probiotics,nuclearreceptorsignaling,and anti … · 2019. 7. 31. · the mechanism of action for...

17
Hindawi Publishing Corporation Gastroenterology Research and Practice Volume 2011, Article ID 971938, 16 pages doi:10.1155/2011/971938 Review Article Probiotics, Nuclear Receptor Signaling, and Anti-Inflammatory Pathways Sonia S. Yoon 1 and Jun Sun 1, 2 1 Division of Gastroenterology and Hepatology, Department of Medicine, Strong Memorial Hospital, University of Rochester Mediacal Center, University of Rochester, Rochester, NY 14642, USA 2 Department of Microbiology and Immunology, University of Rochester, P.O. Box 646, 601 Elmwood Avenue, Rochester, NY 14642, USA Correspondence should be addressed to Jun Sun, jun [email protected] Received 2 February 2011; Revised 28 March 2011; Accepted 19 May 2011 Academic Editor: Genevieve B. Melton-Meaux Copyright © 2011 S. S. Yoon and J. Sun. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. There is increased investigation of the human microbiome as it relates to health and disease. Dysbiosis is implicated in various clinical conditions including inflammatory bowel disease (IBD). Probiotics have been explored as a potential treatment for IBD and other diseases. The mechanism of action for probiotics has yet to be fully elucidated. This paper discusses novel mechanisms of action for probiotics involving anti-inflammatory signaling pathways. We highlight recent progress in probiotics and nuclear receptor signaling, such as peroxisome-proliferator-activated receptor gamma (PPARγ) and vitamin D receptor (VDR). We also discuss future areas of investigation. 1. Introduction Probiotics are ingestible microorganisms with health bene- fits. Increased interest in the intestinal microbiome and its eect on health and disease is evidenced by the concomi- tant increase in peer-reviewed clinical trials investigating probiotics as therapy since 1999 [1]. Studies of the various signaling pathways involved in the response to bacteria and inflammation have led to a more detailed understanding of mechanisms and actions of probiotics. This paper discusses progress in understanding how probiotics contribute to intestinal mucosal function, particularly in relation to anti- inflammatory signaling pathways. 2. Intestinal Microflora The intestinal microflora, as a whole, serves important functions in metabolism, intestinal epithelial cell function and health, immunity, and inflammatory signaling [2, 3]. Recently, there has been increasing interest in the role of the intestinal microflora and its total genetic composition, together referred to as the microbiome in the development, maintenance, and perpetuation of various clinical condi- tions, both intestinal and extraintestinal. Dysbiosis has been implicated in various clinical condi- tions including atopy, irritable bowel syndrome (IBS), col- orectal cancer, alcoholic liver disease in animal and human studies, obesity and other metabolic disorders, and chronic inflammatory diseases such as IBD [411]. Decreased diversity of the intestinal microbiota was seen in fecal samples obtained from children who subsequently developed allergic disease [6, 7]. Altered microbiota composition in colon cancer patients when compared to patients with normal colonoscopies and in patients with IBS compared to unaected patients has also been demonstrated [5, 9]. Alcohol feeding resulted in enteric bacterial overgrowth in a mouse model [8]. The role of the microbiota in obesity has been extensively studied and carefully reviewed in the literature [12, 13]. Microbial composition in IBD patients with ulcerative colitis (UC) or Crohn’s disease (CD) as com- pared to unaected individuals has been studied and shows decreased diversity [4, 1419]. This altered microflora may have significant implications for the intestinal milieu, with as yet incompletely understood eects. The pathogenesis of IBD

Upload: others

Post on 26-Jan-2021

0 views

Category:

Documents


0 download

TRANSCRIPT

  • Hindawi Publishing CorporationGastroenterology Research and PracticeVolume 2011, Article ID 971938, 16 pagesdoi:10.1155/2011/971938

    Review Article

    Probiotics, Nuclear Receptor Signaling, andAnti-Inflammatory Pathways

    Sonia S. Yoon1 and Jun Sun1, 2

    1 Division of Gastroenterology and Hepatology, Department of Medicine, Strong Memorial Hospital,University of Rochester Mediacal Center, University of Rochester, Rochester, NY 14642, USA

    2 Department of Microbiology and Immunology, University of Rochester, P.O. Box 646,601 Elmwood Avenue, Rochester, NY 14642, USA

    Correspondence should be addressed to Jun Sun, jun [email protected]

    Received 2 February 2011; Revised 28 March 2011; Accepted 19 May 2011

    Academic Editor: Genevieve B. Melton-Meaux

    Copyright © 2011 S. S. Yoon and J. Sun. This is an open access article distributed under the Creative Commons AttributionLicense, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properlycited.

    There is increased investigation of the human microbiome as it relates to health and disease. Dysbiosis is implicated in variousclinical conditions including inflammatory bowel disease (IBD). Probiotics have been explored as a potential treatment for IBDand other diseases. The mechanism of action for probiotics has yet to be fully elucidated. This paper discusses novel mechanismsof action for probiotics involving anti-inflammatory signaling pathways. We highlight recent progress in probiotics and nuclearreceptor signaling, such as peroxisome-proliferator-activated receptor gamma (PPARγ) and vitamin D receptor (VDR). We alsodiscuss future areas of investigation.

    1. Introduction

    Probiotics are ingestible microorganisms with health bene-fits. Increased interest in the intestinal microbiome and itseffect on health and disease is evidenced by the concomi-tant increase in peer-reviewed clinical trials investigatingprobiotics as therapy since 1999 [1]. Studies of the varioussignaling pathways involved in the response to bacteria andinflammation have led to a more detailed understanding ofmechanisms and actions of probiotics. This paper discussesprogress in understanding how probiotics contribute tointestinal mucosal function, particularly in relation to anti-inflammatory signaling pathways.

    2. Intestinal Microflora

    The intestinal microflora, as a whole, serves importantfunctions in metabolism, intestinal epithelial cell functionand health, immunity, and inflammatory signaling [2, 3].Recently, there has been increasing interest in the role ofthe intestinal microflora and its total genetic composition,together referred to as the microbiome in the development,

    maintenance, and perpetuation of various clinical condi-tions, both intestinal and extraintestinal.

    Dysbiosis has been implicated in various clinical condi-tions including atopy, irritable bowel syndrome (IBS), col-orectal cancer, alcoholic liver disease in animal and humanstudies, obesity and other metabolic disorders, and chronicinflammatory diseases such as IBD [4–11]. Decreaseddiversity of the intestinal microbiota was seen in fecalsamples obtained from children who subsequently developedallergic disease [6, 7]. Altered microbiota composition incolon cancer patients when compared to patients withnormal colonoscopies and in patients with IBS comparedto unaffected patients has also been demonstrated [5, 9].Alcohol feeding resulted in enteric bacterial overgrowth ina mouse model [8]. The role of the microbiota in obesityhas been extensively studied and carefully reviewed in theliterature [12, 13]. Microbial composition in IBD patientswith ulcerative colitis (UC) or Crohn’s disease (CD) as com-pared to unaffected individuals has been studied and showsdecreased diversity [4, 14–19]. This altered microflora mayhave significant implications for the intestinal milieu, with asyet incompletely understood effects. The pathogenesis of IBD

  • 2 Gastroenterology Research and Practice

    likely involves a combination of factors including intestinaldysbiosis in conjunction with environmental factors in agenetically susceptible host [20].

    Based on the concept of a dysregulated or dysfunctionalmicrobiota in disease, various methods to attenuate theeffects of an altered microbiome have been attempted.

    3. Probiotics

    “Probiotics” were first described in the literature by Lillyand Stillwell in 1965 as growth-promoting factors producedby certain microorganisms [21] although it may have beendescribed as early as 1908 [22]. Recently, probiotics weredefined as “live organisms which, when consumed in ade-quate amounts as part of food, confer a health benefit on thehost” (Report of a Joint FAO/WHO Expert Consultation onEvaluation of Health and Nutritional Properties of Probioticsin Food Including Powder Milk with Live Lactic AcidBacteria (October 2001), “Health and Nutritional Propertiesof Probiotics in Food Including Powder Milk with LiveLactic Acid Bacteria”, Food and Agriculture Organizationof the United Nations, World Health Organization). Themechanisms of action of probiotics include immune modu-lation, direct effect on commensal and pathogenic bacteria toinhibit infection and restore homeostasis, and modificationof pathogenic toxins and host products [23]. The efficacy ofprobiotics in various clinical conditions both in the pediatricand adult patient population has been extensively studiedand carefully reviewed [1, 19, 24–32].

    Rectal infusion of normal stool via enemas to treatpseudomembranous colitis has been described as early as1958 [70]. Infusion of stool via nasogastric tube to thesmall intestine or via colonoscopy to the colon for CDADhas also been described and shows high response rates[71–74]. A recent study showed that fecal bacteriotherapywas effective in relief of clinical symptoms in a patientwith recurrent CDAD and that this was accompanied bythe repopulation of the diseased intestinal microbiota withbeneficial species that were diminished pretreatment [75].Other methods to supply live, nonpathogenic organisms tothe intestinal microbiota in AAD and CDAD include orallyadministered probiotics. The efficacy of various probioticformulations in AAD and CDAD has been extensivelystudied and carefully reviewed [1]. A recent study showedthat the probiotics Lactobacillus acidophilus and Lactobacilluscasei were well tolerated and effective in reducing therisk of the development of AAD and CDAD [76]. Theutility of the probiotic yeast Saccharomyces boulardii for avariety of conditions including traveler’s diarrhea, enteralnutrition-associated diarrhea, AAD, and CDAD has beeninvestigated, and according to a recent meta-analysis, strongevidence exists for advocating its use in traveler’s diarrheaand AAD [77]. Recent trials using Bifidobacterium bifidumand Saccharomyces boulardii demonstrated improvement inclinical IBS symptoms and quality of life [78, 79], and severalreviews of the evidence for the utility of probiotics in IBShave been published [80–82].

    For IBD therapy, treatment with different strains ofprobiotics has shown varied results. Small trials have shown

    promise for probiotic use in the induction and maintenanceof remission in UC. VSL#3 has been shown to be safe andeffective in the treatment of acute mild to moderately activeUC [83]. Patients with mild to moderate UC unresponsive toconventional therapy achieved a combined induction remis-sion/response rate of 77% with treatment with VSL#3 [84].E. coli Nissle 1917 was found to be effective and equivalent tomesalazine in maintaining remission in UC [85]. In anotherstudy, Lactobacillus rhamnosus GG (LGG) was equivalent tomesalazine in the maintenance of remission in UC, however,appeared to be more effective in prolonging the relapse-free time [86]. Evidence also exists for the role of probioticsin prophylaxis of pouchitis after surgery in UC patientsas well as induction of remission in chronic pouchitis[87, 88].

    Studies of probiotic use in induction and maintenanceof remission and prevention of postoperative recurrencein CD have been less consistent than those for UC. Asmall study of LGG for the prevention of recurrence aftersurgery in CD did not show any improvement over placebo[89]; however, Saccharomyces boulardii appears useful inmaintaining remission in CD [90, 91]. The progress in theuse of probiotics for IBD has been carefully reviewed [92, 93];however, there remains a relative lack of well-designed, large,randomized, placebo-controlled trials.

    Several barriers exist to advocating broad use of probi-otics in clinical practice, not least of which is the considerableheterogeneity in the experimental designs with respect tospecies and strains of probiotics and the various animalmodels utilized [94]. Although clinical trials examining therole of probiotics in the treatment and/or prevention of AAD,CDAD, IBD including UC, CD, and pouchitis, necrotizingenterocolitis, infectious gastroenteritis, radiation-inducedenteritis, and colitis, IBS and various atopic diseases havebeen reported [1, 24, 25, 28, 29, 31, 87, 95–97]; in manycases, results have been inconsistent, and large, well-designedtrials are lacking. An additional complicating factor pertainsto issues of quality control. Determining whether a commer-cially available probiotic actually contains the live organismsit purports to contain and determining if there is rationalselection of component probiotic strains in “cocktails” areissues that must be considered [22]. Future research to refinetechniques to accurately identify “normal” and “diseased”microbiota and to further elucidate the specific effects andmechanisms of actions of individual probiotic strains will aidin optimizing therapeutic efficacy.

    4. Mechanisms for Probiotics inAnti-Inflammation

    There has been and continues to be considerable research indelineating the underlying mechanisms by which probioticsexert their beneficial effects. The mechanisms regulating thefunction of probiotics are very diverse. It is well accepted thatprobiotics use distinct cellular and molecular mechanisms,including blocking pathogenic bacterial effects, regulat-ing immune responses, and altering intestinal epithelialhomeostasis by promoting cell survival, enhancing barrierfunction, and stimulating protective responses [32].

  • Gastroenterology Research and Practice 3

    Table 1 outlines representative publications on probioticmechanisms of actions. The probiotic-host interaction iscomplex and further complicated by the fact that certainprobiotic effects appear to be species and strain specific.Different probiotics have been shown to exert both pro-inflammatory [98] and anti-inflammatory effects on den-dritic cells [99]. A recent study demonstrated that the anti-inflammatory effect of certain lactobacilli is via NOD2-mediated signaling [100]. NOD2/CARD15 is a member ofa superfamily of genes involved in intracellular bacterialrecognition and has been identified as an important suscep-tibility gene for CD [101, 102]. The authors speculate thatthe inconsistent clinical results of lactobacilli use in patientswith CD may be related to a relative deficiency of NOD2.Probiotic effect on the innate immune responsive pathwaysincluding toll-like receptor (TLR), nuclear factor kappa B(NF-κB), mitogen-activated protein kinase (MAPK), c-JunNH2-terminal kinase (JNK) has been extensively investigated(Table 1). Activation of specific TLRs also appears to bespecies specific [47, 48]. The action of E. coli Nissle 1917 onCaco-2 cells was found to be mediated by flagellin possiblyvia a TLR pathway [103]. The probiotic-induced effect on theNF-κB signaling pathway is well represented in the literatureand is generally characterized by inhibition (Table 1).

    Defective epithelial barrier function has been implicatedin IBD and can predict relapse during clinical remission[104–109]. One way by which probiotics have been shownexert their action is by stabilizing tight junctions (TJs)and enhancing barrier function of intestinal epithelial cells(Table 1).

    Abnormal STAT/suppressor of cytokine signaling(SOCS) signaling has been demonstrated in CD patients[110], and probiotics are also shown to modulate the JAK-STAT signaling in human placental trophoblast cells [111].Increasing evidence further demonstrates that metabolism,xenobiotics, and nuclear receptor signaling are involved inthe action of probiotics [67, 68].

    Induction of heat shock proteins (HSPs) and endogenousantimicrobial peptides (defensins) via activation of NF-κB,MAPK, and JNK has also been linked to probiotic action [35,41, 43]. Since defensins are implicated in the pathogenesis ofIBD, increased expression by probiotics provides a possiblemechanism for clinical efficacy seen in certain IBD patientsand deserves further study.

    5. Defensins and Nuclear Receptor Signaling

    Defensins are a class of endogenous antimicrobial peptidesinvolved in innate immunity which is highly evolutionarilyconserved and represents a primary line of defense againstvarious microbial pathogens [112–114]. Antimicrobial pep-tides are widely distributed throughout the animal and plantkingdom, and despite their evolutionary heritage, remaineffective antimicrobial agents [114]. This is due, in largepart, to their mechanism of action involving membranedisruption and pore formation, which is not easily exploitedby pathogens to confer resistance [112–115]. Importantantimicrobial peptides in humans include defensins, cathe-licidins, lysozymes, and other antimicrobial antiproteases

    [116]. There are three known defensin subfamilies; α and βdefensins are expressed mainly in immune cells and epithelialcells while the θ defensin is found mainly in immune cellsof the Rhesus macaque [117, 118]. In the gastrointestinaltract, β defensin expression is seen in multiple sites, whereasα defensin expression is largely in the small intestine[119]. In the uninflamed colon, human β defensin 1 isthe predominant defensin and human β defensin 2 and 3are induced with inflammation or infection [120]. In micelacking functional cryptidins (murine α defensins), increasedsurvival and virulence of orally administered bacteria wereseen and intestinal peptide preparations had decreasedantimicrobial activity [121].

    The possible role of a deficiency in defensins in thepathogenesis of IBD has been proposed [116, 122]. ThePaneth cells of the small intestine are the major source ofendogenous antimicrobials, including α defensins [102]. Inaddition, The Paneth cells have been shown to express NOD2[123]. In patients with ileal CD, human α defensin 5 and6 production is reduced, and this effect is magnified inthose patients with a concomitant NOD2 mutation [124].For β defensins, CD patients with colonic disease exhibitnormal levels of β defensin 2 and 3 whereas UC patientshave increased levels, suggesting a role of failure of β defensininduction in the pathogenesis of CD [125]. Constitutivehuman β defensin 1 expression is reduced in CD patientswith colonic involvement independent of inflammation,and recently, the maintenance of constitutive β defensinexpression was shown to be activated by the nuclear receptorperoxisome-proliferator-activated receptor gamma (PPARγ)[122].

    Further contributing to the effect of a defensin defi-ciency in the pathogenesis of IBD may be the diminisheddiversity of the intestinal microbiota seen in IBD patients.The interaction of commensal bacteria with antimicrobialpeptide synthesis is not well understood; however, it hasbeen suggested that commensal bacteria provide chronicstimulation of epithelial cells to produce antimicrobialpeptides at levels sufficient to kill microbial pathogens [114,126].

    Probiotics, but not fecal isolates, have been shown toinduce human β defensin 2 in intestinal epithelial cells [41,42]. Wehkamp et al. and Schlee et al. have reported thatNF-κB and activator protein-1 (AP-1) mediate inductionof human β defensin 2 in intestinal epithelial cells by theprobiotic E. coli Nissle 1917 and VSL#3 [41, 42].

    Interestingly, nuclear receptors are known to regulate theexpressions of defensins [122, 127]. Nuclear receptors rep-resent a class of intracellular transcription factors activatedby ligands which can directly interact with DNA; as a result,nuclear receptors play significant roles in the regulation ofmetabolic, reproductive, developmental, and immune pro-cesses [128–131]. Nuclear receptors regulate transcriptionalactivity by several distinct mechanisms, including “ligand-dependent transactivation, ligand-independent repression,and ligand-dependent transrepression” although the rangeof transcriptional activities of each nuclear receptor variesand even the transcriptional effects of a single nuclearreceptor may be cell specific [132]. A detailed discussion

  • 4 Gastroenterology Research and Practice

    Ta

    ble

    1:Su

    mm

    ary

    for

    mol

    ecu

    lar

    mec

    han

    ism

    san

    dpr

    obio

    tics

    .

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    NF-κB

    (i)

    L.ca

    seiS

    hir

    ota

    (LcS

    )(i

    )T

    HP-

    1(i

    )R

    at(i

    )L-

    lact

    icac

    idan

    dL

    cScu

    ltu

    resu

    pern

    atan

    tin

    hib

    ited

    NF-κB

    acti

    vati

    on,

    TN

    F-α

    mR

    NA

    expr

    essi

    onin

    crea

    se,a

    nd

    TN

    F-α

    prot

    ein

    secr

    etio

    nin

    cells

    trea

    ted

    wit

    hlip

    opol

    ysac

    char

    ide

    (LP

    S)[3

    3]

    NF-κB

    (i)

    LGG

    (i)

    CaC

    o-2

    (i)

    LGG

    redu

    ced

    TN

    F-α

    -in

    duce

    dN

    F-κB

    tran

    sloc

    atio

    n,l

    esse

    ned

    decr

    ease

    inIκ

    B,r

    edu

    ced

    TN

    Fα-i

    ndu

    ced

    inte

    rleu

    kin

    (IL)

    -8pr

    odu

    ctio

    n[3

    4]

    NF-κB

    (i)

    VSL

    3#(i

    )YA

    MC

    (i)

    VSL

    #3in

    hib

    ited

    the

    proi

    nfl

    amm

    ator

    yN

    F-κB

    path

    way

    and

    indu

    ced

    the

    expr

    essi

    onof

    cyto

    prot

    ecti

    veh

    eat

    shoc

    kpr

    otei

    ns

    (HSP

    s)in

    inte

    stin

    alep

    ith

    elia

    lce

    lls[3

    5]

    NF-κB

    (i)

    L.pl

    anta

    rum

    (LP

    )(i

    )YA

    MC

    ,RA

    W26

    4.7

    (ii)

    Mu

    rin

    ede

    ndr

    itic

    cells

    (i)

    LP-c

    ondi

    tion

    edm

    edia

    (CM

    )in

    hib

    ited

    the

    chym

    otry

    psin

    -lik

    eac

    tivi

    tyof

    the

    prot

    easo

    me,

    NF-κB

    bin

    din

    gac

    tivi

    tyas

    wel

    las

    the

    degr

    adat

    ion

    ofIκ

    [36]

    NF-κB

    (i)

    LP-L

    2(i

    )C

    aCo-

    2(i

    )C

    aco-

    2ce

    llspr

    ein

    cuba

    ted

    wit

    hLP

    -L2

    show

    edat

    ten

    uat

    ion

    ofm

    onoc

    yte

    chem

    otac

    tic

    prot

    ein

    -1(M

    CP-

    1)pr

    otei

    npr

    odu

    ctio

    nan

    dm

    RN

    Aex

    pres

    sion

    and

    also

    prev

    ente

    dIκ

    degr

    adat

    ion

    inT

    NF-α

    -sti

    mu

    late

    dC

    aco-

    2ce

    lls[3

    7]

    NF-κB

    (i)

    L.ca

    sei(

    Lc)

    (i)

    HE

    K-2

    93T

    (ii)

    CaC

    o-2

    (i)

    Lcdo

    wn

    regu

    late

    dth

    etr

    ansc

    ript

    ion

    ofge

    nes

    enco

    din

    gpr

    oin

    flam

    mat

    ory

    effec

    tors

    and

    adh

    eren

    cem

    olec

    ule

    sin

    duce

    dby

    inva

    sive

    S.fle

    xner

    ires

    ult

    ing

    inan

    ti-i

    nfl

    amm

    atio

    nm

    edia

    ted

    byth

    ein

    hib

    itio

    nof

    the

    NF-κB

    path

    way

    [38]

    NF-κB

    (i)

    B.l

    acti

    s(i

    )M

    ode-

    K(i

    i)M

    EF

    (i)

    B.l

    acti

    sac

    tiva

    tes

    NF-κB

    Rel

    Aan

    dp3

    8M

    AP

    Kph

    osph

    oryl

    atio

    nin

    IEC

    lines

    [39]

    NF-κB

    MA

    PK

    (ER

    K1/

    2/p3

    8/JN

    K)

    (i)

    S.bo

    ular

    dii(

    Sb)

    (i)

    T84

    (i)

    NIH

    Mic

    e

    (i)

    Invi

    vo,S

    bpr

    otec

    ted

    mic

    efr

    omSa

    lmon

    ella

    ente

    rica

    sero

    var-

    Typh

    imu

    riu

    m-(

    ST-)

    indu

    ced

    deat

    han

    dpr

    even

    ted

    hep

    atic

    bact

    eria

    ltr

    ansl

    ocat

    ion

    (ii)

    InT

    84hu

    man

    colo

    rect

    alca

    nce

    rce

    lls,S

    bin

    cuba

    tion

    abol

    ish

    edSa

    lmon

    ella

    inva

    sion

    ,pre

    serv

    edba

    rrie

    rfu

    nct

    ion

    ,an

    dde

    crea

    sed

    ST-i

    ndu

    ced

    IL-8

    syn

    thes

    is(i

    ii)

    Sbh

    adan

    inh

    ibit

    ory

    effec

    ton

    ST-i

    ndu

    ced

    acti

    vati

    onof

    the

    MA

    PK

    s,ex

    trac

    ellu

    lar

    regu

    late

    dki

    nas

    e(E

    RK

    )1/

    2,p3

    8an

    dJN

    K,a

    nd

    NF-κB

    [40]

    NF-κB

    MA

    PK

    (p38

    /ER

    K)

    SAP

    K/J

    NK

    (i)

    E.c

    oliN

    issl

    e19

    17(E

    CN

    )(i

    i)L.

    ferm

    entu

    m(i

    ii)

    L.ac

    idop

    hilu

    s(L

    a)(i

    v)Pe

    dioc

    occu

    spe

    ntos

    aceu

    s(v

    )L.

    para

    case

    i(v

    i)V

    SL#3

    (i)

    CaC

    o-2

    (i)

    CaC

    o-2

    cells

    trea

    ted

    wit

    hpr

    obio

    tic

    had

    pea

    kof

    hum

    anβ

    defe

    nsi

    ns-

    2(h

    BD

    -2)

    mR

    NA

    expr

    essi

    onat

    6h

    incu

    bati

    on(i

    i)P

    rom

    oter

    acti

    vati

    onvi

    apr

    obio

    tics

    was

    abol

    ish

    edw

    ith

    the

    dele

    tion

    ofN

    F-κB

    -an

    dac

    tiva

    tor-

    prot

    ein

    -1-

    (AP-

    1-)

    bin

    din

    gsi

    tes

    onth

    eh

    BD

    -2pr

    omot

    er(i

    ii)

    Indu

    ctio

    nof

    hB

    D-2

    depe

    nds

    onM

    AP

    K,E

    RK

    1/2,

    p38,

    and

    c-Ju

    nN

    -ter

    min

    alki

    nas

    e(J

    NK

    ),to

    vary

    ing

    degr

    ees

    [41]

  • Gastroenterology Research and Practice 5

    Ta

    ble

    1:C

    onti

    nu

    ed.

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    NF-κB

    SAP

    K/J

    NK

    (i)

    EC

    N(i

    i)V

    ario

    us

    E.c

    olis

    trai

    ns(i

    ii)

    Var

    iou

    sLa

    ctob

    acill

    ist

    rain

    s

    (i)

    CaC

    o-2

    (i)

    Act

    ive

    and

    hea

    t-in

    acti

    vate

    dE

    CN

    and

    seve

    ralo

    ther

    prob

    ioti

    cba

    cter

    iapo

    ten

    tly

    indu

    ced

    hB

    D-2

    inin

    test

    inal

    epit

    hel

    ialc

    ells

    (ii)

    hB

    D-2

    prom

    oter

    acti

    vati

    onw

    asab

    olis

    hed

    bym

    uta

    tion

    ofth

    etw

    oN

    F-kB

    site

    sin

    the

    hB

    D-2

    prom

    oter

    upo

    ntr

    eatm

    ent

    wit

    hE

    CN

    (iii

    )E

    CN

    -in

    duct

    edac

    tiva

    tion

    ofA

    P-1

    may

    bere

    gula

    ted

    byJN

    Kki

    nas

    epa

    thw

    ay

    [42]

    SAP

    K/J

    NK

    MA

    PK

    (p38

    )(i

    )LG

    G(i

    )YA

    MC

    (i)

    LGG

    -CM

    indu

    ced

    Hsp

    25an

    dH

    sp72

    inti

    me-

    and

    con

    cen

    trat

    ion

    -dep

    ende

    nt

    man

    ner

    (ii)

    LGG

    -CM

    -in

    duce

    dH

    SP72

    indu

    ctio

    nw

    asbl

    ocke

    dby

    the

    Inh

    ibit

    ors

    ofp3

    8an

    dJN

    K

    [43]

    Epi

    derm

    algr

    owth

    fact

    orre

    cept

    or(E

    GFR

    )(i

    )Sb

    (i)

    HT

    -29

    (i)

    AP

    Cm

    in

    (i)

    Upo

    nex

    posu

    reto

    Sb,H

    ER

    -2,H

    ER

    -3,i

    nsu

    lin-l

    ike

    grow

    thfa

    ctor

    -1re

    cept

    or,a

    nd

    EG

    FRw

    ere

    inac

    tiva

    ted

    (ii)

    InH

    T-2

    9ce

    lls,S

    bpr

    omot

    edap

    opto

    sis,

    prev

    ente

    dE

    GF-

    indu

    ced

    prol

    ifer

    atio

    n,a

    nd

    redu

    ced

    cell

    colo

    nyfo

    rmat

    ion

    (iii

    )Sb

    decr

    ease

    din

    test

    inal

    tum

    orgr

    owth

    and

    dysp

    lasi

    ain

    C57

    BL/

    6JM

    in/+

    (Apc

    (Min

    ))m

    ice

    [44]

    TL

    RN

    F-κB

    MA

    PK

    (p38

    )

    (i)

    LGG

    (ii)

    B.l

    ongu

    m(i

    )H

    T-2

    9(i

    i)T

    84

    (i)

    Com

    men

    sal-

    orig

    inD

    NA

    enh

    ance

    dex

    pres

    sion

    ofT

    LR9

    inH

    T-2

    9an

    dT

    84ce

    lls(i

    i)T

    his

    was

    asso

    ciat

    edw

    ith

    atte

    nu

    atio

    nof

    TN

    F-α

    -in

    duce

    dN

    F-κB

    acti

    vati

    onby

    redu

    cin

    gIκ

    degr

    adat

    ion

    and

    p38

    phos

    phor

    ylat

    ion

    (iii

    )LG

    GD

    NA

    decr

    ease

    dth

    eT

    NF-α

    -in

    duce

    dre

    duct

    ion

    intr

    anse

    pith

    elia

    lel

    ectr

    ical

    resi

    stan

    ce(T

    ER

    )

    [45]

    TL

    R(i

    )LP

    BFE

    1685

    (ii)

    LGG

    (i)

    HT

    -29

    (i)

    HT

    29ce

    llsin

    cuba

    ted

    wit

    hla

    ctob

    acill

    ish

    owed

    upr

    egu

    lati

    onof

    TL

    R2

    and

    TL

    R9

    tran

    scri

    ptio

    nle

    vels

    (ii)

    Pro

    tein

    expr

    essi

    onle

    vels

    ofT

    LR2

    and

    TLR

    5w

    ere

    enh

    ance

    d[4

    6]

    TLR

    (i)

    VSL

    #3(i

    )M

    ice

    (i)

    Intr

    agas

    tric

    adm

    inis

    trat

    ion

    ofga

    mm

    a-ir

    radi

    ated

    prob

    ioti

    cssi

    gnifi

    can

    tly

    amel

    iora

    ted

    the

    seve

    rity

    ofD

    SS-i

    ndu

    ced

    colit

    isin

    TLR

    2-an

    dT

    LR4-

    defi

    cien

    tm

    ice

    but

    not

    inT

    LR9-

    defi

    cien

    tm

    ice

    [47]

    TL

    R(i

    )E

    CN

    (i)

    Wild

    -typ

    e(W

    T),

    TL

    R2-

    /TL

    R4-

    knoc

    kou

    tm

    ice

    (i)

    EC

    Nde

    crea

    sed

    colit

    isan

    dpr

    oin

    flam

    mat

    ory

    cyto

    kin

    ese

    cret

    ion

    inW

    Tbu

    tn

    otT

    LR2-

    orT

    LR4-

    knoc

    kou

    tm

    ice

    (ii)

    EC

    Nre

    sult

    edin

    redu

    ctio

    nof

    inte

    rfer

    on(I

    FN)-γ

    secr

    etio

    nin

    TL

    R2

    knoc

    kou

    t(i

    ii)

    Cyt

    okin

    ese

    cret

    ion

    was

    alm

    ost

    un

    dete

    ctab

    lean

    dn

    otm

    odu

    late

    dby

    EC

    Nin

    TLR

    -4-k

    noc

    kou

    tm

    ice

    (iv)

    Incr

    ease

    dT

    LR2

    and

    TLR

    4pr

    otei

    nex

    pres

    sion

    and

    NF-κB

    acti

    vity

    via

    TL

    R2

    and

    TL

    R4

    was

    seen

    wit

    hE

    CN

    and

    hum

    anT

    -cel

    lcoc

    ult

    ure

    [48]

  • 6 Gastroenterology Research and Practice

    Ta

    ble

    1:C

    onti

    nu

    ed.

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    MA

    PK

    (p38

    /ER

    K)

    (i)

    LP(i

    i)L

    c(i

    )M

    ice

    per

    iton

    eal

    mac

    roph

    ages

    (i)

    LPst

    ron

    gly

    indu

    ced

    IL-1

    0an

    dw

    eakl

    yin

    duce

    dIL

    -12;

    Lcst

    ron

    gly

    indu

    ced

    IL-1

    2an

    dw

    eakl

    yin

    duce

    dIL

    -10

    (ii)

    LP,c

    ompa

    red

    toLc

    ,dem

    onst

    rate

    dm

    ore

    rapi

    dan

    dst

    ron

    gac

    tiva

    tion

    ofM

    AP

    Ks,

    espe

    cial

    lyof

    ER

    K(i

    ii)

    Blo

    ckin

    gL

    P-in

    duce

    dE

    RK

    acti

    vati

    onre

    sult

    edin

    decr

    ease

    dIL

    -10

    prod

    uct

    ion

    and

    incr

    ease

    dIL

    -12

    prod

    uct

    ion

    (iv)

    Com

    bin

    edst

    imu

    lati

    onw

    ith

    LPan

    dL

    cre

    sult

    edin

    syn

    ergi

    stic

    indu

    ctio

    nof

    IL-1

    0pr

    odu

    ctio

    n;t

    his

    was

    trig

    gere

    dby

    the

    key

    fact

    ors:

    cell

    wal

    ltei

    choi

    cac

    idan

    dlip

    otei

    choi

    cac

    ids

    (v)

    Teic

    hoi

    c-ac

    id-i

    ndu

    ced

    IL-1

    0pr

    odu

    ctio

    nw

    asm

    edia

    ted

    byT

    LR2-

    depe

    nde

    nt

    ER

    Kac

    tiva

    tion

    [49]

    MA

    PK

    (p38

    /ER

    K)

    Tig

    ht

    jun

    ctio

    ns

    (TJ)

    (i)

    LGG

    (i)

    CaC

    o-2

    (i)

    LGG

    -pro

    duce

    dso

    lubl

    epr

    otei

    ns

    (p40

    and

    p75)

    dim

    inis

    hed

    the

    hydr

    ogen

    -per

    oxid

    e-in

    duce

    dde

    crea

    sein

    TE

    Ran

    din

    crea

    sein

    inu

    linpe

    rmea

    bilit

    yan

    din

    duce

    din

    crea

    sein

    mem

    bran

    etr

    ansl

    ocat

    ion

    ofpr

    otei

    nki

    nas

    eC

    (PK

    C)

    beta

    I,P

    KC

    epsi

    lon

    ,an

    dle

    velo

    fph

    osph

    o-E

    RK

    1/2

    inth

    ede

    terg

    ent-

    inso

    lubl

    efr

    acti

    ons

    (ii)

    LGG

    -pro

    duce

    dso

    lubl

    epr

    otei

    ns

    (p40

    and

    p75)

    prev

    ente

    dhy

    drog

    en-p

    erox

    ide-

    indu

    ced

    redi

    stri

    buti

    onof

    occl

    udi

    n,z

    onu

    laoc

    clu

    den

    s(Z

    O)-

    1,E

    -cad

    her

    in,a

    nd

    beta

    -cat

    enin

    from

    inte

    rcel

    lula

    rju

    nct

    ion

    san

    dth

    eir

    diss

    ocia

    tion

    from

    the

    dete

    rgen

    t-in

    solu

    ble

    frac

    tion

    s(i

    ii)

    p40-

    and

    p75-

    med

    iate

    dre

    duct

    ion

    ofhy

    drog

    en-p

    erox

    ide-

    indu

    ced

    tigh

    tju

    nct

    ion

    disr

    upt

    ion

    and

    inu

    linp

    erm

    eabi

    lity

    was

    atte

    nu

    ated

    byU

    0126

    (aM

    AP

    kin

    ase

    inh

    ibit

    or)

    [50]

    MA

    PK

    (p38

    /ER

    K)

    TJ

    (i)

    B.i

    nfan

    tis

    (i)

    T84

    (i)

    IL-1

    0,IL

    -1de

    fici

    ent

    mic

    e

    (i)

    B.i

    nfan

    tis-

    con

    diti

    oned

    med

    ium

    (BiC

    M)

    incr

    ease

    dT

    ER

    ,ZO

    -1,a

    nd

    occl

    udi

    nex

    pres

    sion

    and

    decr

    ease

    dcl

    audi

    n-2

    expr

    essi

    on;t

    his

    was

    asso

    ciat

    edw

    ith

    incr

    ease

    dph

    osph

    o-E

    RK

    and

    decr

    ease

    dph

    osph

    o-p3

    8(i

    i)T

    NF-α

    -an

    dIF

    N-γ

    -in

    duce

    ddr

    ops

    inT

    ER

    and

    rear

    ran

    gem

    ent

    ofT

    Jpr

    otei

    ns

    wer

    epr

    even

    ted

    byB

    iCM

    (iii

    )In

    hib

    itio

    nof

    ER

    Kat

    ten

    uat

    edth

    epr

    otec

    tion

    from

    TN

    F-α

    and

    IFN

    -γan

    dpr

    even

    ted

    BiC

    M-i

    ndu

    ced

    incr

    ease

    inT

    ER

    (iv)

    Invi

    vo,o

    ralB

    iCM

    redu

    ced

    colo

    nic

    perm

    eabi

    lity,

    inIL

    -10-

    defi

    cien

    tm

    ice,

    lon

    g-te

    rmB

    iCM

    decr

    ease

    dco

    lon

    ican

    dsp

    len

    icIF

    N-γ

    secr

    etio

    n,a

    tten

    uat

    edin

    flam

    mat

    ion

    ,an

    dn

    orm

    aliz

    edco

    lon

    icpe

    rmea

    bilit

    y

    [51]

    TJ

    (i)

    LPM

    B45

    2(i

    )C

    aCo-

    2

    (i)

    LPM

    B45

    2in

    crea

    sed

    TE

    Rac

    ross

    Cac

    o-2

    cell

    mon

    olay

    ers

    indo

    se-d

    epen

    den

    tm

    ann

    er(i

    i)A

    lter

    edex

    pres

    sion

    ofse

    vera

    ltig

    ht-

    jun

    ctio

    n-r

    elat

    edge

    nes

    (in

    clu

    din

    goc

    clu

    din

    and

    asso

    ciat

    edpl

    aqu

    epr

    otei

    ns)

    was

    seen

    inre

    spon

    seto

    LPM

    B45

    2(i

    ii)LP

    MB

    452

    cau

    sed

    chan

    ges

    inge

    ne

    expr

    essi

    onle

    vels

    oftu

    bulin

    and

    prot

    easo

    me

    (iv)

    LPM

    B45

    2-tr

    eate

    dce

    llssh

    owed

    incr

    ease

    dfl

    uor

    esce

    nce

    inte

    nsi

    tyof

    the

    fou

    rti

    ght

    jun

    ctio

    npr

    otei

    ns

    wh

    enco

    mpa

    red

    tou

    ntr

    eate

    dco

    ntr

    ols

    [52]

  • Gastroenterology Research and Practice 7T

    abl

    e1:

    Con

    tin

    ued

    .

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    TJ

    PK

    C(i

    )LP

    (i)

    CaC

    o-2

    (i)

    Un

    con

    juga

    ted

    bilir

    ubi

    n(U

    CB

    )ca

    use

    dde

    crea

    sed

    PK

    Cac

    tivi

    ty,s

    erin

    eph

    osph

    oryl

    ated

    occl

    udi

    ng,

    and

    ZO

    -1le

    vels

    (ii)

    Hig

    hco

    nce

    ntr

    atio

    ns

    ofU

    CB

    cau

    sed

    cyto

    toxi

    city

    and

    decr

    ease

    dT

    ER

    (iii

    )Tr

    eatm

    ent

    wit

    hL

    Pm

    itig

    ated

    the

    effec

    tsof

    UC

    Bon

    TE

    Ran

    dap

    opto

    sis,

    prev

    ente

    dab

    erra

    nt

    expr

    essi

    onan

    dre

    arra

    nge

    men

    tof

    TJ

    prot

    ein

    s,an

    dpa

    rtia

    llyre

    stor

    edP

    KC

    acti

    vity

    and

    seri

    ne

    phos

    phor

    ylat

    edT

    Jpr

    otei

    nle

    vels

    [53]

    TJ

    (i)

    LPD

    SM26

    48(i

    )C

    aCo-

    2

    (i)

    LPD

    SM26

    48re

    duce

    dth

    ede

    lete

    riou

    seff

    ect

    ofE

    sche

    rich

    iaco

    li(e

    nte

    ropa

    thog

    enic

    E.c

    oli(

    EP

    EC

    ))O

    127:

    H6

    (E23

    48/6

    9)on

    TE

    Ran

    dad

    her

    ence

    wit

    hsi

    mu

    ltan

    eou

    sor

    prio

    rco

    cult

    ure

    com

    pare

    dw

    ith

    EP

    EC

    incu

    bati

    onal

    one

    [54]

    TJ

    TL

    R(i

    )LP

    (i)

    CaC

    o-2

    (i)

    Hu

    man

    (i)

    LPin

    duce

    dtr

    ansl

    ocat

    ion

    ofZ

    O-1

    toth

    eT

    Jre

    gion

    inan

    invi

    tro

    mod

    el,b

    ut

    the

    effec

    tson

    occl

    udi

    nw

    ere

    min

    orco

    mpa

    red

    wit

    heff

    ects

    seen

    invi

    vo(i

    i)L

    Pac

    tiva

    ted

    TL

    R2

    sign

    alin

    g,an

    dtr

    eatm

    ent

    wit

    hT

    LR

    2ag

    onis

    tPa

    m(3

    )-C

    ys-S

    K4(

    PC

    SK),

    incr

    ease

    dfl

    uor

    esce

    nt

    stai

    nin

    gof

    occl

    udi

    nin

    the

    TJ

    (iii

    )P

    hor

    bol-

    este

    r-in

    duce

    ddi

    sloc

    atio

    nof

    ZO

    -1an

    doc

    clu

    din

    and

    asso

    ciat

    edin

    crea

    sein

    epit

    hel

    ialp

    erm

    eabi

    lity

    wer

    eat

    ten

    uat

    edw

    ith

    pret

    reat

    men

    tw

    ith

    LPor

    PC

    SK

    [55]

    TJ

    (i)

    B.b

    ifidu

    m(i

    )R

    ats

    (i)

    B.b

    ifidu

    mde

    crea

    sed

    the

    inci

    den

    ceof

    nec

    roti

    zin

    gen

    tero

    colit

    is(N

    EC

    ),n

    orm

    aliz

    edIL

    -6,m

    uci

    n-3

    ,an

    dT

    ff3

    leve

    lsin

    the

    ileu

    mof

    NE

    Cra

    ts,a

    nd

    nor

    mal

    ized

    the

    expr

    essi

    onan

    dlo

    caliz

    atio

    nof

    TJ

    and

    adh

    eren

    sju

    nct

    ion

    (AJ)

    prot

    ein

    sin

    the

    ileu

    mco

    mpa

    red

    wit

    han

    imal

    sw

    ith

    NE

    C(i

    i)B

    .bifi

    dum

    did

    not

    affec

    tre

    duce

    dm

    uci

    n-2

    prod

    uct

    ion

    inth

    eN

    EC

    rats

    [56]

    TJ

    (i)

    VSL

    #3(i

    )B

    AL

    B/c

    mic

    e

    (i)

    VSL

    #3tr

    eatm

    ent

    prev

    ente

    dth

    ein

    crea

    sein

    epit

    hel

    ialp

    erm

    eabi

    lity

    inac

    ute

    colit

    is,d

    ecre

    ase

    inex

    pres

    sion

    and

    redi

    stri

    buti

    onof

    occl

    udi

    n,Z

    O-1

    ,an

    dcl

    audi

    n-1

    ,cla

    udi

    n-3

    ,cla

    udi

    n-4

    ,an

    dcl

    audi

    n-5

    ,an

    din

    crea

    seof

    epit

    hel

    ial

    apop

    toti

    cra

    tio

    [57]

    TJ

    (i)

    Lr(i

    i)L

    a(i

    )M

    ice

    (i)

    Pre

    trea

    tmen

    tw

    ith

    com

    bin

    atio

    nof

    Lran

    dL

    asi

    gnifi

    can

    tly

    prev

    ente

    dde

    crea

    sein

    the

    mem

    bran

    e-bo

    un

    dA

    TPa

    ses

    and

    redu

    ced

    expr

    essi

    onof

    tigh

    tju

    nct

    ion

    prot

    ein

    sin

    the

    mem

    bran

    e[5

    8]

    TJ

    (i)

    B.l

    acti

    s42

    0(i

    i)B

    .lac

    tis

    HN

    019

    (iii

    )L

    aN

    CFM

    (iv)

    L.sa

    livar

    ius

    Ls-3

    3

    (i)

    CaC

    o-2

    (i)

    B.l

    acti

    s42

    0an

    dE

    sche

    rich

    iaco

    liO

    157:

    H7

    (EH

    EC

    )su

    per

    nat

    ant

    had

    oppo

    site

    effec

    tsin

    tigh

    tju

    nct

    ion

    inte

    grit

    y;B

    .lac

    tis

    420

    sup

    ern

    atan

    tpr

    otec

    ted

    the

    tigh

    tju

    nct

    ion

    sfr

    omE

    HE

    C-i

    ndu

    ced

    dam

    age

    wh

    enad

    min

    iste

    red

    befo

    reE

    HE

    Csu

    pern

    atan

    t(i

    i)E

    HE

    Can

    dpr

    obio

    tics

    had

    reve

    rse

    effec

    tsu

    pon

    cycl

    o-ox

    ygen

    ase

    expr

    essi

    on

    [59]

    TJ

    (i)

    EC

    N(i

    )B

    AL

    B/c

    mic

    e

    (i)

    EC

    Nco

    lon

    izat

    ion

    ofgn

    otob

    ioti

    cm

    ice

    resu

    lted

    inu

    preg

    ula

    tion

    ofZ

    O-1

    mR

    NA

    and

    prot

    ein

    leve

    lsin

    IEC

    s(i

    i)E

    CN

    adm

    inis

    trat

    ion

    redu

    ced

    loss

    ofbo

    dyw

    eigh

    tan

    dco

    lon

    shor

    ten

    ing

    inD

    SS-t

    reat

    edm

    ice

    (iii

    )E

    CN

    inoc

    ula

    tion

    amel

    iora

    tes

    the

    infi

    ltra

    tion

    ofth

    eco

    lon

    wit

    hle

    uko

    cyte

    s

    [60]

    TJ

    PK

    C(i

    )E

    CN

    (i)

    T84

    (i)

    EP

    EC

    wit

    hE

    CN

    coin

    cuba

    tion

    :add

    itio

    nof

    EC

    Naf

    ter

    EP

    EC

    infe

    ctio

    nre

    stor

    edba

    rrie

    rin

    tegr

    ity

    and

    abol

    ish

    edba

    rrie

    rdi

    sru

    ptio

    n(i

    i)E

    CN

    alte

    red

    the

    expr

    essi

    on,d

    istr

    ibu

    tion

    ofZ

    O-2

    prot

    ein

    and

    ofdi

    stin

    ctP

    KC

    isot

    ypes

    ;ZO

    -2ex

    pres

    sion

    was

    incr

    ease

    din

    para

    llelt

    oit

    sre

    dist

    ribu

    tion

    tow

    ards

    the

    cell

    bou

    nda

    ries

    (iii

    )E

    CN

    indu

    ces

    rest

    orat

    ion

    ofa

    disr

    upt

    edep

    ith

    elia

    lbar

    rier

    ;th

    isis

    tran

    smit

    ted

    byP

    KC

    zeta

    sile

    nci

    ng

    and

    ZO

    -2re

    dist

    ribu

    tion

    [61]

  • 8 Gastroenterology Research and Practice

    Ta

    ble

    1:C

    onti

    nu

    ed.

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    TJ

    (i)

    LGG

    (i)

    MD

    CK

    -1,T

    84

    (i)

    EH

    EC

    -in

    duce

    dde

    crea

    sein

    elec

    tric

    alre

    sist

    ance

    and

    the

    incr

    ease

    inba

    rrie

    rp

    erm

    eabi

    lity

    assa

    ysw

    ere

    atte

    nu

    ated

    bypr

    obio

    tic

    pret

    reat

    men

    t(i

    i)LG

    Gpr

    otec

    ted

    epit

    hel

    ialm

    onol

    ayer

    sag

    ain

    stE

    HE

    C-i

    ndu

    ced

    redi

    stri

    buti

    onof

    clau

    din

    -1an

    dZ

    O-1

    prot

    ein

    s(i

    ii)

    Hea

    t-in

    acti

    vate

    dLG

    Gdi

    dn

    otaff

    ect

    EH

    EC

    bin

    din

    gor

    disr

    upt

    ion

    ofba

    rrie

    rfu

    nct

    ion

    [62]

    JAK

    /STA

    TM

    AP

    K(p

    38/E

    RK

    )

    (i)

    Stre

    ptoc

    occu

    sth

    erm

    ophi

    lus

    (ii)

    La

    (i)

    HT

    29/c

    l.19A

    (ii)

    Cac

    o-2,

    (i)

    Stre

    ptoc

    occu

    sth

    erm

    ophi

    lus

    (ST

    )/La

    orth

    eco

    mm

    ensa

    lBac

    tero

    ides

    thet

    aiot

    aom

    icro

    n(B

    T)

    prev

    ente

    dth

    eT

    NF-α

    -an

    dIF

    N-γ

    -in

    duce

    dre

    duct

    ion

    inT

    ER

    and

    incr

    ease

    inep

    ith

    elia

    lper

    mea

    bilit

    y(i

    i)ST

    /La

    orB

    Tpr

    even

    ted

    IFN

    -γin

    hib

    itio

    nof

    agon

    ist-

    stim

    ula

    ted

    chlo

    ride

    secr

    etio

    n(i

    ii)

    ST/L

    aor

    BT

    rest

    orat

    ion

    ofC

    l(-)

    secr

    etio

    nw

    asbl

    ocke

    dby

    inh

    ibit

    ors

    ofp3

    8M

    AP

    K,E

    RK

    1,2,

    and

    PI3

    K(i

    v)ST

    /La

    pret

    reat

    men

    tre

    vers

    edth

    eIF

    N-γ

    -in

    duce

    ddo

    wn

    regu

    lati

    onof

    the

    cyst

    icfi

    bros

    istr

    ansm

    embr

    ane

    con

    duct

    ance

    regu

    lato

    r(C

    FTR

    )an

    dth

    eN

    KC

    C1

    cotr

    ansp

    orte

    r(v

    )T

    he

    effec

    tsof

    ST/L

    aor

    BT

    onT

    ER

    and

    per

    mea

    bilit

    yw

    ere

    pote

    nti

    ated

    bya

    Jan

    us

    kin

    ase

    (JA

    K)

    inh

    ibit

    orbu

    tn

    otby

    p38,

    ER

    K1,

    2,or

    PI3

    Kin

    hib

    itio

    n(v

    i)R

    edu

    ced

    acti

    vati

    onof

    supp

    ress

    orof

    cyto

    kin

    esi

    gnal

    ing

    (SO

    CS)

    3an

    dST

    AT

    1,3

    was

    seen

    only

    inpr

    obio

    tic-

    trea

    ted

    epit

    hel

    ialc

    ells

    expo

    sed

    tocy

    toki

    nes

    [63]

    JAK

    /STA

    TN

    F-κB

    (i)

    LcS

    (i)

    RA

    W26

    4.7

    (ii)

    LI-

    LP

    MC

    (iii

    )U

    C-P

    BM

    C

    (i)

    SAM

    P1/

    Yit

    mic

    e

    (i)

    LcS

    impr

    oved

    chro

    nic

    ileit

    isin

    SAM

    P1/

    Yit

    mic

    e(i

    i)L

    csim

    prov

    edm

    uri

    ne

    chro

    nic

    colit

    is,a

    nd

    this

    was

    asso

    ciat

    edw

    ith

    decr

    ease

    dIL

    -6pr

    odu

    ctio

    nby

    larg

    ein

    test

    inal

    lam

    ina

    prop

    ria

    mon

    onu

    clea

    rce

    lls(

    LI-L

    PM

    Cs)

    and

    dow

    nre

    gula

    tion

    ofpr

    oin

    flam

    mat

    ory

    cyto

    kin

    essu

    chas

    IL-6

    and

    IFN

    -γpr

    odu

    ctio

    nin

    LPM

    C(i

    ii)

    IL-6

    rele

    ase

    inLP

    S-ac

    tiva

    ted

    LI-L

    PM

    C,R

    AW

    264.

    7,an

    du

    lcer

    ativ

    eco

    litis

    -per

    iph

    eral

    bloo

    dm

    onon

    ucl

    ear

    cells

    (UC

    -PB

    MC

    s)w

    asin

    hib

    ited

    byLc

    S-de

    rive

    dpo

    lysa

    cch

    arid

    e-pe

    ptid

    ogly

    can

    com

    plex

    (PSP

    G)

    (iv)

    Inlip

    opol

    ysac

    char

    ide-

    (LP

    S-)

    stim

    ula

    ted

    LI-

    LP

    MC

    and

    RA

    W26

    4.7

    cells

    ,Lc

    Sin

    hib

    ited

    the

    prod

    uct

    ion

    ofIL

    -6;o

    ther

    stra

    ins

    ofLa

    ctob

    acill

    usdi

    dn

    ot(v

    )N

    ucl

    ear

    tran

    sloc

    atio

    nof

    NF-κB

    was

    dow

    nre

    gula

    ted

    byL

    cS

    [64]

  • Gastroenterology Research and Practice 9

    Ta

    ble

    1:C

    onti

    nu

    ed.

    Invo

    lved

    path

    way

    sP

    robi

    otic

    sIn

    vitr

    osy

    stem

    Invi

    vosy

    stem

    Sum

    mar

    yR

    ef.

    JAK

    /STA

    T(i

    )La

    ctob

    acill

    ushe

    lvet

    icus

    R00

    52

    (i)

    Inte

    stin

    e40

    7(i

    i)H

    Ep-

    2(i

    ii)

    Cac

    o-2

    (i)

    Aft

    erE

    HE

    CO

    157:

    H7

    infe

    ctio

    n,S

    TAT

    -1ac

    tiva

    tion

    was

    redu

    ced

    com

    pare

    dto

    un

    infe

    cted

    cells

    (ii)

    Pre

    incu

    bati

    onw

    ith

    L.he

    lvet

    icus

    R00

    52(b

    utn

    otbo

    iled

    L.he

    lvet

    icus

    R00

    52,

    aneq

    ual

    con

    cen

    trat

    ion

    ofvi

    able

    LrR

    0011

    ,or

    surf

    ace-

    laye

    rpr

    otei

    ns)

    follo

    wed

    byE

    HE

    Cin

    fect

    ion

    abro

    gate

    ddi

    sru

    ptio

    nof

    IFN

    -γ-S

    TAT

    -1si

    gnal

    ing

    [65]

    SAP

    K/J

    NK

    MA

    PK

    (p38

    )N

    F-κB

    (i)

    S.ce

    revi

    siae

    UFM

    G90

    5(i

    )M

    ice

    (i)

    Aft

    erSa

    lmon

    ella

    chal

    len

    ge,S

    .cer

    evis

    iae

    905

    inh

    ibit

    edw

    eigh

    tlo

    ssan

    din

    crea

    sed

    surv

    ival

    rate

    (ii)

    Leve

    lsof

    proi

    nfl

    amm

    ator

    ycy

    toki

    nes

    wer

    ede

    crea

    sed,

    and

    acti

    vati

    onof

    mit

    ogen

    -act

    ivat

    edpr

    otei

    nki

    nas

    es(p

    38an

    dJN

    K,b

    ut

    not

    ER

    K1/

    2),N

    F-κB

    ,an

    dA

    P-1

    was

    mod

    ula

    ted

    byS.

    cere

    visi

    ae90

    5

    [66]

    Lipi

    d/X

    enob

    ioti

    c(i

    )V

    SL#3

    (i)

    IL-1

    0K

    O

    (i)

    Pro

    biot

    ics

    resu

    lted

    indo

    wn

    regu

    lati

    onof

    CX

    CL

    9,C

    XC

    L10

    ,CC

    L5,

    T-c

    ell

    acti

    vati

    onan

    dIR

    GM

    (ii)

    Pro

    biot

    ictr

    eatm

    ent

    decr

    ease

    dth

    en

    um

    ber

    ofC

    CL5

    +C

    D3+

    dou

    ble-

    posi

    tive

    Tce

    llsco

    nsi

    sten

    tw

    ith

    redu

    ctio

    nin

    inte

    grin

    san

    du

    preg

    ula

    ted

    gale

    ctin

    2(i

    ii)

    Lipi

    d-an

    dP

    PAR

    -sig

    nal

    ing-

    asso

    ciat

    edge

    nes

    wer

    eal

    sou

    preg

    ula

    ted

    (iv)

    Alt

    ered

    mic

    robi

    aldi

    vers

    ity

    was

    not

    edin

    prob

    ioti

    c-tr

    eate

    dm

    ice

    (v)

    Infl

    amm

    atio

    nin

    IL10

    -KO

    mic

    esh

    owed

    diff

    eren

    tial

    regu

    lati

    onof

    vari

    ous

    sign

    alin

    gpa

    thw

    ays(

    infl

    amm

    ator

    y,n

    ucl

    ear

    rece

    ptor

    ,lip

    id,a

    nd

    xen

    obio

    tic)

    [67]

    Met

    abol

    ism

    /G

    luco

    seU

    ptak

    e

    (i)

    La(i

    i)L.

    gass

    eri

    (iii

    )L.

    amyl

    ovor

    us(i

    v)L.

    galli

    naru

    m(v

    )L.

    john

    soni

    i

    (i)

    CaC

    o-2

    (i)

    Exp

    osu

    reto

    bact

    eria

    -fre

    esu

    pern

    atan

    tsof

    La

    cult

    ure

    din

    chem

    ical

    lyde

    fin

    edm

    edia

    (CD

    M)

    wit

    h11

    0m

    Mfr

    uct

    ose

    incr

    ease

    dgl

    uco

    seac

    cum

    ula

    tion

    ;ex

    posu

    reto

    asu

    spen

    sion

    ofth

    eba

    cter

    iah

    adn

    oeff

    ect

    (ii)

    Hea

    t-de

    nat

    uri

    ng

    the

    supe

    rnat

    ant

    dim

    inis

    hed

    the

    incr

    ease

    ingl

    uco

    seac

    cum

    ula

    tion

    (iii)

    Supe

    rnat

    ants

    prep

    ared

    wit

    han

    aero

    bic

    cult

    ure

    ofL.

    gass

    eri,

    L.am

    ylov

    orus

    ,L.

    galli

    naru

    m,a

    ndL.

    john

    soni

    iin

    the

    CD

    Mw

    ith

    fru

    ctos

    ein

    crea

    sed

    glu

    cose

    accu

    mu

    lati

    on

    [68]

    Rea

    ctiv

    eO

    xyge

    nSp

    ecie

    s(R

    OS)

    NF-κB

    (i)

    Lr(i

    )FH

    s74I

    nt

    (i)

    C57

    BL/

    6J

    (i)

    LGG

    indu

    ced

    RO

    Sge

    ner

    atio

    nin

    inte

    stin

    alep

    ith

    elia

    invi

    tro

    and

    invi

    vo(i

    i)In

    crea

    sed

    glu

    tath

    ion

    e(G

    SH)

    oxid

    atio

    nan

    dcu

    llin

    -1de

    ned

    dyla

    tion

    was

    seen

    inth

    ein

    test

    ines

    ofm

    ice

    fed

    LGG

    ,sh

    owin

    glo

    calR

    OS

    gen

    erat

    ion

    and

    the

    resu

    ltan

    tU

    bc12

    inac

    tiva

    tion

    (iii

    )P

    refe

    edin

    gLG

    Gpr

    even

    ted

    TN

    F-α

    -in

    duce

    dac

    tiva

    tion

    ofin

    test

    inal

    NF-κB

    [69]

  • 10 Gastroenterology Research and Practice

    of nuclear receptors and their mechanisms of action isbeyond the scope of this article; however, further discussionof two nuclear receptors (peroxisome-proliferator-activatedreceptor gamma (PPARγ) and vitamin D receptor (VDR))with putative roles in inflammation is warranted.

    PPARγ is a member of a class of nuclear receptors thatform obligate heterodimers with the retinoid X receptor(RXR) [129]. The PPAR family has been shown to affectvarious cellular functions including “adipocyte differentia-tion, fatty-acid oxidation, and glucose metabolism” [129].PPARγ is highly expressed in the large intestine [133], and itsactivation has been shown to be protective in animal modelsof colitis [134, 135]. Decreased PPARγ expression in UCpatients has been shown [136], and the anti-inflammatorycompound 5-aminosalicylic acid (5-ASA) commonly uti-lized in IBD therapy was shown to be a PPARγ agonist,thereby establishing a possible mechanism by which it exertsits anti-inflammatory effects [137]. PPARγ also plays a rolein the maintenance of “constitutive epithelial expression of asubset of β defensins in the colon” [122].

    6. Vitamin D and Vitamin D Receptor (VDR)

    Vitamin D receptor (VDR) is a nuclear receptor that medi-ates most known functions of 1,25-dihydroxyvitamin D(1,25(OH)2D3), the active form of vitamin D [138]. VDRheterodimerizes with RXR once VDR is activated by1,25(OH)2D3. VDR binds to the vitamin D response elementin the target gene promoter to regulate gene transcription[139]. VDR downstream target genes include antimicrobialpeptides such as cathelicidin and β defensin.

    VDR is critical in regulating intestinal homeostasis bypreventing pathogenic bacterial invasion, inhibiting inflam-mation, and maintaining cell integrity [140–145]. VitaminD directly modulates the T-cell receptor (TCR) [146], andvitamin D has also been shown to downregulate the expres-sion of proinflammatory cytokines and have regulatoryeffects on autophagy and various immune cells includingT cells, B cells, macrophages, dendritic cells, and epithelialcells [147, 148]. It has been reported that 1,25(OH)2D3suppresses the development of IBD in animal models [149].Deficiency of 1,25(OH)2D3 has been reported in patientswith IBD [150, 151], and, recently, using a novel vitamin Dbioavailability test, vitamin D deficiency or insufficiency wasseen in more than 70% of patients with quiescent CD [152].Given the diverse immune functions of vitamin D, deficientlevels may have important implications for the developmentand maintenance of intestinal homeostasis. A possible roleof vitamin D status and VDR signaling in modulating theeffects of intestinal microflora in other conditions suchas asthma and obesity has been suggested [100]. Whilepresent literature has primarily focused on elucidating theimmunoregulatory effects of vitamin D, there is a paucity ofdata on the status and function of VDR [147]. In addition,probiotic-induced modulation of anti-inflammatory VDRsignaling in colitis remains virtually unexplored.

    Recent studies indicate that VDR−/− mice haveincreased bacterial loading in the intestine [145, 153]. Ourmicroarray data found that VDR signaling responds to

    pathogenic Salmonella in intestinal colitis in vivo [154]. Datafrom a recent study demonstrate that bacterial stimulation,both commensal and pathogenic, regulates VDR expressionand location and that VDR negatively regulates bacterial-induced intestinal NF-κB activation [153]. In general,probiotic-induced nuclear receptor signaling is not well char-acterized. The probiotic VSL3# was associated with nuclearreceptor signaling in the IL10−/− colitis model [67]. Nuclearreceptors have been shown to negatively regulate bacterial-stimulated NF-κB activity in intestinal epithelium [153, 155].Our recent data show probiotic treatment is able to enhanceVDR expression and activity in the host. An increase inVDR expression and a concomitant increase in cathelicidinmRNA in cultured intestinal epithelial cells when treatedwith Lactobacillus plantarum were seen [156]. We used aprobiotic monoassociated pig model to assess the probioticeffect on VDR expression in vivo and found intestinal VDRincreased significantly after probiotic colonization comparedto the ex-germ-free pig. Furthermore, our unpublished dataindicate that probiotics did not inhibit inflammation in micelacking VDR.

    The presence of VDR in various tissues along with itsability to exert diverse actions in differentiation, growth, andanti-inflammation sets the stage for exploitation of VDRligands for the treatment of various inflammatory conditions[157, 158]. Although the potential importance of VDR as atherapeutic target has been appreciated [159], no approachto date has safely and effectively altered VDR’s activity.Hence, understanding VDR’s contribution to probiotic-induced anti-inflammation may provide significant insightin the pathogenesis of inflammatory conditions such as IBD,and thereby, guide the development of novel treatments.Further investigation of the complex interplay of nuclearreceptors, defensins, probiotics, and inflammatory pathwaysmay provide significant insight into the mechanisms ofaction of probiotics in anti-inflammation.

    7. Current Problems and Future Directions

    The individual diversity of the intestinal microflora under-scores the difficulty of identifying the entire human micro-biota and poses barriers to this field of research. In addition,it is apparent that the actions of probiotics are species andstrain specific [19]. It is also apparent that even a single strainof probiotic may exert its actions via multiple, concomitantpathways. Current investigation into the mechanism ofaction of specific probiotics has focused on probiotic-induced changes in the innate immune functions involvingTLRs and its downstream systems including NF-κB, JAK-STAT, MAPK, and SAPK/JNK pathways. Future research onnovel mechanisms of action for probiotics involving nuclearreceptor signaling, including PPARγ and VDR, is needed.With evolving knowledge, effective probiotic therapy will bepossible in the future.

    AbbreviationsAAD: Antibiotic-associated diarrheaAP-1: Activator protein-1CD: Crohn’s disease

  • Gastroenterology Research and Practice 11

    CDAD: Clostridium-difficile-associated diseaseCDM: Chemically defined mediaCM: Conditioned mediaCTFR: Cystic fibrosis transmembrane

    conductance regulatorECN: E. coli Nissle 1917EGFR: Epidermal growth factor receptorEHEC: E. coli O157:H7EPEC: Enteropathogenic E. coliERK: Extracellular-signal-regulated kinaseGSH: GlutathionehBD: Human beta defensinHSP: Heat shock proteinsIBD: Inflammatory bowel diseaseIBS: Irritable bowel syndromeIEC: Intestinal epithelial cellIFN: InterferonIL: InterleukinIκB: Inhibitor of kappa BJAK/STAT: Janus kinase/signal transducers and

    activators of transcriptionLa: Lactobacillus acidophilusLc: Lactobacillus caseiLcS: Lactobacillus casei ShirotaLGG: Lactobacillus rhamnosus GGLI-LPMC: Large intestinal lamina propria

    mononuclear cellsLP: Lactobacillus plantarumLPS: LipopolysaccharideLr: Lactobacillus rhamnosusMAPK: Mitogen-activated protein kinaseMCP: Monocyte chemotactic proteinNEC: Necrotizing enterocolitisNF-κB: Nuclear factor kappa BPKC: Protein kinase CPPARγ: Peroxisome-proliferator-activated

    receptor gammaROS: Reactive oxygen speciesRXR: Retinoid X receptorSAPK/JNK: Stress-activated protein kinase/c-Jun

    NH2-terminal kinaseSb: Saccharomyces boulardiiSOCS: Suppressor of cytokine signalingTCR: T-cell receptorTER: Transepithelial electrical resistanceTJs: Tight junctionsTLR: Toll-like receptorTNF: Tumor necrosis factorUC: Ulcerative colitisUCB: Unconjugated bilirubinVDR: Vitamin D receptorZO: Zonula occludens.

    Acknowledgments

    This work was supported by the National Institutes of Health(DK075386-0251, R03DK089010-01) and the IDEAL awardfrom the New York State’s Empire State Stem Cell Board(N09G-279) to J. Sun.

    References

    [1] L. V. McFarland, “Evidence-based review of probioticsfor antibiotic-associated diarrhea and Clostridium difficileinfections,” Anaerobe, vol. 15, no. 6, pp. 274–280, 2009.

    [2] A. S. Neish, “Microbes in gastrointestinal health and disease,”Gastroenterology, vol. 136, no. 1, pp. 65–80, 2009.

    [3] W. S. Garrett, J. I. Gordon, and L. H. Glimcher, “Homeostasisand Inflammation in the Intestine,” Cell, vol. 140, no. 6, pp.859–870, 2010.

    [4] M. Joossens, G. Huys, M. Cnockaert et al., “Dysbiosis of thefaecal microbiota in patients with Crohn’s disease and theirunaffected relatives,” Gut, vol. 60, no. 5, pp. 631–637, 2011.

    [5] I. Sobhani, J. Tap, F. Roudot-Thoraval et al., “Microbialdysbiosis in colorectal cancer (CRC) patients,” PLoS ONE,vol. 6, no. 1, Article ID e16393, 2011.

    [6] Y. M. Sjögren, M. C. Jenmalm, M. F. Böttcher, B. Björkstén,and E. Sverremark-Ekström, “Altered early infant gut micro-biota in children developing allergy up to 5 years of age,”Clinical and Experimental Allergy, vol. 39, no. 4, pp. 518–526,2009.

    [7] B. Björkstén, P. Naaber, E. Sepp, and M. Mikelsaar, “Theintestinal microflora in allergic Estonian and Swedish 2-year-old children,” Clinical and Experimental Allergy, vol. 29, no.3, pp. 342–346, 1999.

    [8] A. W. Yan, D. E. Fouts, J. Brandl et al., “Enteric dysbiosisassociated with a mouse model of alcoholic liver disease,”Hepatology, vol. 53, no. 1, pp. 96–105, 2011.

    [9] A. Kassinen, L. Krogius-Kurikka, H. Mäkivuokko et al., “Thefecal microbiota of irritable bowel syndrome patients differssignificantly from that of healthy subjects,” Gastroenterology,vol. 133, no. 1, pp. 24–33, 2007.

    [10] R. E. Ley, P. J. Turnbaugh, S. Klein, and J. I. Gordon,“Microbial ecology: human gut microbes associated withobesity,” Nature, vol. 444, no. 7122, pp. 1022–1023, 2006.

    [11] M. Vijay-Kumar, J. D. Aitken, F. A. Carvalho et al., “Metaboliesyndrome and altered gut microbiota in mice lacking toll-likereceptor 5,” Science, vol. 328, no. 5975, pp. 228–231, 2010.

    [12] R. E. Ley, “Obesity and the human microbiome,” CurrentOpinion in Gastroenterology, vol. 26, no. 1, pp. 5–11, 2010.

    [13] R. E. Ley, F. Bäckhed, P. Turnbaugh, C. A. Lozupone, R.D. Knight, and J. I. Gordon, “Obesity alters gut microbialecology,” Proceedings of the National Academy of Sciences ofthe United States of America, vol. 102, no. 31, pp. 11070–11075, 2005.

    [14] C. Manichanh, L. Rigottier-Gois, E. Bonnaud et al., “Reduceddiversity of faecal microbiota in Crohn’s disease revealed bya metagenomic approach,” Gut, vol. 55, no. 2, pp. 205–211,2006.

    [15] D. N. Frank, A. L. S. Amand, R. A. Feldman, E. C. Boedeker,N. Harpaz, and N. R. Pace, “Molecular-phylogenetic char-acterization of microbial community imbalances in humaninflammatory bowel diseases,” Proceedings of the NationalAcademy of Sciences of the United States of America, vol. 104,no. 34, pp. 13780–13785, 2007.

    [16] M. P. Conte, S. Schippa, I. Zamboni et al., “Gut-associatedbacterial microbiota in paediatric patients with inflamma-tory bowel disease,” Gut, vol. 55, no. 12, pp. 1760–1767, 2006.

    [17] R. B. Sartor, “Microbial influences in inflammatory boweldiseases,” Gastroenterology, vol. 134, no. 2, pp. 577–594, 2008.

    [18] C. D. Packey and R. B. Sartor, “Interplay of commensal andpathogenic bacteria, genetic mutations, and immunoregu-latory defects in the pathogenesis of inflammatory bowel

  • 12 Gastroenterology Research and Practice

    diseases,” Journal of Internal Medicine, vol. 263, no. 6, pp.597–606, 2008.

    [19] C. Reiff and D. Kelly, “Inflammatory bowel disease, gutbacteria and probiotic therapy,” International Journal ofMedical Microbiology, vol. 300, no. 1, pp. 25–33, 2010.

    [20] R. B. Sartor, “Mechanisms of disease: pathogenesis ofcrohn’s disease and ulcerative colitis,” Nature Clinical PracticeGastroenterology and Hepatology, vol. 3, no. 7, pp. 390–407,2006.

    [21] D. M. Lilly and R. H. Stillwell, “Probiotics: growth-promoting factors produced by microorganisms,” Science,vol. 147, no. 3659, pp. 747–748, 1965.

    [22] E. M. Quigley, “Probiotics in gastrointestinal disorders,”Hospital Practice, vol. 38, no. 4, pp. 122–129, 2010.

    [23] T. A. Oelschlaeger, “Mechanisms of probiotic actions—areview,” International Journal of Medical Microbiology, vol.300, no. 1, pp. 57–62, 2010.

    [24] B. G. Spyropoulos, E. P. Misiakos, C. Fotiadis, and C.N. Stoidis, “Antioxidant properties of probiotics and theirprotective effects in the pathogenesis of radiation-inducedenteritis and coliti,” Digestive Diseases and Sciences, vol. 52,no. 2, pp. 285–294, 2010.

    [25] F. Yan and D. B. Polk, “Probiotics as functional food in thetreatment of diarrhea,” Current Opinion in Clinical Nutritionand Metabolic Care, vol. 9, no. 6, pp. 717–721, 2006.

    [26] K. Madsen, “Probiotics in critically ill patients,” Journal ofClinical Gastroenterology, vol. 42, supplement 3, pp. S116–S118, 2008.

    [27] C. I. Fotiadis, C. N. Stoidis, B. G. Spyropoulos, and E. D.Zografos, “Role of probiotics, prebiotics and synbiotics inchemoprevention for colorectal cancer,” World Journal ofGastroenterology, vol. 14, no. 42, pp. 6453–6457, 2008.

    [28] R. Francavilla, V. Miniello, A. M. Magistà et al., “A ran-domized controlled trial of lactobacillus GG in children withfunctional abdominal pain,” Pediatrics, vol. 126, no. 6, pp.e1445–e1452, 2010.

    [29] D. W. Thomas, F. R. Greer, J. J. S. Bhatia et al., “Clinicalreport—probiotics and prebiotics in pediatrics,” Pediatrics,vol. 126, no. 6, pp. 1217–1231, 2010.

    [30] D. Damaskos and G. Kolios, “Probiotics and prebioticsin inflammatory bowel disease: microflora ’on the scope’,”British Journal of Clinical Pharmacology, vol. 65, no. 4, pp.453–467, 2008.

    [31] A. Tursi, G. Brandimarte, A. Papa et al., “Treatment ofrelapsing mild-to-moderate ulcerative colitis with the pro-biotic VSL3 as adjunctive to a standard pharmaceuticaltreatment: a double-blind, randomized, placebo-controlledstudy,” American Journal of Gastroenterology, vol. 105, no. 10,pp. 2218–2227, 2010.

    [32] C. Vanderpool, F. Yan, and D. B. Polk, “Mechanisms ofprobiotic action: implications for therapeutic applications ininflammatory bowel diseases,” Inflammatory Bowel Diseases,vol. 14, no. 11, pp. 1585–1596, 2008.

    [33] T. Watanabe, H. Nishio, T. Tanigawa et al., “ProbioticLactobacillus casei strain Shirota prevents indomethacin-induced small intestinal injury: involvement of lactic acid,”American Journal of Physiology, vol. 297, no. 3, pp. G506–G513, 2009.

    [34] L. Zhang, N. Li, R. Caicedo, and J. Neu, “Alive and deadLactobacillus rhamnosus GG decrease tumor necrosis factor-α-induced interleukin-8 production in Caco-2 cells,” Journalof Nutrition, vol. 135, no. 7, pp. 1752–1756, 2005.

    [35] E. O. Petrof, K. Kojima, M. J. Ropeleski et al., “Probioticsinhibit nuclear factor-κB and induce heat shock proteins

    in colonic epithelial cells through proteasome inhibition,”Gastroenterology, vol. 127, no. 5, pp. 1474–1487, 2004.

    [36] E. O. Petrof, E. C. Claud, J. Sun et al., “Bacteria-free solutionderived from Lactobacillus plantarum inhibits multiple NF-kappaB pathways and inhibits proteasome function,” Inflam-matory Bowel Diseases, vol. 15, no. 10, pp. 1537–1547, 2009.

    [37] B. Wang, J. Li, J. Chen, Q. Huang, N. Li, and J. Li, “Effectof live Lactobacillus plantarum L2 on TNF-α-induced MCP-1 production in Caco-2 cells,” International Journal of FoodMicrobiology, vol. 142, no. 1-2, pp. 237–241, 2010.

    [38] M. T. Tien, S. E. Girardin, B. Regnault et al., “Anti-inflammatory effect of Lactobacillus casei on Shigella-infected human intestinal epithelial cells,” Journal ofImmunology, vol. 176, no. 2, pp. 1228–1237, 2006.

    [39] P. A. Ruiz, M. Hoffmann, S. Szcesny, M. Blaut, and D. Haller,“Innate mechanisms for Bifidobacterium lactis to activatetransient pro-inflammatory host responses in intestinalepithelial cells after the colonization of germ-free rats,”Immunology, vol. 115, no. 4, pp. 441–450, 2005.

    [40] F. S. Martins, G. Dalmasso, R. M. E. Arantes et al., “Inter-action of Saccharomyces boulardii with Salmonella entericaserovar typhimurium protects mice and modifies T84 cellresponse to the infection,” PLoS ONE, vol. 5, no. 1, ArticleID e8925, 2010.

    [41] M. Schlee, J. Harder, B. Köten, E. F. Stange, J. Wehkamp,and K. Fellermann, “Probiotic lactobacilli and VSL#3induce enterocyte β-defensin 2,” Clinical and ExperimentalImmunology, vol. 151, no. 3, pp. 528–535, 2008.

    [42] J. Wehkamp, J. Harder, K. Wehkamp et al., “NF-κB- and AP-1-mediated induction of human beta defensin-2 in intestinalepithelial cells by Escherichia coli Nissle 1917: a novel effectof a probiotic bacterium,” Infection and Immunity, vol. 72,no. 10, pp. 5750–5758, 2004.

    [43] Y. Tao, K. A. Drabik, T. S. Waypa et al., “Soluble factors fromLactobacillus GG activate MAPKs and induce cytoprotectiveheat shock proteins in intestinal epithelial cells,” AmericanJournal of Physiology, vol. 290, no. 4, pp. C1018–C1030, 2006.

    [44] X. Chen, J. Fruehauf, J. D. Goldsmith et al., “Saccharomycesboulardii inhibits EGF receptor signaling and intestinaltumor growth in Apc(min) mice,” Gastroenterology, vol. 137,no. 3, pp. 914–923, 2009.

    [45] D. Ghadimi, M. De Vrese, K. J. Heller, and J. Schrezenmeir,“Effect of natural commensal-origin DNA on toll-like recep-tor 9 (TLR9) signaling cascade, chemokine IL-8 expression,and barrier integritiy of polarized intestinal epithelial cells,”Inflammatory Bowel Diseases, vol. 16, no. 3, pp. 410–427,2010.

    [46] M. G. Vizoso Pinto, M. Rodriguez Gómez, S. Seifert, B.Watzl, W. H. Holzapfel, and C. M. A. P. Franz, “Lactobacillistimulate the innate immune response and modulate theTLR expression of HT29 intestinal epithelial cells in vitro,”International Journal of Food Microbiology, vol. 133, no. 1-2,pp. 86–93, 2009.

    [47] D. Rachmilewitz, K. Katakura, F. Karmeli et al., “Toll-likereceptor 9 signaling mediates the anti-inflammatory effectsof probiotics in murine experimental colitis,” Gastroenterol-ogy, vol. 126, no. 2, pp. 520–528, 2004.

    [48] A. Grabig, D. Paclik, C. Guzy et al., “Escherichia coli strainNissle 1917 ameliorates experimental colitis via toll-likereceptor 2- and toll-like receptor 4-dependent pathways,”Infection and Immunity, vol. 74, no. 7, pp. 4075–4082, 2006.

    [49] R. Kaji, J. Kiyoshima-Shibata, M. Nagaoka, M. Nanno, andK. Shida, “Bacterial teichoic acids reverse predominant IL-12 production induced by certain lactobacillus strains into

  • Gastroenterology Research and Practice 13

    predominant IL-10 production via TLR2-dependent ERKactivation in macrophages,” Journal of Immunology, vol. 184,no. 7, pp. 3505–3513, 2010.

    [50] A. Seth, F. Yan, D. B. Polk, and R. K. Rao, “Probioticsameliorate the hydrogen peroxide-induced epithelial barrierdisruption by a PKC- And MAP kinase-dependent mecha-nism,” American Journal of Physiology, vol. 294, no. 4, pp.G1060–G1069, 2008.

    [51] J. B. Ewaschuk, H. Diaz, L. Meddings et al., “Secreted bioac-tive factors from Bifidobacterium infantis enhance epithelialcell barrier function,” American Journal of Physiology, vol.295, no. 5, pp. G1025–G1034, 2008.

    [52] R. C. Anderson, A. L. Cookson, W. C. McNabb et al., “Lac-tobacillus plantarum MB452 enhances the function of theintestinal barrier by increasing the expression levels of genesinvolved in tight junction formation,” BMC Microbiology,vol. 10, article 316, 2010.

    [53] Y. Zhou, H. Qin, M. Zhang et al., “Lactobacillus plantaruminhibits intestinal epithelial barrier dysfunction induced byunconjugated bilirubin,” British Journal of Nutrition, vol. 104,no. 3, pp. 390–401, 2010.

    [54] R. C. Anderson, A. L. Cookson, W. C. McNabb, W. J. Kelly,and N. C. Roy, “Lactobacillus plantarum DSM 2648 is apotential probiotic that enhances intestinal barrier function,”FEMS Microbiology Letters, vol. 309, no. 2, pp. 184–192, 2010.

    [55] J. Karczewski, F. J. Troost, I. Konings et al., “Regulation ofhuman epithelial tight junction proteins by Lactobacillusplantarum in vivo and protective effects on the epithelialbarrier,” American Journal of Physiology, vol. 298, no. 6, pp.G851–G859, 2010.

    [56] L. Khailova, K. Dvorak, K. M. Arganbright et al., “Bifidobac-terium bifidum improves intestinal integrity in a rat modelof necrotizing enterocolitis,” American Journal of Physiology,vol. 297, no. 5, pp. G940–G949, 2009.

    [57] R. Mennigen, K. Nolte, E. Rijcken et al., “Probiotic mixtureVSL#3 protects the epithelial barrier by maintaining tightjunction protein expression and preventing apoptosis in amurine model of colitis,” American Journal of Physiology, vol.296, no. 5, pp. G1140–G1149, 2009.

    [58] G. Moorthy, M. R. Murali, and S. N. Devaraj, “Lactobacillifacilitate maintenance of intestinal membrane integrity dur-ing Shigella dysenteriae 1 infection in rats,” Nutrition, vol. 25,no. 3, pp. 350–358, 2009.

    [59] H. Putaala, T. Salusjärvi, M. Nordström et al., “Effect offour probiotic strains and Escherichia coli O157:H7 on tightjunction integrity and cyclo-oxygenase expression,” Researchin Microbiology, vol. 159, no. 9-10, pp. 692–698, 2008.

    [60] S. N. Ukena, A. Singh, U. Dringenberg et al., “ProbioticEscherichia coli Nissle 1917 inhibits leaky gut by enhancingmucosal integrity,” PLoS ONE, vol. 2, no. 12, Article IDe1308, 2007.

    [61] A. A. Zyrek, C. Cichon, S. Helms, C. Enders, U. Sonnenborn,and M. A. Schmidt, “Molecular mechanisms underlying theprobiotic effects of Escherichia coli Nissle 1917 involve ZO-2 and PKCζ redistribution resulting in tight junction andepithelial barrier repair,” Cellular Microbiology, vol. 9, no. 3,pp. 804–816, 2007.

    [62] K. C. Johnson-Henry, K. A. Donato, G. Shen-Tu, M. Gordan-pour, and P. M. Sherman, “Lactobacillus rhamnosus strainGG prevents enterohemorrhagic Escherichia coli O157:H7-induced changes in epithelial barrier function,” Infection andImmunity, vol. 76, no. 4, pp. 1340–1348, 2008.

    [63] S. Resta-Lenert and K. E. Barrett, “Probiotics and com-mensals reverse TNF-α- and IFN-γ-induced dysfunction in

    human intestinal epithelial cells,” Gastroenterology, vol. 130,no. 3, pp. 731–746, 2006.

    [64] S. Matsumoto, T. Hara, T. Hori et al., “ProbioticLactobacillus-induced improvement in murine chronicinflammatory bowel disease is associated with the down-regulation of pro-inflammatory cytokines in lamina propriamononuclear cells,” Clinical and Experimental Immunology,vol. 140, no. 3, pp. 417–426, 2005.

    [65] N. Jandu, Z. J. Zeng, K. C. Johnson-Henry, and P. M. Sher-man, “Probiotics prevent enterohaemorrhagic Escherichiacoli O157 : H7-mediated inhibition of interferon-γ-inducedtyrosine phosphorylation of STAT-1,” Microbiology, vol. 155,no. Pt 2, pp. 531–540, 2009.

    [66] F. S. Martins, S. D. A. Elian, A. T. Vieira et al., “Oraltreatment with Saccharomyces cerevisiae strain UFMG 905modulates immune responses and interferes with signalpathways involved in the activation of inflammation in amurine model of typhoid fever,” International Journal ofMedical Microbiology, vol. 301, no. 4, pp. 359–364, 2011.

    [67] C. Reiff, M. Delday, G. Rucklidge et al., “Balancing inflam-matory, lipid, and xenobiotic signaling pathways by VSL#3,a biotherapeutic agent, in the treatment of inflammatorybowel disease,” Inflammatory Bowel Diseases, vol. 15, no. 11,pp. 1721–1736, 2009.

    [68] A. K. Rooj, Y. Kimura, and R. K. Buddington, “Metabolitesproduced by probiotic Lactobacilli rapidly increase glucoseuptake by Caco-2 cells,” BMC Microbiology, vol. 10, article16, 2010.

    [69] P. W. Lin, L. E. S. Myers, L. Ray et al., “Lactobacillus rhamno-sus blocks inflammatory signaling in vivo via reactive oxygenspecies generation,” Free Radical Biology and Medicine, vol.47, no. 8, pp. 1205–1211, 2009.

    [70] B. Eiseman, W. Silen, G. S. Bascom, and A. J. Kauvar, “Fecalenema as an adjunct in the treatment of pseudomembranousenterocolitis,” Surgery, vol. 44, no. 5, pp. 854–859, 1958.

    [71] J. Aas, C. E. Gessert, and J. S. Bakken, “Recurrent Clostridiumdifficile colitis: case series involving 18 patients treated withdonor stool administered via a nasogastric tube,” ClinicalInfectious Diseases, vol. 36, no. 5, pp. 580–585, 2003.

    [72] S. E. Persky and L. J. Brandt, “Treatment of recurrentClostridium difficile-associated diarrhea by administrationof donated stool directly through a colonoscope,” AmericanJournal of Gastroenterology, vol. 95, no. 11, pp. 3283–3285,2000.

    [73] S. S. Yoon and L. J. Brandt, “Treatment of refrac-tory/recurrent C. difficile-associated disease by donated stooltransplanted via colonoscopy: a case series of 12 patients,”Journal of Clinical Gastroenterology, vol. 44, no. 8, pp. 562–566, 2010.

    [74] G. Russell, J. Kaplan, M. Ferraro, and I. C. Michelow, “Fecalbacteriotherapy for relapsing Clostridium difficile infectionin a child: a proposed treatment protocol,” Pediatrics, vol.126, no. 1, pp. e239–e242, 2010.

    [75] A. Khoruts, J. Dicksved, J. K. Jansson, and M. J. Sadowsky,“Changes in the composition of the human fecal microbiomeafter bacteriotherapy for recurrent clostridium difficile-associated diarrhea,” Journal of Clinical Gastroenterology, vol.44, no. 5, pp. 354–360, 2010.

    [76] X. W. Gao, M. Mubasher, C. Y. Fang, C. Reifer, and L. E.Miller, “Dose-response efficacy of a proprietary probioticformula of lactobacillus acidophilus CL1285 and lactobacil-lus casei LBC80R for antibiotic-associated diarrhea andclostridium difficile-associated diarrhea prophylaxis in adult

  • 14 Gastroenterology Research and Practice

    patients,” American Journal of Gastroenterology, vol. 105, no.7, pp. 1636–1641, 2010.

    [77] L. V. McFarland, “Systematic review and meta-analysis ofsaccharomyces boulardii in adult patients,” World Journal ofGastroenterology, vol. 16, no. 18, pp. 2202–2222, 2010.

    [78] S. Guglielmetti, D. Mora, M. Gschwender, and K. Popp,“Randomised clinical trial: bifidobacterium bifidumMIMBb75 significantly alleviates irritable bowel syn-dromeand improves quality of life—a double-blind, pla- cebo-controlled study,” Alimentary Pharmacology and Ther-apeutics, vol. 33, no. 10, pp. 1123–1132, 2011.

    [79] C. H. Choi, S. Y. Jo, H. J. Park, S. K. Chang, J.-S. Byeon,and S.-J. Myung, “A randomized, double-blind, placebo-controlled multicenter trial of saccharomyces boulardii inirritablebowel syndrome: effect on quality of life,” Journal ofClinical Gastroenterology. In press.

    [80] G. C. Parkes, J. D. Sanderson, and K. Whelan, “Treatingirritable bowel syndrome with probiotics: the evidence,”Proceedings of the Nutrition Society, vol. 69, no. 2, pp. 187–194, 2010.

    [81] P. Moayyedi, A. C. Ford, N. J. Talley et al., “The efficacy ofprobiotics in the treatment of irritable bowel syndrome: asystematic review,” Gut, vol. 59, no. 3, pp. 325–332, 2010.

    [82] D. M. Brenner, M. J. Moeller, W. D. Chey, and P. S.Schoenfeld, “The utility of probiotics in the treatment ofirritable bowel syndrome: a systematic review,” AmericanJournal of Gastroenterology, vol. 104, no. 4, pp. 1033–1049,2009.

    [83] A. Sood, V. Midha, G. K. Makharia et al., “The probioticpreparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis,” Clinical Gastroen-terology and Hepatology, vol. 7, no. 11, pp. 1202–1209, 2009.

    [84] R. Bibiloni, R. N. Fedorak, G. W. Tannock et al., “VSL#3probiotic-mixture induces remission in patients with activeulcerative colitis,” American Journal of Gastroenterology, vol.100, no. 7, pp. 1539–1546, 2005.

    [85] W. Kruis, P. Frič, J. Pokrotnieks et al., “Maintaining remissionof ulcerative colitis with the probiotic Escherichia coli Nissle1917 is as effective as with standard mesalazine,” Gut, vol. 53,no. 11, pp. 1617–1623, 2004.

    [86] M. A. Zocco, L. Z. Dal Verme, F. Cremonini et al., “Efficacyof Lactobacillus GG in maintaining remission of ulcerativecolitis,” Alimentary Pharmacology and Therapeutics, vol. 23,no. 11, pp. 1567–1574, 2006.

    [87] P. Gionchetti, F. Rizzello, U. Helwig et al., “Prophylaxisof pouchitis onset with probiotic therapy: a double-blind,placebo-controlled trial,” Gastroenterology, vol. 124, no. 5,pp. 1202–1209, 2003.

    [88] T. Mimura, F. Rizzello, U. Helwig et al., “Once daily highdose probiotic therapy (VSL#3) for maintaining remissionin recurrent or refractory pouchitis,” Gut, vol. 53, no. 1, pp.108–114, 2004.

    [89] C. Prantera, M. L. Scribano, G. Falasco, A. Andreoli, and C.Luzi, “Ineffectiveness of probiotics in preventing recurrenceafter curative resection for Crohn’s disease: a randomisedcontrolled trial with Lactobacillus GG,” Gut, vol. 51, no. 3,pp. 405–409, 2002.

    [90] E. Garcia Vilela, M. De Lourdes De Abreu Ferrari, H.Oswaldo Da Gama Torres et al., “Influence of Saccharomycesboulardii on the intestinal permeability of patients withCrohn’s disease in remission,” Scandinavian Journal of Gas-troenterology, vol. 43, no. 7, pp. 842–848, 2008.

    [91] M. Guslandi, G. Mezzi, M. Sorghi, and P. A. Testoni, “Sac-charomyces boulardii in maintenance treatment of Crohn’s

    disease,” Digestive Diseases and Sciences, vol. 45, no. 7, pp.1462–1464, 2000.

    [92] J. B. Ewaschuk and L. A. Dieleman, “Probiotics and prebioticsin chronic inflammatory bowel diseases,” World Journal ofGastroenterology, vol. 12, no. 37, pp. 5941–5950, 2006.

    [93] C. Hedin, K. Whelan, and J. O. Lindsay, “Evidence for the useof probiotics and prebiotics in inflammatory bowel disease: areview of clinical trials,” Proceedings of the Nutrition Society,vol. 66, no. 3, pp. 307–315, 2007.

    [94] S. C. Ng, A. L. Hart, M. A. Kamm, A. J. Stagg, and S.C. Knight, “Mechanisms of action of probiotics: recentadvances,” Inflammatory Bowel Diseases, vol. 15, no. 2, pp.300–310, 2009.

    [95] K. Alfaleh and D. Bassler, “Probiotics for prevention of necro-tizing enterocolitis in preterm infants,” Cochrane Database ofSystematic Reviews, no. 1, article CD005496, 2008.

    [96] A. Bousvaros, S. Guandalini, R. N. Baldassano et al., “Arandomized, double-blind trial of lactobacillus GG versusplacebo i