You are on page 1of 13

Saccharomyces boulardii ameliorates Citrobacter rodentium -induced colitis through actions on bacterial virulence factors

X. Wu, B. A. Vallance, L. Boyer, K. S. B. Bergstrom, J. Walker, K. Madsen, J. R. O'Kusky, A. M. Buchan and K. Jacobson
Am J Physiol Gastrointest Liver Physiol 294:G295-G306, 2008. First published 21 November 2007; doi:10.1152/ajpgi.00173.2007 You might find this additional info useful... This article cites 62 articles, 28 of which can be accessed free at: http://ajpgi.physiology.org/content/294/1/G295.full.html#ref-list-1 This article has been cited by 6 other HighWire hosted articles, the first 5 are: Bacterial infection causes stress-induced memory dysfunction in mice Mlanie G Gareau, Eytan Wine, David M Rodrigues, Joon Ho Cho, Mark T Whary, Dana J Philpott, Glenda MacQueen and Philip M Sherman Gut, March, 2011; 60 (3): 307-317. [Abstract] [Full Text] [PDF] Promising Immunomodulatory Effects of Selected Strains of Dairy Propionibacteria as Evidenced In Vitro and In Vivo Benoit Foligne, Stephanie-Marie Deutsch, Jerome Breton, Fabien J. Cousin, Joelle Dewulf, Michel Samson, Bruno Pot and Gwenael Jan AEM, December, 2 2010; 76 (24): 8259-8264. [Abstract] [Full Text] [PDF] Perinatal lipid nutrition alters early intestinal development and programs the response to experimental colitis in young adult rats Sheila M. Innis, Chuanbin Dai, Xiujuan Wu, Alison M. J. Buchan and Kevan Jacobson Am J Physiol Gastrointest Liver Physiol, December, 2010; 299 (6): G1376-G1385. [Abstract] [Full Text] [PDF] Bacterial infection causes stress-induced memory dysfunction in mice Mlanie G Gareau, Eytan Wine, David M Rodrigues, Joon Ho Cho, Mark T Whary, Dana J Philpott, Glenda MacQueen and Philip M Sherman Gut, October, 21 2010; (): . [Abstract] [Full Text] [PDF] Probiotic bacteria and intestinal epithelial barrier function Christina L. Ohland and Wallace K. MacNaughton Am J Physiol Gastrointest Liver Physiol, June, 2010; 298 (6): G807-G819. [Abstract] [Full Text] [PDF] Updated information and services including high resolution figures, can be found at: http://ajpgi.physiology.org/content/294/1/G295.full.html Additional material and information about AJP - Gastrointestinal and Liver Physiology can be found at: http://www.the-aps.org/publications/ajpgi

Downloaded from ajpgi.physiology.org on March 18, 2011

This infomation is current as of March 18, 2011.

AJP - Gastrointestinal and Liver Physiology publishes original articles pertaining to all aspects of research involving normal or abnormal function of the gastrointestinal tract, hepatobiliary system, and pancreas. It is published 12 times a year (monthly) by the American Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright 2008 by the American Physiological Society. ISSN: 0193-1857, ESSN: 1522-1547. Visit our website at http://www.the-aps.org/.

Am J Physiol Gastrointest Liver Physiol 294: G295G306, 2008. First published November 21, 2007; doi:10.1152/ajpgi.00173.2007.

Saccharomyces boulardii ameliorates Citrobacter rodentium-induced colitis through actions on bacterial virulence factors
X. Wu,1,2 B. A. Vallance,1,2 L. Boyer,1,2 K. S. B. Bergstrom,1,2 J. Walker,3 K. Madsen,3 J. R. OKusky,4 A. M. Buchan,5 and K. Jacobson1,2,5
1

Children and Family Research Institute and 2Division of Gastroenterology, BC Childrens Hospital, Vancouver, British Columbia; 3Department of Medicine, University of Alberta, Edmonton, Alberta; and 4Department of Pathology and Laboratory Medicine and 5Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada

Submitted 18 April 2007; accepted in nal form 10 November 2007

Wu X, Vallance BA, Boyer L, Bergstrom KS, Walker J, Madsen K, OKusky JR, Buchan AM, Jacobson K. Saccharomyces boulardii ameliorates Citrobacter rodentium-induced colitis through actions on bacterial virulence factors. Am J Physiol Gastrointest Liver Physiol 294: G295G306, 2008. First published November 21, 2007; doi:10.1152/ajpgi.00173.2007.Saccharomyces boulardii has received increasing attention as a probiotic effective in the prevention and treatment of infectious and inammatory bowel diseases. The aim of this study was to examine the ameliorating effects of S. boulardii on Citrobacter rodentium colitis in vivo and identify potential mechanisms of action. C57BL/6 mice received 2.5 108 C. rodentium by gavage on day 0, followed by S. boulardii (25 mg; 5 108 live cells) gavaged twice daily from day 2 to day 9. Animal weights were monitored until death on day 10. Colons were removed and assessed for epithelial barrier function, histology, and myeloperoxidase activity. Bacterial epithelial attachment and type III secreted proteins translocated intimin receptor Tir (the receptor for bacterial intimin) and EspB (a translocation apparatus protein) required for bacterial virulence were assayed. In infected mice, S. boulardii treatment signicantly attenuated weight loss, ameliorated crypt hyperplasia (234.7 7.2 vs. 297.8 17.6 m) and histological damage score (0.67 0.67 vs. 4.75 0.75), reduced myeloperoxidase activity (2.1 0.4 vs. 4.7 0.9 U/mg), and attenuated increased mannitol ux (17.2 5.0 vs. 31.2 8.2 nm cm 2 h 1). The ameliorating effects of S. boulardii were associated with signicantly reduced numbers of mucosal adherent C. rodentium, a marked reduction in Tir protein secretion and translocation into mouse colonocytes, and a striking reduction in EspB expression and secretion. We conclude that S. boulardii maintained colonic epithelial barrier integrity and ameliorated inammatory sequelae associated with C. rodentium infection by attenuating C. rodentium adherence to host epithelial cells through putative actions on the type III secretion system. infectious colitis; probiotics; bacterial adherence; Tir; EspB

CITROBACTER RODENTIUM

is a gram-negative bacterium that colonizes the colons of mice, causing attaching/effacing (A/E) lesions and colonic hyperplasia (1, 42, 43, 51, 53). The lesions produced at the epithelial surface are indistinguishable from those caused by enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) infections, which are among the leading bacterial causes of human diarrheal diseases worldwide (57). While EPEC leads to the deaths of thousands of children in developing countries (14), EHEC is responsible for sporadic diarrheal outbreaks and deaths in North America

and Europe (35). C. rodentium, in addition to serving as a small-animal model for EPEC and EHEC infection, can also serve as a murine model for inammatory bowel disease (32, 33). C. rodentium, like EPEC and EHEC, attaches to luminal surfaces of host intestinal epithelial cells, effacing localized regions of microvilli and injecting bacterial effector proteins into the host cell via their locus of enterocyte effacement (LEE)-encoded syringelike type III secretion system (TTSS) (21, 23). Translocator proteins including the E. coli secreted proteins EspA, EspB, and EspD are responsible for the assembly of a translocation conduit in the host cell membrane that assists in the translocation of effector proteins into host cells (21, 41). Effector proteins, including the translocated intimin receptor Tir, which acts as the receptor for the bacterial outer membrane adhesin intimin, anchor the bacteria to host cells, modulate host cell functions, induce cytoskeletal rearrangements, and facilitate bacterial proliferation and disease development (9, 20, 28, 40, 48). In C. rodentium infection, Tir and intimin have been shown to be essential virulence factors (23, 25, 32), while EspB has been shown to be an important colonization factor (46) and component of the translocation apparatus necessary in translocation activity (41, 62). Furthermore, LEE-encoded SepL and SepD have been shown to regulate TTSS hierarchy of translocators and effectors, where SepL and SepD are not only necessary for efcient translocator secretion but also control a switch from translocator to effector secretion in response to environmental signals (21, 22). The ability of A/E pathogens to secrete LEE-encoded proteins into culture medium by using the LEE-encoded TTSS provides an excellent opportunity to examine the impact of potential therapies on these virulence factors (22). C. rodentium, in addition to providing an excellent in vivo model to investigate pathogen-host interactions under physiological conditions, also provides an opportunity to evaluate therapeutic interventions. Probiotics, which are nonpathogenic live microorganisms that promote favorable effects on health by altering indigenous microora, have been shown to be efcacious in C. rodentium infection. To date, in vivo probiotic studies in C. rodentium infection have focused on lactobacillus strains and their impact on host epithelial cell and inammatory responses (13, 34, 60). Pretreatment of mice in early life (13), in young adulthood (34, 60), or concurrently with inoculation of C. rodentium (13) has been shown to effectively alter
The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. G295

Downloaded from ajpgi.physiology.org on March 18, 2011

Address for reprint requests and other correspondence: K. Jacobson, Div. of Gastroenterology, BC Childrens Hospital, 4480 Oak St., Rm. K4-181, Vancouver, BC, Canada V6H 3V4 (e-mail: kjacobson@cw.bc.ca). http://www.ajpgi.org

0193-1857/08 $8.00 Copyright 2008 the American Physiological Society

G296

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

host regulatory immune responses, leading to amelioration of epithelial damage and attenuation of C. rodentium-induced colitis. While a ne balance exists between the effects of probiotics on attenuation of the hosts mucosal proinammatory responses and the induction of intestinal mucosal hyporesponsiveness to pathogenic determinants, a potential concern remains as to whether such modulation can result in adverse consequences such as delayed pathogen clearance. Thus probiotics that target pathogen virulence should also be considered. Some studies suggest that probiotics modulate pathogenhost epithelial cell interactions through antibacterial actions (3, 59), through competitive binding to sites on epithelial cells (54), through bacterial adherence to the probiotic (27), or through degradation of bacterial toxins (11, 12). While such studies suggest benecial effects through a reduction in pathogen-host interaction, it remains unclear whether such effects have potential benet over the potential risks associated with suppression of the hosts mucosal immune response. Saccharomyces boulardii is a thermophilic nonpathogenic yeast that has been used in vivo for both prevention and treatment of infectious and inammatory intestinal diseases (5, 29, 30, 37, 45). The benets of S. boulardii are strengthened by its ability to rapidly achieve high concentrations in the gastrointestinal tract after oral administration and to be maintained at stable levels in a viable form. Moreover, S. boulardii does not permanently colonize the colon and does not usually translocate across the intestinal mucosa (2, 4, 6). Several in vitro studies using S. boulardii have shown protective effects against EHEC and EPEC infection through various mechanisms including bacterial adherence to yeast via adhesions (27), preservation of epithelial barrier function, and modulation of signal transduction pathways (17, 18, 20, 55). Interestingly, while benecial effects were provided in these models, the studies have predominantly focused on host cellular responses, whereas the potential impact of S. boulardii on virulence of these pathogens has received limited attention. Consequently, the aim of the present study was to examine the efcacy of S. boulardii in ameliorating the intestinal disease associated with C. rodentium infection in vivo, and to determine whether its ameliorating effects are due in part to its actions on pathogenic determinants of the enteric pathogen.
MATERIALS AND METHODS

Mice. Six- to eight-week-old C57BL/6 mice weighing 18 20 g were purchased from Charles River Laboratories (St. Constant, QC, Canada) and were maintained on standard laboratory chow and tap water ad libitum. All protocols were approved by the Animal Research Ethics Boards at the University of British Columbia. Bacterial strains and infection of mice. Mice were orally inoculated with wild-type C. rodentium (formerly C. freundii biotype 4280), strain DBS100 (50). For inoculations, bacteria were grown overnight in Luria broth (LB). Mice were infected by oral gavage with 100 l of LB containing 2.5 108 colony-forming units (CFU) of C. rodentium. Control mice received 100 l of fresh LB by oral gavage. The mice were killed on postinfection (p.i.) day 10. Day 10 p.i. was chosen because at this time the infection is well established and colitis is evident (43). For the in vitro studies, the escN mutant strain of C. rodentium (type III apparatus mutant that does not secrete Tir or EspB) was generated as previously described (22) and included as a negative control for assessment of these virulence factors.
AJP-Gastrointest Liver Physiol VOL

Probiotic treatment in vivo. S. boulardii was supplied in lyophilized form by MFI Pharma (Richmond Hill, ON, Canada). Each capsule contained 250 mg of viable S. boulardii cells ( 5 109). Mice received 100 l of 250 mg/ml S. boulardii in saline by oral gavage twice daily from day 2 until death on day 10. The dose chosen was in keeping with doses used previously in murine models (8, 10, 24). Control mice received saline by oral gavage. Preparation of S. boulardii culture supernatant. S. boulardii was rst cultured in Sabouraud dextrose (SD) broth (100 mg/ml) overnight at 37C. The yeast culture was then centrifuged at 5,000 rpm for 30 min at 4C, and the supernatant was collected. The supernatant was then passed twice through a 0.2- m-pore size lter to remove the yeast cells. The supernatant was included to ascertain whether the in vitro actions of S. boulardii were mediated through a secreted factor(s). Assessment of colonic response to infection. Mice were weighed every second day and killed on day 10 p.i. The colons were removed and examined for total and mucosal associated C. rodentium (n 12/group), histological damage score, crypt heights, goblet cell response, myeloperoxidase (MPO) activity (n 6/group), and epithelial ion transport and permeability (n 12/group). Histological analysis: damage score and crypt height. Colons were excised, and distal colonic segments (0.5 cm) were removed, xed with 10% neutral buffered formalin for 24 h, and then transferred to 70% ethanol. Fixed tissues were embedded in parafn. Cross sections of the colon (5 m) were cut and mounted on slides. Tissue sections were stained with hematoxylin and eosin. Cell morphology was observed under light microscopy. Histological damage scoring was determined with criteria adapted from Galeazzi et al. (26). Briey, the damage score consisted of a score for the severity of epithelial injury (graded 0 3, from absent to mild including supercial epithelial injury, moderate including focal erosions, and severe including multifocal erosions), the extent of inammatory cell inltrate (graded 0 3, from absent to transmural), and goblet cell depletion (0 1). In each case a numerical score was assigned. Three tissue sections from each animal were coded and examined by two blinded observers to prevent observer bias. Tissue sections were assessed (each separated by at least 500 m) under a Nikon Eclipse 400 light microscope and averaged to obtain a mean histological damage score. Crypt heights were measured by micrometry, with 10 measurements taken in distal colon sections of each mouse. Only well-oriented crypts and tissue sections with intact muscularis propria were measured. Myeloperoxidase activity. The activity of MPO, an enzyme produced by neutrophils, was measured. Distal colonic segments adjacent to tissues retrieved for histological damage were excised, snap-frozen in liquid nitrogen, and stored at 80C. MPO activity was measured within 7 days by a previously described method (7). Briey, tissues were homogenized in hexadecyltrimethylammonium bromide buffer (Sigma) and centrifuged. The supernatant was added to a solution containing o-dianisidine (Sigma) and hydrogen peroxide. The absorbance of the colorimetric reaction was measured by a spectrophotometer. MPO is expressed in units per milligram of wet tissue, with 1 unit being the quantity of enzyme able to convert 1 mol of hydrogen peroxide to water in 1 min at room temperature. Epithelial function. Colonic epithelial barrier function was measured in accordance with well-established protocols (15, 44). Mice were anesthetized with halothane and then killed by cervical dislocation. Distal colonic segments were removed and mounted in Lucite chambers exposing mucosal and serosal surfaces to 10 ml of oxygenated Krebs buffer (in mM: 115 NaCl, 8 KCl, 1.25 CaCl2, 1.2 MgCl2, 2 KH2PO4, 225 NaHCO3; pH 7.35). The buffer was maintained at 37C by a heated water jacket and circulated with CO2-O2. Fructose (10 mM) was added to the serosal and mucosal surfaces. For measurement of basal mannitol uxes, 1 mM mannitol with 10 Ci of 3H was added to the mucosal side and samples of the buffer from both the serosal and mucosal sides were taken at 5-min intervals. The ux was then calculated with a published protocol (44).
294 JANUARY 2008

Downloaded from ajpgi.physiology.org on March 18, 2011

www.ajpgi.org

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

G297

Spontaneous transepithelial potential difference (PD) was determined, and the tissue was clamped at zero voltage by continuously introducing an appropriate short-circuit current (Isc) with an automatic voltage clamp (DVC 1000, World Precision Instruments, New Haven, CT), except for 510 s every 5 min when PD was measured by removing the voltage clamp. Stimulated increase in Isc was induced by addition of the adenylate cyclase-activating agent forskolin (10 5 M) to the serosal surface. Epithelial responsiveness was dened as the maximal increase in Isc to occur within 5 min of exposure to secretagogue. Transmission electron microscopy. Segments of colon, measuring 6 8 mm in length, were xed by immersion in a xative solution containing 4% paraformaldehyde (PFA) and 1% glutaraldehyde in 0.1 M phosphate buffer (pH 7.4) at 4C for 24 48 h. Tissue blocks were washed in 0.1 M phosphate buffer and postxed for 12 h in 1% buffered osmium tetroxide. After being washed in acetate buffer for 1 h, the blocks were stained with 2% uranyl acetate for 12 h, followed by dehydration in ascending grades of ethanol, equilibration in propylene oxide, and embedding in Epon (Jembed 812, Canemco, Montreal, QC, Canada). Semithin sections were cut at right angles to the long axis of the intestine at a thickness of 0.7 m, mounted on glass slides, and stained with 1% toluidine blue in 0.4% sodium borate. After the blocks were trimmed, ultrathin sections were cut at a thickness of 60 65 nm, mounted on Formvar-coated slot grids, and stained with lead citrate. Sections were examined with a Morgagni transmission electron microscope (FEI). Bacterial counts. Colonic tissues including luminal contents (total bacterial counts) and colonic tissues after colonic contents were vigorously ushed away with sterile phosphate-buffered saline (PBS) (mucosal associated bacterial counts) were homogenized in 1.5 ml of sterile PBS at low speed with a Kinematica tissue homogenizer (Brinkmann). Homogenates were then serially diluted and plated onto MacConkey agar plates selective for gram-negative organisms (PML Microbiologicals). Bacterial colonies were enumerated after overnight incubation at 37C. C. rodentium colonies were easily distinguished from colonies derived from commensal ora by their size and appearance. The validity of this approach was previously veried by PCR analysis for LEE genes (58). Bacterial counts are reported as colonyforming units per gram. Western blot analysis of Tir in colonic tissues. To investigate the effect of S. boulardii on bacterial attachment to epithelial cells, Western blot analysis of Tir, an essential virulence factor needed for bacterial attachment, was performed (23). Briey, colons were opened and stool pellets gently removed, followed by vigorous washing with cold PBS to remove nonadherent bacteria and homogenization with lysis buffer [in mM: 150 NaCl, 20 Tris pH 7.5, 1 EDTA, 2.5 sodium pyrophosphate, 1 -glycerophosphate, 1 phenylmethylsulfonyl uoride (PMSF), 1 sodium orthovanadate, and 1 sodium uoride, with 1% Triton X-100, 1% mammalian protease inhibitor cocktail (Sigma), and 1% bacterial protease inhibitor cocktail (Sigma)]. Thirty micrograms of each protein sample was loaded and resolved by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) after transfer to nitrocellulose and blocking in a 5% solution of dried skim milk. Membranes were then probed with rat polyclonal antibody against His-tagged Citrobacter Tir. Primary antibodies were probed with goat anti-rat horseradish peroxidase-conjugated IgG (Santa Cruz Biotechnology) for 60 min at room temperature. The presence of antibodies was assessed by an enhanced chemiluminescence detection system (Amersham, Little Chalfont, UK). The intensity of bands was quantied with NIH Scion Image software. Immunouorescence staining of LPS and translocated Tir in colonic tissues. To further examine the association between bacterial attachment to epithelial cells and Tir translocation, immunouorescence staining of colonic tissues was performed with standard techniques as described previously (23). Briey, colonic tissues were rinsed with ice-cold PBS, embedded in Shandon cryomatrix (Thermo Electron, Pittsburgh, PA), frozen with isopentane (Sigma) and liquid
AJP-Gastrointest Liver Physiol VOL

N2, and stored at 80C. Serial sections were cut at a thickness of 6 m and xed in 4% PFA for 15 min at room temperature. Tissue sections were directly blocked with 1% bovine serum albumin for 1 h at room temperature, followed by the addition of the rat polyclonal anti-His-tagged Citrobacter Tir sera (kindly provided by Dr. W. Deng, Biotechnology Laboratory, University of British Columbia, Vancouver, BC, Canada) as well as the rabbit polyclonal anti-LPS (E. coli Poly8, Biotech Laboratories) and incubated at 4C for 4 h. After extensive washing with PBS, Alexa Fluor 568-conjugated goat antirat IgG (Molecular Probes, Eugene, OR) as well as Alexa Fluor 488-conjugated goat-anti-rabbit IgG (Molecular Probes) were added and incubated for 1 h at room temperature. The tissues were again washed and mounted with ProLong Gold Antifade (Invitrogen) mounting medium containing 4 ,6-diamidino-2-phenylindole (DAPI) for staining host cell DNA. Coverslips were viewed at 350, 488, and 594 nm on a Zeiss AxioImager microscope. Images were obtained with a Zeiss AxioImager microscope equipped with an AxioCam HRm camera operating through AxioVision software (version 4.4). Western blot analysis of total and secreted proteins from C. rodentium in vitro. To investigate the effect of S. boulardii on secretion of type III proteins by C. rodentium, total and secreted Tir and EspB proteins were assessed as described previously (23) but modied with the addition of S. boulardii. Briey, C. rodentium strain DBS100 was grown overnight in LB at 37C. Eighty microliters of C. rodentium culture was cocultured with 80 l of 100 mg/ml viable S. boulardii or S. boulardii supernatant in 4 ml of plain DMEM (preincubated overnight in a 5% CO2 incubator) to a 1:50 dilution. Cocultures were incubated in upright test tubes in a tissue culture incubator for 6 h to an optical density at 600 nm (OD600) of 0.7. Cocultures were then centrifuged at 13,000 rpm. for 20 min at 4C to remove bacteria, and the supernatant was precipitated with 10% trichloroacetic acid (TCA) to concentrate proteins secreted into the culture medium. The remaining bacteria pellets were dissolved in 2 SDSPAGE buffer and designated as total bacterial proteins. The secreted proteins precipitated from the supernatant were also dissolved in 2 SDS-PAGE buffer, and the residual TCA was neutralized with 1 l of saturated Tris. The volumes of buffer used to resuspend bacterial pellets and secreted proteins were normalized to the OD600 of the subcultures (when cultured with S. boulardii supernatant) or to bacterial numbers counted by the MacConkey agar plate method described above (when cultured with S. boulardii) to ensure equal loading of samples. The proteins were analyzed in 10% SDS-PAGE and processed for Western blotting analysis with rat polyclonal antibodies against the His-tagged Citrobacter Tir and mouse monoclonal antibody against Citrobacter EspB (kindly provided by Dr. W. Deng). RT-PCR and quantitative real-time PCR analysis of expression of tir and espb genes. The effect of S. boulardii on the expression of tir and espb genes in C. rodentium was assessed by RT-PCR as described previously (21) and by real-time PCR analysis (16, 38). C. rodentium was treated similarly to the growth conditions described above for total and secreted protein assays. After induction in DMEM in the absence or presence of S. boulardii or its supernatant for 6 h in a 5% CO2 tissue culture incubator, bacterial cultures were centrifuged and the bacterial pellet used for isolating total RNA. Bacterial RNA was stabilized with RNAprotect Bacteria Reagent (Qiagen). Bacteria were lysed with lysozyme (Sigma) in Tris-EDTA (TE) buffer to exclude interference with S. boulardii, and total RNA was extracted with the RNeasy mini kit (Qiagen) according to the manufacturers instructions. The RNA samples were treated with DNase I (1 U of DNase I per 1 g of RNA) to eliminate any contaminating DNA as described previously (38). Equal amounts of the total RNA (1 g) for each sample were then used for reverse transcription (RT) with Superscript reverse transcriptase and random primers from Invitrogen according to the suppliers instructions. No contamination by genomic DNA was detectable by RT-PCR and real-time PCR for Tir and EspB after DNase I treatment performed with two negative (no RT and no
294 JANUARY 2008

Downloaded from ajpgi.physiology.org on March 18, 2011

www.ajpgi.org

G298

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

cDNA) control reactions (data not shown). Real-time PCR was validated by comparing Tir expression by C. rodentium grown in LB and DMEM media as described previously (38), with a 4.5-fold increase in DMEM over that observed in LB medium. Quantitative real-time PCR was performed with IQ SYBR Green Supermix (Bio-Rad) and a Bio-Rad MJ Mini-Opticon Real-Time PCR System (Bio-Rad). Primers were designed by Primer3 (version 4.0) and tested for degeneracy with the BLAST program. The primers used were EspB: forward 5 -AAACTGATGCGTGAGATGGTC-3 and reverse 5 -CTTCAGAGGCGGTATTGACAG-3 ; Tir: forward 5 GCCGACAGAACAGACAATAGC-3 and reverse 5 -ACATCCAACCTTCAGCATACG-3 ; and 16S forward 5 -aggccttcgggttgtaaagt-3 and reverse 5 -gactcaagcctgccagtttc-3 . Cycling conditions were as follows: denaturation for 5 min at 95C and amplication for 40 cycles at 95C for 30 s, 60C for 30 s, and at 72C for 40 s. After completion of the cycling process, samples were subjected to a temperature ramp (from 53 to 95C) with continuous uorescence monitoring for melting curve analysis. For each PCR product, a single narrow peak was obtained by melting curve analysis at the specic melting temperature, indicating specic amplications. Primer pair efciency was tested by looking at how Ct [the difference between the 2 threshold cycle (Ct) values of 2 PCRs for the same template amount] varied with template dilution, as suggested by the manufacturers instruction guide. Quantication was carried out with Gene Ex Macro OM 3.0 software (Bio-Rad) where PCR efciencies for each of the primer sets were incorporated into the nal calculation. The Ct method was used to calculate the relative amount of specic RNA present in a sample, from which the fold induction of transcription of the gene was estimated by comparison to values relative to the wild-type strain grown in DMEM medium. Data are expressed as means SD. Statistical analysis. Results are expressed as means SE of separate experiments, except for real-time PCR analysis, where results are expressed as means SD. Analyses were conducted with Graph Pad Prism 4 statistical software for Windows (GraphPad Software, San Diego, CA). Differences between means were calculated by one-way analysis of variance or paired t-tests where appropriate. Specic differences were tested with the Student-Neuman-Keuls test. A P value 0.05 was considered statistically signicant.
RESULTS

Fig. 1. Effect of Citrobacter rodentium infection and Saccharomyces boulardii treatment on body weight. Body weights were monitored every other day until death on day 10 in noninfected control animals (CTL), in animals that received S. boulardii (Sb), in C. rodentium-infected animals (CR), and in C. rodentium-infected animals treated with S. boulardii (CR Sb). Compared with noninfected control animals, C. rodentium-infected mice demonstrated signicant weight loss by day 2 postinfection (p.i.), with weights decreasing further to reach their lowest levels at day 4 p.i. and increasing thereafter but remaining signicantly lower at day 10 p.i. Treatment with S. boulardii in C. rodentium-infected animals abrogated weight loss such that weights were signicantly higher than those observed in C. rodentium-infected animals by day 4 p.i. By day 8 p.i. body weight had returned to levels that were not signicantly different from those in control animals. S. boulardii treatment in control animals had no adverse effects on weight gain. Results are expressed as means SE for 12 animals/group. *P 0.05, **P 0.004 compared with control; P 0.01 compared with S. boulardii-treated C. rodentium-infected animals.

Downloaded from ajpgi.physiology.org on March 18, 2011

S. boulardii treatment ameliorates C. rodentium-induced morbidity. C. rodentium-infected mice showed overt clinical signs of infection, including a hunched posture, rapid loss of body weight, and defecation of soft stool. Signicant weight loss was evident by day 2 p.i. (P 0.004, Fig. 1), with weights reaching their lowest levels by day 4 p.i. (P 0.001, Fig. 1). Thereafter, animal weights increased but still remained significantly below control levels at day 10 p.i. (P 0.001). Treatment with S. boulardii abrogated most clinical signs of infection and signicantly attenuated the weight loss such that on day 4 p.i. animals demonstrated a weight reduction of only 13.2% compared with 31.5% in infected untreated mice (Fig. 1, P 0.01). Moreover, weight recovery was signicantly enhanced in S. boulardii-treated animals, with levels increasing more rapidly, resulting in weights that were not signicantly different from those in control animals by day 8 p.i. In contrast, infected untreated animals demonstrated poor weight recovery, with signicant weight differences still evident on day 10 p.i. compared with control (P 0.01) and S. boulardii treated (P 0.01) animals. Weight gain in mice given S. boulardii alone paralleled that seen in control animals. S. boulardii treatment ameliorates C. rodentium-induced colitis. In keeping with previous studies (1, 43), C. rodentium infection on day 10 p.i. was associated with supercial epitheAJP-Gastrointest Liver Physiol VOL

lial damage, goblet cell depletion, colonic crypt hyperplasia, and the presence of a mild transmural inammatory inltrate comprised of neutrophils and mononuclear cells (Fig. 2 and Fig. 3, A and B). These histological changes were more marked in the distal than proximal colon (data not shown), consistent with the predominance of bacterial colonization in the distal colon (58, 61). S. boulardii treatment attenuated the colitic response associated with C. rodentium infection. S. boulardii treatment ameliorated infection-induced epithelial damage and reduced transmural inltration of neutrophils and lymphocytes (Fig. 2, C and D). Where C. rodentium infection was associated with a marked increase in histological damage score (P 0.001 compared with control, Fig. 3A), S. boulardii treatment significantly attenuated the damage score to a level that was not signicantly different from that in control animals. Moreover, the 47-fold increase in MPO activity observed with C. rodentium infection (P 0.001 compared with control, Fig. 3B) was signicantly abrogated by S. boulardii treatment (P 0.001); however, MPO levels remained signicantly elevated above control values (22-fold increase, P 0.01). In contrast to the typical goblet cell distribution along the length of colonic crypts, infected tissues displayed only a few mature goblet cells scattered predominantly at the surface of crypts, whereas S. boulardii treatment abrogated goblet cell depletion, resulting in preservation of goblet cells with the majority having a well-differentiated phenotype and containing abundant mucus (data not shown). In addition, S. boulardii treatment signicantly attenuated C. rodentium-associated crypt hyperplasia (P 0.01, Fig. 3C); however, crypt lengths still remained signicantly elevated above control levels (P 0.05). Notably, on its own, S. boulardii had no demonstrable effects on any of the parameters assessed (Figs. 2B and 3). Together, these data
294 JANUARY 2008

www.ajpgi.org

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

G299

Fig. 2. Histological appearance of tissue sections for noninfected control (A), S. boulardii-treated (B), and C. rodentium-infected (C) animals and S. boulardii treatment in C. rodentium-infected animal (D). A: normal histological appearance. B: normal histological appearance. C: supercial epithelial damage, colonic crypt hyperplasia and goblet cell depletion, thickening of the muscularis propria, and mild transmural inltration of neutrophils and mononuclear cells. D: evidence of a well-organized and intact epithelial cell layer, presence of goblet cells, absence of crypt hyperplasia and thickening of the muscularis propria, and no increase in inammatory cells.

Downloaded from ajpgi.physiology.org on March 18, 2011

demonstrated that oral treatment with S. boulardii signicantly attenuated C. rodentium-induced colitis. S. boulardii treatment attenuates epithelial pathophysiology. Considering that A/E pathogens cause diarrhea, we sought to assess colonocyte function during infection and the impact of S. boulardii on the pathophysiology. Infected mice showed signicant increases in unidirectional mannitol uxes compared with control mice (P 0.01, Fig. 4A). In contrast, treatment with S. boulardii prevented the infection-induced increase in mannitol uxes, resulting in levels that were not signicantly different from those in control mice (Fig. 4A). Similarly, the increase in mannitol ux observed with infection in the proximal colon was attenuated by treatment with S. boulardii (data not shown). S. boulardii treatment on its own had no effect on mannitol uxes. Additionally, C. rodentium caused a signicant reduction in basal Isc (P 0.05, Fig. 4B), a trend toward a decrease in PD (3.55 0.24 mV infected vs. 4.24 0.61 mV control animals), and a signicant 37% reduction in Isc response to forskolin (P 0.05, Fig. 4C), suggesting impairment in adenosine 3 ,5 -cyclic monophosphate-dependent active chloride secretion and signicant impairment in physiological function. Treatment with S. boulardii prevented infection-induced alteration in colonic ionic transport function such that baseline Isc and Isc response to forskolin remained at levels similar to those in control mice (Fig. 4, B and C). Similarly, PD remained at levels similar to baseline (4.28 0.43 infected S. boulardii treated vs. 4.24 0.61 control animals). Likewise, in the proximal colon, the above parameters were reduced with infection, while this reduction was attenuated by treatment with S. boulardii (data not shown). Interestingly, S. boulardii treatment in control mice resulted in a trend toward increased basal Isc and Isc responses to forskolin (Fig. 4, B and C).
AJP-Gastrointest Liver Physiol VOL

On transmission electron microscopy, as demonstrated previously, C. rodentium infection was associated with marked microvillus effacement and intimate attachment of C. rodentium to epithelial cells (A/E lesions, Fig. 5). S. boulardii treatment reduced bacterial attachment accompanied by attenuation in epithelial changes associated with infection (Fig. 5). While the length of microvilli appeared quite variable in each animal in part because of the angle of specimen orientation, S. boulardii treatment led to an obvious attenuation in microvillus effacement (Fig. 5). S. boulardii treatment reduces mucosal adherence of C. rodentium. We next sought to determine whether S. boulardii had an effect on the numbers of total and/or mucosal adherent C. rodentium in the colon. To address this question total colonic and mucosal associated bacterial CFUs were enumerated after overnight incubation of homogenates on MacConkey agar plates. As shown in Table 1, S. boulardii treatment was associated with a 2.4-fold decrease in total C. rodentium counts compared with untreated infected mice; however, the decrease was not signicant. In contrast, S. boulardii treatment signicantly reduced the number of mucosal adherent C. rodentium, associated with a 5.9-fold decrease compared with untreated infected animals (P 0.05, Table 1). S. boulardii had no bactericidal effect. To examine whether the reduction of mucosal adherent bacteria was due to a bactericidal effect of S. boulardii, we investigated its impact on bacterial growth in vitro. C. rodentium was cocultured with S. boulardii in DMEM for 6 h, after which bacteria was serially diluted and plated onto MacConkey agar plates. The number of bacteria was enumerated after overnight incubation at 37C. The number of bacteria did not differ signicantly between C. rodentium cocultured with or without S. boulardii (7.19 0.87 108 vs. 7.04 0.63 108 CFU/ml, means SE, P
294 JANUARY 2008

www.ajpgi.org

G300

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

Downloaded from ajpgi.physiology.org on March 18, 2011

Fig. 3. A: histological damage score measured on day 10 in noninfected control animals (CTL), in animals that received S. boulardii alone (Sb), in C. rodentium-infected animals (CR), and in C. rodentium-infected animals treated with S. boulardii (CR Sb). C. rodentium infection was associated with a signicant increase in histological damage score compared with control. S. boulardii treatment in C. rodentium-infected animals signicantly attenuated the increase in damage score such that the damage score was not signicantly different from control levels. S. boulardii treatment alone was not associated with a signicant change in damage score. Results are expressed as means SE for 6 animals/group. **P 0.001 compared with control; P 0.001 compared with C. rodentium-infected animals. B: myeloperoxidase (MPO) activity measured on day 10. C. rodentium infection was associated with a signicant increase in MPO activity compared with control. S. boulardii treatment in C. rodentium-infected animals signicantly attenuated the increase in MPO activity compared with C. rodentium-infected animals; however, MPO activity remained signicantly elevated above control levels. S. boulardii treatment alone was not associated with a signicant change in MPO activity. Results are expressed as means SE for 6 animals/group. **P 0.001 compared with control; P 0.001 compared with C. rodentiuminfected and control animals. C: crypt height measured on day 10. C. rodentium infection was associated with a signicant increase in crypt height compared with control animals. S. boulardii treatment in C. rodentium-infected animals signicantly attenuated the increase in crypt height; however, crypt height remained signicantly elevated above control levels. S. boulardii treatment alone was not associated with a signicant change in crypt height. Results are expressed as means SE for 6 animals/group. *P 0.05, **P 0.01 compared with control; P 0.05 compared with C. rodentium-infected animals.

Fig. 4. Colonic barrier function and electrical parameters in noninfected control animals (CTL), S. boulardii-treated animals (Sb), C. rodentiuminfected animals (CR), and C. rodentium-infected animals treated with S. boulardii (CR Sb). C. rodentium infection was associated with a signicant increase in unidirectional mannitol ux (A) and a signicant reduction in short-circuit current (Isc; B) and Isc response to forskolin (C). Treatment with S. boulardii in C. rodentium-infected animals prevented the increase in unidirectional mannitol ux (A) and the reduction in Isc (B) and Isc response to forskolin (C) such that these parameters were not signicantly different from control. S. boulardii treatment alone resulted in a trend toward increased basal Isc (B) and Isc response to forskolin (C). Results are expressed as means SE for 12 animals/group. *P 0.05, **P 0.01 compared with control animals; P 0.05 compared with C. rodentium-infected animals.

0.45). Similarly, culture with S. boulardii supernatant prepared with SD broth did not affect bacterial viability (data not shown). S. boulardii treatment reduces Tir translocation in vivo. It was apparent that while high numbers of C. rodentium remained within the lumen of infected mouse colon after treatment with S. boulardii, proportionally fewer bacteria were found adherent to the mucosa. Moreover, S. boulardii was not found to reduce bacterial numbers through a bactericidal action. Thus we next sought to determine whether the action of S. boulardii was mediated through an effect on C. rodentium attachment, through effects on the expression of the TTSS
294 JANUARY 2008

AJP-Gastrointest Liver Physiol VOL

www.ajpgi.org

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

G301

Fig. 5. Transmission electron micrographs of representative mouse colonic epithelium from (from top to bottom) a normal, uninfected mouse, a mouse treated with S. boulardii, a mouse infected with C. rodentium, and a C. rodentium-infected S. boulardii-treated mouse. While the normal uninfected and S. boulardii-treated mice showed healthy microvilli, infected mice demonstrated intimately attached bacteria and effacement of microvilli. In contrast, C. rodentium-infected and S. boulardii-treated mice showed an obvious attenuation in microvillus effacement (bottom). Calibration bars, 0.3 m.

cated Tir in the colonic tissues of C. rodentium-infected mice with or without S. boulardii treatment was performed. To selectively stain for translocated Tir, rather than Tir within bacteria, we stained for Tir for 4 h without using detergent to permeabilize bacteria as previously described (23). In the absence of S. boulardii, we found large numbers of adherent C. rodentium coating the luminal surface of epithelial cells (Fig. 6, BD). Where antisera raised against LPS visualized adherent bacteria (Fig. 6B) attached to epithelial cells, anti-Tir staining demonstrated translocated Tir within colonic epithelial cells (Fig. 6, C and D) beneath the adherent bacteria. In contrast, S. boulardii treatment in most C. rodentium-infected tissues section was associated with a marked reduction in the number of C. rodentium that were attached to the epithelium, as well as the detection of translocated Tir (Fig. 6, EG). Patchy areas where C. rodentium was attached to epithelial cells in association with translocated Tir were occasionally evident; however, most C. rodentium were found within the lumen or mucous layer (conrmed on gram stain, data not shown) and were not associated with Tir expression (Fig. 6, EG). The antisera raised against LPS did not demonstrate adherent bacteria in uninfected mice (data not shown). These results conrm and advance earlier results indicating that S. boulardii, while not associated with a signicant reduction in total bacteria, signicantly reduced adherent bacterial numbers and translocated Tir. S. boulardii treatment reduces total and secreted EspB protein and secreted Tir protein in vitro. Since S. boulardii treatment reduced the number of attached C. rodentium, we next reasoned that the observed action of S. boulardii was mediated through effects on the TTSS. To address this question, we took advantage of previous methodologies using a dened in vitro culture method for assessment of total and secreted Tir and EspB proteins (23). Western blot analysis conrmed Tir protein expression by C. rodentium (Fig. 7, A and C) and Tir protein secretion into the culture medium (Fig. 7, B and D). In contrast, coculture of S. boulardii with C. rodentium was associated with a signicant reduction in secreted Tir protein to almost undetectable levels (Fig. 7, B and D), whereas no effect on bacterial Tir protein expression was observed (Fig. 7, A and C). Consistent with previous studies, the type III secretion system mutant escN demonstrated expression of Tir but no evidence of secretion (Fig. 7) (21, 23). Similarly, C. rodentium was shown to express (Fig. 7, A and C) and secrete (Fig. 7, B and D) EspB protein, whereas coculture of S. boulardii with C. rodentium was associated with a signicant reduction in the secretion of EspB protein (Fig. 7, B and D) but also accompanied by a signicant reduction in bacterial expression of EspB (Fig. 7, A and C). The type III secretion system mutant escN

Downloaded from ajpgi.physiology.org on March 18, 2011

effector Tir. Western blot analysis of C. rodentium-infected mouse colon lysates (after washing away nonadherent bacteria and homogenizing with lysis buffer containing 1% Triton X-100 to determine total Tir) (50) indicated that protein levels of total Tir were markedly downregulated, by approximately threefold, in the presence of S. boulardii (Fig. 6A), suggesting that Tir expression was reduced in the presence of S. boulardii. While Tir is expressed within bacteria, it only impacts on bacterial attachment after its translocation into host cells. To determine whether translocated Tir was reduced during S. boulardii treatment, immunouorescent staining for transloAJP-Gastrointest Liver Physiol VOL

Table 1. Total and mucosal associated bacterial counts in the colons of C57BL/6 mice with or without Saccharomyces boulardii treatment
Total Bacterial Counts, 108 CFU/g Mucosal Associated Bacterial Counts, 108 CFU/g

Treatment Group

C. rodentium C. rodentium

S. boulardii

8.88 1.20 3.76 1.13

7.39 1.78 1.25 0.23*

Values are means SE. *P 0.05 compared with Citrobacter rodentiuminfected mice. CFU, colony-forming unit.
294 JANUARY 2008

www.ajpgi.org

G302

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

Fig. 6. A: colonic lysates (after washing away nonadherent bacteria) were assessed for total Tir protein by Western blot analysis in control (CTL), S. boulardii-treated (Sb), C. rodentiuminfected (CR), and S. boulardii-treated and C. rodentium-infected (CR Sb) animals. -Actin antibody was used as the loading control. Treatment with S. boulardii was associated with a marked reduction in protein levels of translocated Tir, with levels decreasing by 3fold. BG: immunostaining of C. rodentium LPS (green), translocated Tir (red), and DAPIstained epithelial cell nuclei (blue). B and C represent the colon of a CR mouse, with the merged image in D, and EG provide the corresponding images in a CR Sb animal. In the C. rodentium-infected tissue, note the bacteria lining the crypt epithelial cells (B) with Tir staining similarly positioned (C) but situated beneath LPS-stained bacteria (D). In contrast, in the CR Sb tissue, note the bacteria within the lumen (E) and with minimal Tir expression (G, H). Images were taken at 400 magnication, except for insets in D and F, which were taken at 1,000 magnication. Tir was stained for 4 h without any detergent to permeabilize the tissues. As a result, only translocated Tir was labeled.

Downloaded from ajpgi.physiology.org on March 18, 2011

demonstrated expression of total EspB protein, but again no evidence of secretion. To determine whether the action of S. boulardii on the TTSS was mediated by a secreted S. boulardii factor(s), S. boulardii

culture supernatant was added to C. rodentium in DMEM. The culture of S. boulardii supernatant with C. rodentium resulted in a reduction in EspB protein expression and secretion (Fig. 8) and in Tir secretion (Fig. 8, B and D) comparable to that

Fig. 7. Effect of S. boulardii on total and secreted C. rodentium Tir and EspB in vitro. A and B: Western blot analysis of total (A) and secreted (B) Tir and EspB proteins in C. rodentium in the presence (CR Sb) or absence (CR) of S. boulardii and escN (type III apparatus mutant that does not secrete Tir or EspB). C and D: quantication of total (C) and secreted (D) Tir (lled bars) and EspB (open bars) Western blots (representing a mean of 3 separate runs). Protein levels are expressed as % of untreated C. rodentium control. Coculture of C. rodentium with S. boulardii did not signicantly alter total Tir expression in C. rodentium (A, C), however, S. boulardii treatment signicantly inhibited Tir secretion in the culture medium even at low concentrations (B, D). In contrast, S. boulardii treatment signicantly downregulated total EspB expression (A, C), in addition to signicantly inhibiting EspB secretion into the culture medium (B, D). *P 0.001 compared with corresponding C. rodentium control. AJP-Gastrointest Liver Physiol VOL
294 JANUARY 2008

www.ajpgi.org

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

G303

Downloaded from ajpgi.physiology.org on March 18, 2011

Fig. 8. Effect of S. boulardii supernatant (SBS) on total and secreted C. rodentium Tir and EspB in vitro by Western blot analysis. C. rodentium was cocultured with S. boulardii supernatant (SBS) in serial dilutions, from 1:20 to 1:400 with DMEM. Total (A, C) and secreted (B, D) Tir (lled bars) and EspB (open bars) Western blots were quantied (representing a mean of 3 separate runs) and protein levels expressed as % of untreated C. rodentium control. First and second lanes, C. rodentium cultured in DMEM and C. rodentium cultured in DMEM and S. boulardii culture medium [Sabouraud dextrose (SD) broth] at a dilution of 1:50. Total and secreted Tir and EspB proteins were similarly detected in both culture media (A, B). Consistent with the observations in Fig. 7, SBS in culture with C. rodentium signicantly reduced Tir secretion in culture medium even at low concentrations (3rd 6th lanes, B; D) but failed to affect total Tir expression (3rd 6th lanes, A; C). In contrast, SBS signicantly downregulated total EspB expression (3rd 6th lanes, A; C) and secretion into the culture medium (3rd 6th lanes, B; D). Boiling the supernatant obliterated the inhibitory action of SBS. *P 0.001 compared with corresponding C. rodentium cultured in DMEM.

observed when C. rodentium was cocultured with S. boulardii (Fig. 7). The effect of the S. boulardii culture supernatant was dose dependent and evident at low concentrations (Fig. 8). Boiling the supernatant abolished the inhibitory action (Fig. 8), suggesting the presence of a heat-labile protein(s). Together, these data suggest that S. boulardii via a secreted protein(s) attenuated attachment of C. rodentium to host epithelial cells through effects on key virulence factors, namely, a reduction in expression and secretion of EspB and a reduction in secretion and translocation of Tir. S. boulardii inuences secretion but not transcription of EspB or Tir. To further explore the effects of S. boulardii on total and secreted Tir and EspB proteins, RT-PCR and realtime PCR analysis of Tir and EspB in wild-type C. rodentium in the absence or presence of S. boulardii were performed. No signicant difference in transcription of EspB or Tir was evident in C. rodentium in the absence or presence of S. boulardii (Fig. 9A, Table 2). Similarly, S. boulardii supernatant had no effect on the transcription of EspB and Tir in C.
AJP-Gastrointest Liver Physiol VOL

rodentium (Fig. 9B, Table 2). These results suggest that S. boulardii or its supernatant exerts a modulatory effect on EspB at a posttranscriptional level and further conrmed that S. boulardii or its supernatant inuences secretion but not transcription and translation of Tir.
DISCUSSION

To date, the studies of probiotics in ameliorating C. rodentium infection have focused on their impact on host responses, namely, intestinal epithelial cell, mucosal, and systemic inammatory responses (13, 17, 18, 24, 34). While the present study assessed host mucosal responses, the study also evaluated the impact of S. boulardii on C. rodentium pathogenic determinants. Specically, the study demonstrated a potentially novel action of S. boulardii through modulation of the effector protein Tir and the translocator protein EspB with resultant reduction of bacterial attachment to host intestinal epithelial cells. In addition, S. boulardii treatment ameliorated
294 JANUARY 2008

www.ajpgi.org

G304

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

Fig. 9. Effect of S. boulardii on transcription of EspB and Tir in C. rodentium. No signicant difference in transcription of EspB and Tir was found in C. rodentium in the absence and presence of S. boulardii (A). Similarly, S. boulardii supernatant had no effect on transcription of EspB and Tir (B).

bacteria-induced barrier disruption and attenuated the mucosal inammatory response. S. boulardii treatment markedly reduced bacterial adherence to host epithelial cells, enabling us to examine the relationship between epithelial barrier function and bacterial epithelial attachment in vivo. In keeping with recent observations (31), C. rodentium infection was associated with marked disruption in epithelial barrier function that was more pronounced in the distal colon, consistent with the degree of bacterial epithelial adherence and host mucosal response. It was apparent that the effective reduction in bacterial attachment to host epithelia and associated preservation of epithelial barrier function were likely largely responsible for the observed attenuation in mucosal inammatory response associated with C. rodentium infection. However, the additive effects of S. boulardii-mediated enhancement of epithelial barrier function and a shift from a primarily proinammatory Th1 cellular response to a more balanced Th1/Thr (regulatory) host immune response cannot be excluded (13, 1719, 34, 55, 60). The study demonstrated that S. boulardii treatment signicantly reduced Tir translocation into colonocytes of mice infected with C. rodentium in association with a reduction in mucosal associated bacteria, suggesting a potential mechanism of action through reduction in receptor-mediated attachment and anchoring of bacteria to host epithelial cells (23). While a bactericidal effect was ruled out as accounting for these changes, several additional mechanisms of action can be considered, including adherence of C. rodentium to luminal S. boulardii (27), physical interference at the epithelial surface as Table 2. Real-time PCR analysis of EspB and Tir gene expression in wild-type C. rodentium in absence or presence of S. boulardii or S. boulardii supernatant
Culture (in DMEM) EspB Tir

CR CR CR CR CR CR SB

DMEM (boiled) SB SBS SBS (boiled) Sabouraud dextrose broth

1.00 0.13 1.07 0.05 0.96 0.05 0.98 0.02 0.87 0.05 0.89 0.03 ND

1.00 0.11 1.06 0.05 0.97 0.04 1.01 0.02 0.92 0.06 0.91 0.03 ND

Values are means SD. Data indicate relative levels of transcription of EspB and Tir compared with wild-type C. rodentium (CR) grown in DMEM. CR were cocultured with S. boulardii (SB) or S. boulardii supernatant (SBS) in DMEM as described in MATERIALS AND METHODS. ND, not determined. AJP-Gastrointest Liver Physiol VOL

demonstrated in vitro with lactobacillus strains (54), or potential actions on innate defenses, such as increasing effectiveness of the mucus layer and thus reducing and/or preventing C. rodentium from reaching the epithelium, where it is thought that C. rodentium receives the signals to turn on its TTSS. Consistent with observations in conventional and gnotobiotic mice treated with S. boulardii and exposed to enteropathogens (49), S. boulardii treatment in the C. rodentium model was associated with an insignicant reduction in total colonic C. rodentium counts. The reduction in total colonic C. rodentium might be explained on the basis of reduced bacterial attachment to epithelial cells and increased expulsion of nonadherent bacteria in the stool. The in vitro culture system used to induce C. rodentium TTSS (23, 39) demonstrated a differential expression and secretion of Tir and EspB in the presence of S. boulardii. The signicant reduction in protein levels of secreted Tir together with a signicant reduction in expression and secretion of EspB protein suggest that the therapeutic benets of S. boulardii were mediated through actions on A/E pathogen virulence factors (23, 41, 46, 52). The observation that these actions were inhibited and the secretion prole restored by boiling the supernatant suggests the presence of heat-sensitive protein(s) secreted by S. boulardii. A key observation of our study was the demonstration that S. boulardii reduced EspB expression, in association with diminished secretion. The lack of effect on EspB transcription suggests posttranscriptional control at a level of translational regulation. Furthermore, S. boulardii may have modied the equilibrium between protein synthesis and degradation, leading to reduced intracellular levels of EspB associated with reduced secretion. Although SepL and SepD have been shown to modulate secretion of translocators including EspB, the action of S. boulardii was likely not mediated through actions on these proteins because in vitro studies with SepL and SepD mutants demonstrate normal expression of EspB but low levels of secreted EspB (21, 22). Another important nding in our study was that S. boulardii abolished Tir secretion. While S. boulardii may have altered the equilibrium between Tir protein synthesis and degradation with resultant reduced secretion, additional evidence suggests that the reduction in secreted Tir may be secondary to yeast protease digestion (11, 12, 56). Our observations also suggest that the effect of S. boulardii on EspB contributed to the reduced translocation of Tir into
294 JANUARY 2008

Downloaded from ajpgi.physiology.org on March 18, 2011

www.ajpgi.org

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS

G305

host cells in infected and treated mice. The potential mechanism of action is further supported by the observation that EspB in C. rodentium infection is required for colonization (46) and is an essential component of the translocation apparatus necessary in translocation activity (36, 41, 62). Moreover, in vitro studies indicate that while effector molecules can be expressed and secreted by bacteria in the absence of translocator proteins (23), translocation of effectors into host cells does not occur (41, 46, 47) and in vivo colonization fails (46). In conclusion, using a murine model of bacteria-induced colitis, we have demonstrated that S. boulardii is effective in attenuating virulence of C. rodentium by reducing bacterial attachment to host epithelial cells potentially through actions on the translocator protein EspB and the effector protein Tir. Further studies are required to determine the specic mechanisms involved.
ACKNOWLEDGMENTS The authors thank B. Davis from MFI Pharma for the unrestricted supply of S. boulardii and the technical staff of the Morphometry Laboratories (Nutrition Research Program, Child and Family Research Institute) for assistance with the electron microscopy. GRANTS This work was supported by a grant from the Crohns Colitis Foundation of Canada (awarded to K. Jacobson). K. Jacobson is a Senior Clinician Scientist supported by the Children with Intestinal and Liver Disorders (CHILD) and British Columbia Childrens Hospital Foundations. B. A. Vallance is the CHILD Foundation Research Scholar, a Michael Smith Foundation Health Research Scholar, and the Canada Research Chair in Pediatric Gastroenterology. REFERENCES 1. Barthold SW, Coleman GL, Jacoby RO, Livestone EM, Jonas AM. Transmissible murine colonic hyperplasia. Vet Pathol 15: 223236, 1978. 2. Berg R, Bernasconi P, Fowler D, Gautreaux M. Inhibition of Candida albicans translocation from the gastrointestinal tract of mice by oral administration of Saccharomyces boulardii. J Infect Dis 168: 1314 1318, 1993. 3. Bernet-Camard MF, Lievin V, Brassart D, Neeser JR, Servin AL, Hudault S. The human Lactobacillus acidophilus strain LA1 secretes a non-bacteriocin antibacterial substance(s) active in vitro and in vivo. Appl Environ Microbiol 63: 27472753, 1997. 4. Blehaut H, Massot J, Elmer GW, Levy RH. Disposition kinetics of Saccharomyces boulardii in man and rat. Biopharm Drug Dispos 10: 353364, 1989. 5. Bleichner G, Blehaut H, Mentec H, Moyse D. Saccharomyces boulardii prevents diarrhea in critically ill tube-fed patients. A multicenter, randomized double-blind, placebo-controlled trial. Intensive Care Med 23: 517 523, 1997. 6. Boddy AV, Elmer GW, McFarland LV, Levy RH. Inuence of antibiotics on the recovery and kinetics of Saccharomyces boulardii in rats. Pharm Res 8: 796 800, 1991. 7. Boughton-Smith NK, Wallace JL, Whittle BJ. Relationship between arachidonic acid metabolism, myeloperoxidase activity and leukocyte inltration in a rat model of inammatory bowel disease. Agents Actions 25: 115123, 1988. 8. Buts JP, Bernasconi P, Vaerman JP, Dive C. Stimulation of secretory IgA and secretory component of immunoglobulins in small intestine of rats treated with Saccharomyces boulardii. Dig Dis Sci 35: 251256, 1990. 9. Campellone KG, Robbins D, Leong JM. EspFU is a translocated EHEC effector that interacts with Tir and N-WASP and promotes Nck-independent actin assembly. Dev Cell 7: 217228, 2004. 10. Cartwright-Shamoon J, Dickson GR, Dodge J, Carr KE. Uptake of yeast (Saccharomyces boulardii) in normal and rotavirus treated intestine. Gut 39: 204 209, 1996. 11. Castagliuolo I, LaMont JT, Nikulasson ST, Pothoulakis C. Saccharomyces boulardii protease inhibits Clostridium difcile toxin A effects in the rat ileum. Infect Immun 64: 52255232, 1996. AJP-Gastrointest Liver Physiol VOL

12. Castagliuolo I, Riegler MF, Valenick L, LaMont JT, Pothoulakis C. Saccharomyces boulardii protease inhibits the effects of Clostridium difcile toxins A and B in human colonic mucosa. Infect Immun 67: 302307, 1999. 13. Chen CC, Louie S, Shi HN, Walker WA. Preinoculation with the probiotic Lactobacillus acidophilus early in life effectively inhibits murine Citrobacter rodentium colitis. Pediatr Res 58: 11851191, 2005. 14. Clarke SC, Haigh RD, Freestone PPE, Williams PH. Virulence of enteropathogenic Escherichia coli, a global pathogen. Clin Microbiol Rev 16: 365378, 2003. 15. Clarkson TW, Toole SR. Measurement of short-circuit current and ion transport across the ileum. Am J Physiol 206: 658 668, 1964. 16. Cordone A, Mauriello EMF, Pickard DJ, Dougan G, De Felice M, Ricca E. The Irp gene and its role in type I mbriation in Citrobacter rodentium. J Bacteriol 187: 7009 7017, 2005. 17. Czerucka D, Dahan S, Mograbi B, Rossi B, Rampal P. Saccharomyces boulardii preserves the barrier function and modulates the signal transduction pathway induced in enteropathogenic Escherichia coli-infected T84 cells. Infect Immun 68: 5998 6004, 2000. 18. Dahan S, Dalmasso G, Imbert V, Peyrn JF, Rampal P, Czerucka D. Saccharomyces boulardii interferes with enterohemorrhagic Escherichia coli-induced signaling pathways in T84 cells. Infect Immun 71: 766 773, 2003. 19. Dalmasso G, Loubat A, Dahan S, Calle G, Rampal P, Czerucka D. Saccharomyces boulardii prevents TNF- -induced apoptosis in EHECinfected T84 cells. Res Microbiol 157: 456 465, 2006. 20. de Grado M, Abe A, Gauthier A, Steele-Mortimer O, DeVinney R, Finlay BB. Identication of the intimin-binding domain of Tir of enteropathogenic Escherichia coli. Cell Microbiol 1: 717, 1999. 21. Deng W, Li Y, Hardwidge PR, Frey EA, Pfuetzner RA, Lee S, Gruenheid S, Strynakda NCJ, Puente JL, Finlay BB. Regulation of type III secretion hierarchy of translocators and effectors in attaching and effacing bacterial pathogens. Infect Immun 73: 21352146, 2005. 22. Deng W, Puente JL, Gruenheid S, Li Y, Vallance BA, Vazquez A, Barba J, Ibarra JA, ODonnell P, Metalnikov P, Ashman K, Lee S, Goode D, Pawson T, Finlay BB. Dissecting virulence: systematic and functional analyses of a pathogenicity island. Proc Natl Acad Sci USA 101: 35973602, 2004. 23. Deng W, Vallance BA, Li Y, Puente J, Finlay BB. Citrobacter rodentium translocated intimin receptor (Tir) is an essential virulence factor needed for actin condensation, intestinal colonization and colonic hyperplasia in mice. Mol Microbiol 48: 95115, 2003. 24. Dias RS, Bambirra EA, Silva ME, Nicoli JR. Protective effect of Saccharomyces boulardii against the cholera toxin in rats. Braz J Med Biol Res 28: 323325, 1995. 25. Frankel G, Phillips AD, Novakova M, Field H, Candy DCA, Schauer DB, Douce G, Dougan G. Intimin from enteropathogenic Escherichia coli restores murine virulence to a Citrobacter rodentium eaeA mutant: induction of an immunoglobulin A response to intimin and EspB. Infect Immun 64: 53155325, 1996. 26. Galeazzi F, Blennerhassett PA, Qiu B, OByrne PM, Collins SM. Cigarette smoke aggravates experimental colitis in rats. Gastroenterology 117: 877 883, 1999. 27. Gedek BR. Adherence of Escherichia coli serogroup O157 and the Salmonella typhimurium mutant DT 104 to the surface of Saccharomyces boulardii. Mycoses 42: 261264, 1999. 28. Goosney DL, DeVinney R, Finlay BB. Recruitment of cytoskeletal and signaling proteins to enteropathogenic and enterohemorrhagic Escherichia coli pedestals. Infect Immun 69: 33153322, 2001. 29. Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur J Gastroenterol Hepatol 15: 697 698, 2003. 30. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in maintenance treatment of Crohns disease. Dig Dis Sci 45: 14621464, 2000. 31. Guttman JA, Li Y, Wickham ME, Deng W, Vogl AW, Finlay BB. Attaching and effacing pathogen-induced tight junction disruption in vivo. Cell Microbiol 8: 634 645, 2006. 32. Higgins LM, Frankel G, Connerton I, Goncalves NS, Dougan G, MacDonald TT. Role of bacterial intimin in colonic hyperplasia and inammation. Science 285: 588 591, 1999. 33. Higgins LM, Frankel G, Douce G, Dougan G, MacDonald TT. Citrobacter rodentium infection in mice elicits a mucosal Th1 cytokine
294 JANUARY 2008

Downloaded from ajpgi.physiology.org on March 18, 2011

www.ajpgi.org

G306

Saccharomyces boulardii ATTENUATES Citrobacter rodentium COLITIS 48. Phillips N, Hayward RD, Koronakis V. Phosphorylation of the enteropathogenic E. coli receptor by the Src-family kinase c-Fyn triggers actin pedestal formation. Nat Cell Biol 6: 618 625, 2004. 49. Rodrigues AC, Nardi RM, Bambirra EA, Vieira EC, Nicoli JR. Effect of Saccharomyces boulardii against experimental oral infection with Salmonella typhimurium and Shigella exneri in conventional and gnotobiotic mice. J Appl Bacteriol 81: 251256, 1996. 50. Rosenshine I, Donnenberg MS, Kaper JB, Finlay BB. Signal transduction between enteropathogenic Escherichia coli (EPEC) and epithelial cells: EPEC induces tyrosine phosphorylation of host cell proteins to initiate cytoskeletal rearrangement and bacterial uptake. EMBO J 11: 35513560, 1992. 51. Schauer DB, Falkow S. Attaching and effacing locus of a Citrobacter freundii biotype that causes transmissible murine colonic hyperplasia. Infect Immun 61: 2486 2492, 1993. 52. Schauer DB, Falkow S. The eae gene of Citrobacter freundii biotype 4280 is necessary for colonization in transmissible murine colonic hyperplasia. Infect Immun 61: 4654 4661, 1993. 53. Schauer DB, Zabel BA, Pedraza IF, OHara CM, Steigerwalt AG, Brenner DJ. Genetic and biochemical characterization of Citrobacter rodentium sp. nov. J Clin Microbiol 33: 2064 2068, 1995. 54. Sherman PM, Johnson-Henry KC, Yeung HP, Ngo PSC, Goulet J, Tompins TA. Probiotics reduce enterohemorrhagic Escherichia coli O157:H7- and enteropathogenic E. coli O127:H6-induced changes in polarized T84 epithelial cell monolayers by reducing bacterial adhesion and cytoskeletal rearrangements. Infect Immun 73: 51835188, 2005. 55. Sougioultzis S, Simeonidis S, Bhaskar KR, Chen X, Anton PM, Keates S, Pothoulakis C, Kelly CP. Saccharomyces boulardii produces a soluble anti-inammatory factor that inhibits NF- B-mediated IL-8 gene expression. Biochem Biophys Res Commun 343: 69 76, 2006. 56. Tasteyre A, Barc MC, Karjalainen T, Bourlioux P, Collignon A. Inhibition of in vitro cell adherence of Clostridium difcile by Saccharomyces boulardii. Microb Pathog 32: 219 225, 2002. 57. Vallance BA, Finlay BB. Exploitation of host cells by enteropathogenic Escherichia coli. Proc Natl Acad Sci USA 97: 8799 8806, 2000. 58. Vallance BA, Deng W, Jacobson K, Finlay BB. Host susceptibility to the attaching and effacing bacterial pathogen Citrobacter rodentium. Infect Immun 71: 34433453, 2003. 59. Van de Guchte M, Ehrlich SD, Maguin E. Production of growthinhibiting factors by Lactobacillus delbruekii. J Appl Microbiol 91: 147153, 2001. 60. Varcoe JJ, Krejcarek G, Busta F, Brady L. Prophylactic feeding of Lactobacillus acidophilus NCFM to mice attenuates overt colonic hyperplasia. J Food Prot 66: 457 465, 2003. 61. Wiles S, Clare S, Harker J, Huett A, Young D, Dougan G, Frankel G. Organ specicity, colonization and clearance dynamics in vivo following oral challenges with the murine pathogen Citrobacter rodentium. Cell Microbiol 6: 963972, 2004. 62. Wolff C, Nisan I, Hanski E, Frankel G, Rosenshine I. Protein translocation into host epithelial cells by infecting enteropathogenic Escherichia coli. Mol Microbiol 28: 143155, 1998.

34.

35. 36. 37. 38. 39. 40.

41. 42.

43. 44.

45.

46. 47.

response and lesions similar to those in murine inammatory bowel disease. Infect Immun 67: 30313039, 1999. Johnson-Henry KC, Nadja M, Mitchell DJ, Ngan BY, Galindo-Mata E, Jones NL, Sherman PM. Amelioration of the effects of Citrobacter rodentium infection in mice by pretreatment with probiotics. J Infect Dis 191: 2106 2117, 2005. Kaper JB. Enterohemorrhagic Escherichia coli. Curr Opin Microbiol 1: 103108, 1998. Kenny B, DeVinney R, Stein M, Reinscheid DJ, Frey EA, Finlay BB. Enteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into mammalian cells. Cell 91: 511520, 1997. Kimmey KB, Elmer GW, Surawicz CM, McFarland LV. Prevention of further recurrences of Clostridium difcile colitis with Saccharomyces boulardii. Dig Dis Sci 35: 897901, 1990. Leverton LQ, Kaper JB. Temporal expression of enteropathogenic Escherichia coli virulence genes in an in vitro model of infection. Infect Immun 73: 1034 1043, 2005. Li Y, Frey E, Mackenzie AM, Finlay BB. Human response to Escherichia coli O157:H7 infection: antibodies to secreted virulence factors. Infect Immun 68: 5090 5095, 2000. Lommel S, Benesch S, Rohde M, Wehland J, Rottner K. Enterohaemorrhagic and enteropathogenic Escherichia coli use different mechanisms for actin pedestal formation that converge on N-WASP. Cell Microbiol 6: 243254, 2004. Luo W, Donnenberg MS. Analysis of the function of enteropathogenic Escherichia coli EspB by random mutagenesis. Infect Immun 74: 810 820, 2006. Luperchio SA, Newman JV, Dangler CA, Schrenzel MD, Brenner DJ, Steigerwalt AG, Schauer DB. Citrobacter rodentium, the causative agent of transmissible murine colonic hyperplasia, exhibits clonality: synonymy of C. rodentium and mouse pathogenic Escherichia coli. J Clin Microbiol 38: 4343 4350, 2000. Luperchio SA, Schauer DB. Molecular pathogenesis of Citrobacter rodentium and transmissible murine colonic hyperplasia. Microbes Infect 3: 333340, 2001. Madsen K, Cornish A, Soper P, McKaigney C, Jijon H, Yachimec C, Doyle J, Jewell L, De Simone C. Probiotic bacteria enhance murine and human intestinal epithelial barrier function. Gastroenterology 121: 580 591, 2001. McFarland LV, Surawicz CM, Greenberg RN, Fekety R, Elmer GW, Moyer KA, Melcher SA, Bowen KE, Cox JL, Noorani Z. A randomized placebo-controlled trial of Saccharomyces boulardii in combination with standard antibiotics for Clostridium difcile disease. JAMA 2712: 1913 1918, 1994. Newman JV, Zabel BA, Jha SS, Schauer DB. Citrobacter rodentium espB is necessary for signal transduction and for infection of laboratory mice. Infect Immun 67: 6019 6025, 1999. OConnell CB, Creasey EA, Knutton S, Elliott S, Crowther LJ, Luo W, Albert MJ, Kaper JB, Frankel G, Donnenberg MS. SepL, a protein required for enteropathogenic Escherichia coli type III translocation, interacts with secretion component SepD. Mol Microbiol 52: 16131625, 2004.

Downloaded from ajpgi.physiology.org on March 18, 2011

AJP-Gastrointest Liver Physiol VOL

294 JANUARY 2008

www.ajpgi.org

You might also like