10
Role of Naturally Occurring CD4 CD25 Regulatory T Cells in Experimental Atherosclerosis Adi Mor, David Planer, Galia Luboshits, Arnon Afek, Shula Metzger, Tova Chajek-Shaul, Gad Keren, Jacob George Objective—Naturally occurring CD4 CD25 regulatory T cells (Tregs) exert suppressive effects on effector CD4 cells and downregulate experimental autoimmune disorders. We investigated the importance and potential role of Tregs in murine atherogenesis. Methods and Results—Tregs were investigated comparatively between aged and young apolipoprotein E– knockout (ApoE-KO) mice and age-matched C57BL/6 littermates. The effect of oxidized LDL (oxLDL) was tested on the functional suppressive properties of Tregs from ApoE-KO and C57BL/6 mice. Tregs, CD4 CD25 cells, and saline were infused into ApoE-KO mice to study their effects on atherogenesis. Treg numbers were reduced in atherosclerotic compared with nonatherosclerotic ApoE-KO mice. The functional suppressive properties of Tregs from ApoE-KO mice were compromised in comparison with those from their C57BL/6 littermates. Thus, oxLDL attenuated the suppressive properties of Tregs from C57BL/6 mice and more so in ApoE-KO mice. Transfer of Tregs from age-matched ApoE-KO mice resulted in significant attenuation of atherosclerosis compared with that after delivery of CD4 CD25 / T cells or phosphate-buffered saline. Conclusions—CD4 CD25 Tregs may play a protective role in the progression of atherosclerosis and could be considered a therapeutic tool if results from human studies can solidify observations in murine models. (Arterioscler Thromb Vasc Biol. 2007;27:893-900.) Key Words: atherosclerosis T cells immune response adoptive transfer foxp3 A therosclerosis is a multifactorial process that involves interactions among endothelial cells, macrophages, smooth muscle cells, and lymphocytes. 1–3 The outcome of growth of the atherosclerotic plaque is a result of its final influence on organ perfusion, resulting in cerebrovascular events and acute coronary syndromes. The role of the immune system in atherosclerosis has received considerable interest in recent years; however, sufficient knowledge to justify the development of targeted immunomodulatory strat- egies has not yet been obtained. 1–3 Cumulative data based on atherosclerotic experimental models suggest that CD4 lym- phocytes are present within murine plaques from initial stages of the disease, 4 and adoptive transfer of these cells is potentially proatherogenic. 5–7 Moreover, it has been shown that cytokine products of CD4 cells (interferon-, interleukin [IL]-10, IL-4, IL-18, etc) also influence the extent and nature of the atherosclerotic plaque (reviewed in Hansson, 1 Libby, 2 and Binder et al 3 ). These collective findings support the contention that antigen-specific responses may be operable in the evolving atheromatous plaque. In recent years, several T-cell populations have been described that possess predominant regulatory effects (Tregs) on T cells’ responding to infection-driven and self-antigens (reviewed in Sakaguchi 8 and O’Garra and Vieira 9 ). Initial studies addressing these regulatory T-cell populations were reported in experimental animal models, and some of these findings were reproduced subsequently in humans. Accord- ingly, a compromised number or function of Tregs has been associated with immune system–mediated disorders in hu- mans, such as diabetes, 10 rheumatoid arthritis, 11 and multiple sclerosis. 12 In this context, 2 populations of T cells were described: the first appears to respond to antigen-specific, major histocom- patibility complex–restricted triggers with secretion of anti- inflammatory cytokines, 13,14 and the second population of regulatory cells comprises the CD4 CD25 Tregs. 15,16 It was recently found that the transcriptional regulator forkhead/ winged helix transcription factor foxp3 governs mouse CD4 CD25 Treg function. 17,18 This finding was strongly supported by the observation that patients with immune system dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance have a severe inflammatory disease accompanied by a mutation in the FOXP3 gene. 19 Original received June 24, 2006; final version accepted November 19, 2006. From the Department of Cardiology (A.M., G.L., A.A., G.K., J.G.), Tel Aviv Sourasky Medical Center, Sackler School of Medicine, Tel Aviv University, Tel Aviv, and the Department of Medicine (D.P., S.M., T.C.-S.), Hadassah University Hospital, Mount Scopus, Israel. A.M. and D.P. contributed equally to this study. Correspondence and reprint requests to Jacob George, MD, Department of Cardiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel. E-mail [email protected] © 2007 American Heart Association, Inc. Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org DOI: 10.1161/01.ATV.0000259365.31469.89 893 by guest on August 8, 2015 http://atvb.ahajournals.org/ Downloaded from by guest on August 8, 2015 http://atvb.ahajournals.org/ Downloaded from by guest on August 8, 2015 http://atvb.ahajournals.org/ Downloaded from

Role of Naturally Occurring CD4+CD25+ Regulatory T Cells in Experimental Atherosclerosis

Embed Size (px)

Citation preview

Role of Naturally Occurring CD4�CD25� Regulatory T Cellsin Experimental Atherosclerosis

Adi Mor, David Planer, Galia Luboshits, Arnon Afek, Shula Metzger, Tova Chajek-Shaul,Gad Keren, Jacob George

Objective—Naturally occurring CD4�CD25� regulatory T cells (Tregs) exert suppressive effects on effector CD4 cells anddownregulate experimental autoimmune disorders. We investigated the importance and potential role of Tregs inmurine atherogenesis.

Methods and Results—Tregs were investigated comparatively between aged and young apolipoprotein E–knockout(ApoE-KO) mice and age-matched C57BL/6 littermates. The effect of oxidized LDL (oxLDL) was tested on thefunctional suppressive properties of Tregs from ApoE-KO and C57BL/6 mice. Tregs, CD4�CD25� cells, and salinewere infused into ApoE-KO mice to study their effects on atherogenesis. Treg numbers were reduced in atheroscleroticcompared with nonatherosclerotic ApoE-KO mice. The functional suppressive properties of Tregs from ApoE-KO micewere compromised in comparison with those from their C57BL/6 littermates. Thus, oxLDL attenuated the suppressiveproperties of Tregs from C57BL/6 mice and more so in ApoE-KO mice. Transfer of Tregs from age-matched ApoE-KOmice resulted in significant attenuation of atherosclerosis compared with that after delivery of CD4�CD25�/� T cells orphosphate-buffered saline.

Conclusions—CD4�CD25� Tregs may play a protective role in the progression of atherosclerosis and could be considereda therapeutic tool if results from human studies can solidify observations in murine models. (Arterioscler Thromb VascBiol. 2007;27:893-900.)

Key Words: atherosclerosis � T cells � immune response � adoptive transfer � foxp3

Atherosclerosis is a multifactorial process that involvesinteractions among endothelial cells, macrophages,

smooth muscle cells, and lymphocytes.1–3 The outcome ofgrowth of the atherosclerotic plaque is a result of its finalinfluence on organ perfusion, resulting in cerebrovascularevents and acute coronary syndromes. The role of theimmune system in atherosclerosis has received considerableinterest in recent years; however, sufficient knowledge tojustify the development of targeted immunomodulatory strat-egies has not yet been obtained.1–3 Cumulative data based onatherosclerotic experimental models suggest that CD4 lym-phocytes are present within murine plaques from initial stagesof the disease,4 and adoptive transfer of these cells ispotentially proatherogenic.5–7 Moreover, it has been shownthat cytokine products of CD4 cells (interferon-�, interleukin[IL]-10, IL-4, IL-18, etc) also influence the extent and natureof the atherosclerotic plaque (reviewed in Hansson,1 Libby,2

and Binder et al3). These collective findings support thecontention that antigen-specific responses may be operable inthe evolving atheromatous plaque.

In recent years, several T-cell populations have beendescribed that possess predominant regulatory effects (Tregs)

on T cells’ responding to infection-driven and self-antigens(reviewed in Sakaguchi 8 and O’Garra and Vieira9). Initialstudies addressing these regulatory T-cell populations werereported in experimental animal models, and some of thesefindings were reproduced subsequently in humans. Accord-ingly, a compromised number or function of Tregs has beenassociated with immune system–mediated disorders in hu-mans, such as diabetes,10 rheumatoid arthritis,11 and multiplesclerosis.12

In this context, 2 populations of T cells were described: thefirst appears to respond to antigen-specific, major histocom-patibility complex–restricted triggers with secretion of anti-inflammatory cytokines,13,14 and the second population ofregulatory cells comprises the CD4�CD25� Tregs.15,16 It wasrecently found that the transcriptional regulator forkhead/winged helix transcription factor foxp3 governs mouseCD4�CD25� Treg function.17,18 This finding was stronglysupported by the observation that patients with immunesystem dysregulation, polyendocrinopathy, enteropathy, andX-linked inheritance have a severe inflammatory diseaseaccompanied by a mutation in the FOXP3 gene.19

Original received June 24, 2006; final version accepted November 19, 2006.From the Department of Cardiology (A.M., G.L., A.A., G.K., J.G.), Tel Aviv Sourasky Medical Center, Sackler School of Medicine, Tel Aviv

University, Tel Aviv, and the Department of Medicine (D.P., S.M., T.C.-S.), Hadassah University Hospital, Mount Scopus, Israel.A.M. and D.P. contributed equally to this study.Correspondence and reprint requests to Jacob George, MD, Department of Cardiology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel. E-mail

[email protected]© 2007 American Heart Association, Inc.

Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org DOI: 10.1161/01.ATV.0000259365.31469.89

893 by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

In a recent study by Ait-Oufella et al,20 it was suggested forthe first time that Tregs are protective in murine atherogen-esis. Because atherosclerosis is influenced by innate andadaptive immune system responses,1–3 we set out to investi-gate the hypothesis that naturally occurring Tregs are capableof influencing the size and composition of atheroscleroticlesions. Employing the apolipoprotein E–knockout (ApoE-KO) mouse model, we found that Tregs are compromised inatherosclerotic animals and that their delivery attenuatesplaque progression.

MethodsAnimalsApoE-KO mice on a C57BL/6 background21 and their wild-typelittermates were purchased from Jackson Laboratories (Bar Harbor,Me) and maintained at the local animal facility. Mice were fed anormal chow diet containing 4.5% fat by weight (0.02% cholesterol).

Preparation of Copper-Oxidized LDLBlood for lipoprotein isolation was collected in EDTA (1 mg/mL)from normal coronary arteries subjects after a 12-hour fast. LDL(density�1.019 to 1.063 g/L) was isolated from plasma after densityadjustment with KBr and preparative ultracentrifugation at 50 000rpm for 22 hours with a type 50 rotor. LDL preparations werewashed by ultracentrifugation, dialyzed against 0.15 mol/L EDTA(pH 7.4), passed through an Acrodisc filter (0.22-�m pore size) toremove aggregates, and stored under N2 in the dark. Copperoxidation of LDL was performed by incubation of postdialyzed LDL(1 mg protein per mL in EDTA-free phosphate-buffered saline[PBS]) with copper sulfate (10 �mol/L) for 24 hours at 37°C.Lipoprotein oxidation was confirmed by analysis of thiobarbituricacid–reactive substances.

Cell Separation and Flow CytometrySplenocytes were costained with the following monoclonal antibod-ies: fluorescein isothiocyanate–labeled anti-CD4 (7D4; MiltenyiBiotec, Bergisch Gladbach, Germany), phycoerythrin-labeled anti-CD25 (GK1.5, Miltenyi Biotec), fluorescein isothiocyanate–labeledmouse IgG2b� isotypic control (KLH/G2b-1-2, Southern Biotech),and phycoerythrin-labeled mouse IgM� isotypic control (RTK2118,BioLegend). For fluorescence-activated cell sorting (FACS) analysisof foxp3 intracellular expression, we used the fluorescein isothio-cyanate anti-mouse/rat foxp3 staining set (clone FJK-16s,e-Bioscience). Stained cells were analyzed on a FACScan flowcytometer with CellQuest software (Becton-Dickinson).

CD4�CD25� T cells were isolated from total splenocytes by a firststep of negative sorting with a cocktail of hapten-conjugated CD8,CD11b, CD16, CD19, CD36, and CD56 antibodies and microbeadscoupled to an anti-hapten monoclonal antibody (CD4� T-cell isola-tion kit; Miltenyi Biotec). This was followed by positive selection ofCD25� cells by microbead separation (CD25 microbeads; MiltenyiBiotech), providing �95% purity as assessed by FACS analysis.

Isolation, Activation, and Transfer of CD4�CD25�

T-Cell SubsetsPurified CD4� cells were isolated from spleens of ApoE-KO mice byfirst staining non-CD4� T cells with a biotin antibody cocktail (10�L/107 total cells), incubation for 10 minutes at 4°C, magneticlabeling with antibiotin microbeads (20 �L/107 total cells), incuba-tion for 15 minutes at 4°C, 2 washes, and depletion over amagnetically activated cell sorting LD column. CD4�CD25� T cellswere directly labeled with anti-CD25 microbeads (10 �L/107 CD4�

cells), incubated for 15 minutes at 4°C, and positively selected withmagnetically activated cell sorting MS columns according to themanufacturer’s instructions (Miltenyi Biotec). The purity of all cellsubsets processed with this method was �95%, as determined byFACS analysis (data not shown). For intravenous injection, 105 cells

were suspended in PBS, and a total volume of 450 �L per mouse wasinjected.

Coculture of T-Effector and Treg CellsNinety-six-well plates (Nunc) were coated with 1 �g/mL anti-CD3monoclonal antibody (17A2, eBioscience) overnight at 4°C andwashed. CD4�CD25� (responder) and CD4�CD25� (suppressor) Tcells (104 cells/well) were cultured in RPMI medium supplementedwith 10% fetal calf serum at different responder-suppressor ratios(1:1, 1:1/2, 1:1/4, and 1:1/8). All cells were cultured in a finalvolume of 200 �L in the presence of irradiated 104 T cell–depletedaccessory cells per well. After 72 hours, [3H]thymidine (1 �Ci/well)was added for 16 hours before proliferation was assayed by scintil-lation counting (beta counter). Percent inhibition of proliferation wasdetermined from the following formula: 1�(median [3H]thymidineuptake of 1:1 CD4�CD25�:CD4�CD25� coculture/median [3H]thy-midine uptake of CD4�CD25� cells).

Western Blot Analysis of Foxp3Western blotting was performed with rat serum anti-foxp3 (poly-clonal antibody, Bioscience) at a dilution of 1:1000 and a secondaryantibody, peroxidase-conjugated AffiniPure donkey anti-rat IgG(H�L, Jackson Laboratories). Actin served as a control protein.

In Vivo Adoptive Transfer Study DesignIn a preliminary study, we infused 4 groups of ApoE-KO mice with104, 105, and 3�105 Tregs and PBS 3 times until sacrifice, 3 monthsafter the initial injection. We found that delivery of 104 or 105 Tregsdid not influence aortic sinus plaque size, whereas administration of3�105 Tregs led to a 30% reduction in plaque size (P�0.08). Wethus selected the latter protocol, assuming that doubling the numberof animals would produce a statistically significant change in lesionformation.

Three-month-old male ApoE-KO mice were transferred intrave-nously through the tail vein with PBS (n�10), 3�105 CD4�CD25�

Tregs (n�9), or 3�105 CD4�CD25� cells (n�10). The sameprotocol for injection was repeated twice more at 3 weekly intervals.Mice were euthanized 3 months after the initial injection. Plasmawas obtained for assessment of lipid profiles, and aortas as well asaortic sinuses were frozen for assessment of atherosclerosis andimmunohistochemistry.

Lipid ProfileTotal plasma cholesterol and triglyceride levels were determinedwith an automated enzymatic technique (Boehringer Mannheim,Mannheim, Germany).

Assessment of Aortic Sinus AtherosclerosisQuantification of atherosclerotic lesions was done by calculating thelesion size in the aortic sinus as previously described.6,7 The heartand upper section of the aorta were removed from the animals, andthe peripheral fat was cleaned away carefully. The upper section wasembedded in OCT compound and frozen. Every other sectionthroughout the aortic sinus (400 �m) was taken for analysis. Sectionswere evaluated for atherosclerosis after being stained with oil red O.

Histochemical Analysis of Aortic Sinus SectionsImmunohistochemical staining was performed with rat anti-CD4(PharMingen). Staining with Masson’s trichrome was used to delin-eate the fibrous area and was determined by quantitativemorphometry.

Sudan IV Staining of Aortic LesionsThe aortas were dissected from the aortic arch to the iliac bifurcationand washed for 1 hour in PBS, pH 7.4, and 0.5 mmol/L EDTA on arotating table.22 The aorta was then fixed with a formal-sucrosesolution (4% paraformaldehyde, 5% sucrose, 20 mmol/L butylatedhydroxytoluene, and 2 mmol/L EDTA, pH 7.4) overnight. Theadventitial fat was trimmed from the aorta while being viewed under

894 Arterioscler Thromb Vasc Biol. April 2007

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

a microscope, opened longitudinally, rinsed briefly in 70% ethanol,immersed for 6 minutes in a filtered solution of Sudan IV (SigmaChemical Co) in 35% ethanol and 50% acetone for 10 minutes, anddestained in 80% ethanol.22 Sudan IV–stained aortas were assessedfor lesion area by morphometry.

Reverse Transcription–Polymerase Chain Reactionfor Aortic Expression of IL-10For reverse transcription—polymerase chain reaction (RT-PCR),aortas from Treg-, CD4�CD25� T cell–, and PBS-treated mice wereanalyzed for expression of mRNA for IL-10. Primers used were5�-CTGGACAACATACTGCTAACCGAC-3� (sense) and 5�-ATTCATTCATGGCCTTGTAGACACC-3� (antisense).

Statistical AnalysisAll parameters were evaluated by 1-way ANOVA. P�0.05 wasconsidered statistically significant. Results are expressed asmean�SEM unless specified otherwise in the text.

ResultsInitially, we tested the hypothesis that the number and functionalproperties of Tregs are compromised in ApoE-KO mice com-pared with those in wild-type C57BL/6 littermates. We found byFACS analysis of spleen cells from 5 mice in each group at 3months that the number of Tregs (CD4�CD25� of the total CD4population) was significantly reduced in ApoE-KO mice (meanof 4.6�0.2%) compared with C57BL/6 mice (8.4�0.4%,P�0.0001; Figure 1A). Lymph node (pooled inguinal andpara-aortic) Tregs were reduced in ApoE-KO mice (mean of3.9�1.3%) in comparison with C57BL/6 mice (5.1�1.4%,P�0.05). Moreover, thymic Tregs were significantly reduced inApoE-KO mice (1.1�0.3%) compared with those in C57BL/6mice (2.4�0.7%, P�0.05).

To further comparatively validate the identity of the Tregsin ApoE-KO and C57BL/6 mice, we performed double

Figure 1. Comparative analysis of the number and function of Tregs from ApoE-KO and C57BL/6 mice. A, Spleen cells were obtainedfrom 3 month-old ApoE-KO and C57BL/6 littermates and stained with antibodies to CD4 and CD25 as outlined in Methods. Data arepresented for 5 mice in each group. B, Staining of splenocytes from ApoE-KO and C57BL/6 mice was performed for detection ofCD25�Foxp3� cells. C, Different ratios of magnetic bead–sorted Tregs from ApoE-KO and C57BL/6 mice were incubated with effectorCD4�CD25� T cells from the latter animals in the presence of irradiated APC. Figure shows 1 representative experiment of 4. Thymi-dine uptake was used for assessment of effector cell proliferative indices. D, The comparative effect of Tregs from ApoE-KO andC57BL/6 mice expressed as percent inhibition of responder T cells. E, Spleen cells from young nonatherosclerotic and older athero-sclerotic mice were assayed for assessment of the number of Tregs (*P�0.001). Results represent data from 5 mice in each group.Representative FACS sheets are provided in E.

Mor et al Regulatory T Cells and Atherosclerosis 895

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

staining for CD25 and foxp3. We found that in ApoE-KOmice, the number of CD25�foxp3� cells was significantlydiminished (2.5�0.1%) compared with those from theirwild-type littermates (3.1�0.2%, P�0.05; Figure 1B). More-over, we observed differences in the functional propertiesbetween both groups. Isolated CD4�CD25� Tregs fromApoE-KO mice were significantly less potent than those fromtheir C57BL/6 littermates in the suppression of effectorCD4�CD25� cells obtained from the latter animals (Figure1C). Thus, tritiated thymidine uptake by CD4�CD25� cellsfrom C57BL/6 mice was significantly higher when they werecocultured in the presence of Tregs from ApoE-KO mice incomparison with Tregs from C57BL/6 mice (at a responder toTreg ratio of 1:1, P�0.01; at a 1:2 ratio, P�0.05; at a 1:4ratio, P�0.01; and at a 1:8 ratio, P�0.01, respectively;probability values indicate the average of all 5 separateexperiments). A representative experiment of the 5 that wereperformed is provided in Figure 1C. No differences wereevident between responder cell proliferation in C57BL/6mice when the assay was conducted in the presence ofactivated protein C (APC) from ApoE-KO or C57BL/6 mice.We found that when CD4�CD25� cells from ApoE-KO micewere used as responders in the same suppressive assay, Tregsfrom ApoE-KO mice were compromised with regard to theirability to inhibit proliferation compared with Tregs fromC57BL/6 mice (a mean reduction of 38.% over all ratios,P�0.01).

To pursue the question of whether the presence of athero-sclerosis in ApoE-KO mice is associated with a change in theTreg pool, we compared their number and function in thesemice at the age of 6 weeks (when mature plaques are not yetevident) with atherosclerotic ApoE-KO mice at an age of 6months (Figure 1C). We found that the number of spleencell–derived Tregs was significantly reduced in atheroscle-rotic (7.2�013%) compared with nonatherosclerotic(9.4�0.2%) ApoE-KO mice (P�0.01).

OxLDL has been suggested as a leading candidate intriggering atherosclerotic plaque formation.1–3 We hypothe-sized that oxLDL, known to trigger lymphocyte apoptosis,would exert a more profound depleting effect on Tregscompared with effector CD4�CD25� cells. Indeed, incuba-tion of CD4� cells derived from the spleen with oxLDL (1�g/mL) resulted in a significant (35%) relative reduction inthe Treg pool (Figure 2, A and B) compared with a nonsig-nificant lowering of the numbers of effector CD4�CD25�

cells (data not shown).Foxp3 is a key transcriptional regulator of Tregs. We

reasoned that the effect of oxLDL on Tregs could be mediatedby the effect on foxp3. We found that oxLDL incubated in thepresence of murine splenocytes led to a time-dependentdownregulation of foxp3 (Figure 2C) compared with noeffect on a control protein (�-actin).

We then investigated whether oxLDL is capable of inter-fering with the suppressor functions of Tregs. For thispurpose, different ratios of Tregs were incubated with effec-tor cells in the presence of irradiated APC. OxLDL led to asignificant reduction in the suppressive properties of Tregs,evident by enhanced proliferation and thymidine uptake ofeffector CD4�CD25� cells. To study whether a differential

response to oxLDL-mediated suppression exists betweenhyperlipidemic ApoE-KO mice and their wild-type litter-mates, we performed a comparative coculture assay in thepresence of oxLDL (Figure 3). We found that the suppressiveproperties of Tregs from ApoE-KO mice were significantlymore hampered (a mean of 80% inhibition of proliferation ata 1:1 suppressor to responder ratio) compared with sortedTregs from C57BL/6 mice (Figure 3). Under these condi-tions, incubation of oxLDL with either Tregs or responder Tcells alone did not change their proliferative capacity asmeasured in (radioactive) counts per minute (data not shown).To prove that cell viability was not compromised in thisassay, representative samples were tested by the trypan blueexclusion test, showing no evident changes.

Aiming to establish the role of Tregs in the progression ofexperimental atherosclerosis, we performed an in vivo adop-tive transfer study. We found that ApoE-KO recipients of

Figure 2. OxLDL downregulates Tregs and foxp3 protein con-tent in murine splenocytes. OxLDL was incubated in 2 concen-trations with spleen cells obtained from ApoE-KO mice. A repre-sentative figure of 4 is shown, and a combined analysis of 4collective separate experiments is displayed in B. P�0.01. C,Protein content of foxp3 by Western blotting compared with acontrol protein (actin) and densitometric analysis performed withthe indicated concentrations of oxLDL.

896 Arterioscler Thromb Vasc Biol. April 2007

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

Tregs exhibited a significantly reduced extent of atheroscle-rosis as evident by smaller aortic sinus plaques comparedwith those in ApoE-KO mice treated with PBS or witheffector CD4�CD25� T cells (Figures 4A and 5). This findingwas validated by aortic en face analysis of atherosclerosissurface coverage area, showing decreased lesions in Treg-transferred ApoE-KO mice compared with effector T cell–and PBS-treated recipients (Figures 4B and 5).

We further explored the nature of plaque development inresponse to the transfer of Tregs. Whereas PBS and Tregtransfer led to a similar distribution of a fibrous cap and lipidcore (mean fibrous areas of 61�2% and 65�3%, respec-tively) as evidenced by Masson’s trichrome staining, infusionof CD4�CD25� T cells resulted in lesions that were moreunstable, exhibiting larger lipid cores and smaller fibrouscaps (mean fibrous area of 44�3%, P�0.005 for both

Figure 3. OxLDL induces a differential inhibitor effect on Tregs from ApoE-KO and C57BL/6 mice. Coculture of Tregs with responderCD4�CD25� cells in the presence of irradiated APC was performed in cells isolated and sorted from 3-month-old mice in the presenceand absence of 1 �g/mL oxLDL. Thymidine incorporation was assayed to evaluate the proliferative properties of the responder T cells.Figure shows a representative experiment from 4 performed. Lower panel shows the differential effect of oxLDL expressed as percentinhibition of responder T cells.

Figure 4. Adoptive transfer of Tregsinfluences plaque size and compositionin ApoE-KO mice. Recipient ApoE-KOmice received 3 injections of Tregs,CD4�CD25� cells, and PBS at 3 weeklyintervals. At sacrifice, 4 months after ini-tial cell transfer, the hearts wereremoved for assessment of aortic sinusatherosclerosis (A), and the aortas werestripped longitudinally for en face evalua-tion of atherosclerotic surface coverageafter Sudan IV staining (B). To study sta-bility indices, morphometric analyseswere performed to measure the frac-tional area occupied by fibrous tissue(C). CD4 infiltration of the plaques wasassayed with the appropriate monoclonalantibody (D). Collective results of A–Drepresent mean�SEM from sections ofmice in each group.

Mor et al Regulatory T Cells and Atherosclerosis 897

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

comparisons; Figures 4C and 5). We then further character-ized the nature of the plaques by staining for CD4� and foundthat Treg and PBS transfer led to a similar extent ofinfiltration of the plaques compared with increased numbersof CD4� cells observed in plaques from effector T cell–transferred mice (Figures 4D and 5). To exclude the possi-bility that Treg transfer was associated with a change in lipidprofile, we performed a plasma metabolic assessment, whichshowed that no differences were evident between the groupswith respect to total cholesterol and triglyceride levels (datanot shown). Importantly, we also analyzed the numbers ofTregs in the spleens of 3 recipient groups and found nodifferences.

IL-10 expression is considered 1 of the mediators ofsuppression, either in naturally occurring or in antigen-specific Tregs (reviewed in O’Garra and Vieira9). We thustested the hypothesis that IL-10 expression within atheroscle-rotic aortas could have been altered by cell transfer. Interest-ingly, we found that aortas from mice that received Tregsexhibited significantly increased mRNA expression of IL-10(online Figure I), attesting to a possible mediating effect onplaque progression. No differences in mRNA expression byRT-PCR were evident between aortas of PBS- and Treg-treated ApoE-KO mice.

DiscussionIn the current study, we investigated the functional role ofCD4�CD25� Tregs in atherosclerosis by using the referenceexperimental model of the ApoE-KO mouse. Atherosclerosisis a progressive disorder in which the role of regulatory Tcells has been suggested by several observations: (1) adoptivetransfer studies of lymphocytes, which have been shown toaccelerate murine atheromas5–7; (2) T-helper 2 cytokines,such as IL-10 that are produced by Tregs, that have beenshown to attenuate lesion formation as opposed to T-helper 1cytokines, which have been demonstrated to enhance lesionformation (reviewed in Hansson,1 Libby,2 and Binder et al3);

and (3) induction of oral tolerance with respective generationof antigen-specific Tregs that has been suggested to attenuateexperimental atherogenesis.20,23,24

Tregs comprise several cell populations. Antigen-specificTregs are operable via secretion of cytokines such as IL-10and transforming growth factor-�, whereas naturally occur-ring Tregs act by cell-to-cell contact but predominantlythrough secretion of transforming growth factor-�.8,9,25–27

Despite the lack of a unifying and acceptable membraneantigen that characterizes Tregs, the most likely molecule onCD4 used for phenotyping is CD25. We therefore assayed thenumber of CD4�CD25� cells in the spleen of atheroscleroticApoE-KO mice and compared them with those of controlC57BL/6 littermates. Indeed, we found that ApoE-KO micehad a significantly reduced number of splenic Tregs. To furtherconfirm the identity of these Tregs, we made a comparativeanalysis of CD25�foxp3� cells in these same mice. We observeda reduction in the number of CD25�foxp3� cells in ApoE-KOmice that mirrored the differences in CD4�CD25� cells.

When compared with younger, nonatherosclerotic littermates,6-month-old ApoE-KO mice exhibited a lower number ofCD4�CD25� Tregs. Collectively, these findings suggest that thedevelopment of atheromatous plaques in the mouse could beassociated with depletion of a peripheral Treg pool, although nocause-and-effect relation could be inferred. We then comparedthe functional suppressive properties of Tregs from atheroscle-rotic ApoE-KO mice with those in C57BL/6 littermates. Inter-estingly, we found that the suppressive effect of Tregs on theproliferative capacity of the effector CD4�CD25� populationwas significantly hampered in ApoE-KO mice compared withC57BL/6 mice. The inhibitory action of Tregs has been shownto be associated with their ability to suppress experimentalautoimmune disorders,28 and their transfer could correct therespective syndromes.29,30 Thus, we hypothesize that Treg dys-regulation in the atherosclerotic mouse could promote lesionprogression.

Figure 5. Atherosclerosis in recipients ofregulatory and responder T cells. Repre-sentative photomicrographs from aorticsinus sections of Treg- (A, D, G), PBS-(B, E, H), and CD4�CD25� (C, F, I) trans-ferred ApoE-KO mice stained with Mas-son’s trichrome (A, B, C; magnification�50), CD4 (D, E, F; magnification �200;arrows indicate representative cells) andoil red O (G, H, I; magnification �50).

898 Arterioscler Thromb Vasc Biol. April 2007

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

There are sufficient data to support the presence and roleof oxLDL in the development of the atheroscleroticplaque.31 We sought to explore the influence of oxLDL oneffector and Treg relative numbers in mice. The relativepercentage of Tregs of the total CD4 population wassignificantly reduced by incubation with oxLDL comparedwith a nonsignificant depleting effect on CD4�CD25�

cells. Moreover, the compromising effect of oxLDL on thesuppressive effects of Tregs from atheroscleroticApoE-KO mice appeared to be more robust than in theirC57BL/6 littermates. The current study design, however,did not allow us to rule out the possibility that ApoEdeficiency by itself was causally associated with thedifferential effect of oxLDL. This hypothesis could betested by performing the assays in vitro in the presence orabsence of ApoE or its derivatives.

Unanswered questions remain with regard to the cause ofdepletion and functional compromise of Tregs in ApoE-KOmice. Because some degree of LDL oxidation has been notedin the plasma of subjects with hyperlipidemia32 and may alsobe evident in hypercholesterolemic mice, we speculate thatthe Treg pool may contact modified LDL in the circulation,not just within the atheroma. The effects of oxLDL to whichthese Tregs were exposed could well have been maintainedduring the 3-day culture of the functional suppressive assays.

Foxp3 has been shown to govern the development andfunction of Tregs, and evidence supports its presence in bothCD25� Tregs and CD25� cells with regulatory activity.33

Loss-of-function mutations of foxp3 eliminate CD25� Tregsand result in autoimmune disorders in both humans andmice.34,35 We thus explored whether the suppressive functionof oxLDL was associated with its effect on foxp3. We foundthat oxLDL triggered a time-dependent downregulatory ef-fect on the expression of foxp3. Collectively, these findingssuggest that the oxLDL that is present within the atheromaand possibly in minor concentrations in serum could becausally associated with the compromised population ofTregs in the atherosclerotic mouse.

To pursue a cause-and-effect relation between Tregs andatherosclerosis, we performed adoptive transfer assays, as hasbeen done in models of experimental autoimmune encepha-lomyelitis and diabetes.29,30 We found that transfer of Tregsresulted in a significant reduction in atherosclerotic lesionformation compared with the delivery of either PBS oreffector CD4�CD25� cells. This finding was validated byseparately measuring plaque size in the aortic sinus and theentire longitudinally stripped aortas, with essentially similarfindings. Transfer of CD4�CD25� did not significantly influ-ence plaque formation when compared with PBS. It should betaken into consideration that we infused a total of 9�105

Tregs and effector T cells divided into 3 injections, yet therelative number of Tregs accounts for a significantly higherproportion of the peripheral population in the mouse, as Tregsrepresent only 2% to 5% of total lymphocytes. Thus, wecannot exclude the possibility that infusion of effector T cellsin larger numbers would have promoted murine atherogene-sis, as may be inferred by previous observations.5–7 Bystudying the nature of the plaques induced after cell transfer,we found that delivery of effector CD4�CD25� cells resulted

in lesions that were more unstable, exhibiting a smallerfibrous area and a higher number of inflammatory cellscompared with the situation in Treg- or PBS-injected mice.

The exact mechanisms mediating the suppressive proper-ties of naturally occurring Tregs have not yet been elucidated.As mentioned, cell-to-cell contact could be a mechanism,supported by our in vitro coculture experiments. However,there is also evidence to suggest that a population of naturallyoccurring Tregs are IL-10 producers.9 This would be consis-tent with an antiatherogenic effect, as IL-10 is causallyassociated with attenuation of experimental atherogenesis(reviewed in Hansson,1 Libby,2 and Binder et al3). We foundby RT-PCR that expression of mRNA for IL-10 in aortas ofTreg-transferred ApoE-KO mice was significantly enhanced.In view of the strength of the evidence in favor of anantiatherogenic role for IL-10 in experimental atherogenesis,we reason that this is 1 of the plausible mechanisms for theplaque-attenuating effect of Treg transfer. We also analyzedthe Treg pool in the 3 recipient groups and found nodifferences in their numbers. However, despite similar num-bers in the 3 groups, a systemic elevation of Tregs as apossible protective mechanism could not be ruled out, be-cause analysis was performed several weeks after the finaldelivery of Tregs.

It was recently observed by Ait-Oufella et al20 that Tregshave a protective role in atherogenesis by successfully block-ing CD25 monoclonal antibodies in the ApoE-KO mousemodel. Our results provide complementary data that furthersupport the potential role of Tregs in the pathogenesis ofatherosclerosis in the mouse. These findings are also consis-tent with our recent report,36 showing that patients with acutecoronary syndromes exhibit a reduced number and functionalcompromise of their peripheral Tregs.

In conclusion, we found that atherosclerotic mice exhibit acompromised pool of naturally occurring Tregs and thattransfer of these CD4�CD25� Tregs significantly reducesplaque progression in the ApoE-KO mouse model. Thesefindings suggest a novel role for naturally occurring Tregs inatherogenesis and open new potential therapeutic avenues.

Sources of FundingThis study was supported in part by a grant from the Israeli Sciencefoundation (to J.G.; grant No. 832/06) and the Chief Scientist ofIsrael (to J.G.).

DisclosuresNone.

References1. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease.

N Engl J Med. 2005;352:1685–1695.2. Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874.3. Binder CJ, Chang MK, Shaw PX, Miller YI, Hartvigsen K, Dewan A,

Witztum JL. Innate and acquired immunity in atherogenesis. Nat Med.2002;8:1218–1226.

4. Zhou X, Stemme S, Hansson GK. Evidence for a local immune responsein atherosclerosis: CD� T cells infiltrate lesions of apoprotein-E miceAm J Pathol. 1996;149:359–367.

5. Zhou X, Nicoletti A, Elhage R, Hansson GK. Transfer of CD4� T cellsaggravates atherosclerosis in immunodeficient apolipoprotein E knockoutmice. Circulation. 2000;102:2919–2922.

Mor et al Regulatory T Cells and Atherosclerosis 899

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

6. George J, Harats D, Gilburd B, Afek A, Shaish A, Levy Y, Kopolovic J,Shoenfeld Y. Adoptive transfer of �2-glycoprotein I reactive lym-phocytes promotes early atherosclerosis in LDL-receptor deficient mice.Circulation. 2000;102:1822–1827.

7. George J, Afek A, Gilburd B, Shoenfeld Y, Harats D. Cellular andhumoral immune responses to heat shock protein 65 are both involved inpromoting fatty-streak formation in LDL-receptor deficient mice. J AmColl Cardiol. 2001;38:900–905.

8. Sakaguchi S. Naturally arising CD4� regulatory T cells for immunologicself-tolerance and negative control of immune responses. Annu RevImmunol. 2004;22:531–562.

9. O’Garra A, Vieira P. Regulatory T cells and mechanisms of immunesystem control. Nat Med. 2004;10:801–805.

10. Lindley S, Davan CS, Bishop A, Roep BO, Peakman M, Tree TI.Defective suppressor function in CD4�CD25� T-cells from patients withtype 1 diabetes. Diabetes. 2005;54:92–99.

11. Cao D, Malmstrom V, Baecher-Allan C, Hafler D, Klareskog L, TrollmoC. Isolation and functional characterization of regulatory CD25brightCD4�

T cells from the target organ of patients with rheumatoid arthritis. EurJ Immunol. 2003;33:215–223.

12. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of functionalsuppression by CD4�CD25� regulatory T cells in patients with multiplesclerosis. J Exp Med. 2004;199:971–979.

13. Chen Y, Kuchroo VK, Inobe I, Hafler D, Weiner HL. Regulatory T cellclones induced by oral tolerance: suppression of autoimmune encephalo-myelitis. Science. 1994;265:1237–1240.

14. Hafler DA, Kent SC, Pietrusewicz MJ, Khoury SJ, Weiner HL, FukauraH. Oral administration of myelin induces antigen-specific TGF-�1secreting T cells in patients with multiple sclerosis. Ann N Y Acad Sci.1997;835:120–131.

15. Levings MK, Sangregorio R, Roncarolo MG. Human CD25�CD4� Tregulatory cells suppress naive and memory T cell proliferation and canbe expanded in vitro without loss of function. J Exp Med. 2001;193:1295–1302.

16. Baecher-Allan C, Brown JA, Freeman GJ, Hafler DA. CD4�CD25high

regulatory cells in human peripheral blood. J Immunol. 2001;167:1245–1253.

17. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the devel-opment and function of CD4�CD25� regulatory T cells. Nat Immunol.2003;4:330–336.

18. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell developmentby the transcription factor Foxp3. Science. 2003;299:1057–1061.

19. Gambineri E, Torgerson TR, Ochs HD. TI-immune dysregulation, poly-endocrinopathy, enteropathy, and X-linked inheritance (IPEX), asyndrome of systemic autoimmunity caused by mutations of FOXP3, acritical regulator of T-cell homeostasis. Curr Opin Rheumatol. 2003;15:430–435.

20. Ait-Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, ZollJ, Merval R, Esposito B, Cohen JL, Fisson S, Flavell RA, Hansson GK,Klatzmann D, Tedgui A, Mallat Z. Natural regulatory T cells control thedevelopment of atherosclerosis in mice. Nat Med. 2006;12:178–180.

21. Plump AS, Smith JD, Hayek T, Aalto-Setala K, Walsh A, Verstuyft JG,Rubin EM, Breslow JL. Severe hypercholesterolemia and atherosclerosisin apolipoprotein-E deficient mice created by homologous recombinationin ES cells. Cell. 1992;71:343–353.

22. George J, Afek A, Shaish A, Levkovitz H, Bloom N, Cyrus T, Zhao L,Funk CD, Sigal E, Harats D. 12/15-Lipoxygenase gene disruptionattenuates atherogenesis in LDL receptor-deficient mice. Circulation.2001;104:1646–1650.

23. Maron R, Sukhova G, Faria AM, Hoffman E, Mach F, Libby P, WeinerHL. Mucosal administration of heat shock protein-65 decreases athero-sclerosis and inflammation in aortic arch of low-density lipoproteinreceptor-deficient mice. Circulation. 2002;106:1708–1715.

24. Harats D, Yacov N, Gilburd, Shoenfeld Y, George J. Oral tolerance withheat shock protein 65 attenuates Mycobacterium tuberculosis-inducedand high-fat-diet-driven atherosclerotic lesions. J Am Coll Cardiol. 2002;40:1333–1338.

25. George J, Yacov N, Breitbart E, Bangio L, Shaish A, Gilburd B,Shoenfeld Y, Harats D. Suppression of early atherosclerosis in LDL-receptor deficient mice by oral tolerance with �2-glycoprotein I. Car-diovasc Res. 2004;62:603–609.

26. Piccirillo CA, Thornton AM. Cornerstone of peripheral tolerance: nat-urally occurring CD4�CD25� regulatory T cells. Trends Immunol. 2004;25:374–380.

27. Baecher-Allan C, Viglietta V, Hafler DA. Human CD4�CD25� regu-latory T cells. Semin Immunol. 2004;16:89–98.

28. Alyanakian MA, You S, Damotte D, Gouarin C, Esling A, Garcia C,Havouis S, Chatenoud L, Bach JF. Diversity of regulatory CD4� T cellscontrolling distinct organ-specific autoimmune diseases. Proc Natl AcadSci U S A. 2003;100:15806–15811.

29. Tang Q, Henriksen KJ, Bi M, Finger EB, Szot G, Ye J, Masteller EL,McDevitt H, Bonyhadi M, Bluestone JA. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med.2004;199:1455–1465.

30. McGeachy MJ, Stephens LA, Anderton SM. Natural recovery and pro-tection from autoimmune encephalomyelitis: contribution ofCD4�CD25� regulatory cells within the central nervous system.J Immunol. 2005;175:3025–3032.

31. Miller YI, Chang MK, Binder CJ, Shaw PX, Witztum JL. Oxidized lowdensity lipoprotein and innate immune receptors. Curr Opin Lipidol.2003;14:437–445.

32. Holvet P, Harris P, Tracy R, Verhamme P, Newman A, Rubin S,Simonsick E, Colbert L, Kritchvesky S. Association of high coronaryheart disease risk status with circulating oxidized LDL in the well-functioning elderly: findings from the Health, Aging, and Body Compo-sition study. Arterioscler Thromb Vasc Biol. 2003;23:1444–1448.

33. Ochs HD, Ziegler SF, Torgerson TR. FOXP3 acts as a rheostat of theimmune response. Immunol Rev. 2005;203:156–164.

34. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, WhitesellL, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregu-lation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) iscaused by mutations of FOXP3. Nat Genet. 2001;27:20–21.

35. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, YasaykoSA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F. Disruption of a newforkhead/winged-helix protein, scurfin, results in the fatal lymphoprolif-erative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73.

36. Mor A, Luboshits G, Planer D, Keren G, Geoge J. Altered status ofCD4�CD25� regulatory T cells in patients with acute coronary syn-dromes. Eur Heart J. 2006;27:2530–2537.

900 Arterioscler Thromb Vasc Biol. April 2007

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from

Fig. I

Fig. I: Assessment of mRNA expression of IL-10.

Upon sacrifice, 5 aortas were obtained from Treg, CD4+CD25

- cells and PBS injected

mice and studied by RT-PCR for the expression of mRNA for IL-10. Bars display

mean±SEM of densitometric expression of IL-10 in each group.* p<0.05.

0

50

100

150

200

250

PBS CD4CD25- CD4CD25+rela

tive a

mo

un

t o

f IL

10 m

RN

A

Keren and Jacob GeorgeAdi Mor, David Planer, Galia Luboshits, Arnon Afek, Shula Metzger, Tova Chajek-Shaul, Gad

Atherosclerosis Regulatory T Cells in Experimental+CD25+Role of Naturally Occurring CD4

Print ISSN: 1079-5642. Online ISSN: 1524-4636 Copyright © 2007 American Heart Association, Inc. All rights reserved.

Greenville Avenue, Dallas, TX 75231is published by the American Heart Association, 7272Arteriosclerosis, Thrombosis, and Vascular Biology

doi: 10.1161/01.ATV.0000259365.31469.892007;27:893-900; originally published online February 1, 2007;Arterioscler Thromb Vasc Biol. 

http://atvb.ahajournals.org/content/27/4/893World Wide Web at:

The online version of this article, along with updated information and services, is located on the

http://atvb.ahajournals.org/content/suppl/2007/02/05/01.ATV.0000259365.31469.89.DC1.htmlData Supplement (unedited) at:

  http://atvb.ahajournals.org//subscriptions/

at: is onlineArteriosclerosis, Thrombosis, and Vascular Biology Information about subscribing to Subscriptions:

  http://www.lww.com/reprints

Information about reprints can be found online at: Reprints: 

document. Question and AnswerPermissions and Rightspage under Services. Further information about this process is available in the

which permission is being requested is located, click Request Permissions in the middle column of the WebCopyright Clearance Center, not the Editorial Office. Once the online version of the published article for

can be obtained via RightsLink, a service of theArteriosclerosis, Thrombosis, and Vascular Biologyin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

by guest on August 8, 2015http://atvb.ahajournals.org/Downloaded from