You are on page 1of 6

Nutrition in Clinical Practice http://ncp.sagepub.

com/

Unraveling Mechanisms of Action of Probiotics


Philip M. Sherman, Juan C. Ossa and Kathene Johnson-Henry
Nutr Clin Pract 2009 24: 10
DOI: 10.1177/0884533608329231

The online version of this article can be found at:


http://ncp.sagepub.com/content/24/1/10

Published by:

http://www.sagepublications.com

On behalf of:

The American Society for Parenteral & Enteral Nutrition

Additional services and information for Nutrition in Clinical Practice can be found at:

Email Alerts: http://ncp.sagepub.com/cgi/alerts

Subscriptions: http://ncp.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

>> Version of Record - Feb 24, 2009

What is This?

Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014
Nutrition in Clinical Practice
Invited Review Volume 24 Number 1
February/March 2009 10-14

Unraveling Mechanisms of Action © 2009 American Society for


Parenteral and Enteral Nutrition
10.1177/0884533608329231
of Probiotics http://ncp.sagepub.com
hosted at
http://online.sagepub.com

Philip M. Sherman, MD, FRCPC; Juan C. Ossa, MD;


and Kathene Johnson-Henry, BSc
Financial disclosure: Research efforts in the laboratory of the authors are supported by a Fay Shapiro-Cutler Research Grant-in-Aid
from the Crohn’s and Colitis Foundation of Canada and by Institut Rosell-Lallemand, Inc (Montreal, Quebec, Canada). PMS is the
recipient of a Canada Research Chair in Gastrointestinal Disease.

Probiotics are defined as living organisms that, when adminis- regulatory T cells, interleukin-10). Some probiotics have the
tered in sufficient numbers, are of benefit to the host. Current capacity to activate receptors in the enteric nervous system, which
evidence indicates that varying probiotic strains mediate their could be used to promote pain relief in the setting of visceral
effects by a variety of different effects that are dependent on the hyperalgesia. Future development of the effective use of probi-
dosage employed as well as the route and frequency of delivery. otics in treating various gastroenterological disorders in human
Some probiotics act in the lumen of the gut by elaborating anti- participants should take advantage of this new knowledge. The
bacterial molecules such as bacteriocins; others enhance the creation of novel formulations of probiotics could be directed to
mucosal barrier by increasing the production of innate immune effectively target certain mechanisms of actions that are altered
molecules, including goblet cell–derived mucins and trefoil fac- in specific disease states. (Nutr Clin Pract. 2009;24:10-14)
tors and defensins produced by intestinal Paneth cells; and
other probiotics mediate their beneficial effects by promoting Keywords: lactobacillus; probiotics; intestines; diarrhea;
adaptive immune responses (secretory immune globulin A, mucins; defensins; tight junctions

T
he World Health Organization defines probiotics as effects in a variety of conditions involving the gastrointesti-
live organisms (bacteria, yeast) that, when ingested nal (GI) tract.5 Figure 1 summarizes potential mechanisms
in sufficient amounts, have a beneficial effect on of action of probiotics. As considered in greater detail
the overall health of the host.1 An array of products is cur- below, it is now apparent that not all probiotic strains exert
rently sold to consumers,2 but availability varies from the same mechanisms of action. The separations consid-
country to country. In fact, there are concerns about ered below are somewhat artificial in nature but are simply
batch-to-batch consistency in viable numbers of organ- presented as a conceptual framework by which to consider
isms present in many of the probiotics now available in the just how probiotic agents might work. Certainly, some pro-
marketplace. Current enthusiasm for implementing the biotic agents will have multiple activities and can affect
use of probiotics has been hampered, at least in part, by various stages that are considered in the figure.
concerns about precisely how the various organisms pur-
ported as probiotic agents mediate their beneficial effects.
A wide array of strains employed and a bewildering com- Lumenal Effects
plexity of proposed mechanisms of action have raised a
certain level of skepticism in the biomedical research com- Enhancing the Microbial Flora:
munity as a whole. Nevertheless, recent advances in the Creating a Balance
field are helping to sort out facts from fiction.3,4
Multiple randomized, controlled clinical trials and Probiotics possess the ability to transiently colonize the GI
meta-analyses demonstrate that probiotics elicit beneficial tract, increase the concentration of beneficial microbes,
and thereby create a balance in the gut microbiota to the
ultimate benefit of the host. A front line of defense against
From the Research Institute, Hospital for Sick Children,
the adverse effects of pathogens is provided by probiotics
University of Toronto, Toronto, Ontario, Canada. that exert a direct antimicrobial effect. For instance, some
probiotic strains elaborate antibacterial products referred to
Address correspondence to: Philip M. Sherman, MD, FRCPC,
Gastroenterology and Nutrition, Room 8409, Hospital for Sick
as bacteriocins. These antimicrobial factors inhibit the
Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, growth and virulence of enteric bacterial pathogens. Isogenic
Canada; e-mail: philip.sherman@sickkids.ca. mutants deficient in the gene coding for bacteriocin are less

10
Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014
Actions of Probiotics / Sherman et al 11

Figure 1. Schematic depiction of potential mechanisms of action by which probiotics can promote GI health. Probiotics (PB) and
surface-layer proteins (slp) competitively exclude microbial pathogens from mucosal surfaces. Tight junction proteins, such as zona
occludins (ZO)-1 and claudin1, remain intact and thereby prevent both uptake of intact macromolecules and translocation of viable
organisms to mesenteric lymph nodes. Probiotic strains possess the ability to modulate signal transduction pathways to block activation
and translocation to the nucleus of the transcription factor, nuclear factor-kappa B (NF-κB), interferon-γ (IFNγ), and mitogen-activated
protein kinases (MAPK). Through a cascade of signaling events, probiotics can enhance production and secretion of anti-inflammatory
cytokines, including interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) by a subset of immune cells, referred to as T reg-
ulatory cells (Tregs). Humoral immune system responses to probiotics include enhanced production of immune globulins, including
secretory immunoglobin A (sIgA). Innate immune responses to probiotics include increased mucin and trefoil factor production by
goblet cells (G) and enhanced production of antibacterial defensins by Paneth cells (PC) and intestinal epithelia. DC, dendritic cell;
P, pathogen; TC, T lymphocyte.

effective as probiotics, at least when tested in a mouse secreted factors from Lactobacillus acidophilus (strain La-5)
model of invasive Listeria monoctyogenes infection.6 Lactic affect virulence gene expression by enterohemorrhagic
acid–producing probiotics may also exert antimicrobial Escherichia coli of the serotype O157:H7.9 The probiotic
effects on pathogenic organisms by reducing the local pH reduces the secretion of autoinducer-2 molecules by the
of the microenvironment in the gut lumen. Such specula- pathogenic E. coli, which results in reduced expression of
tion is supported by in vitro studies demonstrating that co- genes contained in the locus of enterocyte effacement
culture of Lactobacillus species with pathogens results in (LEE) pathogenicity island that is critical for mediating
reduced growth of the virulent organism in a pH-dependent intimate bacterial binding to host cell surfaces—the so-
manner.7 called attaching and effacing lesion.10
Quorum sensing refers to the mechanisms by which
single-cell organisms communicate with each other.8 It
Colonization Resistance
has been suggested that some strains of probiotics may
influence gene expression of microbial pathogens and Although some probiotic strains have the ability to secrete
thereby reduce their virulence potential. For instance, factors that affect the luminal microbiotia, other probiotics

Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014
12 Nutrition in Clinical Practice / Vol. 24, No. 1, February/March 2009

interact with epithelial cells lining the GI tract to prevent enhancing the integrity of the epithelial barrier, which
the binding and downstream effects of enteric pathogens. serves as the surface lining of the GI tract.21,22 Probiotics
Probiotics, such as Lactobacillus helveticus (strain may do so by preventing changes in tight junction pro-
R0052), possess relatively hydrophobic cell surface prop- teins (such as occludins and claudins) and by enhancing
erties and therefore are able to bind nonspecifically to the electrical resistance of tight junctions contained in
host cell surfaces, including the apical microvillus mem- the apical junction complexes between adjacent polarized
brane of epithelial cells.11 An attribute of this probiotic epithelia, which occur in a protein kinase C and mitogen-
strain is that it possesses a thick surface-layer protein, activated protein (MAP) kinase-dependent fashion.23
which has the ability to adhere to the surface of epithelial Reduced programmed cell death (apoptosis) also appears
cells and reduce the effects of bacterial enteropathogens.11 to enhance barrier resistance to injurious agents, including
As a result, probiotics and nonviable products derived proinflammatory cytokines.24 Decreased apoptosis may
from certain probiotic strains can obscure receptor bind- serve to maintain epithelial barrier integrity and resist-
ing sites to prevent pathogens from binding and invading ance to injurious agents, including enteric pathogens, by
the infected host. reducing breaks in the mucosal barrier.
Probiotics may also compete for a limited niche in
the complex microbiota of the large bowel, thereby exclud-
ing a site for replication by pathogens.12 This mechanism Extending Findings to Relevant Animal
of action is often referred to as colonization resistance. Models of Intestinal Injury
Saccharomyces boulardii also has the potential to indi-
rectly block the attachment of enterohemorrhagic E. coli, The protective effects of probiotics on intestinal function
serotype O157:H7, to mucosal surfaces by binding in vitro have been confirmed in some in vivo studies by
directly to the pathogen.13 using the Citrobacter rodentium infection of mice as a model
of bacterial-induced infectious colitis.25 Complementary
studies in rodent models of stress-induced intestinal
Mucosal Effects barrier dysfunction, including use of water avoidance,26
physical restraint,27 and maternal separation of rat pups,28
Probiotics, such as Lactobacillus plantarum (strain 299v), also demonstrate the benefit of probiotics in enhancing
have the capacity to enhance the production and secretion epithelial cell barrier integrity, including a reduction in
of mucins (MUC2 and MUC3) from human intestinal (HT- both the uptake of intact macromolecules (including, for
29) epithelial cells.14 Probiotic mixtures also increase example, fluorescein-tagged dextran, horseradish peroxi-
MUC2 gene expression and mucin protein secretion in rat dase, and chromium-labeled EDTA) and the translocation
colon.15 The enhanced mucus layer overlying the epithelial of viable bacteria from the intestinal lumen to mesenteric
lining of the gut can serve as an antibacterial shield that pre- lymph nodes.
vents the binding of enteric pathogens, such as enteropath-
ogenic E. coli,16 to mucosal surfaces and increase clearance
of the pathogen from the GI tract.17 Trefoil factors are anti- Submucosal Effects
bacterial peptides, which are also secreted from mucin-
producing cells in response to various noxious stimuli, Through bacterial-epithelial cell cross-talk, probiotics are
including microbial pathogens. These trefoil factors appear able to affect both innate and adaptive arms of the host
to act in concert with mucins to reduce binding of pathogens immune system.29 For instance, some probiotic strains
to the epithelial cell surface.18 have the ability to promote the differentiation of B cells
Paneth cells are located near the base of the crypt cell into plasma cells and increase the production of secretory
compartment in the distal small intestine. These special- immunoglobulin A.30 Polymeric IgA sticks to the mucus
ized cells produce antibacterial cationic peptides referred layer overlying the gut epithelium and binds to patho-
to as defensins (and cryptdins, in mice). At least some genic microorganisms, thereby reducing their ability to
probiotics enhance release of defensins from intestinal gain access and bind to the microvillus membrane glyco-
epithelia, which may explain, at lease in part, the benefi- calyx of enterocytes and colonocytes.
cial effects of probiotics in the setting of acute infectious Probiotics can also prevent activation of the proin-
enteritis.19 For instance, the probiotics Lactobacillus fermen- flammatory nuclear transcription factor, nuclear factor-
tum and E. coli (strain Nissle 1917) each stimulate, in both kappa B (NF-κB).31,32 This results in decreased secretion
a time- and dose-dependent manner, human β-defensin of the chemokine interleukin (IL)-8, which is a potent
mRNA expression and protein secretion in intestinal neutrophil chemoattractant. Probiotics and nonpathogenic
(Caco2) epithelial cells grown in tissue culture.20 organisms prevent NF-κB translocation from the cytosol
In addition to promoting the production of antibac- to the nucleus by blocking phosphorylation and subse-
terial substances, probiotics also have a direct effect on quent degradation by ubiquitination of I-κB.33 Although

Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014
Actions of Probiotics / Sherman et al 13

some probiotics are able to inhibit activation of this Conclusions


nuclear transcription factor, there are also reports that
other strains of probiotics are able to activate NF-κB and There has been an explosion in our knowledge related to
increase levels of the proinflammatory cytokine IL-6.3 how probiotics exert their beneficial effects. Current evi-
These discrepancies serve to emphasize further the dence suggests that we should remain cautiously optimistic
strain-specific effects of probiotics on the host. about the increasingly widespread use of these agents as
Certain T cell immune responses are also affected by therapeutics. However, there is also reason to pause and
probiotics. For instance, using adoptive transfer of reflect on the potential for adverse effects when using
CD4+CD45RBhi T lymphocytes from healthy mice into viable organisms. For instance, the results of a prospective,
SCID (severe combined immunodeficiency) mice as a randomized trial in humans with severe acute pancreatitis
reproducible model of colitis, the yeast probiotic S. described a 2.5-fold increased relative risk of death among
boulardii was shown to reduce intestinal inflammation.34 those entered into the probiotic treatment arm of the
The mechanism of action relates to a yeast-related secre- study.40 Although the precise mechanisms accounting for
tion product, which reduces the trafficking of T helper-1 this adverse outcome are not known, the authors of the
cells, producing the proinflammatory cytokine interferon-γ report suggest that reduced splanchnic blood flow in
from mesenteric lymph nodes to the inflamed colon. In severely ill patients cared for in an intensive care unit set-
a model of hapten-induced colitis in mice, probiotics ting may impair clearance of the large numbers of viable
(VSL #3) reduced the severity of a second induction of bacteria employed as a probiotic agent. Relative tissue
intestinal inflammation, which was associated with the hypoxia and impaired gut barrier function both could
presence of regulatory T cells expressing transforming increase the risk of bacterial translocation. Evidence of
growth factor (TGF)-β.35 In another study, the subset bowel ischemia in 9 patients treated with probiotics,
of regulatory T cells expressing CD4 and Foxp3 was including 8 who died, vs none of the patients in the placebo
increased following challenge with Lactococcus lactis.36 group supports such considerations. Accordingly, even
These biological effects are mediated by TGF-β and the though probiotics generally can be considered safe for use
anti-inflammatory cytokine IL-10. in human participants,41 enthusiasm for testing probiotics
It has also been demonstrated that varying probiotic in high dosages, in combination regimens, and in extremely
strains can have different effects on the secretion of anti- ill patients must be tempered with the potential for causing
inflammatory (eg, IL-10) and proinflammatory (eg, IL-12) harm. As a result, initial animal studies and subsequent
cytokines by immune cells. The relative secretion profiles applications to humans must be conducted with rigor
are reported to predict the ability of the probiotic strains to employing the best available research methodologies.
have an impact on disease outcome—for instance, in Ongoing studies should continue to rely on comple-
experimental models of hapten-induced colitis in mice.37 In mentary experimental approaches, including reductionist
humans with irritable bowel syndrome (IBS), participants systems employing eukaryotic cells grown in tissue cul-
randomized to treatment with Bifodobacterium infantis ture and relevant animal models of human disease.
(strain 35624) resulted in an improvement in clinical Increasing evidence demonstrates that probiotics have
symptoms in association with a normalization of the ratio multiple mechanisms of action that are related to effects
of IL-10:IL-12 secretion by peripheral blood mononuclear in the lumen of the gut, the integrity of the mucosal bar-
cells. By contrast, another probiotic, Lactobacillus salivar- rier, and both innate and adaptive immune host
ius (strain UCC4331), had less effect on abdominal symp- responses. However, such effects are not likely to be
toms and had no discernable effect on the abnormally low caused by all probiotics strains tested. Thus, strain selec-
ratio of IL-10:IL-12 in patients with IBS.38 These observa- tion and consideration of the merits of using combina-
tions serve to highlight the strain-specific effects of various tions of probiotics may well provide a strategy in the
agents proposed for use as probiotics. future to develop targeted interventions.

Impact of Probiotics on the


Acknowledgments
Enteric Nervous System We thank Ms Aleah Henry for the production of the fig-
A recent interesting and novel study provides compelling ure employed in this work.
evidence that some probiotics also have the capacity to
activate specific opioid and cannibinoid receptors in the
References
gut.39 Such strains could prove particularly useful as
agents in settings where visceral pain is a prominent man- 1. Agostoni C, Axelsson I, Braegger C, et al. Probiotic bacteria in dietetic
ifestation of disease—for instance, in patients with IBS products for infants: a commentary by the ESPGHAN committee on
and chronic inflammatory bowel diseases. nutrition. J Pediatr Gastroenterol Nutr. 2004;38:365-374.

Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014
14 Nutrition in Clinical Practice / Vol. 24, No. 1, February/March 2009

2. Reid G, Anukam K, Koyama T. Probiotic products in Canada with changes in polarized T84 epithelial cell monolayers by reducing
clinical evidence: what can gastroenterologists recommend? Can J bacterial adhesion and cytoskeletal rearrangements. Infect Immun.
Gastroenterol. 2008;22:169-175. 2005;73:5183-5188.
3. Boirivant M, Strober W. The mechanism of action of probiotics. 22. Johnson-Henry KC, Donato KA, Shen-Tu G, Gordanpour M,
Curr Opin Gastroenterol. 2007;23:679-692. Sherman PM. Lactobacillus rhamnosus strain GG prevents entero-
4. Saavedra JM. Use of probiotics in pediatrics: rationale, mecha- hemorrhagic Escherichia coli O157:H7-induced changes in epithe-
nisms of action, and practical aspects. Nutr Clin Pract. 2007;22: lial barrier function. Infect Immun. 2008;76:1340-1348.
351-365. 23. Seth A, Yan F, Polk DB, Rao RK. Probiotics ameliorate the hydro-
5. Michail S, Sylvester F, Fuchs G, Issenman R, for the NASPGHAN gen peroxide-induced epithelial barrier disruption by a PKC- and
Nutrition Report Committee. Clinical efficacy of probiotics: review MAP kinase dependent mechanism. Am J Physiol. 2008;294:
of the evidence with focus on children. J Pediatr Gastroenterol G1060-G1069.
Nutr. 2006;43:550-557. 24. Yan F, Cao H, Cover TL, Whitehead R, Washington MK, Polk DB.
6. Corr SC, Li Y, Riedel CU, O’Toole PW, Hill C, Gaha CG. Soluble proteins produced by probiotic bacteria regulate intestinal
Bacteriocin production as a mechanism for the antiinfective activ- epithelial cell survival and growth. Gastroenterology. 2007;132:
ity of Lactobacillus salivarius UCC118. Proc Natl Acad Sci USA. 562-575.
2007;104:7617-7621. 25. Johnson-Henry KC, Nadjafi M, Avitzur Y, et al. Amelioration of the
7. Fayol-Messaoudi D, Berger CN, Coconnier Polter M-H, Lievin-Le effects of Citrobacter rodentium infection in mice by pretreatment
Moal V, Servin AL. pH-, lactic acid-, and non-lactic acid-dependent with probiotics. J Infect Dis. 2005;191:2106-2117.
activities of probiotic lactobacilli against Salmonella enterica 26. Zareie M, Johnson-Henry K, Jury J, et al. Probiotics prevent bacte-
serovar Typhimurium. Appl Environ Microbiol. 2005;71:6008-6013. rial translocation and improve intestinal barrier function in rats
8. Hughes DT, Sperandio V. Inter-kingdom signalling: communica- following chronic psychological stress. Gut. 2006;55:1553-1560.
tion between bacteria and their hosts. Nat Rev Microbiol. 2008;6: 27. Ait-Belgnaoui A, Han W, Lamine F, et al. Lactobacillus farciminis
111-120. treatment suppresses stress induced visceral hypersensitivity: a
9. Medellin-Pena MJ, Wang H, Johnson R, Anand S, Griffiths MW. possible action through interaction with epithelial cell
Probiotics affect virulence-related gene expression in Escherichia cytoskeleton contraction. Gut. 2006;55:1090-1094.
coli O157:H7. Appl Environ Microbiol. 2007;73:4259-4267. 28. Gareau MG, Jury J, MacQueen G, Sherman PM, Perdue MH.
10. Russell RM, Sharp FC, Rasko DA, Sperandio V. QseA and Probiotic treatment of rat pups normalizes corticosterone release
GrlR/GrlA regulation of the locus of enterocyte effacement genes and ameliorates colonic dysfunction induced by maternal separa-
in enterohemorrhagic Escherichia coli. J Bacteriol. 2007;189: tion. Gut. 2007;56:1522-1528.
5387-5392. 29. Walker WA. Mechanisms of action of probiotics. Clin Infect Dis.
11. Johnson-Henry KC, Hagen KE, Gordonpour M, Tompkins TA, 2008;48(suppl 2):S87-S91.
Sherman PM. Surface-layer protein extracts from Lactobacillus 30. Corthesy B, Gaskins HR, Mercenier A. Cross-talk between probiotic
helveticus inhibit enterohaemorrhagic Escherichia coli O157:H7 bacteria and the host immune system. J Nutr. 2007;137:781S-790S.
adhesion to epithelial cells. Cell Microbiol. 2007;9:356-367. 31. Fiocchi C. Probiotics in inflammatory bowel disease: yet another
12. Wu X, Vallance BA, Boyer L, et al. Saccharomyces boulardii ame- mechanism of action? Gastroenterology. 2006;131:2009-2012.
liorates Citrobacter rodentium–induced colitis through actions on 32. Artis D. Epithelial-cell recognition of commensal bacteria and
bacterial virulence factors. Am J Physiol. 2008;294:G295-G306. maintenance of immune homeostatis in the gut. Nat Rev Immunol.
13. Gedek BR. Adherence of Escherichia coli serogroup O157 and the 2008;8:411-420.
Salmonella typhimurium mutant DT104 to the surface of 33. Neish AS, Gewirtz AT, Zeng H, et al. Prokaryotic regulation of
Saccharomyces boulardii. Mycoses. 1999;42:261-264. epithelial responses by inhibition of IκB-α ubiquitination. Science.
14. Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA. 2000;289:1560-1563.
Probiotics inhibit enteropathogenic E. coli adherence in vitro by 34. Dalmasso G, Cottrez F, Imbert V, et al. Saccharomyces boulardii
inducing intestinal mucin gene expression. Am J Physiol. 1999;276: inhibits inflammatory bowel disease by trapping T cells in mesen-
G941-G950. teric lymph nodes. Gastroenterology. 2006;131:1812-1825.
15. Caballero-Franco C, Keller K, De Simone C, Chadee K. The VSL#3 35. Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M.
probiotic formula induces mucin gene expression and secretion in Probiotics ameliorate recurrent Th1-mediated murine colitis by
colonic epithelial cells. Am J Physiol. 2007;292:G315-G322. inducing IL-10 and IL-10-dependent TGF-β-bearing regulatory
16. Mack DR, Ahrne S, Hyde L, Wei S, Hollingsworth MA. Extracellular cells. J Immunol. 2005;174:3237-3246.
MUC3 mucin secretion follows adherence of Lactobacillus strains 36. Huibregste IL, Snoeck V, De Creus A, et al. Induction of ovalbu-
to intestinal epithelial cells in vitro. Gut. 2003;52:827-833. min-specific tolerance by oral administration of Lactococcus lactis
17. Linden SK, Sutton P, Karlsson NG, Korolik V, McGuckin MA. secreting ovalbumin. Gastroenterology. 2007;133:517-528.
Mucins in the mucosal barrier to infection. Nat Mucosal Immunol. 37. Foligne B, Nutten S, Grangette C, et al. Correlation between in
2008;1:183-197. vitro and in vivo immunomodulatory properties of lactic acid bac-
18. Clyne M, Dillon P, Daly S, et al. Helicobacter pylori interacts with teria. World J Gastroenterol. 2007;13:236-243.
the human single-domain trefoil protein TFF1. Proc Natl Acad Sci 38. O’Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and
USA. 2004;101:7409-7414. Bifidobacterium in irritable bowel syndrome: symptom responses
19. Wehkamp J, Harder J, Wehkamp K, et al. NF-κB and AP-1- and relationship to cytokine profiles. Gastroenterology. 2005;128:
mediated induction of human beta defensin-2 in intestinal 541-551.
epithelial cells by Escherichia coli Nissle 1917: a novel effect of a 39. Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus
probiotic bacterium. Infect Immun. 2004;72:5750-5758. modulates intestinal pain and induces opioid and cannabinioid
20. Schlee M, Harder J, Koten B, Stange EF, Wehkamp J, Fellermann receptors. Nat Med. 2007;13:35-37.
K. Probiotic lactobacilli and VSL#3 induce enterocyte β-defensin 40. Besselink MGH, van Santvoor HC, Buskens E, et al. Probiotic pro-
2. Clin Exp Immunol. 2008;151:4528-4535. phylaxis in predicted severe acute pancreatitis: a randomized, double-
21. Sherman PM, Johnson-Henry KC, Yeung HP, Ngo PSC, Goulet J, blind, placebo-controlled trial. Lancet. 2008;371:651-659.
Tompkins TA. Probiotics reduce enterohemorrhagic Escherichia 41. Boyle RJ, Robins-Browne RM, Tang MLK. Probiotic use in clinical
coli O157:H7- and enteropathogenic E. coli O127:H7-induced practice: what are the risks? Am J Clin Nutr. 2006;83:1256-1264.

Downloaded from ncp.sagepub.com at Scientific library of Moscow State University on January 6, 2014

You might also like