, 137.
[55] Morrissey, P.J.; Charrier, K.; Braddy, S.; Liggitt, D.; [83] Perdigon, G.; Vintini, E.; Alvarez, S.; Medina, M.;
Watson, J.D. J. Exp. Med., 1993, 178, 237. Medici, M. J. Dairy Sci., 1999, 82, 1108.
[56] Cong, Y.; Brandwein, S.L.; McCabe, R.P.; Lazenby, A.; [84] Vitini, E.; Alvarez, S.; Medina, M.; Medici, M.; de
Birkenmeier, E.H.; Sundberg, J.P.; Elson, C.O. J. Exp. Budeguer, M.V.; Perdigon, G. Biocell., 2000, 24, 223.
Med., 1998, 187, 855. [85] Maassen, C.B.; van Holten-Neelen, C.; Balk, F.; den Bak-
[57] Schultz, M.; Veltkamp, C.; Dieleman, L.A.; Grenther, Glashouwer, M.J.; Leer, R.J.; Laman, J.D.; Boersma, W.J.;
W.B.; Wyrick, P.B.; Tonkonogy, S.L.; Sartor, R.B. Claassen, E. Vaccine, 2000, 18, 2613.
Inflamm. Bowel Dis., 2002, 8, 71. [86] Cross, M.L. FEMS Immunol. Med. Microbiol., 2002, 34,
245.
Probiotics and Immune Regulation of Inflammatory Bowel Diseases Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 153

[87] Cross, M.L.; Mortensen, R.R.; Kudsk, J.; Gill, H.S. Med. [119] Barkla, D.H.; Gibson, P.R. Pathology, 1999, 31, 230.
Microbiol. Immunol. (Berl), 2002, 191, 49. [120] Dwinell, M.B.; Lugering, N.; Eckmann, L.; Kagnoff, M.F.
[88] Herias, M.V.; Hessle, C.; Telemo, E.; Midtvedt, T.; Gastroenterol., 2001, 120, 49.
Hanson, L.A.; Wold, A.E. Clin. Exp. Immunol., 1999, 116, [121] Mennechet, F.J.; Kasper, L.H.; Rachinel, N.; Li, W.;
283. Vandewalle, A.; Buzoni-Gatel, D. J. Immunol., 2002, 168,
[89] Kaila, M.; Isolauri, E.; Soppi, E.; Virtanen, E.; Laine, S.; 2988.
Arvilommi, H. Pediatr. Res., 1992, 32, 141. [122] Cerf-Bensussan, N.; Quaroni, A.; Kurnick, J.T.; Bhan,
[90] Fukushima, Y.; Kawata, Y.; Hara, H.; Terada, A.; A.K. J. Immunol., 1984, 132, 2244.
Mitsuoka, T. Int. J. Food Microbiol., 1998, 42, 39. [123] Panja, A.; Goldberg, S.; Eckmann, L.; Krishen, P.; Mayer,
[91] Fukushima, Y.; Kawata, Y.; Mizumachi, K.; Kurisaki, J.; L. J. Immunol., 1998, 161, 3675.
Mitsuoka, T. Int. J. Food Microbiol., 1999, 46, 193. [124] Kraehenbuhl, J.P.; Neutra, M.R. Physiol. Rev., 1992, 72,
[92] Perdigon, G.; Rachid, M.; De Budeguer, M.V.; Valdez, 853.
J.C. J. Dairy Res., 1994, 61, 553. [125] Neutra, M.R.; Mantis, N.J.; Kraehenbuhl, J.P. N a t .
[93] de Waard, R.; Garssen, J.; Snel, J.; Bokken, G.C.; Sako, T.; Immunol., 2001, 2, 1004.
Veld, J.H.; Vos, J.G. Clin. Diagn. Lab. Immunol., 2001, 8, [126] Kraehenbuhl, J.P.; Hopkins, S.A.; Kerneis, S.; Pringault,
762. E. Behring Inst. Mitt., 1997, 24.
[94] Pestka, J.J.; Ha, C.L.; Warner, R.W.; Lee, J.H.; Ustunol, Z. [127] Hunziker, W.; Kraehenbuhl, J.P. J. Mammary Gland Biol.
J. Food Prot., 2001, 64, 392. Neoplasia., 1998, 3, 287.
[95] Aattour, N.; Bouras, M.; Tome, D.; Marcos, A.; [128] Heppner, F.L.; Christ, A.D.; Klein, M.A.; Prinz, M.; Fried,
Lemonnier, D. Br. J. Nutr., , 2002, 87, 367. M.; Kraehenbuhl, J.P.; Aguzzi, A. Nat. Med., 2001, 7, 976.
[96] Shida, K.; Makino, K.; Morishita, A.; Takamizawa, K.; [129] Didierlaurent, A.; Sirard, J.C.; Kraehenbuhl, J.P.; Neutra,
Hachimura, S.; Ametani, A.; Sato, T.; Kumagai, Y.; Habu, M.R. Cell Microbiol., 2002, 4, 61.
S.; Kaminogawa, S. Int. Arch. Allergy Immunol., 1998, [130] Woolverton, C.J.; Holt, L.C.; Mitchell, D.; Sartor, R.B.
115, 278. Vet. Immunol. Immunopathol., 1992, 34, 127.
[97] Kirjavainen, P.V.; El-Nezami, H.S.; Salminen, S.J.; [131] Helgeland, L.; Vaage, J.T.; Rolstad, B.; Midtvedt, T.;
Ahokas, J.T.; Wright, P.F. FEMS Immunol. Med. Brandtzaeg, P. Immunol., 1996, 89, 494.
Microbiol., 1999, 26, 131. [132] Brandtzaeg, P.; Berstad, A.E.; Farstad, I.N.; Haraldsen, G.;
[98] Kirjavainen, P.V.; ElNezami, H.S.; Salminen, S.J.; Helgeland, L.; Jahnsen, F.L.; Johansen, F.E.; Natvig, I.B.;
Ahokas, J.T.; Wright, P.F. Clin. Diagn. Lab. Immunol., Nilsen, E.M.; Rugtveit, J. Behring Inst. Mitt., 1997, 1.
1999, 6, 799. [133] Madsen, K.L.; Lewis, S.A.; Tavernini, M.M.; Hibbard, J.;
[99] Suri-Payer, E.; Amar, A.Z.; Thornton, A.M.; Shevach, E.M. Fedorak, R.N. Gastroenterol., 1997, 113, 151.
J. Immunol., 1998, 160, 1212. [134] Ichikawa, H.; Kuroiwa, T.; Inagaki, A.; Shineha, R.;
[100] Groux, H.; Powrie, F. Immunol. Today, 1999, 20, 442. Nishihira, T.; Satomi, S.; Sakata, T. Dig. Dis. Sci., 1999,
[101] Roncarolo, M.G.; Levings, M.K. Curr. Opin. Immunol., 44, 2119.
2000, 12, 676. [135] Planchon, S.M.; Martins, C.A.; Guerrant, R.L.; Roche, J.K.
[102] Sakaguchi, S.; Sakaguchi, N.; Shimizu, J.; Yamazaki, S.; J. Immunol., 1994, 153, 5730.
Sakihama, T.; Itoh, M.; Kuniyasu, Y.; Nomura, T.; Toda, [136] Jung, S.; Fehr, S.; Harder-d'Heureuse, J.; Wiedenmann,
M.; Takahashi, T. Immunol. Rev., 2001, 182, 18. B.; Dignass, A.U. Scand. J. Gastroenterol., 2001, 36,
[103] Shevach, E.M.; McHugh, R.S.; Thornton, A.M.; Piccirillo, 963.
C.; Natarajan, K.; Margulies, D.H. Adv. Exp. Med. Biol., [137] Dignass, A.U. Inflamm. Bowel Dis., 2001, 7, 68.
2001, 490, 21. [138] Miller, A.; Lider, O.; Weiner, H.L. J. Exp. Med., 1991, 174,
[104] Battaglia, M.; Blazar, B.R.; Roncarolo, M.G. Microbes 791.
Infect., 2002, 4, 559. [139] Miller, A.; Lider, O.; Roberts, A.B.; Sporn, M.B.; Weiner,
[105] Belkaid, Y.; Piccirillo, C.A.; Mendez, S.; Shevach, E.M.; H.L. Proc. Natl. Acad. Sci. USA, 1992, 89, 421.
Sacks, D.L. Nature, 2002, 420, 502. [140] Chen, W.; Jin, W.; Cook, M.; Weiner, H.L.; Wahl, S.M. J.
[106] Cong, Y.; Weaver, C.T.; Lazenby, A.; Elson, C.O. J. Immunol., 1998, 161, 6297.
Immunol., 2002, 169, 6112. [141] Weiner, H.L. Microbes Infect., 2001, 3, 947.
[107] von der Weid, T.; Bulliard, C.; Schiffrin, E.J. Clin. Diagn. [142] Weiner, H.L. Nat. Immunol., 2001, 2, 671.
Lab. Immunol., 2001, 8, 695. [143] Weiner, H.L. Immunol. Rev., 2001, 182, 207.
[108] Groux, H.; O'Garra, A.; Bigler, M.; Rouloux, M.; [144] Sartor, R.B. Gastroenterol., 1994, 106, 533.
Antonenko, S.; deVries, J.E.; Roncarolo, M.G. Nature, [145] Sartor, R.B. Am. J. Gastroenterol., 1997, 92, 5S.
1997, 389, 737. [146] Schreiber, S. Gut, 1997, 41, 274.
[109] Herberman, R.B.; Ortaldo, J.R. Science, 1981, 214, 24. [147] Hara, M.; Kingsley, C.I.; Niimi, M.; Read, S.; Turvey, S.E.;
[110] Hercend, T.; Schmidt, R.E. Immunol. Today, 1988, 9, 291. Bushell, A.R.; Morris, P.J.; Powrie, F.; Wood, K.J. J.
[111] Trinchieri, G.; Perussia, B.; Santoli, D.; Cerottini, J.C. Immunol., 2001, 166, 3789.
Transplant Proc., 1979, 11, 807. [148] Cottrez, F.; Groux, H. J. Immunol., 2001, 167, 773.
[112] Santoli, D.; Koprowski, H. Immunol. Rev., 1979, 44, 125. [149] Groux, H.; Cottrez, F.; Rouleau, M.; Mauze, S.;
[113] Gill, H.S.; Rutherfurd, K.J.; Cross, M.L. J. Clin. Immunol., Antonenko, S.; Hurst, S.; McNeil, T.; Bigler, M.;
2001, 21, 264. Roncarolo, M.G.; Coffman, R.L. J. Immunol., 1999, 162,
[114] Hessle, C.; Hanson, L.A.; Wold, A.E. Clin. Exp. Immunol., 1723.
1999, 116, 276. [150] Wakkach, A.; Cottrez, F.; Groux, H. Eur. Cytokine Netw.,
[115] Fehniger, T.A.; Shah, M.H.; Turner, M.J.; VanDeusen, 2000, 11, 153.
J.B.; Whitman, S.P.; Cooper, M.A.; Suzuki, K.; Wechser, [151] Leach, M.W.; Davidson, N.J.; Fort, M.M.; Powrie, F.;
M.; Goodsaid, F.; Caligiuri, M.A. J. Immunol., 1999, 162, Rennick, D.M. Toxicol. Pathol., 1999, 27, 123.
4511. [152] Asseman, C.; Powrie, F. Gut, 1998, 42, 157.
[116] Haller, D.; Serrant, P.; Granato, D.; Schiffrin, E.J.; Blum, [153] Kucharzik, T.; Lugering, N.; Pauels, H.G.; Domschke, W.;
S. Clin. Diagn. Lab. Immunol., 2002, 9, 649. Stoll, R. Clin. Exp. Immunol., 1998, 111, 152.
[117] Kagnoff, M.F.; Eckmann, L. J. Clin. Invest., 1997, 100, 6. [154] Lammers, K.M.; Helwig, U.; Swennen, E.; Rizzello, F.;
[118] Strober, W. Ann. N. Y. Acad. Sci., 1998, 859, 37. Venturi, A.; Caramelli, E.; Kamm, M.A.; Brigidi, P.;
154 Current Drug Targets - Inflammation & Allergy, 2003, Vol. 2, No. 2 Cong et al.

Gionchetti, P.; Campieri, M. Am. J. Gastroenterol., 2002, [171] Maassen, C.B.; van Holten, J.C.; Balk, F.; Heijne den
97, 1182. Bak-Glashouwer, M.J.; Leer, R.; Laman, J.D.; Boersma,
[155] Neish, A.S.; Gewirtz, A.T.; Zeng, H.; Young, A.N.; Hobert, W.J.; Claassen, E. Vet. Quart., 1998, 20, S81.
M.E.; Karmali, V.; Rao, A.S.; Madara, J.L. Science, 2000, [172] Miettinen, M.; Lehtonen, A.; Julkunen, I.; Matikainen, S.
289, 1560. J. Immunol., 2000, 164, 3733.
[156] Yan, F.; Polk, D.B. J. Biol. Chem., 2002, [173] Gill, H.S.; Rutherfurd, K.J.; Prasad, J.; Gopal, P.K. Br. J.
[157] Banchereau, J.; Steinman, R.M. Nature, 1998, 392, 245. Nutr., 2000, 83, 167.
[158] Kronin, V.; Winkel, K.; Suss, G.; Kelso, A.; Heath, W.; [174] Borruel, N.; Carol, M.; Casellas, F.; Antolin, M.; de Lara,
Kirberg, J.; von Boehmer, H.; Shortman, K. J. Immunol., F.; Espin, E.; Naval, J.; Guarner, F.; Malagelada, J.R. Gut,
1996, 157, 3819. 2002, 51, 659.
[159] Suss, G. and Shortman, K. J. Exp. Med., 1996, 183, 1789. [175] Medzhitov, R.; Janeway, C., Jr. Immunol. Rev., 2000, 173,
[160] Jonuleit, H.; Schmitt, E.; Schuler, G.; Knop, J.; Enk, A.H. 89.
J. Exp. Med., 2000, 192, 1213. [176] Thoma-Uszynski, S.; Kiertscher, S.M.; Ochoa, M.T.;
[161] Steinbrink, K.; Wolfl, M.; Jonuleit, H.; Knop, J.; Enk, Bouis, D.A.; Norgard, M.V.; Miyake, K.; Godowski, P.J.;
A.H. J. Immunol., 1997, 159, 4772. Roth, M.D.; Modlin, R.L. J. Immunol., 2000, 165, 3804.
[162] Steinbrink, K.; Graulich, E.; Kubsch, S.; Knop, J.; Enk, [177] Hirschfeld, M.; Weis, J.J.; Toshchakov, V.; Salkowski,
A.H. Blood, 2002, 99, 2468. C.A.; Cody, M.J.; Ward, D.C.; Qureshi, N.; Michalek,
[163] Muller, G.; Muller, A.; Tuting, T.; Steinbrink, K.; Saloga, S.M.; Vogel, S.N. Infect. Immun., 2001, 69, 1477.
J.; Szalma, C.; Knop, J.; Enk, A.H. J. Invest. Dermatol., [178] Tanamoto, K.; Azumi, S.; Haishima, Y.; Kumada, H.;
2002, 119, 836. Umemoto, T. J. Immunol., 1997, 158, 4430.
[164] Kelsall, B.L.; Strober, W. S p r i n g e r Semin. [179] Yoshimura, A.; Lien, E.; Ingalls, R.R.; Tuomanen, E.;
Immunopathol., 1997, 18, 409. Dziarski, R.; Golenbock, D. J. Immunol., 1999, 163, 1.
[165] Kelsall, B.L.; Strober, W. Res. Immunol., 1997, 148, 490. [180] Muroi, M.; Ohnishi, T.; Tanamoto, K. J. Biol. Chem.,
[166] Christensen, H.R.; Frokiaer, H.; Pestka, J.J. J. Immunol., 2002, 277, 42372.
2002, 168, 171. [181] Martin, M.; Katz, J.; Vogel, S.N.; Michalek, S.M. J .
[167] Viney, J.L.; Mowat, A.M.; O'Malley, J.M.; Williamson, E.; Immunol., 2001, 167, 5278.
Fanger, N.A. J. Immunol., 1998, 160, 5815. [182] Pulendran, B.; Kumar, P.; Cutler, C.W.; Mohamadzadeh,
[168] Asseman, C.; Mauze, S.; Leach, M.W.; Coffman, R.L.; M.; Van Dyke, T.; Banchereau, J. J. Immunol., 2001, 167,
Powrie, F. J. Eex. Med., 1999, 190, 995. 5067.
[169] Morrissette, N.; Gold, E.; Aderem, A. Trends Cell Biol., [183] Ogawa, T.; Uchida, H.; Amino, K. Microbiol., 1994, 140 (
1999, 9, 199. Pt 5), 1209.
[170] Smith, P.D.; Smythies, L.E.; Mosteller-Barnum, M.; [184] Netea, M.G.; van Deuren, M.; Kullberg, B.J.; Cavaillon,
Sibley, D.A.; Russell, M.W.; Merger, M.; Sellers, M.T.; J.M.; Van der Meer, J.W. Trends Immunol., 2002, 23, 135.
Orenstein, J.M.; Shimada, T.; Graham, M.F.; Kubagawa, H. [185] Sutas, Y.; Hurme, M.; Isolauri, E. Scand. J. Immunol.,
J. Immunol., 2001, 167, 2651. 1996, 43, 687.

You might also like