Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate (DSS)-Induced Experimental...

2
AGA Abstracts led to the significant reduction of both IL-17-producing Lin- cKit- Thy1+ Sca1+ innate lymphoid cells and IL-17-producing CD4+ T cells in the colon of Muc1-deficient TCR alpha double KO mice. Since Muc1 was proposed previously to play an inflammatory role in a Th1-mediated colitis of IL-10 KO mice, we next examined the role of Muc1 in another Th1- mediated colitis model, CD45RB model. As seen in TCR alpha KO mice, absence of Muc1 further enhanced Th17 responses and exacerbated the colitis in recipient Muc1-deficient RAG1 double KO mice (as compared to recipient RAG1 KO mice) after transfer of naïve CD4+ T cells from WT mice. Conclusion: A membrane-bound mucin functions as a key determinant of colitis-associated Th17 responses and seems to contribute to the suppression of both Th2- and Th1-mediated colitis. 921 IL28Ralpha Deficiency Enforces T Cell-Dependent Experimental Colitis via Suppression of Regulatory T-Cells Heike Dornhoff, Sean Doyle, Markus F. Neurath, Jürgen Siebler Introduction: Type III IFNs were recently discovered and denoted as IFNλ1 and IFNλ2/3 (also called IL29 and IL28A/IL28B). Whereas IL29 occurred in human, in mice it is not functional and represents a pseudogene. IL28 signals through two different receptor com- plexes, the IL28Rα specific for IL29 and IL28A/B and IL10Rb. Interaction of these cytokines with the receptors leads to the phosphorylation of the main components of the signalling STAT1 and 2, which form a trimeric complex together with IRF9, the ISGF3 complex, that drives transcription of ISGs. Type III IFNs belong to the IL10 superfamily and have antiviral activity similar to the Type I IFNs. But the immunomodulatory function of this new class of cytokines was not been investigated, until now. The aim of this study was to determine the functional role of IL28Rα expression on T-cells in the pathogenesis of experimental colitis. Methods: To analyse whether the receptor has a regulatory role in colitis we adoptively transferred naïve T-cells from wild type (WT) and IL28Rα knock out (KO) animals into RagKO mice, which were deficient for T- and B-cells. After three to five weeks, dependent on the transferred T cell number, the inflammation of the gut of every mouse was monitored by colonoscopy. Results: We could show that the IL28Rα is also being expressed on CD4+ T-cells, which was previously described mostly for non-haematopoietic cells. Comparison between naïve and nTregs revealed a markedly downregulation of the receptor in regulatory T cells but show an upregulation by TGFβ (TiTreg)-induced peripheral Tregs in WT T- cells. Interestingly, KO T-cells demonstrated a significant reduction in the expression of Foxp3 in TiTregs compared to WT. There is evidence that these KO cells have an increased apoptosis rate compared to the counterparts. Analysis of these T-cells in an adoptive transfer colitis model indicate a significant heightened colitis in KO reconstituted RAG mice, whereas WT reconstituted RAG mice were more protected with regard to the severity of the inflamma- tion of the gut. Moreover, they show a massive diminishment of Foxp3 and CD25+ expressing regulatory T cells In-Vivo, which resulted in a higher production of proinflammatory TH1/ TH17 cytokines. Furthermore, reconstitution of RAG mice with naïve WT T-cells together with either in-vitro generated WT TiTreg or KO TiTreg resulted in a dramatic defect in the healing of the colitis by KO TiTregs in comparison to WT TiTregs. Discussion/Conclusion: Here, we describe for the first time the immunomodulatory function of IL28Rα on T-cells. Beside the antiviral activity of IL28 on epithelial cells, IL28 seems to have survival functions in generating peripheral TGFβ induced T regulatory cells. Therefore, IL28 emerges as an attractive target for modulating chronic intestinal inflammation. 922 CD4 + CD25 + Regulatory T Cells Suppress the Developmental Pathway From TH17 to Alternative TH1 Cells via TH17/TH1 and TH1-Like Cells Tomohisa Sujino, Takanori Kanai, Yuuichi Ono, Yohei Mikami, Atsushi Hayashi, Tomomitsu Doi, Katsuyoshi Matsuoka, Tadakazu Hisamatsu, Hiromasa Takaishi, Haruhiko Ogata, Akihiko Yoshimura, Toshifumi Hibi BACKGROUND/AIMS: Although accumulating evidence suggests that both Th17 and Th1 cells are positively involved in the pathogenesis of intestinal inflammation, we here aim to clarify not only the developmental pathway of Th17, Th17/Th1, and Th1 cells in the pathogenesis of chronic colitis, but also the position of CD4 + CD25 + Foxp3 + regulatory T (T R ) cells in their developmental pathway using In Vivo adoptive transfer experiments. METHODS: We here conducted series of adoptive transfer experiments for the induction and suppression of colitis using naïve CD4 + CD45RB high T cells and/or CD4 + CD25 + T R cells obtained from RORγt gfp/+ reporter mice or Ly5.1- and Ly5.2-derived mice. RESULTS: We found that there are three types of colitogenic CD4 + Th1 cells (IL-17A - IFN-γ + ): 1) RORγt + T- bet + classical Th1 cells directly differentiated from naïve T cells, 2) RORγt + T-bet + Th1-like cells, and 3) RORγt + T-bet + alternative Th1 cells terminally differentiated via the pathway of RORγt + T-bet + Th17 (IL-17A + IFN-γ + ),RORγt + T-bet + Th17/Th1 (IL-17A + IFN-γ + ),RORγt + T- bet + Th1-like (IL-17A + IFN-γ + ) cells. In such a developmental pathway, CD4 + CD25 + T R cells suppressed the development of not only classical Th1 cells but also that of alternative Th1 cells at the step Th17/Th1alternative Th1 cells, resulting in the accumulation of Th17 and Th17/Th1 cells in mice in which the development of colitis is suppressed. Furthermore, CD4 + CD25 + T R cells modulate the established balance of Th17 and Th1 cells in colitic condition to yield a high ratio of Th17 and Th17/Th1 cells to Th1 cells in non-colitic condition. CONCLUSIONS: The present study demonstrates that Th17 and Th17/Th1 cells themselves become colitogenic RORγt + T-bet + alternative Th1 cells via RORγt + T-bet + Th1- like cells independently of classical Th1 cells, suggesting T-bet rather than RORγt as an ideal target of intestinal inflammation. S-150 AGA Abstracts 923 Loss of TNFR2 Exacerbates Colitis in Interleukin-10 Knockout Mice Shivesh Punit, Philip E. Dubé, Kay Washington, D Brent Polk Tumor necrosis factor (TNF) elicits a wide range of responses in both the mucosal immune system and colon epithelium. The biological activities of TNF are mediated by two receptors (TNFR1 and TNFR2). TNFR1 is ubiquitously expressed and contains a death domain that mediates apoptosis through caspase activation. TNFR2 is expressed on hematopoietic cells and its expression is induced on intestinal epithelial cells during inflammation in human IBD patients and in murine colitis models (Mizoguchi et al). Previous work from our lab has shown that TNFR2 enhances intestinal epithelial proliferation, migration and wound closure In Vitro. Yet, the role of TNFR2 mediated effects during intestinal inflammation is not well understood. Therefore, we hypothesized that TNFR2 deletion exacerbates colitis in the interleukin-10 knockout (IL-10-/-) colitis model. METHODS: IL10-/- mice with TNFR2 deletion (IL10-/-/TNFR2-/-) were compared to IL10-/- and TNFR2-/- mice (n=4-5). Colonic permeability was assessed using FITC dextran enemas. Colon mucosal tissues were analyzed by flow cytometry and explant supernatants were analyzed by cytokine array to determine the profile of immune cell infiltration and the cytokine milieu, respectively. Epithelial proliferation was determined by Ki67 staining and the number of proliferating cells per 100 crypts was quantified. Colon histological sections were examined and the degree of colitis was blindly scored. Statistical differences were determined by Mann Whitney test. RESULTS: IL10-/-/TNFR2-/- mice developed more severe colitis compared to IL-10-/- mice by 16 weeks of age (median colitis score of 12 vs. 9; p<0.05) characterized by markedly increased crypt inflammation and hyperplasia. TNFR2-/- colons were histologically normal. Colonic permeability was similar between IL10-/-/TNFR2-/- and IL10-/- mice. Increased epithelial hyperproliferation was observed in IL10-/-/TNFR2-/- colons compared to IL-10-/- colons (Avg. Ki67 positive cells/crypt 48 vs. 26; P<0.001). IL10-/-/TNFR2-/- colons had increased cytotoxic T-cell and dendritic cell infiltration. TNF was significantly elevated in IL10-/-/TNFR2-/- colonic explant culture compared to IL10-/- explants and this difference increased with mouse age. CONCLUSION: TNFR2 is protective in IL10-/- colitis. Loss of TNFR2 on the IL10-/- background results in increased production of TNF, which could be due to increased colonic infiltration of cytotoxic T-cells and dendritic cells; however, the cell type responsible has not yet been identified. Further studies are warranted to examine the specific role of TNFR2 in inflammation-associated alteration of the intestinal epithelium. This study emphasizes the role of TNFR2 signaling in the pathology of immune-mediated colitis and may lead to the development of novel therapeutics. 924 Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate (DSS)-Induced Experimental Colitis Rizwan Ahmad, Rupesh Chaturvedi, Mohammad Asim, D Brent Polk, Robert D. Beauchamp, Keith T. Wilson, Punita Dhawan, Amar B. Singh Increased mucosal permeability is a key characteristic of inflammatory bowel disease (IBD). The claudin family of proteins constitutes the tight junctions (TJs), which are the sole determinants of the paracellular permeability in an intact epithelium. Importantly, a common finding from the analysis of IBD (Crohn's disease and ulcerative colitis) patient samples and related animal models is that claudin-2 expression is highly upregulated in IBD. Within the claudin family, claudin-2 is unique as its expression correlates with the epithelial leakiness. However, it is also noteworthy that colonic claudin-2 is expressed among undifferentiated colonocytes at the crypt base (the proliferative zone) and is a target of the Wnt/β-catenin signaling. However, the role of claudin-2 in the pathogenesis of IBD is not known. To investigate the potential role of claudin-2 in IBD, we generated Villin-claudin-2 transgenic (Cl-2Tg) mice that overexpress claudin-2 in the colon, a condition similar to that found in IBD. Our studies using these mice have shown that the colonic epithelial permeability (using rectal delivery of FITC-Dextran, 4 kDa) in Cl-2Tg mice (12.35+2.9) is significantly increased compared to WT littermates (1.10+0.44; p<0.05). Interestingly, the colon and crypt lengths in Cl-2Tg mice (8.25+0.13 cm and 230.8+3.31 μm respectively) are also significantly increased compared to WT littermates [7.10+0.19 cm and 193.50+2.48 μm respectively; p<0.01 (colon) and p<0.001 (crypt)]. Most importantly, irrespective of the increased colonic permeability Cl-2TG mice did not develop symptoms associated with colonic inflammation and in fact were significantly protected when subjected to the dextran sodium sulfate (DSS)- induced colitis. This protection was evident at the clinical and histological levels and the overall injury score in the DSS-treated Cl-2TG mice was 7.53+1.09 compared to 19.27+1.66

Transcript of Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate (DSS)-Induced Experimental...

Page 1: Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate (DSS)-Induced Experimental Colitis

AG

AA

bst

ract

sled to the significant reduction of both IL-17-producing Lin- cKit- Thy1+ Sca1+ innatelymphoid cells and IL-17-producing CD4+ T cells in the colon of Muc1-deficient TCR alphadouble KO mice. Since Muc1 was proposed previously to play an inflammatory role in aTh1-mediated colitis of IL-10 KO mice, we next examined the role of Muc1 in another Th1-mediated colitis model, CD45RB model. As seen in TCR alpha KO mice, absence of Muc1further enhanced Th17 responses and exacerbated the colitis in recipient Muc1-deficientRAG1 double KO mice (as compared to recipient RAG1 KO mice) after transfer of naïveCD4+ T cells from WT mice. Conclusion: A membrane-bound mucin functions as a keydeterminant of colitis-associated Th17 responses and seems to contribute to the suppressionof both Th2- and Th1-mediated colitis.

921

IL28Ralpha Deficiency Enforces T Cell-Dependent Experimental Colitis viaSuppression of Regulatory T-CellsHeike Dornhoff, Sean Doyle, Markus F. Neurath, Jürgen Siebler

Introduction: Type III IFNs were recently discovered and denoted as IFNλ1 and IFNλ2/3(also called IL29 and IL28A/IL28B). Whereas IL29 occurred in human, in mice it is notfunctional and represents a pseudogene. IL28 signals through two different receptor com-plexes, the IL28Rα specific for IL29 and IL28A/B and IL10Rb. Interaction of these cytokineswith the receptors leads to the phosphorylation of the main components of the signallingSTAT1 and 2, which form a trimeric complex together with IRF9, the ISGF3 complex, thatdrives transcription of ISGs. Type III IFNs belong to the IL10 superfamily and have antiviralactivity similar to the Type I IFNs. But the immunomodulatory function of this new classof cytokines was not been investigated, until now. The aim of this study was to determinethe functional role of IL28Rα expression on T-cells in the pathogenesis of experimentalcolitis. Methods: To analyse whether the receptor has a regulatory role in colitis we adoptivelytransferred naïve T-cells from wild type (WT) and IL28Rα knock out (KO) animals intoRagKO mice, which were deficient for T- and B-cells. After three to five weeks, dependenton the transferred T cell number, the inflammation of the gut of every mouse was monitoredby colonoscopy. Results: We could show that the IL28Rα is also being expressed on CD4+T-cells, which was previously described mostly for non-haematopoietic cells. Comparisonbetween naïve and nTregs revealed a markedly downregulation of the receptor in regulatoryT cells but show an upregulation by TGFβ (TiTreg)-induced peripheral Tregs in WT T-cells. Interestingly, KO T-cells demonstrated a significant reduction in the expression ofFoxp3 in TiTregs compared to WT. There is evidence that these KO cells have an increasedapoptosis rate compared to the counterparts. Analysis of these T-cells in an adoptive transfercolitis model indicate a significant heightened colitis in KO reconstituted RAG mice, whereasWT reconstituted RAGmice were more protected with regard to the severity of the inflamma-tion of the gut. Moreover, they show a massive diminishment of Foxp3 and CD25+ expressingregulatory T cells In-Vivo, which resulted in a higher production of proinflammatory TH1/TH17 cytokines. Furthermore, reconstitution of RAG mice with naïve WT T-cells togetherwith either in-vitro generated WT TiTreg or KO TiTreg resulted in a dramatic defect in thehealing of the colitis by KO TiTregs in comparison to WT TiTregs. Discussion/Conclusion:Here, we describe for the first time the immunomodulatory function of IL28Rα on T-cells.Beside the antiviral activity of IL28 on epithelial cells, IL28 seems to have survival functionsin generating peripheral TGFβ induced T regulatory cells. Therefore, IL28 emerges as anattractive target for modulating chronic intestinal inflammation.

922

CD4+CD25+ Regulatory T Cells Suppress the Developmental Pathway FromTH17 to Alternative TH1 Cells via TH17/TH1 and TH1-Like CellsTomohisa Sujino, Takanori Kanai, Yuuichi Ono, Yohei Mikami, Atsushi Hayashi,Tomomitsu Doi, Katsuyoshi Matsuoka, Tadakazu Hisamatsu, Hiromasa Takaishi,Haruhiko Ogata, Akihiko Yoshimura, Toshifumi Hibi

BACKGROUND/AIMS: Although accumulating evidence suggests that both Th17 and Th1cells are positively involved in the pathogenesis of intestinal inflammation, we here aim toclarify not only the developmental pathway of Th17, Th17/Th1, and Th1 cells in thepathogenesis of chronic colitis, but also the position of CD4+CD25+Foxp3+ regulatory T(TR) cells in their developmental pathway using In Vivo adoptive transfer experiments.METHODS: We here conducted series of adoptive transfer experiments for the inductionand suppression of colitis using naïve CD4+CD45RBhigh T cells and/or CD4+CD25+ TR cellsobtained from RORγtgfp/+ reporter mice or Ly5.1- and Ly5.2-derived mice. RESULTS: Wefound that there are three types of colitogenic CD4+ Th1 cells (IL-17A-IFN-γ+): 1) RORγt+T-bet+ classical Th1 cells directly differentiated from naïve T cells, 2) RORγt+T-bet+ Th1-likecells, and 3) RORγt+T-bet+ alternative Th1 cells terminally differentiated via the pathwayof RORγt+T-bet+ Th17 (IL-17A+IFN-γ+),RORγt+T-bet+ Th17/Th1 (IL-17A+IFN-γ+),RORγt+T-bet+ Th1-like (IL-17A+IFN-γ+) cells. In such a developmental pathway, CD4+CD25+ TR cellssuppressed the development of not only classical Th1 cells but also that of alternative Th1cells at the step Th17/Th1→alternative Th1 cells, resulting in the accumulation of Th17and Th17/Th1 cells in mice in which the development of colitis is suppressed. Furthermore,CD4+CD25+ TR cells modulate the established balance of Th17 and Th1 cells in coliticcondition to yield a high ratio of Th17 and Th17/Th1 cells to Th1 cells in non-coliticcondition. CONCLUSIONS: The present study demonstrates that Th17 and Th17/Th1 cellsthemselves become colitogenic RORγt+T-bet+ alternative Th1 cells via RORγt+T-bet+ Th1-like cells independently of classical Th1 cells, suggesting T-bet rather than RORγt as anideal target of intestinal inflammation.

S-150AGA Abstracts

923

Loss of TNFR2 Exacerbates Colitis in Interleukin-10 Knockout MiceShivesh Punit, Philip E. Dubé, Kay Washington, D Brent Polk

Tumor necrosis factor (TNF) elicits a wide range of responses in both the mucosal immunesystem and colon epithelium. The biological activities of TNF are mediated by two receptors(TNFR1 and TNFR2). TNFR1 is ubiquitously expressed and contains a death domain thatmediates apoptosis through caspase activation. TNFR2 is expressed on hematopoietic cellsand its expression is induced on intestinal epithelial cells during inflammation in humanIBD patients and in murine colitis models (Mizoguchi et al). Previous work from our labhas shown that TNFR2 enhances intestinal epithelial proliferation, migration and woundclosure In Vitro. Yet, the role of TNFR2 mediated effects during intestinal inflammation isnot well understood. Therefore, we hypothesized that TNFR2 deletion exacerbates colitisin the interleukin-10 knockout (IL-10-/-) colitis model. METHODS: IL10-/- mice withTNFR2 deletion (IL10-/-/TNFR2-/-) were compared to IL10-/- and TNFR2-/- mice (n=4-5).Colonic permeability was assessed using FITC dextran enemas. Colon mucosal tissues wereanalyzed by flow cytometry and explant supernatants were analyzed by cytokine array todetermine the profile of immune cell infiltration and the cytokine milieu, respectively.Epithelial proliferation was determined by Ki67 staining and the number of proliferatingcells per 100 crypts was quantified. Colon histological sections were examined and thedegree of colitis was blindly scored. Statistical differences were determined by Mann Whitneytest. RESULTS: IL10-/-/TNFR2-/- mice developed more severe colitis compared to IL-10-/-mice by 16 weeks of age (median colitis score of 12 vs. 9; p<0.05) characterized by markedlyincreased crypt inflammation and hyperplasia. TNFR2-/- colons were histologically normal.Colonic permeability was similar between IL10-/-/TNFR2-/- and IL10-/- mice. Increasedepithelial hyperproliferation was observed in IL10-/-/TNFR2-/- colons compared to IL-10-/-colons (Avg. Ki67 positive cells/crypt 48 vs. 26; P<0.001). IL10-/-/TNFR2-/- colons hadincreased cytotoxic T-cell and dendritic cell infiltration. TNF was significantly elevated inIL10-/-/TNFR2-/- colonic explant culture compared to IL10-/- explants and this differenceincreased with mouse age. CONCLUSION: TNFR2 is protective in IL10-/- colitis. Loss ofTNFR2 on the IL10-/- background results in increased production of TNF, which could bedue to increased colonic infiltration of cytotoxic T-cells and dendritic cells; however, thecell type responsible has not yet been identified. Further studies are warranted to examinethe specific role of TNFR2 in inflammation-associated alteration of the intestinal epithelium.This study emphasizes the role of TNFR2 signaling in the pathology of immune-mediatedcolitis and may lead to the development of novel therapeutics.

924

Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate(DSS)-Induced Experimental ColitisRizwan Ahmad, Rupesh Chaturvedi, Mohammad Asim, D Brent Polk, Robert D.Beauchamp, Keith T. Wilson, Punita Dhawan, Amar B. Singh

Increased mucosal permeability is a key characteristic of inflammatory bowel disease (IBD).The claudin family of proteins constitutes the tight junctions (TJs), which are the soledeterminants of the paracellular permeability in an intact epithelium. Importantly, a commonfinding from the analysis of IBD (Crohn's disease and ulcerative colitis) patient samples andrelated animal models is that claudin-2 expression is highly upregulated in IBD. Within theclaudin family, claudin-2 is unique as its expression correlates with the epithelial leakiness.However, it is also noteworthy that colonic claudin-2 is expressed among undifferentiatedcolonocytes at the crypt base (the proliferative zone) and is a target of the Wnt/β-cateninsignaling. However, the role of claudin-2 in the pathogenesis of IBD is not known. Toinvestigate the potential role of claudin-2 in IBD, we generated Villin-claudin-2 transgenic(Cl-2Tg) mice that overexpress claudin-2 in the colon, a condition similar to that found inIBD. Our studies using these mice have shown that the colonic epithelial permeability (usingrectal delivery of FITC-Dextran, 4 kDa) in Cl-2Tg mice (12.35+2.9) is significantly increasedcompared to WT littermates (1.10+0.44; p<0.05). Interestingly, the colon and crypt lengthsin Cl-2Tg mice (8.25+0.13 cm and 230.8+3.31 μm respectively) are also significantlyincreased compared to WT littermates [7.10+0.19 cm and 193.50+2.48 μm respectively;p<0.01 (colon) and p<0.001 (crypt)]. Most importantly, irrespective of the increased colonicpermeability Cl-2TG mice did not develop symptoms associated with colonic inflammationand in fact were significantly protected when subjected to the dextran sodium sulfate (DSS)-induced colitis. This protection was evident at the clinical and histological levels and theoverall injury score in the DSS-treated Cl-2TG mice was 7.53+1.09 compared to 19.27+1.66

Page 2: Claudin-2 Transgenic Mice Are Protected From the Dextran Sodium Sulfate (DSS)-Induced Experimental Colitis

in the WT littermates (n#15; p<0.001). Further studies using real time PCR showed, asexpected, several fold increase in the expressions of proinflammatory cytokines includingIL-1α, IL-1β, TNF-α, IFN-γ in the DSS-treated WT mice but not in the DSS-treated Cl-2TGmice suggesting potential immune tolerance/adaptation. Further assessment of prolifera-tion and apoptosis in isolated colonic epithelial cells using BrdU and cleaved caspase-3staining and FACS analysis showed decreased apoptosis (p<0.05) and increased proliferation(p<0.05) in the DSS-treated Cl-2Tg mice compared to the WT littermates. Taken together,our data supports that claudin-2 has a key role in the regulation of colonic homeostasis.Our data further suggests that immune tolerance/adaptation possibly due to the constitutiveincrease in colonic permeability and increased colonic epithelial cell viability underlie theprotection from the DSS-induced colitis in claudin-2 TG mice.

Claudin-2 TG mice are protected from the DSS-colitis. A. % change in body weight in theDSS-treated (4% DSS wt./vol in drinking water) Cl-2TG and WT mice. Body weight ispresented relative to the weight of respective group exposed to water alone; B. Injury scorefor DSS treated WT & Cl-2TG mice (n#15). Injury score for mice exposed to water (eithergroup) was zero; and C. H&E staining showing histological details . mean+SEM,**p<0.01***p<0.001

925

In Vivo Constitutive Expression of an IBD Associated Gene TNFSF15 CausesSevere Inflammation and Induces Fibrostenotic Disease in 2 Murine Models ofChronic ColitisRobert Barrett, Xiaolan Zhang, Nicole Yeager, Hon Wai Koon, Mary Ann Nguyen,Piangwarin Phaosawasdi, Chenyang Li, Charalabos Pothoulakis, Stephan R. Targan, DavidQ. Shih

Background: Elevated expression of TNFSF15 (TL1A) is found in inflamed colonic andintestinal mucosa of IBD patients. Furthermore, TL1A haplotype B is associated with itsenhanced production and is also characterized by fibrostenotic disease and need for surgeryin IBD patients. Transgenic (tg) mice with constitutive expression of TL1A in either T cellsor antigen presenting cells (APC) lead to spontaneous ileitis. Aim: To assess whether InVivo constitutive TL1A expression in myeloid and lymphoid cells exacerbate inflammationand cause fibrostenotic disease in 2 murine models of chronic colitis. Methods: Chronicdextran sodium sulfate (DSS) and adoptive T-cell transfer models were used. Disease activityindex (DAI: stool blood, consistency and weight loss), gross inflammation, Histologicalanalyses of H&E, Masson Trichrome, and vimentin stained gut sections were analyzed forinflammation and fibrosis in a blinded fashion. mRNA was measured by RT-PCR and proteinexpression was measured by flow cytometry and ELISA. Results: Compared to wildtype(WT) littermates, overexpression of Tl1a in lymphoid or myeloid cells lead to significantlyenhanced weight loss and worsened DAI. We observed enhanced gross inflammatory scorein the intestine of myeloid Tl1a tg (3.2 for vs. 1.4, p<0.05) and T cell tg (3.5 vs. 1.4, p<0.05)in the chronic DSS model and in the T cell transfer model (3.6 vs. 1, p<0.01). Tl1a tg miceshowed increased histologic inflammation in the intestine, particularly in the ileum of myeloidTl1a tg (8.9 vs. 5.9, p<0.05) and T cell Tl1a tg (9.7 vs. 5.7, p<0.05) for the chronic DSSmodel and adoptive transfer model (11.2 vs. 5.8, p<0.05). Mucosal Tl1a tg T cells or APCwere found to have a more activated phenotype (WT 21%, myeloid tg 37%, myeloid tg46%) and higher IFN-γ expression (WT 1.2%, myeloid tg 2%, T cell tg 3.2%) in bothmodels of chronic colitis. Notably, gross strictures were only present in the Tl1a tg mice:small intestine (30%, up to 3 cm), colon (20%, up to 1 cm) and ureter (10%, 2 mm) withresultant hydronephrosis. Enhanced histologic fibrosis in the Tl1a tg mice were confirmedby increased Masson Trichrome (marker for collagen formation) and vimentin (marker forfibroblast) staining. Consistent with increased fibrosis, increased expression of fibrogenicfactors TGF-β1 and IGF-1 were found in the intestine and colon of tg mice. Conclusion:Tl1a modulates the development of both severe gut mucosal inflammation and markedfibrosis through enhancing Th1 effector functions and expression of pro-fibrogenic factors.

926

Pyrosequencing Reveals Irritable Bowel Syndrome Subtype Defined bySpecies-Specific Alterations in the Microbial Gut Environment.Ian B. Jeffery, Paul W. O' Toole, Lena Ohman, Marcus J. Claesson, Jennifer Deane,Eamonn M. Quigley, Magnus Simren

Introduction: Irritable bowel syndrome (IBS) is a common functional gastrointestinal dis-order that may be triggered by enteric pathogens and has also been linked to alterations inthe microbiota and the host immune response. Aim: To perform a detailed analysis of thefecal microbiota in IBS and control subjects and to correlate findings with key clinical andphysiological parameters. Methods: We included 20 patients with IBS (mean age 38 years;14 females; 9 IBS-D, 4 IBS-C, 7 IBS NonC-NonD). GI and psychological symptom severityand quality of life were evaluated with validated questionnaires. Rectal sensitivity was testedwith a rectal barostat, and colonic transit time (radiopaque markers) was measured. Fecalsamples were obtained from the patients and 20 age- and gender-matched healthy subjects.We applied pyrosequencing of over 32,000 16S rRNA gene V4 region amplicons per subjectto characterize the fecal microbiota. Results: The majority of patients had GI symptomseverity ranging from moderate to severe, and 8/20 patients had clinically significant anxietyand/or depression. Rectal hypersensitivity was detected in 6/19 patients, whereas colonictransit was rapid in 6 and slow in 2 patients. Reduced quality of life (mental and/or physical)was seen in 18/19 patients. The microbial data had no association with the clinical or

S-151 AGA Abstracts

physiological phenotype, except for a number of OTUs that were significantly associatedwith a rapid colonic transit time. Clustering analysis of the pyrosequencing data revealedtwo subgroups of IBS patients. These groups were significantly different from each other(p-value: 0.0025) and did not correlate with Rome II or Rome III subtypes or other clinicaldata. Group 1 clustered with the control samples and had no significantly associatedmicrobialfeatures when compared to normal subjects. Group 2 was defined by large microbiome-wide changes characterized by increased prevalence in the proportion of Phylum Firmicutescompared to control subjects, and a depletion of Bacteroidetes. Statistically associated withthis high Firmicutes/ Bacteroidetes (FB) ratio subset, were 114 operational taxonomic units(OTUs) of the phylum Firmicutes. 110 of these were of the Order Clostridiales and 47 and27 of these were of the Genera Blautia and Papillibacter, respectively. These Genera belongto Clostridium cluster XIVa, which, as a cluster, was over-represented in the High FB IBSassociated OTUs (p-value: < 0.001). Conclusions: This detailed analysis of a well-character-ized cohort of IBS patients does not support a pronounced or reproducible IBS-relatedmicrobiome, but rather identified a discrete subset of IBS patients with alterations in theiroverall microbial ecology. No clear association with the clinical and physiological phenotypeof the patients could be detected apart from a modest association with colonic transit.

927

The Cellular Distribution and Expression of ZO-1, Occludin and Claudin-1Were Differentially Altered During Irritable Bowel Syndrome According to theSub-Types and SymptomsNathalie Bertiaux-Vandaële, Stéphanie Beutheu Youmba, Belmonte Liliana, StephaneLecleire, Michel Antonietti, Guillaume Gourcerol, Leroi Anne Marie, Pierre M. Déchelotte,Philippe R. Ducrotté, Moïse Coëffier

Background & aims: Recent studies have suggested that increased intestinal permeabilitymay be involved in the pathophysiology of irritable bowel syndrome (IBS). However thedifferential expression of tight junction proteins according to IBS sub-types and symptomsremained unknown. Thus, we aimed to study the expression of ZO-1, occludin and claudin-1 in the colonic mucosa of patients with IBS. Methods: Fifty IBS patients fulfilling the RomeIII criteria and 31 controls were included. IBS patients were with predominant diarrhea(IBS-D, n=19), with predominant constipation (IBS-C, n=14), with constipation alternatingwith diarrhea (IBS-A, n=15) or unclassified (IBS-U, n=2). IBS symptom intensity was quanti-fied on 10-cm VAS. Tight junction proteins (claudin-1, ZO-1, occludin) were studied byqRT-PCR, western blot and immunofluorescence in immunohistochemistry. The results,expressed as mean ± SD, were compared by nonparametric Mann-Whitney or Kruskal-Wallis with Dunn post-tests, p <0.05 was considered the threshold of significance. Results:ZO-1 and occludin expression was lower in IBS patients compared with controls (0.25±0.27 vs 0.50 ±0.54 and 0.50 ±0.34 vs 1.22 ±1.76; p<0.05), whereas only a trend for adecrease of claudin-1 was observed (0.29 ±0.36 vs 0.32 ±0.23 ; p=0.08). The mRNA levelsremained unaffected. In the subgroup analyses, occludin and claudin-1 expression wasdecreased in IBS-D patients (p<0.05) but not in IBS-C and IBS-A patients whereas ZO-1expression was not modified. The subcellular distribution of these 3 proteins was alteredin IBS-C and IBS-D patients. While the 3 tight junction proteins staining appeared mainlyat the apical of epithelial cells with the typical reticular pattern in the colonic samples ofcontrols, occludin and claudin-1 was mainly cytosolic in the samples of IBS-C patients andcompleted abolished in IBS-D patients. In IBS-C and IBS-D patients, ZO-1 staining was lessapical and more diffuse in the cytoplasm. Occludin (r=0.40, p<0.01) and claudin-1 (r=0.46,p<0.01) expression was correlated with the duration of symptoms. The expression of occludinwas lower in patients with an abdominal pain intensity higher than 6 on the VAS (p<0.05).Conclusions: Occludin and claudin-1 appeared markedly affected in IBS with predominantdiarrhea. In addition, our results suggest that alteration of tight junction proteins may beinvolved in the initiation of IBS and contribute to visceral hypersensitivity.

928

The Potency of Constipated IBS Fecal Supernatant to Increase ColonicPermeability in Mice Involves Occludin Enzymatic Degradation Linked toCysteine-Protease ActivityAnita Annahazi, Valérie Bézirard, Mathilde Leveque, Helene Eutamene, Laurent Ferrier,Moïse Coëffier, Philippe R. Ducrotté, Richárd Róka, Krisztina Gecse, Andras I. Rosztoczy,Tamas Molnar, Thierry Piche, Vassilia Theodorou, Tibor Wittmann, Lionel Bueno

Background and aims: We have previously shown that (i) fecal cysteine-protease (CP)activity is elevated only in constipated IBS (IBS-C) patients among IBS subtypes and (ii)repeated mucosal exposure to this luminal factor enhances colorectal sensitivity and increasesintestinal permeability in mice. Since CP are involved in the increase of airway permeabilityby digesting epithelial tight junction proteins, we evaluated the ability of CP in the IBS-Cfecal supernatant to degrade tight junction proteins in mice and T84 cells as well as recombin-ant occludin In Vitro. Methods: Fecal samples and colonic biopsies were collected fromhealthy subjects (n=20 & 5 respectively) and IBS-C patients (n=25 & 5 respectively). CPactivity was assayed using a selective substrate and controlled by a selective CP inhibitor,E64. IBS-C and healthy controls fecal supernatants (FSN) pre-incubated or not with E64,or papain solution (as a control CP) or vehicle (saline) were (i) infused intracolonically (IC)in C57/BL6 mice (0.3 ml for 3h, twice at 12 h interval) or, (ii) added to the apical site of T84cells seeded (5x105 cells/well) in 24-well Fluoblock® plate system. Intestinal permeability tooral FITC-4kD dextran was assessed 4h after the end of the second IC infusion. Permeabilityto FITC-4kD dextran was measured every hour over 24 hours in T84 cells. Expression ofoccludin in mice, human biopsies and T84 cells was evaluated by, western blotting andimmunostaining. Finally, recombinant occludin was incubated in presence of IBS-C, healthycontrols FSN or papain during 15 min, and occludin degradation was assessed by westernblotting. Results: CP activity in IBS-C FSN was significantly increased (p<0.05) vs healthycontrols (2420±718 and 319±91 Δfluo/mg prot). IBS-C FSN and papain IC infusion increasedcolonic permeability in mice compared to healthy controls or saline (403±53 vs 333±43;562±73 vs 341±40 Δfluo/μl respectively) as well as in T84 cells (34% increase vs controlFSN, starting at 15h after application). This effect was markedly reduced by E64 both inmice and T84 cells. Occludin protein levels were 2-fold lower in IBS-C colonic biopsies vs

AG

AA

bst

ract

s