Skip main navigation

Oral Anti-CD3 Antibody Treatment Induces Regulatory T Cells and Inhibits the Development of Atherosclerosis in Mice

Originally publishedhttps://doi.org/10.1161/CIRCULATIONAHA.109.863431Circulation. 2009;120:1996–2005

Abstract

Background— Accumulating evidence suggests that several subsets of regulatory T cells that actively mediate immunologic tolerance play crucial roles in atherogenesis. Recently, orally administered anti-CD3 monoclonal antibody has been shown as an inducer of novel regulatory T cells expressing latency-associated peptide (LAP) on their surface, which potently prevents systemic autoimmunity. In the present study, we hypothesized that oral anti-CD3 antibody treatment may inhibit atherosclerosis in mice.

Methods and Results— Six-week-old apolipoprotein E–deficient mice on a standard diet were orally given anti-CD3 antibody or control immunoglobulin G on 5 consecutive days, and atherosclerosis was assessed at age 16 weeks. Oral administration of anti-CD3 antibody significantly reduced atherosclerotic lesion formation and accumulations of macrophages and CD4+ T cells in the plaques compared with controls. We observed a significant increase in LAP+ cells and CD25+Foxp3+ cells in the CD4+ T-cell population in anti-CD3–treated mice, in association with increased production of the antiinflammatory cytokine transforming growth factor-β and suppressed T-helper type 1 and type 2 immune responses. Neutralization of transforming growth factor-β in vivo abrogated the preventive effect of oral anti-CD3 antibody.

Conclusions— Our findings indicate the atheroprotective role of oral anti-CD3 antibody treatment in mice via induction of a regulatory T-cell response. These findings suggest that oral immune modulation may represent an attractive therapeutic approach to atherosclerosis.

Atherosclerosis is an inflammatory condition of the arterial wall involving cells of innate and adaptive immunity.1,2 It leads to severe clinical events caused by the rupture of plaques and thrombotic occlusion of the artery and is the main cause of acute coronary syndrome and stroke, which account for approximately half of the deaths in Western countries. It is now widely recognized that chronic inflammation via T-cell–mediated pathogenic immune responses plays an important role in atherogenesis.2 Accumulating evidence suggests that several subsets of regulatory T cells (Tregs), which have been shown to maintain immunologic unresponsiveness to self-antigens,3 inhibit atherosclerosis development through the downregulation of activated T-cell responses.4–6 These studies imply that promotion of an endogenous regulatory immune response has a therapeutic potential to suppress atherosclerotic diseases.

Clinical Perspective on p 2005

Anti-CD3–specific antibodies strongly suppress immune responses by antigenic modulation of the T-cell receptor/CD3 complex.7 In 1985, parenteral anti-CD3 antibody was available for use in acute transplant rejection in humans. However, its long-term use is limited because of severe side effects such as mitogenicity and antiglobulin responses.7 The mitogenicity of anti-CD3 antibody with Fc portion is caused by interacting with Fc receptors (FcRs) on monocytes or macrophages. Therefore, humanized Fc-mutated antibodies were designed and demonstrated to be effective in human autoimmune diabetes for a long period, although they still induce low levels of cytokine release because of some degree of T-cell activation.8 This long-term beneficial effect of FcR-nonbinding anti-CD3 antibody is partly explained by the recent study showing that FcR-nonbinding anti-CD3 antibody induces transforming growth factor (TGF)-β–producing CD4+CD25+ Tregs.9 Notably, it has been shown that intravenously administered FcR-nonbinding CD3-specific antibody induces a regulatory immune response and inhibits atherosclerosis development and progression.10 Recent studies have revealed that oral or nasal anti-CD3 antibody with or without Fc portion is biologically active and induces CD4+LAP+ (latency-associated peptide) Tregs that suppress experimental autoimmune encephalitis, autoimmune diabetes mellitus, and lupus in a TGF-β–dependent fashion and that this therapy would not be expected to have side effects even though FcR-binding CD3-specific antibody is used.11–13

Mucosal tolerance induction has been shown to inhibit various autoimmune diseases in mice14 and, importantly, atherosclerotic diseases in mice15,16 and therefore has been gaining much interest clinically. High doses of oral antigen lead to anergy or depletion of antigen-specific T cells. On the other hand, low doses of oral antigen induce many types of antigen-specific Tregs, including T-helper type 3 (Th3) cells, regulatory T-cell type 1 (Tr1) cells, naturally occurring CD4+CD25+Foxp3+ Tregs, and CD4+LAP+ Tregs,14 indicating that mucosal tolerance induction seems to be an effective way to treat atherosclerosis. As a possible explanation for the effects of oral anti-CD3 antibody treatment, a mechanism similar to that described in mucosal tolerance is supposed because autoimmune diseases are shown to be suppressed by low doses of oral anti-CD3 antibody but not by high doses.11 However, there is no evidence of antigen specificity with oral anti-CD3 antibody.

Given these backgrounds, we hypothesized that induction of CD4+LAP+ Tregs by oral anti-CD3 antibody treatment would inhibit atherosclerosis in apolipoprotein E–deficient (ApoE−/−) mice. Our findings provide for the first time evidence that oral anti-CD3 antibody treatment induces CD4+LAP+ Tregs and CD4+CD25+Foxp3+ Tregs, which suppress pathogenic immune processes pivotal for atherogenesis through a TGF-β–dependent mechanism and consequently inhibit atherosclerotic plaque formation.

Methods

For expanded Methods, please see the online-only Data Supplement.

Experimental Design

We fed 6-week-old or 16-week-old male ApoE−/− mice 5 μg hamster CD3-specific antibody (clone 145 to 2C11; R&D Systems, Minneapolis, Minn) or 5 μg hamster immunoglobulin G (IgG; Jackson Immuno Research Laboratories Inc, West Grove, Pa) dissolved in 0.2 mL phosphate-buffered saline by gastric intubation with a plastic tube once a day for 5 consecutive days. Mice were killed at 16 weeks of age, and atherosclerotic lesions were assessed. In some experiments, mice were fed control IgG or anti-CD3 antibody daily for 5 days and were injected with 100 μg of neutralizing anti-TGF-β antibody (clone 1D11; Bio Express Inc, West Lebanon, NH) or control rat IgG (Bio Express Inc). Injections were repeated once a week from age 7 weeks to age 16 weeks.

Atherosclerotic Lesion Assessment

For aortic root lesion analysis, 5 consecutive sections (10-μm thickness), spanning 550 μm of the aortic sinus, were collected from each mouse, stained with Oil Red O (Sigma, St Louis, Mo), and quantified with the use of the Image J (National Institutes of Health) as described previously.17 Immunohistochemistry was performed with the use of antibodies to identify macrophages (MOMA-2, 1:400; BMA Biomedicals, Augst, Switzerland), T cells (CD4, 1:100; BD Biosciences, San Jose, Calif), smooth muscle cells (α-smooth muscle actin, 1:400; Sigma), and natural Tregs (Foxp3, 1:100; eBioscience, San Diego, Calif). For en face lesion analysis, the thoracic aorta was opened longitudinally, stained with Oil Red O, and analyzed as described previously.17

Flow Cytometry Analysis

Mesenteric lymph node (MLN) cells and splenocytes were isolated 2 days or 10 weeks after the final oral anti-CD3 antibody administration. Fluorescent-activated cell sorter analysis was performed by FACSCalibur with the use of CellQuest Pro software (BD Biosciences).

Cell Proliferation, Purification, and Cytokine Assays

Lymphocytes from MLNs or spleen were prepared and stimulated with concanavalin A (Sigma) in vitro. Cytokine levels in supernatants were examined by ELISA or a Mouse Cytokine Array kit (R&D Systems). CD4+CD25LAP+ Tregs, CD4+CD25LAP cells, CD4+CD25+ Tregs, and CD4+CD25 cells were purified from spleens or MLNs with the use of a MoFlo cell sorter (DakoCytomation, Denmark). Cell proliferation assay was performed by assessing [3H]thymidine incorporation.

Real-Time Reverse Transcription Polymerase Chain Reaction Analysis

Total RNA was extracted from MLN cells harvested after 2 days of oral antibody treatment or from the aortas after perfusion with RNA later (Ambion, Austin, Tex) with the use of TRIzol reagent (Invitrogen, Carlsbad, Calif). Quantitative polymerase chain reaction (PCR) was performed as described previously.18

Results

Effects of Oral Anti-CD3 Antibody on CD4+LAP+ Tregs or CD4+CD25+Foxp3+ Tregs in MLNs and Spleens

We fed 6-week-old male ApoE−/− mice 5 μg hamster anti-CD3–specific antibody or 5 μg hamster IgG by gastric intubation once a day for 5 consecutive days. Previous studies suggested a fundamental immunologic difference between intravenous and oral anti-CD3 antibody administration.11,12 In fact, oral anti-CD3 antibody treatment did not cause T-cell receptor downmodulation or depletion of T cells (Figure I in the online-only Data Supplement). To investigate the effects of oral anti-CD3 antibody treatment on CD4+LAP+ Treg levels in ApoE−/− mice, we performed flow cytometry studies. Similar to the previous report,11 2 days after the last feeding of anti-CD3 antibody, we found a significant increase in LAP+ Tregs in the CD4+ T-cell population in MLNs and spleens of anti-CD3–treated mice (P<0.05; Figure 1A and 1B). Notably, we observed a significant increase in LAP+ Tregs in the CD4+CD25 T-cell population in spleens (P<0.05) but not in the CD4+CD25+ T-cell population (Figure 1A and 1C), suggesting that oral anti-CD3 antibody treatment results in an increase in CD4+CD25LAP+ Tregs, which are distinct from naturally occurring Tregs expressing CD25+ on their surface. We also examined the effect of oral anti-CD3 treatment on natural CD4+CD25+Foxp3+ Tregs by fluorescence-activated cell sorter analyses. Interestingly, we found a modest but significant increase in CD4+CD25+Foxp3+ Tregs in MLNs of anti-CD3–treated mice compared with control mice (P<0.05) but not in spleens (Figure 1D and 1E).

Figure 1. Effects of oral anti-CD3 antibody administration on CD4+LAP+ Tregs and natural CD4+CD25+Foxp3+ Tregs in spleens and MLNs. ApoE−/− mice were fed control IgG or anti-CD3 antibody daily for 5 days. Two days after the last feeding, lymphoid cells from spleen and MLNs were prepared and stained with biotin-LAP followed by streptavidin-APC, FITC-anti-CD4, and PE-anti-CD25 or stained with FITC-anti-CD4, PE-anti-CD25, and APC-anti-Foxp3. A, Representative results of CD4, CD25, and LAP expression in MLNs assessed by flow cytometry. The graphs represent the percentage of LAP+ cells within the CD4+ population (B) and CD25LAP+ cells within the CD4+ population (C) in spleens and MLNs; n=6 to 7 per group. D, Representative results of CD4, CD25, and Foxp3 expression in MLNs assessed by flow cytometry. E, The graph represents the percentage of CD25+Foxp3+ cells within the CD4+ population in spleens and MLNs. n=8 to 9 per group. Two days after the last antibody treatment, MLN cells (F) and spleen cells (G) were prepared, and proliferation of these cells in vitro in response to anti-CD3 antibody was assessed by [3H]thymidine incorporation (mean±SEM of triplicate wells). Data are representatives of 2 experiments with similar results. *P<0.05 vs control IgG-treated mice.

To determine whether an increased number of Tregs by oral anti-CD3 contributes to the more suppressive potential of lymphoid cells, we performed in vitro proliferation assays of MLN cells or splenocytes in response to anti-CD3 antibody. As shown in Figure 1F and 1G, stimulation with anti-CD3 antibody resulted in the proliferation of MLN cells or splenocytes in control IgG-treated mice in a dose-dependent manner. In contrast, we observed a significant reduction in MLN or spleen cell proliferation of anti-CD3–treated mice (P<0.05), indicating the suppressed immune responses of lymphoid cells in anti-CD3–treated mice on polyclonal stimulation (Figure 1F and 1G). We additionally performed in vitro suppression assays to reveal whether oral anti-CD3 antibody treatment affects the suppressive function of Tregs. These experiments demonstrated that the suppressive function of CD4+CD25LAP+ Tregs or CD4+CD25+ Tregs was not altered by oral anti-CD3 treatment (Figure IIA and IIB in the online-only Data Supplement).

Oral Anti-CD3 Antibody Treatment Inhibits Atherosclerotic Plaque Formation and Reduces Inflammatory Cell Recruitment Into Plaques in ApoE−/− Mice

To determine the effect of oral anti-CD3 treatment on the development of atherosclerosis, we orally treated 6-week-old male ApoE−/− mice with a 5-μg dose of anti-CD3 antibody or control IgG daily for 5 consecutive days. During the experiments, no adverse effect was observed in either group. No statistical differences in body weight or plasma lipid profiles were detected between the 2 groups (Table). At 16 weeks of age, the mice were euthanized, and cryosections of the aortic root of control and anti-CD3–treated mice were stained with Oil Red O and analyzed quantitatively. Surprisingly, anti-CD3–treated mice showed ≈50% reduction in atherosclerotic lesion formation in the aortic root compared with control IgG-treated mice (aortic sinus mean plaque area of 11 3854±20 329 μm2 versus 54 565±5937 μm2 in control IgG-treated and anti-CD3–treated mice, respectively; P=0.007; Figure 2A). In parallel with the cross-sectional studies, en face analysis of thoracic aortas was performed. En face analysis of thoracic aortas revealed a significant 33% reduction in aortic plaque burden at 16 weeks in anti-CD3–treated mice (2.31±0.33%; P<0.05 versus controls) compared with control mice (3.44±0.33%) (Figure 2B). To further evaluate the effect of anti-CD3 antibody treatment on the progression of atherosclerosis, we treated 16-week-old male ApoE−/− mice, euthanized them at 24 weeks, and evaluated the atherosclerotic lesion formation. The results indicated that oral anti-CD3 antibody treatment did not prevent atherosclerosis progression (Figure III in the online-only Data Supplement).

Table. Analysis of Plasma Lipid Profile and Body Weight at 16 Weeks

Control CD3-Ab
Mean±SEM n Mean±SEM n
HDL indicates high-density lipoprotein.
Body weight, g 26.2±0.8 11 26.5±0.7 14
Total cholesterol, mg/dL 416.3±47.0 8 453.0±45.1 8
HDL cholesterol, mg/dL 12.6±1.0 8 14.5±1.6 8
Triglycerides, mg/dL 74.6±18.0 8 70.0±13.0 8

Figure 2. Oral anti-CD3 antibody administration inhibits atherosclerotic plaque formation. A, Representative photomicrographs of Oil Red O staining and quantitative analysis of atherosclerotic lesion size in the aortic sinus of male ApoE−/− mice treated with control IgG or anti-CD3 antibody. A black bar represents 200 μm. B, Representative photomicrographs of Oil Red O staining and quantitative analysis of atherosclerotic lesion size in the thoracic aortas of male ApoE−/− mice treated with control IgG or anti-CD3 antibody. Horizontal bars represent means. *P<0.05 and **P<0.01 vs control IgG-treated mice.

To determine the effects of oral anti-CD3 antibody treatment on plaque component, we next performed immunohistochemical studies of atherosclerotic lesions in the aortic sinus. Interestingly, the atherosclerotic lesions of anti-CD3–treated mice showed a marked 38% reduction in the accumulation of macrophages (P<0.01) and also a 30% decrease in CD4+ T-cell infiltration (P<0.01) compared with control mice (Figure 3A and 3B). In addition, immunohistochemical analysis of the plaques showed that relative smooth muscle cell and collagen contents in the aortic sinus plaques tended to be increased in anti-CD3–treated mice, but this did not reach statistical significance (Figure 3A and 3B). These results suggest that oral anti-CD3 antibody treatment reduces plaque inflammation, which may lead to decreased lesion development.

Figure 3. Effects of oral anti-CD3 treatment on atherosclerotic phenotype. A, Representative sections of aortic sinuses stained with antibodies specific for MOMA-2 for macrophages, CD4+ T cells, and α-smooth muscle actin (α-SMA) for smooth muscle cells in male ApoE−/− mice treated with control IgG or anti-CD3 antibody. The fibrous area was stained by Masson trichrome. A black bar represents 200 μm. B, Quantitative analyses of MOMA-2 (macrophage content), CD4+ T cells, α-actin (smooth muscle cell content), and Masson trichrome staining (collagen) in the aortic sinus of male ApoE−/− mice treated with control IgG or anti-CD3 antibody. Horizontal bars represent means. *P<0.01 vs control IgG-treated mice.

Expression of Inflammatory Markers and Regulatory T-Cell Markers in Atherosclerotic Plaques

To reveal the mechanisms of reduced plaque development and inflammatory cell recruitment, we examined mRNA expressions of the proinflammatory molecules in atherosclerotic aortas by quantitative reverse transcription (RT)-PCR. We found that proinflammatory cytokines such as interferon (IFN)-γ and interleukin (IL)-6, adhesion molecules such as intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1, chemokine monocyte chemoattractant protein (MCP)-1, and macrophages detected by CD68 were markedly reduced in anti-CD3–treated mice compared with control mice (P<0.05; Figure 4A and data not shown).

Figure 4. A, Total RNA was extracted from aortas of 16-week-old control IgG or anti-CD3–treated mice. Expression of IFN-γ, MCP-1, adhesion molecules (ICAM-1, VCAM-1), and Treg-associated markers (TGF-β, Foxp3, and CD25) in atherosclerotic aortas was quantified by quantitative real-time RT-PCR and normalized to GAPDH. Fold change relative to each control group is shown. n=4 to 5 per group. B, Representative section and quantitative analysis of Foxp3+ cells in the plaques of the aortic sinus from control and anti-CD3–treated mice. A white arrow indicates the Foxp3-positive cell. A white bar represents 100 μm. Horizontal bars represent means. *P<0.05 vs control IgG-treated mice.

Recent studies suggest that antigen presentation may occur within the atherosclerotic plaque in addition to the lymphoid organs and that Tregs migrate to the atherosclerotic lesions to suppress local immune responses,19,20 although actual roles of intraplaque Tregs in atherogenesis have not yet been clarified. To determine whether increased accumulation of Tregs into the plaques is involved in the reduction of atherosclerotic lesion formation, we analyzed the expression of Treg-associated markers in the lesions by quantitative reverse transcription PCR (RT-PCR). We found no significant differences in the relative mRNA expressions of CD4+LAP+ Treg-derived cytokine TGF-β or natural Treg-specific markers Foxp3 and CD25 in the plaques among the 2 groups (Figure 4A). Furthermore, immunohistochemical studies of atherosclerotic lesions with the use of anti-Foxp3 antibody demonstrated an increase in the number of natural Tregs within the plaque of anti-CD3–treated mice (Figure 4B). When it is taken into consideration that anti-CD3–treated mice have less plaque in the aorta than control mice, this result is consistent with the quantitative RT-PCR analysis of Treg-specific markers. These data indicate that oral anti-CD3 antibody treatment could inhibit the migration of effector T cells into the lesion selectively and relatively increase the proportion of natural Tregs in atherosclerotic plaque that might suppress pathogenic T-cell immune responses or macrophage activation as well as in the lymphoid organs.

Effects of Oral Anti-CD3 Antibody on Cytokine Production in Lymphocytes From Spleen and MLNs

Next, to determine whether induction of various Tregs by oral anti-CD3 antibody changed the cytokine production profile from lymphocytes, we examined cytokine secretion from spleen or MLN lymphocytes in response to concanavalin A using cytokine arrays and ELISA. On day 7 after the treatment of oral anti-CD3 antibody, we tried to assess the changes in cytokine production from MLN cells; however, the cytokine levels (IFN-γ, IL-4, and IL-10) were below the detectable levels. Therefore, we next investigated cytokine mRNA expression of unstimulated MLN cells by using quantitative RT-PCR. Interestingly, anti-CD3 antibody treatment resulted in a significant decrease in mRNA expressions of T-helper type 1 (Th1) cytokine (IFN-γ) and T-helper type 2 (Th2) cytokines (IL-4 and IL-10) in MLN cells (P<0.05; Figure 5A). Consistent with previous studies, significantly increased production of antiinflammatory cytokine TGF-β, which was the only detectable cytokine in our experiments, was observed in MLN cells of anti-CD3–treated mice (P<0.05; Figure 5B). CD4+LAP+ Tregs induced by oral anti-CD3 antibody reach mesenteric lymph nodes and other lymphoid organs such as spleens through the bloodstream, where they may induce deactivation of pathogenic T cells and macrophages. Therefore, we semiquantitatively analyzed various cytokine or chemokine productions in spleen lymphocytes on day 7 after the treatment. Lymphocytes from anti-CD3–treated mice secreted fewer Th1 cytokines (IFN-γ and IL-2), Th2 cytokines (IL-4, IL-10, and IL-13), and inflammation-related cytokines or chemokines (IL-17, MIP-1α, MIP-2, RANTES, and TNF-α) under stimulation with concanavalin A than those from control mice (Figure 5C), indicating suppressed Th1 and Th2 immune responses in anti-CD3–treated mice. ELISA data confirmed that TGF-β production was markedly increased and cytokine levels such as IFN-γ, IL-10, and IL-17 were significantly decreased in spleen lymphocyte supernatants from anti-CD3–treated mice compared with controls (P<0.05; Figure 5D). Although we could not detect IL-6 expression in the cytokine array analysis, ELISA data for lymphocyte IL-6 production revealed lower production in spleen lymphocytes of anti-CD3–treated mice than controls (P<0.05; Figure 5D). We also assessed cytokine production from lymphocytes 10 weeks after the treatment (at 16 weeks). Surprisingly, we found markedly increased TGF-β secretion in spleen lymphocytes of anti-CD3–treated mice (P<0.05), with no differences in IFN-γ or IL-10 levels between the groups (Figure 5E and data not shown), suggesting the long-term protective effect of TGF-β. In contrast to the increased TGF-β production at 16 weeks, we detected no significant difference in the number of CD4+LAP+ Tregs among the 2 groups, although CD25LAP+ Tregs in the CD4+ T cell population tended to be slightly increased in spleens of anti-CD3–treated mice (1.78±0.14% versus 2.11±0.15% in control IgG-treated and anti-CD3–treated mice, respectively; P=0.068; data not shown). Previous studies have shown that ani-CD3 antibody treatment not only increases the number of CD4+LAP+ Tregs but also enhances the regulatory function of these cells.11,12 Although we did not observe the enhanced suppressive function of Tregs in our in vitro experiments (Figure IIA in the online-only Data Supplement), our data imply that the prolonged TGF-β secretion in lymphocytes of anti-CD3–treated mice may reflect the altered function of these Tregs, which might be maintained for a long time, and suggest an important prolonged contribution of this antiinflammatory cytokine to a reduction in atherosclerosis development.

Figure 5. Immune responses after oral anti-CD3 antibody treatment. ApoE−/− mice were fed control IgG or CD3-specific antibody from day 1 to day 5 and euthanized on day 7. A, Cytokine mRNA levels (IFN-γ, IL-4, and IL-10) in unstimulated MLN cells collected from 4 mice in each group were determined by quantitative RT-PCR. Results are expressed as mean±SEM of triplicates. B, Lymphocytes from MLN cells were stimulated with concanavalin A in vitro for 72 hours. TGF-β production in supernatants was measured by ELISA. n=4 per group. C, Lymphocytes from spleen were prepared and stimulated with concanavalin A in vitro. Levels of various cytokines and chemokines in cell supernatants collected from 4 mice in each group were determined semiquantitatively by a cytokine array kit. Data represent 1 of 2 experiments. D, Cytokine levels (TGF-β, IFN-γ, IL-10, IL-6, and IL-17) in supernatants from spleen lymphocytes were examined quantitatively by ELISA. n=4 per group. E, Six-week-old ApoE−/− mice were fed control IgG or CD3-specific antibody daily for 5 days and killed at 16 weeks of age. TGF-β production from spleen lymphocytes was measured as described above. n=5 per group. *P<0.05 vs control IgG-treated mice.

Suppressive Function of Oral Anti-CD3 Antibody Is Mediated by TGF-β

Previous data suggest that TGF-β plays a key role in the suppressive function of both natural Tregs21 and CD4+LAP+ Tregs.22 In addition, our ELISA data for cytokine production in lymphocytes show that TGF-β is the only cytokine with an antiinflammatory property that is enhanced by anti-CD3 antibody treatment. To determine whether TGF-β is directly involved in preventing plaque formation after anti-CD3 treatment, we performed an in vivo TGF-β neutralization study. Anti-CD3–treated mice that received anti-TGF-β neutralizing antibody have significantly increased atherosclerotic lesion formation and accumulations of macrophages and CD4+ T cells in the plaques compared with anti-CD3–treated mice receiving isotype-matched antibody (Figure 6A and 6B). In addition, when mice were injected with neutralizing antibody, there were no significant differences in aortic root plaque size and inflammatory cell infiltration between control and anti-CD3–treated mice (Figure 6A and 6B). Together, these findings suggest that the suppressive function of oral anti-CD3 antibody in vivo is mediated by TGF-β, at least in part.

Figure 6. Suppression of atherosclerosis and inflammatory changes is TGF-β dependent. After being fed with control IgG or CD3-specific antibody daily for 5 days, mice were injected intraperitoneally with 100 μg of neutralizing anti-TGF-β antibody or isotype-matched antibody once a week from age 7 weeks to age 16 weeks. A, Representative photomicrographs of Oil Red O staining and quantitative analysis of atherosclerotic lesion size in the aortic sinus. A black bar represents 200 μm. B, Quantitative analyses of MOMA-2 (macrophage content) and CD4+ T cells in the aortic sinus. Horizontal bars represent means. *P<0.05 vs anti-CD3–treated mice receiving control rat IgG.

Discussion

Recent work has clearly demonstrated that natural Tregs, which show a high expression of CD25 on their surface23 and also express the transcription factor forkhead box P3 (Foxp3),24 play a protective role in atherogenesis in mice.4 Besides naturally occurring Tregs, previous studies have implicated the protective role of other types of Tregs including Tr1 or Th3 cells in the development of atherosclerosis in mice through the production of antiinflammatory cytokines such as IL-10 or TGF-β, respectively.16,25 Recently, CD4+LAP+ Treg has been identified as a new subset of Tregs that suppresses autoimmune diseases in mice.11–13,22 In the present study, we show that oral anti-CD3 antibody treatment attenuates atherosclerotic lesion formation and that this reduction is associated with an increase in the number of Tregs, including CD4+LAP+ Tregs and CD4+CD25+Foxp3+ Tregs, which are possibly induced in the gut and migrate into other lymphoid organs, subsequently leading to suppression of Th1 and Th2 immune responses through the production of TGF-β. Our data are the first report demonstrating a possible role of CD4+LAP+ Tregs in atherogenesis, although further studies are required to provide more direct evidence for the relationship between an enhancement in CD4+LAP+ Treg number or function and a reduction of atherosclerosis.

Importantly, unlike the case of intravenous FcR-binding anti-CD3 antibody treatment, we observed no evidence of adverse responses of oral FcR-binding anti-CD3 antibody during the experiment, which may be related to the fact that orally administered anti-CD3 antibody is very small in amount (10 times less than that of the usual intravenous administration) and for the most part does not seem to enter the bloodstream.11 It has been reported that immunosuppressive effects by intravenous FcR-binding anti-CD3 antibody may involve mechanisms such as depletion of pathogenic T cells from the bloodstream or lymphoid organs and modulation of the T-cell receptor on T cells.7 In contrast to intravenous FcR-binding anti-CD3 antibody administration, we never observed the T-cell receptor downmodulation or depletion of T cells (Figure I in the online-only Data Supplement), which is consistent with the previous report.11 Ochi et al11 speculated that low doses of oral anti-CD3 antibody administration may result in the induction of Tregs by delivering a weak signal to T cells, which is observed similarly in mucosal tolerance induction. They demonstrated that experimental autoimmune encephalitis was suppressed in mice fed 5 μg anti-CD3 antibody with or without Fc portion but not in mice fed 50 μg or 500 μg antibody.11 Consistent with its effect on suppression of experimental autoimmune encephalitis, it was also shown that suppression of proliferation in spleens and MLN cells was observed with 5 μg anti-CD3 antibody but not with 50 μg or 500 μg. In consideration of this, we decided to use a 5-μg dose of FcR-binding anti-CD3 antibody for the treatment of ApoE−/− mice and found that this treatment results in a dramatic reduction in atherosclerotic lesion formation, associated with a significant increase in CD4+LAP+ Tregs. Intriguingly, we also detected an increased proportion of CD4+CD25+Foxp3+ Tregs in MLNs of anti-CD3–treated mice.

Administration of FcR-nonbinding anti-CD3 monoclonal antibody has been shown to induce long-lasting immune tolerance and to be an effective treatment for autoimmune diabetes in mice and in humans,8,9 acute transplant rejection in humans,7 and, notably, atherosclerosis in mice.10 These studies have indicated that the long-term beneficial effect of FcR-nonbinding anti-CD3 antibody may possibly be caused by increasing TGF-β–producing Tregs. When parenteral FcR-nonbinding anti-CD3 antibody is compared with oral FcR-binding anti-CD3 antibody, there may be some common mechanisms underlying suppression of autoimmunity because both appear to work mainly by inducing TGF-β–producing Tregs rather than eliminating pathogenic T cells. In consideration of this, it is possible that after oral administration, FcR-binding anti-CD3 antibody might lose its Fc portion in the gut and consequently produce F(ab′)2 (FcR-nonbinding anti-CD3 antibody), a small amount of which might enter the bloodstream, leading to induction of Tregs.

Several in vitro and in vivo studies demonstrated that naive T cells in the periphery can differentiate into CD4+Foxp3+ Tregs in the presence of TGF-β.26,27 Recent studies also suggest that differentiation of CD4+Foxp3+ Tregs may be facilitated by dendritic cells in the gut-associated lymphoid tissue in the presence of TGF-β and that retinoic acid produced by dendritic cells plays an important role in their differentiation.28 In this study, we found a significant increase in CD4+ LAP+ Tregs in MLNs and spleens of anti-CD3–treated mice, in association with markedly increased TGF-β secretion by CD4+LAP+ Tregs, which have been reported to secrete much more TGF-β than CD4+LAP T cells.11 Therefore, it is highly possible that upregulated TGF-β secretion in the gut from anti-CD3–induced CD4+LAP+ Tregs could lead to induction of CD4+CD25+Foxp3+ Tregs.

Because previous studies have reported that oral anti-CD3 treatment results in conversion of Th1 immune responses into Th2 or Th3 immune responses in the periphery associated with decreased proliferation in spleen cells and MLN cells,11,12 it seems possible that similar changes in the cytokine production profile from lymphocytes could contribute to the observed antiatherogenic effect of oral anti-CD3 antibody in our study. Unlike previous studies, however, our ELISA and cytokine array analyses clearly demonstrated significant suppression in both Th1 and Th2 immune responses, as well as markedly increased TGF-β production from lymphocytes in oral anti-CD3–treated mice. In this experiment, we used atherosclerosis-prone ApoE−/− mice on C57BL/6 background, which are known to show Th1-prone phenotype. Therefore, the difference in mouse genetic background used might be a possible explanation for the distinct immune responses by oral anti-CD3 antibody between our study and others. Accumulating evidence suggests that T-helper 17 (Th17) cells, which produce the inflammatory cytokine IL-17, play an important role in promotion of many autoimmune diseases.29 In addition, it has been reported that TGF-β stimulation leads to differentiation of naive T cells to Th17 cells or CD4+Foxp3+ Tregs in mice in the presence or absence of IL-6, respectively.30 Our data indicate a shift from Th17 cells to CD4+Foxp3+ Tregs in oral anti-CD3–treated mice, which might partially affect the atheroprotective effects of oral anti-CD3, although whether Th17 cells accelerate atherosclerosis as well as other autoimmune diseases thus far remains unclear.

TGF-β has been attracting much attention as a potent antiatherosclerotic cytokine.31 TGF-β, which is produced by many types of cells in atherosclerotic plaques, suppresses pathogenic immune responses such as recruitment of inflammatory cells into the plaques or foam cell formation and increases collagen biosynthesis.32,33 In particular, TGF-β signaling in T cells appears to be indispensable for the regulation of atherosclerotic disease progression because ApoE−/− mice with specific deletion of TGF-β signaling in T cells show remarkably accelerated plaque formation and inflammatory cell infiltration, associated with enhancement of both Th1 and Th2 immune responses.34 In consideration of this, our data suggest that increased TGF-β production from T cells may be a key mechanism for the preventive effect of oral anti-CD3 antibody on atherosclerosis through the downregulation in both Th1 and Th2 immune responses.

We are somewhat surprised because only 5-day oral treatment with a small amount of anti-CD3 antibody has a prolonged effect on the immune system and results in a dramatic atheroprotective phenotype. This long-term protective effect of oral anti-CD3 antibody was observed similarly in other autoimmune diseases such as experimental autoimmune encephalitis and autoimmune diabetes.11,12 TGF-β appears to play a crucial role in the prolonged antiatherogenic effect because we detected a significant increase in TGF-β production in spleen lymphocytes of oral anti-CD3–treated mice at 10 weeks after the antibody administration. It is reported that CD4+LAP+ Tregs secrete TGF-β and are also activated by TGF-β produced by themselves,22 indicating that they may act in autocrine and paracrine manners. We speculate that a transient increase in CD4+LAP+ Tregs may lead to a positive-feedback loop of activation of CD4+LAP+ Tregs through the production of TGF-β. On the basis of our observed findings, this novel method, which directly governs the inflammatory immune responses in atherogenesis, could be a hopeful strategy to prevent cardiovascular diseases.

In summary, we have demonstrated that oral anti-CD3 antibody treatment inhibits atherosclerosis development via inducing various Tregs and that TGF-β, which may be derived mainly from CD4+LAP+ Tregs, possibly plays an important role in this suppressive mechanism. Although the mechanism by which oral anti-CD3 antibody increases Tregs remains to be clarified, our data indicate that induction of Tregs in the intestine by oral anti-CD3 antibody could be used as a promising therapeutic approach for atherosclerotic vascular disorders. Recent studies have demonstrated no toxicity of orally administered anti-CD3 antibody not only in mice but also in humans.35 Clinical studies in human are required to identify the efficacy of oral anti-CD3 antibody in the prevention of atherosclerotic diseases.

We thank Tomoyuki Yamaguchi for helpful suggestions and Michiyo Nakata for technical assistance.

Sources of Funding

This work was supported by a Grant-in-Aid for Scientific Research in Japan and by research grants from the Jinsenkai Medical Foundation, Kanae Medical Foundation, Suzuken Memorial Foundation, and Uehara Medical Foundation.

Disclosures

None.

Footnotes

Correspondence to Tomoya Yamashita, Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan. E-mail

References

  • 1 Ross R. Atherosclerosis: an inflammatory disease. N Engl J Med. 1999; 340: 115–126.CrossrefMedlineGoogle Scholar
  • 2 Hansson GK, Libby P. The immune response in atherosclerosis: a double-edged sword. Nat Rev Immunol. 2006; 6: 508–519.CrossrefMedlineGoogle Scholar
  • 3 Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008; 133: 775–787.CrossrefMedlineGoogle Scholar
  • 4 Ait-Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, Zoll J, Merval R, Esposito B, Cohen JL, Fisson S, Flavell RA, Hansson GK, Klatzmann D, Tedgui A, Mallat Z. Natural regulatory T cells control the development of atherosclerosis in mice. Nat Med. 2006; 12: 178–180.CrossrefMedlineGoogle Scholar
  • 5 Gotsman I, Grabie N, Gupta R, Dacosta R, MacConmara M, Lederer J, Sukhova G, Witztum JL, Sharpe AH, Lichtman AH. Impaired regulatory T-cell response and enhanced atherosclerosis in the absence of inducible costimulatory molecule. Circulation. 2006; 114: 2047–2055.LinkGoogle Scholar
  • 6 Mor A, Planer D, Luboshits G, Afek A, Metzger S, Chajek-Shaul T, Keren G, George J. Role of naturally occurring CD4+ CD25+ regulatory T cells in experimental atherosclerosis. Arterioscler Thromb Vasc Biol. 2007; 27: 893–900.LinkGoogle Scholar
  • 7 Chatenoud L, Bluestone JA. CD3-specific antibodies: a portal to the treatment of autoimmunity. Nat Rev Immunol. 2007; 7: 622–632.CrossrefMedlineGoogle Scholar
  • 8 Herold KC, Hagopian W, Auger JA, Poumian-Ruiz E, Taylor L, Donaldson D, Gitelman SE, Harlan DM, Xu D, Zivin RA, Bluestone JA. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes mellitus. N Engl J Med. 2002; 346: 1692–1698.CrossrefMedlineGoogle Scholar
  • 9 Belghith M, Bluestone JA, Barriot S, Megret J, Bach JF, Chatenoud L. TGF-beta-dependent mechanisms mediate restoration of self-tolerance induced by antibodies to CD3 in overt autoimmune diabetes. Nat Med. 2003; 9: 1202–1208.CrossrefMedlineGoogle Scholar
  • 10 Steffens S, Burger F, Pelli G, Dean Y, Elson G, Kosco-Vilbois M, Chatenoud L, Mach F. Short-term treatment with anti-CD3 antibody reduces the development and progression of atherosclerosis in mice. Circulation. 2006; 114: 1977–1984.LinkGoogle Scholar
  • 11 Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso AS, Wu H, Chen ML, Gandhi R, Miller A, Maron R, Weiner HL. Oral CD3-specific antibody suppresses autoimmune encephalomyelitis by inducing CD4+ CD25- LAP+ T cells. Nat Med. 2006; 12: 627–635.CrossrefMedlineGoogle Scholar
  • 12 Ishikawa H, Ochi H, Chen ML, Frenkel D, Maron R, Weiner HL. Inhibition of autoimmune diabetes by oral administration of anti-CD3 monoclonal antibody. Diabetes. 2007; 56: 2103–2109.CrossrefMedlineGoogle Scholar
  • 13 Wu HY, Quintana FJ, Weiner HL. Nasal anti-CD3 antibody ameliorates lupus by inducing an IL-10-secreting CD4+ CD25- LAP+ regulatory T cell and is associated with down-regulation of IL-17+ CD4+ ICOS+ CXCR5+ follicular helper T cells. J Immunol. 2008; 181: 6038–6050.CrossrefMedlineGoogle Scholar
  • 14 Faria AM, Weiner HL. Oral tolerance. Immunol Rev. 2005; 206: 232–259.CrossrefMedlineGoogle Scholar
  • 15 Maron R, Sukhova G, Faria AM, Hoffmann E, Mach F, Libby P, Weiner HL. Mucosal administration of heat shock protein-65 decreases atherosclerosis and inflammation in aortic arch of low-density lipoprotein receptor-deficient mice. Circulation. 2002; 106: 1708–1715.LinkGoogle Scholar
  • 16 van Puijvelde GH, Hauer AD, de Vos P, van den Heuvel R, van Herwijnen MJ, van der Zee R, van Eden W, van Berkel TJ, Kuiper J. Induction of oral tolerance to oxidized low-density lipoprotein ameliorates atherosclerosis. Circulation. 2006; 114: 1968–1976.LinkGoogle Scholar
  • 17 Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama M. Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE-deficient mice. J Clin Invest. 2002; 110: 331–340.CrossrefMedlineGoogle Scholar
  • 18 Sasaki N, Yamashita T, Takaya T, Shinohara M, Shiraki R, Takeda M, Emoto N, Fukatsu A, Hayashi T, Ikemoto K, Nomura T, Yokoyama M, Hirata K, Kawashima S. Augmentation of vascular remodeling by uncoupled endothelial nitric oxide synthase in a mouse model of diabetes mellitus. Arterioscler Thromb Vasc Biol. 2008; 28: 1068–1076.LinkGoogle Scholar
  • 19 Robertson AK, Hansson GK. T cells in atherogenesis: for better or for worse? Arterioscler Thromb Vasc Biol. 2006; 26: 2421–2432.LinkGoogle Scholar
  • 20 Mallat Z, Ait-Oufella H, Tedgui A. Regulatory T-cell immunity in atherosclerosis. Trends Cardiovasc Med. 2007; 17: 113–118.CrossrefMedlineGoogle Scholar
  • 21 Wan YY, Flavell RA. TGF-beta and regulatory T cell in immunity and autoimmunity. J Clin Immunol. 2008; 28: 647–659.CrossrefMedlineGoogle Scholar
  • 22 Oida T, Zhang X, Goto M, Hachimura S, Totsuka M, Kaminogawa S, Weiner HL. CD4+CD25- T cells that express latency-associated peptide on the surface suppress CD4+CD45RBhigh-induced colitis by a TGF-beta-dependent mechanism. J Immunol. 2003; 170: 2516–2522.CrossrefMedlineGoogle Scholar
  • 23 Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25): breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995; 155: 1151–1164.CrossrefMedlineGoogle Scholar
  • 24 Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003; 299: 1057–1061.CrossrefMedlineGoogle Scholar
  • 25 Mallat Z, Gojova A, Brun V, Esposito B, Fournier N, Cottrez F, Tedgui A, Groux H. Induction of a regulatory T cell type 1 response reduces the development of atherosclerosis in apolipoprotein E-knockout mice. Circulation. 2003; 108: 1232–1237.LinkGoogle Scholar
  • 26 Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady G, Wahl SM. Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J Exp Med. 2003; 198: 1875–1886.CrossrefMedlineGoogle Scholar
  • 27 Apostolou I, von Boehmer H. In vivo instruction of suppressor commitment in naive T cells. J Exp Med. 2004; 199: 1401–1408.CrossrefMedlineGoogle Scholar
  • 28 Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007; 204: 1775–1785.CrossrefMedlineGoogle Scholar
  • 29 Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, Hood L, Zhu Z, Tian Q, Dong C. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005; 6: 1133–1141.CrossrefMedlineGoogle Scholar
  • 30 Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006; 441: 235–238.CrossrefMedlineGoogle Scholar
  • 31 Grainger DJ. Transforming growth factor beta and atherosclerosis: so far, so good for the protective cytokine hypothesis. Arterioscler Thromb Vasc Biol. 2004; 24: 399–404.LinkGoogle Scholar
  • 32 Mallat Z, Gojova A, Marchiol-Fournigault C, Esposito B, Kamate C, Merval R, Fradelizi D, Tedgui A. Inhibition of transforming growth factor-beta signaling accelerates atherosclerosis and induces an unstable plaque phenotype in mice. Circ Res. 2001; 89: 930–934.CrossrefMedlineGoogle Scholar
  • 33 Lutgens E, Gijbels M, Smook M, Heeringa P, Gotwals P, Koteliansky VE, Daemen MJ. Transforming growth factor-beta mediates balance between inflammation and fibrosis during plaque progression. Arterioscler Thromb Vasc Biol. 2002; 22: 975–982.LinkGoogle Scholar
  • 34 Robertson AK, Rudling M, Zhou X, Gorelik L, Flavell RA, Hansson GK. Disruption of TGF-beta signaling in T cells accelerates atherosclerosis. J Clin Invest. 2003; 112: 1342–1350.CrossrefMedlineGoogle Scholar
  • 35 Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso A, Wu H, Chen ML, Gandhi R, Miller A, Maron R, Weiner HL. New immunosuppressive approaches: oral administration of CD3-specific antibody to treat autoimmunity. J Neurol Sci. 2008; 274: 9–12.CrossrefMedlineGoogle Scholar
circulationaha Circulation Circulation Circulation 0009-7322 1524-4539 Lippincott Williams & Wilkins
CLINICAL PERSPECTIVE 17 11 2009

Atherosclerosis is a chronic inflammatory condition of the arterial wall involving immune cells and their releasing molecules. This important notion has led to a quest to develop an antiinflammatory and immune therapy for prevention of atherosclerosis in clinical patients. Accumulating evidence suggests that several subsets of regulatory T cells (Tregs) inhibit atherosclerosis development through the downregulation of activated T-cell responses. Recent studies have demonstrated that parenteral administration of anti-CD3–specific antibody restores long-lasting self-tolerance and is effective in autoimmune diabetes in mice and humans, partly because of the expansion of CD4+CD25+ Tregs. Interestingly, orally administered anti-CD3 antibody has been shown to prevent systemic autoimmunity through induction of novel Tregs expressing latency-associated peptide (LAP) on their surface in the gut. In the present study, we first show that oral anti-CD3 antibody treatment attenuates atherosclerotic lesion formation and plaque inflammation and that this suppression is associated with an increase in the number of CD4+CD25LAP+ Tregs and, unexpectedly, CD4+CD25+Foxp3+ Tregs, which are possibly induced in the gut-associated lymphoid tissue and migrate into other lymphoid organs or atherosclerotic plaques, subsequently leading to suppression of T-helper type 1 and type 2 immune responses through a transforming growth factor-β–dependent mechanism. Our data indicate that oral administration of anti-CD3 antibody seems to be safe and easy to apply and therefore could be used as a promising therapeutic approach for atherosclerotic vascular disorders. Clinical studies are required.

The online-only Data Supplement is available with this article at http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.109.863431/DC1.

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.