Sie sind auf Seite 1von 5

SUPPLEMENT ARTICLE

Mechanisms of Action of Probiotics


W. Allan Walker
Division of Nutrition and Department of Pediatrics, Harvard Medical School, Boston, and Mucosal Immunology Laboratory, Massachusetts General
Hospital for Children, Charlestown, Massachusetts

At birth, the newborn leaves the germ-free intrauterine environment and enters a highly contaminated ex-
trauterine world, which requires potent host defenses to prevent disease. Intestinal defenses develop during
gestation and have the capacity to respond but first must be exposed to colonizing bacteria. I review the
importance of bacterial colonization for the appearance of normal mucosal immune function and the clinical
consequences of inadequate colonization with regard to development of disease. For example, we now know

Downloaded from http://cid.oxfordjournals.org/ at NERL on March 5, 2013


that an imbalance in T-helper (Th) cells (e.g., Th2 levels greater than Th1 levels) can predispose to autoimmune
disease and gut inflammation or disease, such as necrotizing enterocolitis. As we determine the role of bacterial
colonization in the gut (bacterial-epithelial “cross talk”), we should have more-appropriate ways to modulate
the gut immune responses—for example, by use of probiotics to prevent the expression of these gastrointestinal
diseases.

If we define probiotics as “live microorganisms that interact with molecular patterns on both pathogens and
when ingested have a positive effect on health” [1], we commensal bacteria [3], a molecular and cellular basis
can begin to include as probiotics certain commensal for communication could be appreciated. The TLR that
bacteria found in the human gut. During the past de- has been studied the most is TLR-4, whose primary
cade, microbiologists, immunologists, and gastroenter- ligand is gram-negative lipopolysaccharide (LPS) (i.e.,
ologists have actively studied the mechanism by which endotoxin). This receptor interacts with LPS as an LPS-
commensal bacteria improve mucosal defenses of the binding protein complex after being anchored to the
gastrointestinal tract. This article will review selected cell surface by a surface molecule, CD14. With this
aspects of these observations. interaction, a series of signaling molecules is activated
in the cell to release the transcription factor nuclear
BACTERIAL-EPITHELIAL “CROSS TALK” factor kB (NFkB) into the nucleus, which in turn tran-
scribes inflammatory cytokines—for example, IL-8 and
It has been known that colonizing bacteria that interact
IL-6—to provide the basis for an acute innate inflam-
with the gastrointestinal mucosa can communicate with
matory response to an invading pathogen. Since the
the underlying epithelial and mucosal lymphoid ele-
initial discovery of TLR-4, ten additional TLRs have
ments and that such interaction stimulates host de-
been cloned, and a variety of microbial molecular pat-
fenses in the gut [2]. However, it was not until recently
terns have been identified as ligands, including patterns
that investigators began to understand the so-called
on commensal bacteria [4]. The appreciation of mi-
bacterial-epithelial cross talk at the cellular level. With
crobial patterns that interact with pattern-recognition
the discovery of toll-like receptors (TLRs) on eukaryotic
receptors on eukaryotic cells has been the basis of our
epithelial, endothelial, and lymphoid cells, which could
understanding of bacterial-epithelial cross talk and its
role in both innate and adaptive mucosal immunity.
Reprints or correspondence: Dr. W. Allan Walker, Mucosal Immunology
Laboratory, Massachusetts General Hospital for Children, 114 16th St. (114-3503), DEVELOPMENT OF HOST DEFENSE
Charlestown, MA 02129 (wwalker@partners.org).
Clinical Infectious Diseases 2008; 46:S87–91 A vital practical application of bacterial-epithelial cross
 2008 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2008/4603S2-0008$15.00
talk is the communication between colonizing bacteria
DOI: 10.1086/523335 and the germ-free neonatal gastrointestinal tract im-

Probiotic Mechanisms • CID 2008:46 (Suppl 2) • S87


mediately after birth. The nature and composition of these Table 1. Probiotic effects on the development of host defense.
colonizing bacteria are important contributors to the devel-
opment of gastrointestinal host defenses against infection and Generalized mucosal immune response
allergic reaction. Table 1 lists several important components of Balanced T-helper cell response
Self-limited inflammatory response
host defenses affected by this initial colonization. At birth, the
Polymeric IgA secretion
full-term neonate has the developmental capacity to mount an
adequate host defense. However, for the efferent (i.e., active)
component of that defense to occur, the gut lymphoid tissue gut is the capacity of epithelial and lymphoid cells, after an
must first be stimulated by colonizing bacteria [5]. This in- acute innate immune response, to turn off the inflammatory
cludes expression of follicular epithelial (i.e, microfold) cells response in the absence of a self-limited manner. In the absence
over Peyer’s patches in the ileum and colon. These cells facilitate of a self-limited and innate inflammatory response, chronic
interaction between microbes and lymphoid cells [6]. In ad- clinical conditions such as inflammatory bowel disease and
dition, intraepithelial and lamina propria sites are activated to necrotizing enterocolitis (NEC) can occur. Accordingly, several
produce protective cytokines [7], and mesenteric lymph nodes mechanisms have evolved to control chronic inflammation of
secrete polymeric IgA (pIgA) [8]. Recently, it has been dem- the gut. These include the interaction between CpG DNA of
onstrated that dendritic cells in the lamina propria can extend bacteria and the intracellular receptor TLR-9, to which the
their appendices between epithelial cells, and, via TLR-2 and bacteria bind and thus activate T regulatory cells by the pro-

Downloaded from http://cid.oxfordjournals.org/ at NERL on March 5, 2013


TLR-4 on their surface, they can sample commensal-bacterial duction of anti-inflammatory cytokines (e.g., IL-10) (figure 2)
molecular patterns (figure 1) [9]. The interaction leads to mat- [11]. Commensal bacteria can also secrete small molecules that
uration of the dendritic cells and to the release of cytokines, can enter intestinal epithelial cells to inhibit activation of NFkB
which orchestrate the conversion of naive T-helper cells (Th0) [12]. In addition, it has been reported recently that prolonged
into a mature, balanced response of T-helper cells (Th1, Th2, exposure to molecular patterns (of peptidoglycans and LPS)
and Th3/Tr1), an important component in the prevention of that interact with TLR-2 and TLR-4 can activate cell surface
disease. Furthermore, commensal bacteria can cross microfold and intracellular negative regulators, respectively, which, in
cells and interact with antigen-presenting cells in mesenteric turn, can turn off transcription factors, leading to a reduction
lymph nodes to activate naive plasma cells into becoming pIgA- in the production of inflammatory cytokines and chemokines
producing B cells [10]. pIgA, in turn, coats the mucosal surface [13]. These cellular mechanisms are critical to both intestinal
to control subsequent microbial and antigen penetration. host defense and the prevention of chronic disease in the gas-
An important component of mucosal host defense in the trointestinal tract. With an inadequate initial bacterial coloni-

Figure 1. The direct effect of commensal bacteria on a balanced T-helper cell response. Immature dendritic cells in the lamina propria extend an
appendage between enterocytes into the intestinal lumen. Through toll-like receptor 2 (TLR-2) and TLR-4 on this appendage, they sample luminal
commensal bacteria, and, through this interaction, the dentritic cell matures and releases cytokines, which create a microenvironment for naive T-
helper cells (Th0) to mature into balanced Th1 and Th2/Tr1 helper subsets. TREG, T regulatory cells. Data are from [9].

S88 • CID 2008:46 (Suppl 2) • Walker


Downloaded from http://cid.oxfordjournals.org/ at NERL on March 5, 2013
Figure 2. The interaction of commensal bacteria CpG DNA with the intracellular toll-like receptor 9 (TLR-9) in dendritic cells, which stimulates the
production of Th3/Tr1 cells. Commensal bacterial penetrate microfold cells to enter Peyer’s patches and interact with dendritic cells, or they interact
with dendritic cells directly. CpG DNA from these bacteria interacts with the TLR-9 intracellular receptor in dendritic cells and triggers a cytokine
response that facilitates the maturation of T regulatory (TREG) cells that down-regulate acute inflammatory responses to prevent chronic intestinal
inflammation. TGF, transforming growth factor. Data are from [11].

zation, probiotics can be used to accomplish the same balanced was directly correlated with the inhibition of pathogenic Esch-
immune response [14]. erichia coli attachment and of damage to the intestinal tract.

OTHER PROTECTIVE EFFECTS OF PROBIOTICS PROBIOTIC MECHANISMS OF ACTION THAT


ARE DISEASE SPECIFIC
Probiotics have been demonstrated to have an adjuvant effect
on immunologic responses. As mentioned above, their inter- Oral tolerance, allergy, and immunity. The capacity to de-
action with mesenteric lymph nodes can result in an up-reg- velop oral tolerance is an important deterrent to the expression
ulation of pIgA against intestinal pathogens and food antigens of allergy and autoimmune disease. Oral tolerance relates to
[10]. In addition, Saccharomyces boulardii, a probiotic fungal commensal bacteria and innocuous antigens in the gut and
organism, has been demonstrated to enhance the specific IgG prevents excessive immunologic responses to increased
and IgA antibody response to Clostridium difficile toxin A after amounts of antigen stimulation of the gut. Oral tolerance oc-
Clostridium infection [15]. Infants with rotaviral gastroenteritis curs when innocuous antigens and commensal bacteria cross
who were given Lactobacillus rhamnosus strain GG had an in- the mucosal surface and interact with mucosal regulatory T
creased IgA response to the virus [16]. Finally, a pilot study cells that release a cytokine (i.e., transforming growth factor
involving healthy adults about to receive typhoid vaccine for b), which is thought to mediate the down-regulation of humeral
foreign travel showed that those given Lactobacillus GG for 10 and cellular immune responses to those antigens. A recent ob-
days before vaccination had significantly higher levels of anti- servation suggests that oral tolerance cannot occur in the ab-
typhoid antibodies than did those who received a placebo [17]. sence of colonized bacteria. In a study that compared oral
Furthermore, the effect of Lactobacillus GG was specific because tolerance between germ-free and colonized animals exposed
levels of antibodies to antigens in other vaccines received pre- orally to ovalbumin and that measured specific IgE levels to
viously were not increased. quantify the humeral systemic response, it was noted that oral
The use of probiotics has also been effective in enhancing tolerance, seen as a down-regulation of the IgE anti-ova re-
the mucosal barrier to pathogens and antigen presentation. A sponse, could not be achieved in the germ-free state [19]. In
recent study reported that known probiotics, Lactobacillus subsequent studies, both humeral and cellular systemic toler-
strains, could stimulate up-regulation of mucous genes in in- ance to oral antigens could be achieved in germ-free animals
testinal goblet cells [18]. Furthermore, the effect of these pro- if they were conventionalized during the newborn period but
biotics on the activation and secretion of mucus in the intestine not if conventionalized as adults, which suggests that early ex-

Probiotic Mechanisms • CID 2008:46 (Suppl 2) • S89


posure to colonizing bacteria is necessary to achieve oral tol- cross talk, which results in both innate and acquired immune
erance [20]. Finally, recent reports have suggested that the ex- responses. Different probiotics can function differently in the
pression of TLR-4 on enterocytes is necessary to achieve development of intestinal host defenses, as an adjuvant of im-
tolerance [21] and that extensive use of antibiotics would re- mune responses or to strengthen the mucosal barrier. There-
duce levels of colonizing bacteria and thereby cause a loss of fore, one needs to be careful when selecting a probiotic for a
oral tolerance [22]. Finally, probiotics can be used to regain specific function in the gastrointestinal tract.
tolerance after extensive use of antibiotics [23].
NEC. A major condition that affects premature infants is Acknowledgments
NEC. It is thought that necrotic inflammation of the distal Financial support. National Institutes of Health (R37 HD12437, RO1
small intestine is due to an inadequate, immature, excessive DK70260, P30 DK40561, and PO1 DK33506).
inflammatory response to both pathogenic and commensal bac- Supplement sponsorship. This article was published as part of a sup-
plement entitled “Developing Probiotics as Foods and Drugs: Scientific and
teria [24, 25]. A recent clinical study in Taiwan showed that Regulatory Challenges,” sponsored by the Drug Information Association,
the introduction of probiotics (Lactobacillus lactis and Bifido- the National Institutes of Health National Center for Complementary and
bacteria infantum) a few days after birth in premature babies Alternative Medicine (1R13AT003805-01 to Patricia L. Hibberd), the Cal-
ifornia Dairy Research Foundation, Chr. Hansen, the Dannon Company,
with body weight !1500 g can prevent the expression of NEC
General Mills, Institut Rosell, and Yakult International.
[26]. A meta-analysis of different organisms used as probiotics Potential conflicts of interest. W.A.W. is part of the Dannon/Yakult
in this situation has shown that results are generally positive Scientific Advisory Board and is a consultant for the Dannon Company.

Downloaded from http://cid.oxfordjournals.org/ at NERL on March 5, 2013


[27]. We reported elsewhere that IkB, the inhibitor of NFkB,
is underdeveloped in premature enterocytes, which in part ac- References
counts for the NFkB-mediated, excessive, immature increase in 1. Joint FAO/WHO Working Group. Guidelines for the evaluation of
IL-8 production and for the inflammation in NEC [28]. In probiotics in food: report of a joint FAO/WHO working group on
subsequent studies using a known enterocyte-maturation fac- drafting guidelines for the evaluation of probiotics in food, London,
Ontario, Canada. 2002. Available at: http://www.fermented-foods.net/
tor, corticosteroids, we showed that pretreatment with this wgreport2.pdf. Accessed 19 December 2007.
trophic factor in fetal enterocytes can reduce the IL-8 response 2. Shi HN, Walker WA. Bacterial colonization and the development of
to inflammatory stimuli by increased IkB expression, suggesting intestinal defenses. Can J Gastroenterol 2004; 18:493–500.
3. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking
that developmental reduction in the expression of genes that innate and acquired immunity. Nat Immunol 2001; 2:675–80.
regulate the innate immune response may account for the ex- 4. Kawai T, Akira S. TLR signaling. Cell Death Differ 2006; 13:816–25.
pression of NEC in premature infants [29]. Most recently, using 5. Cherayil BJ, Walker WA. Ontogeny of the host response to enteric
microbial infection. In: Hecht G, ed. Microbial pathogenesis and the
DNA microarrays of fetal enterocyte RNA and biopsy of in-
intestinal epithelial cell. Washington, DC: ASM Press, 2003:333–49.
testinal specimens from older children, colleagues and I have 6. Kerneis S, Bogdanova A, Kraehenbuhl JP, Pringault E. Conversion by
shown that TLRs and their signaling-molecule genes are up- Peyer’s patch lymphocytes of human enterocytes into M cells that
regulated, and the negative regulators of innate inflammatory transport bacteria. Science 1997; 277:910–1.
7. Kohler H, McCormick BA, Walker WA. Bacterial-enterocyte crosstalk:
response are down-regulated, again suggesting the develop- cellular mechanisms in health and disease. J Pediatr Gastroenterol Nutr
mental basis for the extensive inflammatory response (N. Nan- 2003; 36:175–85.
thakumar and A. Walker, personal communication). We have 8. Macpherson AJS, Lamarre A, McCoy K, et al. IgA production without
m or d chain expression in developing B cells. Nat Immunol 2001; 2:
also shown that culture media from the same probiotics shown 625–31.
to prevent clinical NEC (i.e., L. lactis and B. infantum [26]) 9. Rescigno M, Urbano M, Valzasina B, et al. Dendritic cells express tight
can modify expression of genes of the innate immune response junction proteins and penetrate gut epithelial monolayers to sample
bacteria. Nat Immunol 2001; 2:361–7.
(e.g., TLRs and signaling molecules were decreased and negative
10. Macpherson AJ, Uhr T. Induction of protective IgA by intestinal den-
regulators were increased) and, at the same time, can strikingly dritic cells carrying commensal bacteria. Science 2004; 303:1662–5.
reduce the IL-8 response (N. Nanthakumar and A. Walker, 11. Rachmilewitz D, Katakara K, Karmeli F, et al. Toll-like receptor 9
signaling mediates the anti-inflammatory effects of probiotics in mu-
personal communication), which further suggests a mechanism
rine experimental colitis. Gastroenterology 2004; 126:520–8.
for protection against NEC. 12. Neish AS, Gewirtz AT, Zeng H, et al. Prokaryotic regulation of epithelial
responses by inhibition of IkBa degradation. Science 2000; 289:1560–3.
13. Otte JM, Podolsky DK. Functional modulation of enterocytes by gram-
CONCLUSIONS
positive and gram-negative microorganisms. Am J Physiol Gastrointest
Liver Physiol 2004; 286:G613–26.
In this review, I have suggested that, on the basis of the strict
14. Kalliomaki MA, Walker WA. Physiologic and pathologic interactions
definition of probiotics, certain human commensal bacteria of bacteria with gastrointestinal epithelium. Gastroenterol Clin North
may qualify as probiotics. The demonstration of pattern-rec- Am 2005; 34:383–99.
ognition receptors (e.g., TLRs) on eukaryotic cells, which in- 15. Qamar A, Aboudola A, Warny M, et al. Saccharomyces boulardii stim-
ulates intestinal immunoglobulin A immune response to Clostridium
teract with molecular patterns on pathogens and commensal difficile toxin A in mice. Infect Immun 2001; 69:2762–5.
bacteria, can provide the molecular basis for bacterial-epithelial 16. Isolauri E, Juntunen M, Rautanen T, et al. Lactobacillus strain (Lac-

S90 • CID 2008:46 (Suppl 2) • Walker


tobacillus casei strain GG) promotes recovery from acute diarrhea in 24. Claud EC, Savidge T, Walker WA. Modulation of human intestinal
children. Pediatrics 1991; 88:90–7. epithelial cell interleukin-8 secretion by human milk factors. Pediatr
17. Teitelbuam JE, Walker WA. Nutritional impact of pre- and probiotics Res 2003; 53:419–25.
as protective gastrointestinal organisms. Annu Rev Nutr 2002; 22: 25. Nanthakumar N, Fusunyan RD, Sanderson IR, Walker WA. Inflam-
107–38. mation in the developing human intestine: a possible pathophysiologic
18. Mack DR, Michail S, Wei S, et al. Probiotics inhibit enteropathogenic
basis for necrotizing enterocolitis. Proc Natl Acad Sci U S A 2000; 97:
E. coli adherence in vitro by inducing intestinal mucin gene expression.
6043–8.
Am J Physiol 1999; 276:G941–50.
19. Sudo N, Sawamura S, Tanaka Y, et al. The requirement of intestinal 26. Lin HC, Su BH, Chen AC, et al. Oral probiotics reduce the incidence
bacterial flora for the development of an IgE production system fully and severity of necrotizing enterocolitis in very low birth weight infants.
susceptible to oral tolerance induction. J Immunol 1997; 159:1739–45. Pediatrics 2005; 115:1–4.
20. Spiekermann G, Walker WA. Oral tolerance and its role in clinical 27. Deshpande G, Rao S, Patole S. Probiotics for prevention of necrotizing
disease. J Pediatr Gastroenterol Nutr 2001; 32:237–55. enterocolitis in preterm neonates with very low birthweight: a system-
21. Bashir MEH, Andersen P, Fuss IJ, et al. An enteric helminth infection atic review of randomized controlled trials. Lancet 2007; 369:1614–20.
protects against an allergic response to dietary antigen. J Immunol 28. Claud EC, Lu L, Anton PM, Savidge T, Walker WA, Cherayil BJ. De-
2002; 169:3284–92. velopmentally-regulated IkB expression in intestinal epithelium and
22. Bashir MEH, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor susceptibility to flagellin-induced inflammation. Proc Natl Acad Sci U
4 signaling by intestinal microbes influences susceptibility to food al-
S A 2004; 101:7404–8.
lergy. J Immunol 2004; 172:6978–87.
29. Nanthakumar NN, Young C, Ko JS, et al. Glucocorticoid responsiveness
23. Braat H, van den Brande J, van Tol E, Hommes D, Peppelenbosch M,
van Deventer S. Lactobacillus rhamnosus induces peripheral hypore- in the developing human intestine: possible role in the prevention of
sponsiveness in stimulated CD4+ T cells via modulation of dendritic necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol
cell function. Am J Clin Nutr 2004; 80:1618–25. 2005; 288:G85–92.

Downloaded from http://cid.oxfordjournals.org/ at NERL on March 5, 2013

Probiotic Mechanisms • CID 2008:46 (Suppl 2) • S91

Das könnte Ihnen auch gefallen