Ameliorating effect of anti-Fas ligand MAb on wasting disease in murine model of chronic colitis

N. Dan,1 T. Kanai,1 T. Totsuka,1 R. Iiyama,1 M. Yamazaki,1 T. Sawada,1 T. Miyata,1 H. Yagita,2 K. Okumura,2 and M. Watanabe1

1Department of Gastroenterology and Hepatology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519; and 2Department of Immunology, Juntendo University School of Medicine, Tokyo 113-8421, Japan.

Submitted 21 February 2003 ; accepted in final form 28 May 2003


    ABSTRACT
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Fas/Fas ligand (FasL) interaction has been implicated in the pathogenesis of various diseases. To clarify the involvement of Fas/FasL in the pathogenesis of intestinal inflammation, we investigated the preventive and therapeutic effects of neutralizing anti-FasL monoclonal antibody (MAb) on the development of chronic colitis induced by adaptive transfer of CD4+CD45RBhigh T cells to SCID mice. Administration of anti-FasL MAb from 1 day after T cell transfer (prevention study) resulted in a significant improvement of clinical manifestations such as wasting and diarrhea. However, histological examination showed that mucosal inflammation in the colon, such as infiltration of T cells and macrophages, was not improved by the anti-FasL MAb treatment. In vitro studies showed that anti-FasL MAb did not inhibit IFN-{gamma} production by anti-CD3/CD28-stimulated lamina propria CD4+ T cells but suppressed TNF-{alpha} and IL-1{beta} production by lamina propria mononuclear cells. Therapeutic administration of anti-FasL MAb from 3 wk after T cell transfer also improved ongoing wasting disease but not intestinal inflammation. These results suggest that the Fas/FasL interaction plays a critical role in regulating systemic wasting disease but not local intestinal inflammation.

Fas/FasL; murine model; Crohn's disease; therapy


INFLAMMATORY BOWEL diseases, including Crohn's disease and ulcerative colitis, are chronic and wasting diseases whose etiology and pathogenesis are poorly understood. However, increasing evidence indicated an important role of immunological mechanisms (16). These diseases are characterized by a massive infiltration of T cells and macrophages in the inflamed mucosa and the production of proinflammatory cytokines such as IFN-{gamma}, TNF-{alpha}, and IL-1{beta} by these cells (5). In fact, TNF-{alpha} has been implicated in the pathogenesis of chronic colitis models (17), and recent clinical application of anti-TNF-{alpha} MAb (Infliximab) has shown a dramatic improvement of clinical symptoms in patients with Crohn's disease (18, 24), although the precise mechanism of its action has not been fully understood.

Fas and its ligand (FasL) are members of the TNF receptor and ligand families (14, 20). Fas is constitutively expressed on various cells and in tissues, including lamina propria (LP) lymphocytes, macrophages, and intestinal epithelial cells (IEC), and is dramatically upregulated at the site of inflammation (1, 15). FasL is expressed on activated T cells and may be involved in T cell-mediated cytotoxity against IEC (2, 15). In addition, nonlymphoid cells, such as IEC, can also express FasL, which may induce apoptosis in neighboring IEC and lead to breakdown of the epithelial barrier function (23). On the other hand, the Fas/FasL system also plays a critical role in downregulation of excessive immune responses (20, 21) and has been implicated in regulation of T cell apoptosis in the mucosa (3, 12). Therefore, it remains still controversial whether the Fas/FasL interaction plays an aggressive or a regulatory role in the pathogenesis of inflammatory bowel disease. In the present study, to clarify the role of Fas/FasL interaction, we evaluated the effect of a neutralizing anti-FasL MAb (MFL) on the development of chronic colitis by utilizing a murine model induced by adoptive transfer of CD4+CD45RBhigh T cells to SCID mice.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Mice. Eight-to-ten-week-old female BALB/c SCID mice and normal BALB/c mice were purchased from Japan Clea (Tokyo, Japan) and maintained under specific pathogen-free conditions in our animal facilities. Mice were treated according to the ethical guidelines of our institution.

Induction of colitis and MAb treatment. Colitis was induced by adoptive transfer of CD4+CD45RBhigh T cells from normal BALB/c mice to BALB/c SCID mice, essentially as described previously (25). CD4+ T cells were isolated from the spleen of BALB/c mice by positive selection using an anti-CD4 (L3T4) MACS magnetic separation system (Miltenyi Biotec, Auburn, CA). The isolated CD4+ T cells were labeled with FITC-conjugated anti-CD45RB MAb (model 16A; BD Pharmingen, San Diego, CA) and phycoerythrin (PE)-conjugated anti-CD4 MAb (model RM4-5; BD Pharmingen) and then fractionated into CD4+CD45RBhigh and CD4+CD45RBlow populations by cell sorting on a FACS Vantage (Becton Dickinson, San Jose, CA). BALB/c SCID mice were intraperitoneally injected with the isolated CD4+CD45RBhigh T cells (3–5 x 105/mouse). The recipient mice were then given 250 µg ip of MFL1 (hamster IgG) (7) in 250 µl PBS three times per week for 8 wk, starting 1 day after T cell transfer, in the preventive protocol. An equivalent amount of hamster IgG (BioTrend, Cologne, Germany) was administered in control mice. Because clinical and histopathological manifestations of colitis were evident from 3–5 wk after T cell transfer in our preliminary experiments, we treated another group of mice by injection of 250 µg ip of MFL or control IgG three times per week for 5 wk, starting from 3 wk after T cell transfer, in the therapeutic protocol. All mice were killed at 8 wk after T cell transfer for histological examination and preparation of LP mononuclear cells (LPMC).

Clinical scoring. Mice were weighed weekly and monitored for gross appearance and signs of soft stool and/or diarrhea. Clinical score was determined as previously described (25). Briefly, we assessed the gross appearance (no hunching or wasting as 0 points, mild-to-moderate hunching or wasting as 1 point, and severe hunching or wasting as 2 points), the stool consistency (well-formed pellets as 0 points, loose stool as 1 point, and liquid stool or bloody stool as 2 points). These scores were summed for individual mice resulting in a total clinical score ranging from 0 (healthy) to 4 (maximal activity of colitic disease).

Histological examination and immunohistochemistry. Tissue samples were fixed in PBS containing 6% neutral-buffered formalin. Paraffin-embedded sections (5 µm) were stained with hematoxylin and eosin. The degree of inflammation (histological score) was determined as previously described (6). Tissue samples for immunohistochemistry were embedded in optimal cutting temperature compound and snap frozen in liquid nitrogen. Six-micrometer sections were incubated with biotinylated anti-mouse CD4 MAb (model RM4-5), biotinylated anti-mouse F4/80 MAb (Serotek, Oxford, UK), and biotinylated isotype-matched control IgG (BD Pharmingen). Biotinylated antibodies were detected by using Vectastain ABC kit (Vector, Burlingame, CA). The sections were finally counterstained with hematoxylin.

Preparation of LPMC. The entirely dissected colon was opened longitudinally, washed with PBS, cut into small pieces, incubated with Ca2+- and Mg2+-free Hanks' balanced salt solution containing 1 mM dithiothreitol (Sigma, St. Louis, MO) for 30 min to remove mucus, and then serially incubated two times in medium containing 0.75 mM EDTA (Sigma) for 60 min. The supernatants from these incubations were collected and treated with 1 mg/ml collagenase A (Roche, Indianapolis, IN) in medium for 2 h. The cells were pelleted two times through a 40% isotonic Percoll solution, and then mononuclear cells were isolated by Ficoll-Hypaque density gradient centrifugation (40/70%) at the interface. CD4+ LP T cells were isolated from the LPMC by positive selection using the anti-CD4 (L3T4) MACS magnetic separation system. The cells were >95% CD4+ as analyzed by flow cytometry.

Flow cytometry. Isolated LPMC were preincubated with an Fc-{gamma} receptor-blocking MAb (model 2.4G2; BD Pharmingen) for 20 min, followed by incubation with FITC-conjugated anti-CD4 MAb for 30 min on ice. Flow cytometric analysis was performed on FACS Calibur (Becton Dickinson) equipped with CellQuest software.

To assess the production of TNF-{alpha} by CD4+ T cells and CD11b+ macrophages, we analyzed intracellular TNF-{alpha} by flow cytometry. Freshly isolated LPMCs were cultured with PMA (50 ng/ml; Sigma) and ionomycin (250 ng/ml; Sigma) to activate CD4+ T cells or LPS (1 µg/ml; Sigma) to activate macrophages in the presence of MFL (10 µg/ml) or control IgG for 6 h. Brefeldin A (10 µg/ml; Sigma) was added for the last5hto induce accumulation of cytokines in the Golgi body. The cells were then stained with anti-CD4-FITC (model L3T4; BD Pharmingen) or anti-CD11b-FITC (model M1/70; BD Pharmingen). After being washed, the cells were fixed and permeabilized for 15 min with 4% formaldehyde containing 0.1% saponin. For intracellular staining, the cells were incubated with anti-TNF-{alpha}-PE (model MP6-XT22; BD Pharmingen) and analyzed on a FACS Calibur.

To exclude the possibility that the MFL treatment induces nonspecific killing of FasL-expressing cells, we treated the colitic mice at 6 wk after the transfer of CD4+ CD45RBhigh cells with a single dose of MFL or control IgG (250 µg). After isolating LPMCs, we determined the number of apoptotic/dead cells by staining with annexin V-FITC/propidium (PI; MBL, Nagoya, Japan).

Cytokine ELISA. To measure IFN-{gamma} production, LP CD4+ T cells (1 x 105) were cultured in 200 µl of complete medium supplemented with 1 µg/ml anti-CD28 MAb (model 37.51; BD Pharmingen) in 96-well flat-bottomed plates (Costar, Cambridge, MA) precoated with 10 µg/ml anti-CD3 MAb (model 145–2C11; BD Pharmingen) for 48 h. To measure TNF-{alpha} and IL-1{beta} production, LPMC (5 x 105) were cultured in 200 µl of complete medium supplemented with 10 µg/ml anti-CD3 MAb and/or 10 µg/ml MFL in 96-well flat-bottomed plates for 24 h. IFN-{gamma}, TNF-{alpha}, and IL-1{beta} concentrations in the culture supernatant were determined by specific ELISA according to the manufacturer's instructions (R&D Systems, Minneapolis, MN).

Statistical analysis. Results were expressed as means ± SE. Data were statistically analyzed by the Mann-Whitney U-test. P values <0.05 were considered to be statistically significant.


    RESULTS
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
Administration of MFL ameliorates wasting disease but not intestinal inflammation. We induced chronic colitis in BALB/c SCID mice by adoptive transfer of CD4+ CD45RBhigh T cells from normal BALB/c mice. The recipient mice manifested a progressive weight loss from 3 wk after T cell transfer and clinical symptoms of colitis such as diarrhea with increased mucus in the stool, anorectal prolapse, and hunched posture by 6–8 wk. The colon from these mice was enlarged and had a greatly thickened wall due to severe inflammation. To explore the contribution of Fas/FasL interaction to the development of chronic colitis, the recipient mice were treated with a neutralizing MFL or control hamster IgG for 8 wk from 1 day after T cell transfer. As shown in Fig. 1A, the control IgG-treated mice manifested progressive weight loss (wasting disease) during 3–8 wk after T cell transfer. In contrast, the MFL-treated mice did not show the progressive weight loss. The average body weight at 8 wk relative to the initial weight was 103.6 ± 3.5% for the MFL-treated mice and 87.5 ± 3.7% for the control IgG-treated mice (P = 0.009). The MFL-treated mice looked healthy during the observation period, and the assessment of colitic symptoms by clinical scoring showed a significant difference (P = 0.01) between the MFL-treated mice and the control IgG-treated mice (Fig. 1B). Dissection at 8 wk showed that the colon from the control IgG-treated mice was enlarged (Fig. 2A) and had a greatly thickened wall (Fig. 2B). In contrast to the improvement of wasting and clinical symptoms by the MFL treatment (Fig. 1), these changes in the colon were not improved in the MFL-treated mice (Fig. 2, A and B). Histological examination showed massive inflammatory infiltrates composed of a large number of lymphocytes and macrophages/dendritic cells and a small number of neutrophils and eosinophils in the colon from the control IgG- or MFL-treated mice. No significant difference was observed by histological scoring of multiple colon sections, which was 6.5 ± 0.7 for the MFL-treated mice vs. 7.0 ± 0.4 for the control IgG-treated mice (Fig. 2C). Immunohistochemical staining also showed no apparent difference in the infiltration of CD4+ T cells and F4/80+ macrophages in the colon between the control IgG- and MFL-treated mice (Fig. 3). A further quantitative evaluation of CD4+ T cell infiltration was made by isolating LPMC from the whole colon and flow cytometric analysis. The average CD4+ T cell number at 8 wk was 30.4 ± 5.9 x 105 cells/colon for the MFL-treated mice vs. mice given 34.5 ± 7.2 x 105 cells/colon for the control IgG-treated mice (P = 0.67, not significantly different). Because the numbers of CD4+ T cells recovered from the colon of these mice were comparably higher than the number of injected cells (3–5 x 105), these results suggested that neither expansion nor deletion of CD4+ cells was affected by the MFL treatment. Finally, to exclude the possibility that the MFL treatment induced nonspecific killing of FasL-expressing cells, we treated the colitic mice at 6 wk after the transfer of CD4+ CD45RBhigh T cells with a single dose of MFL or control IgG (250 µg) and determined the percentages of apoptotic or dead cells in LP cells by the PI/annexin V staining. We confirmed that there was no significant difference between anti-FasL-treated and control IgG-treated LPMCs (anti-FasL vs. control IgG; annexin V+ PI cells, 9.4 ± 0.5 vs. 8.5 ± 1.0, P = 0.31; annexin V+ PI+ cells, 22.5 ± 0.9 vs. 20.9 ± 0.8, P = 0.56).



View larger version (15K):
[in this window]
[in a new window]
 
Fig. 1. Ameliorating effect of anti-Fas/Fas ligand (FasL) MAb treatment on wasting and clinical manifestations. Recipient mice were administered with anti-FasL MAb or control IgG for 8 wk starting 1 day after CD4+ CD45RBhigh T cell transfer. A: change in body weight over time is expressed as the percent of original weight. Data are indicated as the means ± SE of 6 mice in each group. *P < 0.05 compared with control IgG. B: clinical score was determined at 8 wk after T cell transfer. Data are indicated as the means ± SE of 6 mice in each group. *P < 0.05. Ab, antibody.

 


View larger version (35K):
[in this window]
[in a new window]
 
Fig. 2. Macro- and microscopic findings of the colon. A: gross appearance of the colon from normal BALB/c mice and control IgG- or anti-FasL MAb-treated colitic mice at 8 wk after T cell transfer. B: histological appearance of the colon from normal BALB/c mice and control IgG- or anti-FasL MAb-treated colitic mice at 8 wk after T cell transfer. Original magnification, x40. (C) histological scoring of colitis in control IgG- or anti-FasL MAb-treated mice at 8 wk after T cell transfer. Data are indicated as the means ± SE of 4 mice in each group. NS, not significantly different.

 


View larger version (124K):
[in this window]
[in a new window]
 
Fig. 3. Infiltration of CD4+ T cells and F4/80+ macrophages in the colon. Frozen sections of the colon from control IgG- or anti-FasL MAb-treated colitic mice at 8 wk after T cell transfer and normal BALB/c mice were stained with anti-CD4 or F4/80 MAb. Original magnification, x200.

 

Effect of MFL on proinflammatory cytokine production. To explore the mechanism by which the MFL treatment ameliorated the wasting disease and clinical symptoms without affecting histological inflammation, we next examined the effect of MFL on the function of LPMC in vitro. As shown in Fig. 4A, LP CD4+ T cells from the control IgG-treated mice and those from the MFL-treated mice produced a comparable level of IFN-{gamma} on stimulation with anti-CD3 and CD28 MAbs in vitro, indicating that the development and infiltration of IFN-{gamma}-producing Th1 cells was not affected by the MFL treatment. Interestingly, production of TNF-{alpha} and IL-1{beta} by LPMC from colitic mice was significantly inhibited by the addition of MFL (Fig. 4B). We next examined the effect of MFL on TNF-{alpha} production by CD4+ T cells and CD11b+ macrophages by intracellular staining. As shown in Fig. 4C, TNF-{alpha} production by both CD4+ T cells and CD11b+ macrophages were significantly decreased by MFL to control IgG. These results suggested that the MFL treatment prevented the wasting disease possibly by inhibiting the TNF-{alpha} release from both LP CD4+ T cells and CD11b+ macrophages.



View larger version (23K):
[in this window]
[in a new window]
 
Fig. 4. Effect of anti-FasL MAb on cytokine production. A: lamina propria CD4+ T cells were isolated from normal BALB/c mice and control-IgG- or anti-FasL MAb-treated mice at 8 wk after T cell transfer, cultured for 48 h in the presence of anti-CD3 and anti-CD28 MAbs, and then culture supernatants were analyzed for concentration of IFN-{gamma} by ELISA. Data are indicated as the means ± SE of 6 mice in each group. B: lamina propria mononuclear cells were isolated from control IgG-treated colitic mice at 8 wk after T cell transfer, cultured for 24 h in the presence of anti-CD3 and/or anti-FasL MAb, and then culture supernatants were analyzed for concentration of TNF-{alpha} and IL-1{beta} by ELISA. Data are indicated as the means ± SE of 7 mice in each group. *P < 0.05. C: lamina propria mononuclear cells were isolated from control IgG-treated colitic mice at 6 wk after T cell transfer, stimulated with PMA + ionomycin or LPS for 6 h in the presence of anti-FasL MAb or control IgG, and then stained for cell-surface CD4 or CD11b and intracellular TNF-{alpha}. The percentages in the quadrants represent means ± SE of 4 mice.

 

Therapeutic effect of MFL. We next evaluated whether a delayed MFL treatment could improve ongoing wasting disease. Since the wasting disease started 2–3 wk after T cell transfer and the infiltration of lymphocytes was already detectable at 2 wk in our model, we started the MFL treatment from 3 wk after T cell transfer. As shown in Fig. 5A, the delayed MFL treatment significantly improved weight loss compared with control IgG treatment at 8 wk after T cell transfer (P = 0.034). The clinical score at 8 wk was also significantly improved by the delayed MFL treatment (Fig. 5B, P = 0.015). Consistent with the prevention study, however, there was no histological difference in colonic sections between the MFL-treated mice and the control IgG-treated mice (data not shown).



View larger version (15K):
[in this window]
[in a new window]
 
Fig. 5. Therapeutic effect of anti-FasL MAb. Recipient mice were administered with anti-FasL MAb or control IgG for 5 wk starting at 3 wk after T cell transfer (arrow). A: change in body weight over time is expressed as the percent of original weight. Data are indicated as the means ± SE of 4 mice in each group. *P < 0.05 compared with control IgG. B: clinical score was determined at 8 wk after T cell transfer. Data are indicated as the means ± SE of 4 mice in each group. *P < 0.05.

 


    DISCUSSION
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
In this study, we demonstrated that the administration of neutralizing MFL ameliorated wasting disease and clinical symptoms without affecting local inflammation in a murine model of chronic colitis. In vitro experiments suggested that the ameliorating effect of MFL might be mediated by suppression of TNF-{alpha} production by both LP CD4+ T cells and CD11b+ macrophages.

It has been suggested that the Fas/FasL interaction may regulate T cell apoptosis in the mucosa (3, 12). On the other hand, it has been reported that the Fas/FasL interaction is not only involved in T cell apoptosis but also in T cell costimulation (8, 21). Our present results showed that the MFL treatment did not affect the number of CD4+ LP T cells collected from the inflamed colon. In addition, IFN-{gamma} production by LP CD4+ T cells was not also affected by the MFL treatment. These results suggested that the Fas/FasL interaction does not play a critical role in either costimulating T cell expansion and Th1 development or depleting infiltrating T cells in the mucosa.

Recent studies (9, 11, 19) have shown that FasL could induce TNF-{alpha} and IL-1{beta} production by activated CD4+ T cells, macrophages, and dendritic cells in vitro. Therefore, the inhibitory effect of MFL on TNF-{alpha} and IL-1{beta} production by LPMC in vitro is likely mediated by interruption of the interaction between FasL-expressing T cells and Fas-expressing macrophages and/or dendritic cells. Since both TNF-{alpha} and IL-1{beta} have been implicated in the pathogenesis of wasting disease associated with colitis (5), the ameliorating effect of MFL on wasting disease might be mediated by suppression of TNF-{alpha} and IL-1{beta} production in vivo. Consistent with this notion, we recently demonstrated that therapeutic administration of neutralizing anti-TNF-{alpha} MAb improved the ongoing wasting disease without improving the intestinal inflammation (26), just like MFL in the present study. Further studies are needed to determine the effect of MFL on the production of these cytokines in vivo.

It has been reported that FasL expression is massively upregulated in Fas-deficient T cells from lpr mice, and transplantation of bone marrow cells from lpr mice to syngeneic wild-type recipients induces graft-vs.-host disease-like wasting disease (4). In contrast, transplantation of Fas/FasL double-deficient bone marrow cells did not induce the wasting disease (27). This substantiates a critical contribution of the Fas/FasL interaction among bone marrow-derived leukocytes to the pathogenesis of wasting disease, which is consistent with our present observation in a murine colitis model.

Previous studies (2, 15, 23) have suggested that the Fas/FasL interaction may be involved in the IEC damage, since activated T cells or IEC expressed FasL and killed IEC. Although we have not directly assessed the IEC damage in this study, the ameliorating effect of MFL on some clinical symptoms, such as diarrhea, might be partly mediated by improvement of the IEC damage. Further studies using an oxazolone colitis model, which is characterized by the presence of epithelial cell loss and patchy ulceration, are needed to address this point. Alternatively, the perforin pathway should also be addressed, because it was previously reported (10, 22) to be, at least in part, involved in the T cell-mediated mucosal injury and might explain the minor therapeutic effects of MFL in this study.

In the present study, we also demonstrated that the MFL treatment could ameliorate ongoing wasting disease and improve clinical symptoms in a therapeutic protocol. This suggests that the FasL blockade with a humanized MFL (13) may be a novel strategy to improve the quality of life of the patient with chronic colitis such as Crohn's disease.


    DISCLOSURES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 
This work was supported, in part, by grants-in-aid from the Japanese Ministry of Education, Culture, and Science and the Japanese Ministry of Health and Welfare.


    ACKNOWLEDGMENTS
 
The authors express special thanks to Hanae Fujimoto and Hiroshi Nishikawa for technical assistances.


    FOOTNOTES
 

Address for reprint requests and other correspondence: T. Kanai, Dept. of Gastroenterology and Hepatology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyoku, Tokyo 113-8519, Japan (E-mail address: taka.gast{at}tmd.ac.jp).

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.


    REFERENCES
 TOP
 ABSTRACT
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 DISCLOSURES
 REFERENCES
 

  1. Ashany D, Song X, Lacy E, Nikolic-Zugic J, Friedman SM, and Elkon KB. Th1 CD4+ lymphocytes delete activated macrophages through the Fas/APO-1 antigen pathway. Proc Natl Acad Sci USA 92: 11225–11229, 1995.[Abstract]
  2. Bonhagen K, Thoma S, Bland P, Bregenholt S, Rudolphi A, Claesson MH, and Reimann J. Cytotoxic reactivity of gut lamina propria CD4+ alpha beta T cells in SCID mice with colitis. Eur J Immunol 26: 3074–3083, 1996.[ISI][Medline]
  3. Bregenholt S, Petersen TR, and Claesson MH. The majority of lamina propria CD4+ T-cells from SCID mice with colitis undergo Fas-mediated apoptosis in vivo. Immunol Lett 78: 7–12, 2001.[ISI][Medline]
  4. Chu JL, Ramos P, Rosendorff A, Nikolic-Zugic J, Lacy E, Matsuzawa A, and Elkon KB. Massive upregulation of the Fas ligand in lpr and gld mice: implications for Fas regulation and the graft-versus-host disease-like wasting syndrome. J Exp Med 181: 393–398, 1995.[Abstract]
  5. Fiocchi C. Inflammatory bowel disease etiology and pathogenesis. Gastroenterology 115: 182–205, 1998.[ISI][Medline]
  6. Kanai T, Watanabe M, Okazawa A, Sato T, Yamazaki M, Okamoto S, Ishii H, Totsuka T, Iiyama R, Okamoto R, Ikeda M, Kurimoto M, Takeda K, Akira S, and Hibi T. Macrophage-derived IL-18-mediated intestinal inflammation in the murine model of Crohn's disease. Gastroenterology 121: 875–888, 2001.[ISI][Medline]
  7. Kayagaki N, Yamaguchi N, Nagao F, Matsuo S, Maeda H, Okumura K, and Yagita H. Polymorphism of murine Fas ligand that affects the biological activity. Proc Natl Acad Sci USA 94: 3914–3919, 1997.[Abstract/Free Full Text]
  8. Kennedy NJ, Kataoka T, Tschopp J, and Budd RC. Caspase activation is required for T cell proliferation. J Exp Med 190: 1891–1896, 1999.[Abstract/Free Full Text]
  9. Lu B, Wang L, Medan D, Toledo D, Huang C, Chen F, Shi X, and Rojanasakul Y. Regulation of Fas (CD95)-induced apoptosis by nuclear factor-{kappa}B and tumor necrosis factor-{alpha} in macrophages. Am J Physiol Cell Physiol 283: C831–C838, 2002.[Abstract/Free Full Text]
  10. Merger M, Viney JL, Borojevic R, Steele-Norwood D, Zhou P, Clark DA, Riddell R, Maric R, Podack ER, and Croitoru K. Defining the roles of perforin, Fas/FasL, and tumour necrosis factor alpha in T cell induced mucosal damage in the mouse intestine. Gut 51: 155–63, 2002.[Abstract/Free Full Text]
  11. Miwa K, Asano M, Horai R, Iwakura Y, Nagata S, and Suda T. Caspase 1-independent IL-1{beta} release and inflammation induced by the apoptosis inducer Fas ligand. Nat Med 4: 1287–1292, 1998.[ISI][Medline]
  12. Neurath MF, Finotto S, Fuss I, Boirivant M, Galle PR, and Strober W. Regulation of T-cell apoptosis in inflammatory bowel disease: to die or not to die, that is the mucosal question. Trends Immunol 22: 21–26, 2001.[ISI][Medline]
  13. Nisihara T, Ushio Y, Higuchi H, Kayagaki N, Yamaguchi N, Soejima K, Matsuo S, Maeda H, Eda Y, Okumura K, and Yagita H. Humanization and epitope mapping of neutralizing anti-human Fas ligand monoclonal antibodies: structural insights into Fas/Fas ligand interaction. J Immunol 167: 3266–3275, 2001.[Abstract/Free Full Text]
  14. Orlinick JR, Vaishnaw AK, and Elkon KB. Structure and function of Fas/Fas ligand. Int Rev Immunol 18: 293–308, 1999.[Medline]
  15. Pinkoski MJ, Brunner T, Green DR, and Lin T. Fas and Fas ligand in gut and liver. Am J Physiol Gastrointest Liver Physiol 278: G354–G366, 2000.[Abstract/Free Full Text]
  16. Podolsky DK. Inflammatory bowel disease. N Engl J Med 325: 928–937, 1991.[ISI][Medline]
  17. Powrie F, Leach MW, Mause S, Menon S, Caddle LB, and Coffman RL. Inhibition of Th1 responses prevents inflammatory bowel disease in SCID mice reconstituted with CD45Rbhi CD4+ T cells. Immunity 1: 553–562, 1994.[ISI][Medline]
  18. Present DH, Rutgeerts P, Targan S, Hanauer SB, Mayer L, van Hogezand RA, Podolsky DK, Sands BE, Braakman T, DeWoody KL, Schaible TF, and van Deventer SJ. Infliximab for the treatment of fistulas in patients with Crohn's disease. N Engl J Med 340: 1398–1405, 1999.[Abstract/Free Full Text]
  19. Rescigno M, Piguet V, Valzasina B, Lens S, Zubler R, French L, Kindler V, Tschopp J, and Ricciardi-Castagnoli P. Fas engagement induces the maturation of dendritic cells (DCs), the release of interleukin (IL)-1{beta}, and the production of interferon gamma in the absence of IL-12 during DC-T cell cognate interaction: a new role for Fas ligand in inflammatory responses. J Exp Med 192: 1661–1668, 2000.[Abstract/Free Full Text]
  20. Sabelko-Downes KA, and Russell JH. The role of Fas ligand in vivo as a cause and regulator of pathogenesis. Curr Opin Immunol 12: 330–335, 2000.[ISI][Medline]
  21. Siegel RM, Chan FK, Chun HJ, and Lenardo MJ. The multifaceted role of Fas signaling in immune cell homeostasis and autoimmunity. Nat Immun 1: 469–474, 2000.[ISI]
  22. Simpson SJ, De Jong YP, Shah SA, Comiskey M, Wang B, Spielman JA, Podack ER, Mizoguchi E, Bhan AK, and Terhorst C. Consequences of Fas-ligand and perforin expression by colon T cells in a mouse model of inflammatory bowel disease. Gastroenterology 115: 849–55, 1998.[ISI][Medline]
  23. Strater J, Wellisch I, Riedl S, Walczak H, Koretz K, Tandara A, Krammer PH, and Moller P. CD95 (APO-1/Fas)-mediated apoptosis in colon epithelial cells: a possible role in ulcerative colitis. Gastroenterology 113: 160–167, 1997.[ISI][Medline]
  24. Targan SR, Hanauer SB, van Deventer SJH, Mayer L, Present DH, Braakman T, Dewoody KL, Schaible TF, and Rutgeerts PJ. A short term study of chimeric monoclonal antibody (cA2) to tumor necrosis factor alpha for Crohn's disease. N Engl J Med 337: 1029–1035, 1997.[Abstract/Free Full Text]
  25. Totsuka T, Kanai T, Iiyama R, Uraushihara K, Yamazaki M, Okamoto R, Hibi T, Tezuka K, Azuma M, Akiba H, Yagita H, Okumura K, and Watanabe M. Ameliorating effect of anti-inducible costimulator monoclonal antibody in a murine model of chronic colitis. Gastroenterology 124: 410–421, 2003.[ISI][Medline]
  26. Totsuka T, Kanai T, Uraushihara K, Iiyama R, Akiba H, Yagita H, Okumura K, and Watanabe M. Preventive and therapeutic effects of chronic colitis in CD4+CD45RB+ T cell-transferred SCID mice by anti-OX40L MAb administration. Am J Physiol Gastrointest Liver Physiol 284: G595–G603, 2003.[Abstract/Free Full Text]
  27. Zhu B, Beaudette BC, Rifkin IR, and Marshak-Rothstein A. Double mutant MRL-lpr/lpr-gld/gld cells fail to trigger lprgraft-versus-host disease in syngeneic wild-type recipient mice, but can induce wild-type B cells to make autoantibody. Eur J Immunol 30: 1778–1784, 2000.[ISI][Medline]




This Article
Abstract
Full Text (PDF)
Alert me when this article is cited
Alert me if a correction is posted
Citation Map
Services
Email this article to a friend
Similar articles in this journal
Similar articles in ISI Web of Science
Similar articles in PubMed
Alert me to new issues of the journal
Download to citation manager
Search for citing articles in:
ISI Web of Science (1)
Google Scholar
Articles by Dan, N.
Articles by Watanabe, M.
Articles citing this Article
PubMed
PubMed Citation
Articles by Dan, N.
Articles by Watanabe, M.


HOME HELP FEEDBACK SUBSCRIPTIONS ARCHIVE SEARCH TABLE OF CONTENTS
Visit Other APS Journals Online
Copyright © 2003 by the American Physiological Society.