To understand the perpetuation of inflammatory bowel disease (IBD), it is important to clarify whether the colitogenic CD4+ T cells are self-limited effector or long-lived memory T cells. We here investigate the latency of colitogenic CD4+ T cells in the remission stage of colitis under germfree (GF) conditions. We isolated splenic (SP) CD4+ T cells from colitic CD4+CD45RBhigh T cell-injected SCID mice maintained under specific pathogen-free (SPF) conditions and transferred them into SPF or GF SCID mice. Donor colitic SP CD4+ T cells have a characteristic CD44highCD62LIL-7Rαhigh effector-memory T-type phenotype. Six weeks after transfer of cells to GF SCID mice, one group of mice was continued in GF conditions (GF→GF), and the other was transferred into SPF conditions (GF→SPF). GF→SPF but not GF→GF SCID mice developed colitis with elevated production of Th1 and Th17 cytokines at 4 wk after transfer. Surprisingly, a large number of CD4+ effector-memory T cells and a small but substantial number of central-memory T cells remained resident in SP and bone marrow, but not in lamina propria, of the GF→GF SCID recipients. Consistent with this, GF→SPF but not GF→GF SCID mice rapidly developed colitis. Taken together, these findings suggest that long-lived colitogenic memory CD4+ cells can be established even in the presence of commensal Ags, reside outside the intestine in the absence of commensal bacteria, and participate in the perpetuation of colitis. Thus, blocking a stimulus of colitogenic memory CD4+ cells such as IL-7 may have therapeutic benefit for treatment of inflammatory bowel disease.

Studies of Ag exposure in murine models of acute virus infection have provided much information about the dynamics of naive, effector, and memory CD8+ T cell responses (1, 2, 3). Acute virus infection elicits massive proliferation of viral Ag-specific CD8+ T cells, which acquire effector functions (effector phase). After the peak of T cell proliferation, most of the effector CD8+ T cells are eliminated (contraction phase). Following virus (Ag) clearance, a small proportion of the remaining cells differentiate into memory CD8+ T cells that are maintained by cytokine (IL-7 and/or IL-15)-dependent homeostatic proliferation and survive in the absence of their corresponding Ags (memory phase). Thus, Ag clearance is essential for the emergence of memory CD8+ T cells.

In contrast to the generation of CD8+ memory T cells, it is controversial whether Ag clearance is needed for the generation of CD4+ memory T cells. Indeed, there is evidence that persistence of the Ag itself is essential for the maintenance of CD4+ memory T cells (4, 5, 6). Given that Ag-specific effector T cells are thought to be terminally differentiated and short-lived cells, there must be cellular mechanisms by which memory T cells specific for persistent Ags are maintained in the host as “memory-stem cells.” In support of the idea that persistent Ag helps maintain CD4+ memory T cells, rats immunized with irradiated sporozoites are protected from malaria infection, but this protection is lost after drug treatment to remove the remaining parasites (5). A similar phenomenon is found in Leishmania major infection in mice where CD4+CD25+ regulatory T cells and IL-10 are thought to prevent a Th1 immune response from clearing the infection, thus allowing the host to retain CD4+ memory T cells specific to the organism (6). Furthermore, more recent data have shown that in the absence of MHC class II signals that are essential for Ag presentation to CD4+ T cells, surviving memory T cells are functionally impaired, suggesting that TCR signaling is involved in the maintenance of CD4+ memory T cells (7).

All complex metazoans, including humans and mice, are colonized with microbial organisms that comprise an indigenous microflora (8). Although the host evidently benefits from the resident microflora in the gut (9), the presence of commensal bacteria appears to be of crucial importance in the development of human inflammatory bowel disease (IBD)3 and in almost all animal models of IBD (10, 11). For instance, results from most IBD models showing that in germfree (GF) rodents intestinal inflammation is absent indicate that commensal bacteria are indispensable contributors to the pathogenesis of chronic immune-mediated intestinal inflammation.

IBD is caused by excessive and tissue-damaging chronic inflammatory responses, which are thought to be due to inappropriate activation of the immune system in many cases and which commonly take a persistent, disabling course (10, 11). In some patients, disease progresses steadily, whereas in others, it follows a relapsing-remitting course. According to present understanding, the disease is caused and controlled by colitogenic effector and memory CD4+ T cells presumably reacting to commensal bacterial Ags. Importantly, however, it is not known whether different effector CD4+ T cells are recruited at each relapse or whether sequential memory CD4+ T cells are derived from members of the initial attack cohort throughout the entire course of disease. In other words, the nature and regulation of colitogenic effector and memory CD4+ T cells in the host-commensal interaction over time and the correlation between colitogenic effector and memory CD4+ T cells in chronic colitis remain largely unknown.

Given the possible importance of the microflora in intestinal inflammation, we conducted a series of experiments to test the in vivo effect of commensal bacteria using a GF system for the establishment and maintenance of colitogenic CD4+ T cells. We also attempted to assess whether colitogenic CD4+ T cells are self-limited effector cells or long-lived memory cells in a host-commensal bacteria mutualism to understand the perpetuation of IBD and to develop a strategy for IBD treatment.

C3H/HeN mice (6–8 wk old) and C57BL/6-Ly5.2 mice were purchased from CLEA Japan. C57BL/6-Ly5.1 and C57BL/6-Ly5.2 RAG-2−/− mice were obtained from Taconic Laboratory and the Central Experimental Animal Institute. Specific pathogen-free (SPF) and GF breeding colonies of C3H-SCID mice (C3Smnc Prkdc scid/J; The Jackson Laboratory) were maintained at the Animal Facilities (SPF) and the Gnotobiotic Facilities (GF), respectively, of our institute. Sterility in the Gnotobiotic Facilities was tested monthly by culturing of feces and bedding as well as Gram staining. The Institutional Committees on Animal Research of both Tokyo Medical and Dental University and Yakult Central Institute approved the experiments.

The following mAbs other than biotin-conjugated anti-mouse IL-7Rα (A7R34; eBioscience) and anti-CCR7 (EBI-1; eBioscience) were obtained from BD Pharmingen and used for purification of cell populations and flow cytometric analysis: Fcγ (CD16/CD32)-blocking mAb (2.4G2); PE-, PerCP-, and PECy5-conjugated anti-mouse CD4 (RM4-5); FITC-conjugated anti-mouse CD3 (145-2C11); PE- and allophycocyanin-conjugated anti-mouse CD44 (IM7); FITC- and PE-conjugated anti-mouse L-selectin (CD62L) (MEL-14); FITC-conjugated anti-mouse CD69 (H1.2F3); PE-conjugated anti-mouse α4β7 (DATK32); FITC-conjugated anti-mouse CD45RB (16A); PE-conjugated anti-mouse Ly5.1 (CD45.1); PE-conjugated streptavidin; biotin-conjugated rat IgG2; PE-conjugated mouse IgG; and PE-conjugated rat IgG.

To assess the role of commensal bacteria in the persistence of T cell-mediated chronic colitis, CD4+ T cells (5 × 105 cells/mouse) isolated from spleen (SP) of colitic mice induced by an adoptive transfer of CD4+CD45RBhigh T cells into C3H-SCID mice under SPF conditions were injected into new GF (n = 16) or SPF (n = 8) C3H-SCID mice. Six weeks after the transfer, one group of GF SCID recipients was maintained in GF conditions (GF→GF group, n = 8), and the other was moved into SPF conditions (GF→SPF group, n = 8). All groups (SPF, GF→GF, and GF→SPF) were kept for an additional 4 wk and sacrificed 10 wk after cell transfer. They were observed for clinical signs such as hunched posture, piloerection, diarrhea, and blood in the stool. After sacrifice, mice were given a clinical score defined as the sum of four parameters: hunching and wasting, 0 or 1; colon thickening, 0–3 (0, no colon thickening; 1, mild thickening; 2, moderate thickening; 3, extensive thickening); and stool consistency, 0–3 (0, normal beaded stool; 1, soft stool; 2, diarrhea; 3, bloody stool) (12). To assess the histological scores, two parts of the colon were evaluated. The proximal section was defined as 1 cm anal to the cecum, and the distal section was defined as 1 cm oral from the anus. Both tissue samples were cut 5 mm long and fixed in PBS containing 10% neutral-buffered formalin. Paraffin-embedded sections (5 μm) were stained with H&E. The sections were analyzed without prior knowledge of the type of T cell reconstitution or recipient. The mean degree of inflammation in the colon was calculated using a modification of a previously described scoring system as the sum of three parameters: crypt elongation, 0–3; mononuclear cell infiltration, 0–3; and frequency of crypt abscesses, 0–3 (13).

CD4+ T cells were isolated from lamina propria (LP) of colitic RAG2−/− mice previously injected with Ly5.1+ CD4+CD45RBhigh T cells and were labeled with CFSE (Invitrogen) at a concentration of 5 μM according to the manufacturer’s instructions; then, CFSE-labeled LP CD4+ T cells (1 × 107/mouse) were injected into new Ly5.2+ wild-type (WT) C57BL/6J mice.

C57BL/6J mice in the antibiotic-treated group were given drinking water, including ampicillin (1 g/L), vancomycin (500 mg/L), neomycin sulfate (1 g/L), and metronidazole (1 g/L), 2 wk before beginning the adoptive transfer and during the course of the experiment. Control mice received drinking water without antibiotics.

Anti-IL-7Rα mAb (A7R34) was described previously (14). GF SCID mice were treated with rat anti-murine IL-7Rα mAb by i.p. injection of a 1-mg dose once per week for 2 wk (0, 1, and 2 wk after transfer). Control mice were treated with the same amounts of rat control IgG (Sigma-Aldrich). All mice were killed on the day after the last treatment.

To detect the surface expression of a variety of molecules, isolated bone marrow (BM), SP, mesenteric lymph nodes (MLN), or colonic LP mononuclear cells were preincubated with an FcR-blocking mAb (CD16/32 and 2.4G2; BD Pharmingen) for 20 min before incubation with specific FITC-, PE-, PerCP-, and APC-labeled Abs for 30 min on ice. When biotin-conjugated Abs were used, cells were incubated with Abs for 30 min, then with PE-labeled streptavidin for 30 min on ice. Standard four-color flow cytometric analyses were performed using a FACSCalibur (BD Biosciences) and analyzed by CellQuest software (BD Biosciences). Background fluorescence was assessed by staining with irrelevant isotype-matched mAbs.

To measure cytokine production, 1 × 105 CD4+ T cells from LP were cultured in 200 μl of culture medium at 37°C in a humidified atmosphere containing 5% CO2 in 96-well plates (Costar) precoated with 5 μg/ml hamster anti-mouse CD3ε mAb (145-2C11; BD Pharmingen) and 2 μg/ml hamster anti-mouse CD28 mAb (37.51; BD Pharmingen) in PBS overnight at 4°C. Culture supernatants were removed after 48 h and assayed for cytokine production. Cytokine concentrations were determined by specific ELISA as per the manufacturer’s recommendation (R&D Systems).

Consecutive cryostat BM and colon sections were used in all studies. Immunohistochemistry was performed using purified mAb against mouse CD4 (RM4-5; BD Pharmingen) or biotin-conjugated polyclonal IL-7 Ab (BAF407; R&D Systems). In brief, fresh frozen tissue samples were cut into serial sections 6 μm thick, placed on coated slides, and fixed with 4% paraformaldehyde phosphate buffer solution for 10 min. Slides were incubated with the primary Ab at 4°C overnight, then stained for 60 min at room temperature with AlexaFluor 488 goat anti-rat IgG for CD4 detection or AlexaFluor 488 streptavidin (Molecular Probes) for IL-7 detection. All slides were counterstained with 4,6-diamidino-2-phenylindole (Vector Laboratories) and observed under a BioZERO BZ8000 microscope (Keyence).

The results were expressed as the mean ± SEM. Groups of data were compared by the Mann-Whitney U test. Differences were considered to be statistically significant when p < 0.05.

In research into antiviral CD8+ memory T cells, memory T cells are classically defined as cells that persist in the organism once the viral Ag has been cleared and that mediate a much quicker and stronger response when the Ag is met again (1, 2, 3). However, current evidence suggests that maintenance of a memory CD4+ T cell population responding to chronic infection is dependent on the persistent presence of Ag (4, 5, 6), and it is controversial whether CD4+ memory T cells can be maintained in the absence of Ag. In particular, to understand the persistence of IBD, it is important to know whether the colitogenic CD4+ T cells are self-limited effector cells or are long-lived memory cells within the host-commensal bacteria mutualism. We attempted to clarify this issue using a well-known chronic colitis model induced by adoptive transfer of CD4+CD45RBhigh T cells into SCID mice, which characteristically involves the differential activation of Th1/Th17 cells (15, 16). We first checked the phenotype of CD4+ T cells in colonic LP, MLN, SP, and BM of colitic C3H-SCID mice that had previously been injected with syngeneic CD4+CD45RBhigh T cells. Consistent with our previous reports using BALB/c/C.B.17 or C57BL/6J strains (12, 13), LP CD4+ T cells, as well as colitic SP, MLN, and BM CD4+ T cells, are exclusively CD44highCD62LCCR7IL-7Rαhigh TEM cells (Fig. 1). CD69 was also expressed by a high proportion of the CD4+ T cells in various site of colitic mice (Fig. 1). Consistent with this, we have previously shown that in sharp contrast to IL-7+/+ × RAG-1−/− recipients, IL-7−/− × RAG-1−/− recipients given these colitic CD4+ T cells do not develop colitis (13), indicating that these CD4+ T cells could be categorized as TEM cells.

FIGURE 1.

Colitic SP CD4+ donor T cells are CD44highCD62LCCR7IL-7Rαhigh TEM cells. Expression of CD44, CD62L, IL-7Rα (CD127), and CD69 on CD4+ T cells obtained from SP, MLN, LP, and BM in colitic C3H-SCID mice induced by adoptive transfer of CD4+CD45RBhigh T cells (6 wk after transfer). Freshly isolated cells from colitic mice were stained with FITC-labeled anti-CD4 and PE-labeled anti-CD44, anti-CD62L, anti-IL-7Rα, or anti-CD69. Samples were analyzed by flow cytometry. Lymphocytes were identified by characteristic forward angle and side scatter profiles. Data are displayed as dotted plot (four-decade log scale), and quadrant markers were positioned to include >98% of control Ig-stained cells in the bottom left. Percentages in each quadrant are indicated. Representatives of three mice in each group.

FIGURE 1.

Colitic SP CD4+ donor T cells are CD44highCD62LCCR7IL-7Rαhigh TEM cells. Expression of CD44, CD62L, IL-7Rα (CD127), and CD69 on CD4+ T cells obtained from SP, MLN, LP, and BM in colitic C3H-SCID mice induced by adoptive transfer of CD4+CD45RBhigh T cells (6 wk after transfer). Freshly isolated cells from colitic mice were stained with FITC-labeled anti-CD4 and PE-labeled anti-CD44, anti-CD62L, anti-IL-7Rα, or anti-CD69. Samples were analyzed by flow cytometry. Lymphocytes were identified by characteristic forward angle and side scatter profiles. Data are displayed as dotted plot (four-decade log scale), and quadrant markers were positioned to include >98% of control Ig-stained cells in the bottom left. Percentages in each quadrant are indicated. Representatives of three mice in each group.

Close modal

Although it is well established that the existence of commensal bacteria is required for the initial development of most animal models of chronic colitis (10, 11, 17), it is still unclear whether commensal bacteria are also needed for the persistence of colitis and/or the maintenance of colitogenic CD4+ T cells after the T cell-priming process. To this end, we conducted an adoptive retransfer experiment under SPF or GF conditions (Fig. 2,A). We used SP not LP CD4+ T cells isolated from colitic SCID mice as donor cells for two reasons: 1) we wanted to exclude possible contamination by commensal bacteria from colitic LP or MLN samples, and 2) we have previously shown that most of the SP CD4+ T cells in this mouse colitis model express the TEM cell phenotype with a capacity to induce colitis similar to that of LP CD4+ T cells (12). Thus, we injected these SP cells into new SPF or GF C3H-SCID mice (hereafter called SPF or GF SCID mice, respectively) in a retransfer. Furthermore, to assess whether GF SCID recipients develop colitis when exposed to resident commensal bacteria, one group of the GF SCID recipients was moved into the SPF environment 6 wk after the retransfer and kept for an additional 4 wk (GF→SPF SCID mice), and the other group was kept in GF conditions for 10 wk (GF→GF SCID mice) (Fig. 2,A). Consistent with previous reports (12, 13) and in sharp contrast to GF SCID mice, SPF SCID mice showed an enlarged colon with a greatly thickened wall (Fig. 2,B) and manifested progressive weight loss from 3 wk after retransfer (data not shown). Consistent with this, the clinical score of SPF SCID mice at 10 wk after retransfer was significantly increased compared with that of GF SCID mice, which showed no clinical signs of colitis or weight loss throughout the observation period (Fig. 2,C). It was notable that GF→SPF SCID recipients also showed an enlarged and thickened colon, and their clinical scores were significantly increased compared with those of GF→GF SCID recipients, being comparable to those of SPF SCID recipients. As previously reported (18), a markedly enlarged cecum was also reproducibly observed in the GF→GF SCID-recipient mice (Fig. 2 B). The integrity of the GF conditions was confirmed by repeated stool culture tests in the GF groups (data not shown). SP and colon weights of SPF SCID mice and GF→SPF SCID mice were significantly greater than those of GF→GF SCID mice (data not shown).

FIGURE 2.

Commensal bacteria are not essential for survival of colitogenic memory CD4+ T cells. A, C3H-SCID mice were injected i.p. with normal splenic CD4+CD45RBhigh T cells. Six weeks after transfer, mice developed chronic colitis, and then colitic CD4+ T cells were isolated from SP. Doses of 3 × 105 SP CD4+ T cells were injected into new SPF C3H-SCID mice (called SPF SCID mice) or GF C3H-SCID mice (called GF SCID mice). Six weeks after transfer, GF SCID mice were divided two groups, one group was left in GF condition (called GF→GF SCID mice), and the other group was moved to SPF condition (GF→SPF SCID mice). Each group contained eight mice. All mice were sacrificed 10 wk after the retransfer. B, Gross appearance of the colon from SPF (top), GF→GF (middle), and GF→SPF (bottom) SCID mice. C, Clinical scores were determined at 10 wk after the retransfer as described in Materials and Methods. SPF and GF→SPF SCID mice showed the severe clinical signs of colitis, whereas GF→GF SCID mice showed no signs of colitis. Data are indicated as the mean ± SEM of seven mice in each group. *, p < 0.05. D, Colon and SP weight of SPF and GF→SPF SCID mice were significantly larger than those of GF→GF SCID mice.

FIGURE 2.

Commensal bacteria are not essential for survival of colitogenic memory CD4+ T cells. A, C3H-SCID mice were injected i.p. with normal splenic CD4+CD45RBhigh T cells. Six weeks after transfer, mice developed chronic colitis, and then colitic CD4+ T cells were isolated from SP. Doses of 3 × 105 SP CD4+ T cells were injected into new SPF C3H-SCID mice (called SPF SCID mice) or GF C3H-SCID mice (called GF SCID mice). Six weeks after transfer, GF SCID mice were divided two groups, one group was left in GF condition (called GF→GF SCID mice), and the other group was moved to SPF condition (GF→SPF SCID mice). Each group contained eight mice. All mice were sacrificed 10 wk after the retransfer. B, Gross appearance of the colon from SPF (top), GF→GF (middle), and GF→SPF (bottom) SCID mice. C, Clinical scores were determined at 10 wk after the retransfer as described in Materials and Methods. SPF and GF→SPF SCID mice showed the severe clinical signs of colitis, whereas GF→GF SCID mice showed no signs of colitis. Data are indicated as the mean ± SEM of seven mice in each group. *, p < 0.05. D, Colon and SP weight of SPF and GF→SPF SCID mice were significantly larger than those of GF→GF SCID mice.

Close modal

Histological examination showed a prominent epithelial hyperplasia with glandular elongation and massive infiltration of mononuclear cells in the LP of proximal and distal colon from SPF SCID and GF→SPF SCID mice (Fig. 3,A). In contrast, the glandular elongation was mostly abrogated, and fewer mononuclear cells were observed in the LP of the colon from the recipient GF→GF SCID mice (Fig. 3,A). This difference was also confirmed by histological scoring of multiple proximal and distal colon sections: 3.80 ± 0.37 and 4.40 ± 0.43, respectively, in SPF SCID mice; 4.00 ± 0.30 and 5.17 ± 0.056 in GF→SPF SCID mice; and 0.125 ± 0.001 and 0.125 ± 0.001 in GF→GF SCID mice (SPF SCID mice or GF→SPF SCID mice vs GF→GF SCID mice; p < 0.01) (Fig. 3 B).

FIGURE 3.

Commensal bacteria are needed to sustain the colitis. A, Proximal and distal colon from SPF, GF→GF, and GF→SPF SCID mice were stained with H&E. Original magnification: ×40 (top) and ×100 (bottom). B, Histological scores were determined at 10 wk after transfer as described in Materials and Methods. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

FIGURE 3.

Commensal bacteria are needed to sustain the colitis. A, Proximal and distal colon from SPF, GF→GF, and GF→SPF SCID mice were stained with H&E. Original magnification: ×40 (top) and ×100 (bottom). B, Histological scores were determined at 10 wk after transfer as described in Materials and Methods. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

Close modal

A further quantitative evaluation of CD4+ T cell infiltration was made by isolating LP, SP, and BM CD3+CD4+ T cells. As shown in Fig. 4, the number of CD4+ T cells recovered from LP and SP of SPF and GF→SPF SCID mice was significantly higher than that from GF→GF SCID mice. Surprisingly, however, a substantial number of CD4+ T cells remained resident in BM and SP in GF→GF SCID mice, and the number of CD4+ T cells recovered from BM of colitic SPF and GF→SPF SCID mice was not significantly higher that that recovered from noncolitic GF→GF SCID mice. This suggests that even in the absence of commensal bacteria, the colitogenic SP CD4+ T donor cells are not all short-lived effector cells but, in accordance with high expression of IL-7Rα, are long-lived TEM cells or a mixture of effector cells and TEM cells. Alternatively, it is possible that most persisting CD4+ T cells in GF SCID mice respond to environmental Ags derived from food and bedding.

FIGURE 4.

Commensal bacteria are needed for the expansion of CD4+ T cells. LP, SP, and BM CD4+ T cells were isolated from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer, and the number of CD3+CD4+ cells was determined by flow cytometry. Data are indicated as the mean ± SEM of seven mice in each group. *, p < 0.05.

FIGURE 4.

Commensal bacteria are needed for the expansion of CD4+ T cells. LP, SP, and BM CD4+ T cells were isolated from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer, and the number of CD3+CD4+ cells was determined by flow cytometry. Data are indicated as the mean ± SEM of seven mice in each group. *, p < 0.05.

Close modal

We next examined the cytokine production by isolated LP CD4+ T cells from each group. As shown in Fig. 5, LP CD4+ T cells from the GF SCID recipients produced significantly smaller amounts of IFN-γ, TNF-α, and IL-17 than those from the SPF SCID recipients after in vitro stimulation by anti-CD3/CD28 mAbs.

FIGURE 5.

Commensal bacteria are needed for the expansion of Th1 and Th17 CD4+ T cells. Th1 and Th17 cytokine production by anti-CD3/CD28 mAbs-stimulated LP CD4+ T cells isolated from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer was measured by ELISA. Data are indicated as the mean ± SD of six mice in each group. *, p < 0.05.

FIGURE 5.

Commensal bacteria are needed for the expansion of Th1 and Th17 CD4+ T cells. Th1 and Th17 cytokine production by anti-CD3/CD28 mAbs-stimulated LP CD4+ T cells isolated from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer was measured by ELISA. Data are indicated as the mean ± SD of six mice in each group. *, p < 0.05.

Close modal

Flow cytometry analysis revealed that the cell-surface phenotype of LP, SP, and BM CD4+ T cells isolated from SPF or GF→SPF SCID recipients was very similar to that from the primarily injected colitic mice, that is, TEM cell type with CD44highCD62LCD69+IL-7Rαhigh cells (Figs. 1 and 6). Again, the expression level of CD69 on SP CD4+ T cells was significantly lower than that on LP and BM cells in the SPF or GF→SPF SCID recipients (percentage of positive cells in SPF: SP 18.9 ± 3.40, LP 60.4 ± 1.79, and BM 61.8 ± 3.37; SP vs LP or BM, p < 0.05; in GF→SPF, SP 9.83 ± 0.95, LP 55.8 ± 4.21, and BM 53.8 ± 6.28; SP vs LP or BM p < 0.05), suggesting the presence of activation mechanisms in LP and BM. Importantly, the expression pattern of all examined markers in all sites was not significantly different between SPF and GF→SPF SCID recipients (Fig. 6, right panels).

FIGURE 6.

Colitogenic CD4+ TCM cells with the down-modulated IL-7Rα expression were significantly increased in GF recipients. Expression of CD44 and CD62L (C), IL-7Rα (CD127) (A), and CD69 (B) on CD3+CD4+ T cells obtained from LP, SP, and BM from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer. Freshly isolated cells were stained with allophycocyanin-labeled anti-CD4, PerCP-labeled anti-CD3, PE-labeled anti-CD44, FITC-labeled anti-CD62L, biotin-labeled anti-IL-7Rα, FITC-labeled anti-CD69 mAb, or streptavidin PE. Samples were analyzed by flow cytometry. Lymphocytes were identified by characteristic forward angle and side scatter profiles. Data are displayed as dotted plot (four-decade log scale), and quadrant markers were positioned to include >98% of control Ig-stained cells in the bottom left. Percentages in each quadrant are indicated. Representatives of five mice in each group.

FIGURE 6.

Colitogenic CD4+ TCM cells with the down-modulated IL-7Rα expression were significantly increased in GF recipients. Expression of CD44 and CD62L (C), IL-7Rα (CD127) (A), and CD69 (B) on CD3+CD4+ T cells obtained from LP, SP, and BM from SPF, GF→GF, and GF→SPF SCID mice at 10 wk after retransfer. Freshly isolated cells were stained with allophycocyanin-labeled anti-CD4, PerCP-labeled anti-CD3, PE-labeled anti-CD44, FITC-labeled anti-CD62L, biotin-labeled anti-IL-7Rα, FITC-labeled anti-CD69 mAb, or streptavidin PE. Samples were analyzed by flow cytometry. Lymphocytes were identified by characteristic forward angle and side scatter profiles. Data are displayed as dotted plot (four-decade log scale), and quadrant markers were positioned to include >98% of control Ig-stained cells in the bottom left. Percentages in each quadrant are indicated. Representatives of five mice in each group.

Close modal

In contrast, the expression pattern on cells from GF→GF SCID recipients was quite different. First, surprisingly, IL-7Rα expression on LP but not SP and BM CD4+ T cells was significantly down-modulated compared with the paired LP cells of SPF or GF→SPF SCID recipients (Fig. 6,A). Second, and as expected, CD69 expression on cells from all sites was also reduced compared with the matching cell source from SPF or GF→SPF SCID recipients (Fig. 6,B). Third, and quite interestingly, the proportion of CD3+CD4+CD44highCD62L+ TCM cells was significantly increased in SP and BM but not in LP of GF→GF SCID recipients (SP 2.41 ± 0.85%; BM 5.21 ± 0.95%; LP 3.09 ± 0.95%) compared with SPF SCID recipients (SP 1.14 ± 0.1%, p < 0.05; BM 2.80 ± 0.48%, p < 0.05; LP 0.35 ± 0.059%, p = 0.10) and GF→SPF SCID recipients (SP 1.31 ± 0.17%, p < 0.05; BM 2.23 ± 0.33%, p < 0.05; LP 0.18 ± 0.040%, p = 0.28), suggesting the conversion of cells from TCM to TEM after movement of mice from GF→SPF (Fig. 6 C).

The TCR Vβ repertoires of SP CD4+ T cells from SPF, GF→GF, and GF→SPF SCID mice differed only slightly and only in such Vβ families as Vβ7, Vβ9, and Vβ11, which are not major groups in this model (Fig. 7). This suggested that long-lived colitogenic memory CD4+ T cells are broadly polyclonal rather than specifically oligoclonal. Furthermore, this finding suggests that in GF conditions, colitogenic memory CD4+ T cells may be maintained by homeostatic cytokines that would lead to polyclonal proliferation, rather than by response to a specific Ag such as food or bedding that may lead to a specific oligoclonal expansion.

FIGURE 7.

TCR Vβ repertoires show little difference in the presence or absence of commensal bacteria. Flow cytometric analysis of Vβ families on the surface of the splenic CD4+ T cells in SPF, GF→GF, and GF→SPF mice as described in Fig. 2. To analyze the TCR Vβ family repertoire, splenic cells were four-color-stained with PerCP-conjugated anti-CD3mAb, allophycocyanin-conjugated anti-CD4 mAb, PE-conjugated anti-Ly5.1 or Ly5.2 mAb, and a panel of 15 FITC-conjugated Vβ mAbs. Each percentage value indicates the frequency of each Vβ (n = 6). Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

FIGURE 7.

TCR Vβ repertoires show little difference in the presence or absence of commensal bacteria. Flow cytometric analysis of Vβ families on the surface of the splenic CD4+ T cells in SPF, GF→GF, and GF→SPF mice as described in Fig. 2. To analyze the TCR Vβ family repertoire, splenic cells were four-color-stained with PerCP-conjugated anti-CD3mAb, allophycocyanin-conjugated anti-CD4 mAb, PE-conjugated anti-Ly5.1 or Ly5.2 mAb, and a panel of 15 FITC-conjugated Vβ mAbs. Each percentage value indicates the frequency of each Vβ (n = 6). Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

Close modal

We have previously reported that colitogenic memory CD4+ T cells are retained in an IL-7-dependent manner in BM of SPF-conditioned CD4+CD45RBhigh T cell-injected colitic mice (19). Thus, we hypothesized that BM IL-7 may maintain colitogenic memory CD4+ T cells in noncolitic GF→GF SCID mice even in the absence of commensal bacteria. To test this hypothesis, frozen sections of colon and BM from each group were stained with polyclonal anti-IL-7 Ab (green) and anti-CD4 mAb (red). First, Fig. 8 clearly demonstrates marked infiltration of CD4+ T cells in the colon of colitic SPF and GF→SPF SCID recipients, although the expression of IL-7 in epithelial cells was markedly decreased in these two groups (Fig. 8, left top and bottom). In sharp contrast, only a scattering of CD4+ T cells were found in the LP of noncolitic GF→GF SCID recipients despite the presence of epithelial IL-7 (Fig. 8, left middle). However, consistent with the constant expression of IL-7 in BM, substantial numbers of CD4+ T cells were resident in BM of all three groups, regardless of the presence or absence of commensal bacteria (noncolitic or colitic) (Fig. 8, right panel).

FIGURE 8.

Substantial number of CD4+ T cells was resident in BM of noncolitic SCID recipients in the absence of commensal bacteria. Frozen sections of colon from each group (SPF, GF→GF, and GF→SPF) were stained with polyclonal anti-IL-7 Abs (green) and anti-CD4 Abs (red). Representative of five separate samples in each group. Original magnification: ×100.

FIGURE 8.

Substantial number of CD4+ T cells was resident in BM of noncolitic SCID recipients in the absence of commensal bacteria. Frozen sections of colon from each group (SPF, GF→GF, and GF→SPF) were stained with polyclonal anti-IL-7 Abs (green) and anti-CD4 Abs (red). Representative of five separate samples in each group. Original magnification: ×100.

Close modal

To confirm the importance of IL-7 for the maintenance of colitogenic memory CD4+ T cells even in GF conditions, we next blockaded IL-7. GF SCID mice injected with colitogenic SP CD4+ T cells were separated in two groups: one group was treated with anti-IL-7Rα mAb, and the other group was treated with control rat IgG (Fig. 9,A). Two weeks after transfer, all mice were sacrificed, and LP, MLN, SP, and BM cells were analyzed by flow cytometry to determine the number of CD3+CD4+ T cells recovered (Fig. 9,B). As expected, the numbers of CD4+ T cells recovered from sites other than LP of GF SCID mice treated with anti-IL-7Rα mAb were significantly reduced compared with the control IgG-treated group (Fig. 9 B). However, although the number of CD4+ T cells recovered from LP of mice treated with anti-IL-7Rα mAb tended to be lower than that from LP of mice treated with control IgG, the difference was not significant, suggesting the possibility of additional stimuli, such as food and/or bedding Ags and pathogen-associated molecular patterns included in sterile foods, maintaining CD4+ T cells in LP. Collectively, this result supports our hypothesis that IL-7 may maintain colitogenic memory CD4+ T cells outside the intestine of the injected GF SCID mice even in the absence of commensal bacteria.

FIGURE 9.

Neutralization of IL-7 reduced the recovered cell number of CD4+ T cells in various site of GF SCID mice transferred with colitic CD4+ T cells. A, GF SCID mice transferred with colitogenic SP CD4+ T cells previously transferred with CD4+CD45RBhigh T cells were treated with anti-IL-7Rα mAb or control rat IgG at 0, 1, and 2 wk after transfer. B, Recovered CD3+CD4+ T cell number from LP, SP, MLN, and BM were analyzed by a flow cytometry. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

FIGURE 9.

Neutralization of IL-7 reduced the recovered cell number of CD4+ T cells in various site of GF SCID mice transferred with colitic CD4+ T cells. A, GF SCID mice transferred with colitogenic SP CD4+ T cells previously transferred with CD4+CD45RBhigh T cells were treated with anti-IL-7Rα mAb or control rat IgG at 0, 1, and 2 wk after transfer. B, Recovered CD3+CD4+ T cell number from LP, SP, MLN, and BM were analyzed by a flow cytometry. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05.

Close modal

Finally, we asked whether colitogenic memory CD4+ T cells can be retained in lymphocyte-sufficient normal mice, because it is very important to know whether our findings are applicable to the pathogenesis of human IBD where the patients are always immunosufficient. To this end, we injected CFSE-labeled colitogenic LP Ly5.1+CD4+ T cells into Ly5.2+ WT mice treated with either a mixture of antibiotics in distilled water or distilled water alone (Fig. 10,A). One week and 4 wk after transfer, cell numbers were recovered from LP, SP, and BM from injected WT mice, and their numbers of cell divisions were assessed by a flow cytometry (Fig. 10, B and C). As shown in Fig. 10 B, a substantial number of Ly5.1+CD4+ T cells were recovered from LP, SP, and BM of lymphocyte-replete mice at 1 and 4 wk after cell transfer, regardless of antibiotic treatment. Notably, there is almost no difference between antibiotic-treated and control mice in the number of CD4+ T cells in various sites. Consistent with this, a CFSE dilution assay revealed that Ly5.1+CD4+ T cells divided well in LP, SP, and BM at the indicated time points regardless of antibiotic treatment. Collectively, these results suggest that in the noncolitic immunosufficient condition, the maintenance of the colitogenic memory CD4+ T cells is entirely independent of the presence of commensal bacterial Ag but is presumably dependent on homeostatic cytokines such as IL-7.

FIGURE 10.

Colitogenic memory CD4+ T cells were retained outside the intestine even in the normal immunosufficient condition. A, CFSE-labeled LP CD4+ T cells obtained from colitic RAG2−/− mice previously transferred with Ly5.1+CD4+CD45RBhigh T cells were transferred into Ly5.2+ WT mice treated with antibiotics or DW. B, One week and 4 wk after transfer, LP, SP, and BM cells from transferred WT mice were analyzed by a flow cytometry, and the recovered cell number of CD3+CD4+ T cells in LP, SP and BM was calculated. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05. C, Cell division assay. At the indicated time points after transfer (1 and 4 wk after transfer), CFSE incorporation was determined by flow cytometry. Histograms are gated on CD4+ T cells. These data were representative from six experiments.

FIGURE 10.

Colitogenic memory CD4+ T cells were retained outside the intestine even in the normal immunosufficient condition. A, CFSE-labeled LP CD4+ T cells obtained from colitic RAG2−/− mice previously transferred with Ly5.1+CD4+CD45RBhigh T cells were transferred into Ly5.2+ WT mice treated with antibiotics or DW. B, One week and 4 wk after transfer, LP, SP, and BM cells from transferred WT mice were analyzed by a flow cytometry, and the recovered cell number of CD3+CD4+ T cells in LP, SP and BM was calculated. Data are indicated as the mean ± SEM of five mice in each group. *, p < 0.05. C, Cell division assay. At the indicated time points after transfer (1 and 4 wk after transfer), CFSE incorporation was determined by flow cytometry. Histograms are gated on CD4+ T cells. These data were representative from six experiments.

Close modal

The present study has demonstrated that latent long-lived colitogenic memory CD4+ T cells not only reside outside the intestine (for example in the IL-7-sufficient BM environment of noncolitic mice that had been previously injected with colitogenic CD4+ T cells under GF conditions) but also participate in the recurrence of colitis in the presence of commensal bacteria. In other words, these data clearly showed that colitogenic CD4+ memory T cells are established during the process of development of colitis even in the presence of commensal bacteria and can be maintained outside the intestine, possibly in an IL-7-dependent manner, in the remission stage of chronic colitis. Thus, our present results may explain why IBD is an intractable lifelong disease if it depends on the persistence of latent colitogenic CD4+ memory T cells.

Immunologically, “memory” is now generally believed to be a phenomenon that occurs after Ags have been eliminated (1, 2, 3). More specifically, Ags must be eliminated from the body in order for CD8+ memory T cells to form. For example, in the lymphocytic choriomeningitis virus model of acute viral infection, lymphocytes differentiate into CD8+ memory T cells for the first time after the virus has been eliminated (20, 21). In contrast, in a chronic viral infection model using a mutant lymphocytic choriomeningitis virus, it is thought that no CD8+ memory T cells are produced and that the CD8+ effector T cells are ultimately destroyed as a result of “exhaustion” (22, 23). The fact that the Ags responsible for IBD are derived from commensal bacteria that can never be eliminated suggests the possibility that effector T cells continue to emerge and become exhausted without memory T cells ever being produced. However, a requirement for constant production of colitogenic CD4+ effector cells from naive CD4+ T cells that are continuously supplied by the thymus may be difficult to explain in view of the involution of the thymus in both mice and humans. We therefore hypothesized that even though commensal bacteria may permanently reside in the intestine, colitogenic CD4+ T cells could be retained as memory cells. Consistent with this hypothesis, we previously demonstrated that IL-7−/− × RAG-1−/− mice injected with colitogenic CD4+ T cells never develop colitis, whereas evidence of disease was seen in IL-7+/+ × RAG-1−/− recipients (13). These findings demonstrated that IL-7 is essential for the development and maintenance of chronic colitis. Accordingly, it is now assumed that IL-7-dependent memory T cells make up at least part of the population of colitogenic CD4+ T cells and contribute to the overall maintenance of colitis even in the presence of enteric bacteria.

In contrast, our group has previously reported that IL-7 is produced by epithelial cells in the intestine, especially by intestinal goblet cells (24), and it is known that the pathology of IBD is characterized by a decrease in goblet cells when it becomes chronic (25). There was also a marked decrease in strongly staining Alcian blue-positive goblet cells at the sites of the chronic colitis lesions in the CD4+CD45RBhigh T cell transfer model that we used, and we discovered that there was a concomitant decrease in IL-7 production by the epithelial cells (Ref. 26 ; Fig. 8). Why then does IL-7 derived from inflamed intestine decrease when IL-7 is essential to the development of chronic colitis? We hypothesized that intestinal IL-7 is not required to maintain chronic colitis and that colitogenic CD4+ memory T cells are maintained by IL-7 outside the intestine. We previously performed parabiosis surgery that connected the flanks of colitic RAG-2−/− mice into which CD4+CD45RBhigh T cells had been injected with the flanks of untreated IL-7+/+ × RAG-1−/− mice or IL-7−/− × RAG-1−/− mice. We allowed the hemodynamics of the two mice to be shared for several days after the parabiosis, and even though the intestinal epithelial cells of the IL-7−/− × RAG-1−/− host mice do not produce IL-7, they can be viewed as mice that produce IL-7 outside the intestine because of the shared hemodynamics. As expected, ∼4 wk after the parabiosis, the transfer of colitogenic CD4+ T cells from the colitic RAG-2−/− donor mice was associated with the development of chronic colitis in the IL-7+/+ × RAG-1−/− host mice, which was accompanied by marked CD4+ T cell infiltration of the large intestine. Surprisingly, despite the deficiency of intestinal IL-7, frank colitis also developed in the IL-7−/− × RAG-1−/− host mice in the same way as in the control IL-7+/+ × RAG-1−/− host mice (26). These previous findings demonstrated that intestinal IL-7 is not essential for the maintenance of colitogenic CD4 memory T cells in mice with chronic colitis.

In this study, we further attempted to resolve the following two issues. 1) How are colitogenic CD4+ T cells maintained in inflamed mucosa despite the lack of epithelium-derived IL-7? 2) Can colitogenic CD4+ T cells be maintained for a long period in the absence of commensal bacteria in IL-7-sufficient conditions, as is the case for memory CD8+ T cells? If so, are latent colitogenic CD4+ memory T cells involved in the recurrence of colitis after moving from a GF to an SPF environment?

First, we showed that almost all CD4+ T cells in LP of noncolitic GF→GF SCID mice disappeared despite the presence of epithelium-derived IL-7 (Fig. 8). In contrast, a substantial number of CD4+ T cells still resided outside the intestine of GF→GF SCID mice, showing that the number of CD4+ T cells recovered from these noncolitic mice was comparable with that from colitic SPF or GF→SPF SCID mice, especially in BM (Fig. 4). This result suggests that colitogenic CD4+ T cells may be reciprocally regulated in intestine and in the BM so that colitogenic LP and BM CD4+ T cells are stimulated via commensal bacterial Ags and via IL-7 signaling, respectively. Consistent with this idea, we previously showed that: 1) synchronous stimulation of human LP CD4+ T cells by anti-CD3 mAb and IL-7 induces apoptosis of those cells (16), and 2) colitogenic CD4+ T cells actively circulate in colitic mice and the blockade of the circulation by FYT720 treatment ameliorates the colitis in this model (27). This suggests that epithelial IL-7 plays a physiologically important role in intestinal immune homeostasis that prevents the triggering of IBD, whereas the absence of epithelium-derived IL-7 itself may be needed for the persistence of chronic colitis. Further study using IL-7-deficient recipient mice under GF conditions will be needed to test this hypothesis.

Second, and surprisingly, we showed that colitogenic SP CD4+ T cells survived for 6 wk in GF conditions and are involved in the recurrence of colitis after their movement to SPF conditions. Because effector T cells are believed to be short lived in contrast to long-lived memory T cells, this result indicates that the colitogenic SP CD4+ T cells contained at least some fraction of colitogenic memory CD4+ T cells. As an additional explanation for the perpetuation of IBD, the ratio of TCM/TEM cells in GF→GF SCID mice may support the idea that colitogenic “memory stem”-like TCM cells are generated in the process of development and/or persistence of chronic colitis.

The results of our current project show some differences from the findings published by Veltkamp et al. (17). Using their original model of chronic colitis induced by transplantation of BM from normal mice into CD3ε transgenic (BM→Tgε26) mice, they showed that BM→Tgε26 mice do not develop colitis in GF conditions whereas control BM→Tgε26 in SPF conditions do. However, they also showed that viable intestinal bacteria are not necessary for the survival of CD4+ T cells that retain their functional integrity in noncolitic BM→Tgε26 mice in GF conditions and induce colitis when mice are moved to SPF conditions and that Tgε26 mice develop colitis in SPF conditions when MLN cells from BM→Tgε26 mice maintained in GF conditions are injected. Thus, they concluded that continuous stimulation by commensal bacteria is essential for the development of colitis. Although the results of their study seem to be similar to ours, the interpretation of each study is distinct. First, it is very likely that peripheral MLN cells isolated from noncolitic BM→Tgε26 mice in GF conditions contain a substantial number of naive CD4+ T cells, and thus it is very possible that lymphopenic Tgε26 mice injected with these cells develop colitis when transferred to SPF conditions. In contrast, we first isolated colitogenic SP CD4+ memory T cells from colitic mice, and as shown in Fig. 1, these cells all have the phenotype of memory CD4+CD44high T cells. Also, it is very important that naive CD4+ T cells are continuously generated in their BM transplantation system but not in our adoptive transfer system using T cell-deficient SCID mice that lack the ability to generate naive CD4+ T cells continuously from the thymus.

Finally, it should be noted that the expression of IL-7Rα was significantly reduced on CD4+ T cells from all sites of noncolitic GF→GF SCID mice compared with the paired sites in colitic SPF or GF→SPF SCID mice. This was very surprising, being inconsistent with the recent immunological dogma that memory CD4+ T cells express high levels of IL-7Rα. Although it makes sense that the high expression of IL-7Rα on CD4+ T cells of noncolitic GF→GF SCID mice would be maintained because the significant decrease in expression of the activation marker CD69 on those memory T cells suggests that they are in the resting state, we also obtained the same result using antibiotic-treated mice that had previously been injected with CD4+CD45RBhigh T cells. Although further study will be needed to address this issue, colitogenic memory CD4+ T cells may be quite different from conventional memory CD4+ T cells.

Taken together, these findings suggest that long-lived colitogenic memory CD4+ cells can be established as CD4+ TEM or TCM cells during the process of development of colitis, even in the presence of commensal Ags, and that these cells participate in the perpetuation of colitis. Thus, blocking a stimulus of colitogenic memory CD4+ cells such as IL-7, or alternatively an immunological reset by BM transplantation to remove these colitogenic memory CD4+ T cells (28), may have therapeutic benefit for treatment of IBD.

The authors have no financial conflict of interest.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

This study was supported in part by grants-in-aid for Scientific Research, Scientific Research on Priority Areas, Exploratory Research and Creative Scientific Research from the Japanese Ministry of Education, Culture, Sports, Science, and Technology; the Japanese Ministry of Health, Labor, and Welfare; the Japan Medical Association; the Foundation for Advancement of International Science; the Terumo Life Science Foundation; the Ohyama Health Foundation; the Yakult Bio-Science Foundation; and the Research Fund of Mitsukoshi Health and Welfare Foundation.

3

Abbreviations used in this paper: IBD, inflammatory bowel disease; BM, bone marrow; CD62L, L-selectin; LP, lamina propria; MLN, mesenteric lymph node; SP, spleen or splenic; TEM, effector-memory T; TSLP, thymic stromal lymphopoietin; WT, wild type.

1
Sprent, J., C. D. Surh.
2002
. T cell memory.
Annu. Rev. Immunol.
20
:
551
-579.
2
Sallusto, F., J. Geginat, A. Lanzavecchia.
2004
. Central memory and effector memory T cell subsets: function, generation and maintenance.
Annu. Rev. Immunol.
22
:
745
-763.
3
Seder, R. A., R. Ahmed.
2003
. Similarities and differences in CD4+ and CD8+ effector and memory T cell generation.
Nat. Immunol.
9
:
835
-842.
4
Robertson, J. M., M. MacLeod, V. S. Marsden, J. W. Kappler, P. Marrack.
2006
. Not all CD4+ memory T cells are long lived.
Immunol. Rev.
211
:
49
-57.
5
Scheller, L. F., A. F. Azad.
1995
. Maintenance of protective immunity against malaria by persistent hepatic parasites derived from irradiated sporozoites.
Proc. Natl. Acad. Sci. USA
92
:
4066
-4068.
6
Belkaid, Y., C. A. Piccirillo, S. Mendez, E. M. Shevach, D. L. Sacks.
2002
. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity.
Nature
420
:
502
-507.
7
Kassiotis, G., S. Garcia, E. Simpson, B. Stockinger.
2002
. Impairment of immunological memory in the absence of MHC despite survival of memory T cells.
Nat. Immunol.
3
:
244
-250.
8
Hooper, L. V., J. I. Gordon.
2001
. Commensal host-bacterial relationships in the gut.
Science
292
:
1115
-1118.
9
Berg, R. D..
1996
. The indigenous gastrointestinal microflora.
Trends Microbiol.
4
:
430
-435.
10
Podolsky, D. K..
2002
. Inflammatory bowel disease.
N. Engl. J. Med.
347
:
417
-429.
11
Sartor, R. B..
2006
. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis.
Nat. Clin. Pract. Gastroenterol. Hepatol.
3
:
390
-407.
12
Totsuka, T., T. Kanai, R. Iiyama, K. Uraushihara, M. Yamazaki, R. Okamoto, T. Hibi, K. Tezuka, M. Azuma, H. Akiba, et al
2003
. Ameliorating effect of anti-ICOS monoclonal antibody in a murine model of chronic colitis.
Gastroenterology
124
:
410
-421.
13
Totsuka, T., T. Kanai, Y. Nemoto, S. Makita, M. Watanabe.
2007
. IL-7 is essential for the development and the persistence of chronic colitis.
J. Immunol.
178
:
4737
-4748.
14
Yamazaki, M., T. Yajima, M. Tanabe, K. Fukui, E. Okada, R. Okamoto, S. Oshima, T. Nakamura, T. Kanai, M. Uehira, et al
2003
. Mucosal T cells expressing high levels of IL-7 receptor are potential targets for treatment of chronic colitis.
J. Immunol.
171
:
1556
-1563.
15
Powrie, F., M. W. Leach, S. Mauze, L. B. Caddle, R. L. Coffman.
1993
. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C.B-17 scid mice.
Int. Immunol.
5
:
1461
-1471.
16
Yen, D., J. Cheun, H. Scheerens, F. Poulet, T. McClanahan, B. McKenzie, M. A. Kleinschek, A. Owyang, J. Mattson, W. Blumenschein, et al
2006
. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6.
J. Clin. Invest.
116
:
1310
-1316.
17
Veltkamp, C., S. L. Tonkonogy, Y. P. De Jong, C. Albright, W. B. Grenther, E. Balish, C. Terhorst, R. B. Sartor.
2001
. Continuous stimulation by normal luminal bacteria is essential for the development and perpetuation of colitis in Tg (ε26) mice.
Gastroenterology
120
:
900
-913.
18
Thompson, G. R., P. C. Trexler.
1971
. Gastrointestinal structure and function in germ-free or gnotobiotic animals.
Gut
12
:
230
-235.
19
Nemoto, Y., T. Kanai, S. Makita, R. Okamoto, T. Totsuka, K. Takeda, M. Watanabe.
2007
. Bone marrow retaining colitogenic CD4+ T cells may be a pathogenic reservoir for chronic colitis.
Gastroenterology
132
:
176
-189.
20
Wherry, E. J., V. Teichgräber, T. C. Becker, D. Masopust, S. M. Kaech, R. Antia, U. H. von Andrian, R. Ahmed.
2003
. Lineage relationship and protective immunity of memory CD8 T cell subsets.
Nat. Immunol.
4
:
225
-234.
21
Kaech, S. M., E. J. Wherry, R. Ahmed.
2002
. Effector and memory T cell differentiation: implications for vaccine development.
Nat. Rev. Immunol.
2
:
251
-262.
22
Barber, D. L., E. J. Wherry, D. Masopust, B. Zhu, J. P. Allison, A. H. Sharpe, G. J. Freeman, R. Ahmed.
2006
. Restoring function in exhausted CD8 T cells during chronic viral infection.
Nature
439
:
682
-687.
23
Blackburn, S. D., E. J. Wherry.
2007
. IL-10, T cell exhaustion and viral persistence.
Trends Microbiol.
15
:
143
-146.
24
Watanabe, M., Y. Ueno, T. Yajima, Y. Iwao, M. Tsuchiya, H. Ishikawa, S. Aiso, T. Hibi, H. Ishii.
1995
. Interleukin 7 is produced by human intestinal epithelial cells and regulates the proliferation of intestinal mucosal lymphocytes.
J. Clin. Invest.
95
:
2945
-2953.
25
Riddell, R. H..
2004
. Pathology of idiopathic inflammatory bowel diseases. S. B. Sartor, and W. J. Sandborn, eds.
Kirsner’s Inflammatory Bowel Diseases
6th Ed.
399
-424. Saunders, London.
26
Tomita, T., T. Kanai, Y. Nemoto, T. Totsuka, R. Okamoto, K. Tsuchiya, N. Sakamoto, M. Watanabe.
2008
. Systemic, but not intestinal, IL-7 is essential for the persistence of chronic colitis.
J. Immunol.
180
:
383
-390.
27
Fujii, R., T. Kanai, Y. Nemoto, S. Makita, S. Oshima, R. Okamoto, K. Tsuchiya, T. Totsuka, M. Watanabe.
2006
. FTY720 suppresses CD4+CD44highCD62L effector memory T cell-mediated colitis.
Am. J. Physiol.
291
:
G267
-G274. 2006.
28
Oyama, Y., R. M. Craig, A. E. Traynor, K. Quigley, L. Statkute, A. Halverson, M. Brush, L. Verda, B. Kowalska, N. Krosnjar, et al
2005
. Autologous hematopoietic stem cell transplantation in patients with refractory Crohn’s disease.
Gastroenterology
128
:
552
-563.