Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response

Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response

ARTICLE IN PRESS G Model IMLET 5317 1–13 Immunology Letters xxx (2013) xxx–xxx Contents lists available at SciVerse ScienceDirect Immunology Lette...

572KB Sizes 0 Downloads 69 Views

ARTICLE IN PRESS

G Model IMLET 5317 1–13

Immunology Letters xxx (2013) xxx–xxx

Contents lists available at SciVerse ScienceDirect

Immunology Letters journal homepage: www.elsevier.com/locate/immlet

Review

1

Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response

2 3

4

Q1

Dimitry A. Chistiakov a,b,∗ , Igor A. Sobenin b,c,d , Alexander N. Orekhov b,c

5

a

6

b

7

c

8

Pirogov Russian State Medical University, Department of Medical Nanobiotechnology, Ulitsa Ostrovityanova 1, 117997 Moscow, Russia Institute for Atherosclerosis, Skolkovo Innovative Center, Ulutsa Ivana Franko 4/1, 121355 Moscow, Russia Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Baltiyskaya Ulitsa 8, 125315 Moscow, Russia d Russian Cardiology Research and Production Complex, 3rd Cherepkovskaya Ulitsa, 121552 Moscow, Russia

9

10

a r t i c l e

i n f o

a b s t r a c t

11 12 13 14 15

Article history: Received 20 October 2012 Received in revised form 28 January 2013 Accepted 31 January 2013 Available online xxx

16 17 18 19 20 21 22

Keywords: Atherosclerosis Inflammation Regulatory T cells Dendritic cells Immunomodulation

Atherosclerosis is a chronic inflammatory disease, in which multiple types of immune cells are involved. Th1 and Th17 cells play a prominent role in induction and progression of local inflammation in the atherosclerotic plaque. Regulatory T cells (Tregs) can be also found in the plaque but their numbers are decreased and function may be impaired. Tregs are the master modulators of the immune system possessing the immunosuppressive capacity to prevent unfavorable immune responses and maintain tolerance to self-antigens. These cells play the atheroprotective role by inhibiting Th1/Th17-mediated proinflammatory response and down-regulating the antigen-presenting function of dendritic cells (DCs). Tregs mediate the immune response through the cell-to-cell contacts and secretion of anti-inflammatory cytokines IL-10 and TNF-beta. In addition to the natural CD4+ CD25+ Foxp3+ Tregs presented in the thymus, there are several subtypes of inducible Tregs that can be induced from naïve CD4+ T cells by tolerogenic DCs in the periphery. Thus, stimulation of the immunosuppressive activity of Tregs and increasing numbers of Tregs and immunocompetent DCs has a great clinical potential in prevention and treatment of atherosclerosis and its vascular complications. A promising strategy to induce the anti-atherogenic immune response is an oral administration of anti-inflammatory immunomodulators capable to activate the intestine immune tolerance by recruiting mucosal tolerogenic DCs and inducing Tregs. Induced Tregs can then migrate to the inflamed vascular sites and reduce atherogenesis. © 2013 Published by Elsevier B.V.

Abbreviations: LDL, low density lipoprotein; Tregs, regulatory T cells; IL, interleukin; TGF-␤, transforming growth factor beta; Foxp3, forkhead box 3; IL-2R, interleukin-2 receptor; CTLA-4, cytotoxic T-lymphocyte antigen 4; NK, natural killer; DCs, dendritic cells; TCR, T-cell receptor; Tr1, Tregs type 1; Th, T helper cell; Bcl-xL, B-cell lymphoma-extra large; ROR␥t, RAR-related orphan receptor gamma; apoE, apolipoprotein E; LDLR, LDL receptor; oxLDL, oxidized LDL; Scid, severe combined immunodeficiency syndrome; CCL, C-C motif chemokine ligand; IFN, interferon; VSMC, vascular smooth muscle cell; HSP, heat shock protein; IDO, indoleamine 2,3-dioxigenase; PDL-1, programmed death ligand 1; LFA1, lymphocyte function-associated antigen 1; GM-CSF, granulocyte-macrophage colony-stimulating factor; TNF-␣, tumor necrosis factor alpha; CCR, C-C motif chemokine receptor; ECM, extracellular matrix; Ig, immunoglobulin; Rag2, recombination activating gene 2; ICOS, inducible costimulator; LAP, latency-associated peptide; S1P, lysosphingolipid sphingosine 1-phosphate; STAT6, signal transducer and activator of transcription 6; COX, cyclooxygenase; NSAID, non-steroid anti-inflammatory drug; DMARD, disease-modifying anti-rheumatic drug; MTX, methotrexate; RA, rheumatoid arthritis. ∗ Corresponding author at: Department of Medical Nanobiotechnology Pirogov Russian State Medical University, Ulitsa Ostrovityanova 1, 117997 Moscow, Russia. Tel.: +7 495 434 13 01; fax: +7 495 434 14 22. E-mail address: [email protected] (D.A. Chistiakov).

1. Introduction

23

Although low-density lipoprotein (LDL) remains the most important risk factor for atherosclerosis, immune and inflammatory mechanisms play significant and non-redundant role in atherogenesis. The presence of leukocytes within atherosclerotic arteries was discovered in the late 1970s [1]. Multiple leukocyte types were then reported in the atherosclerotic plaque [2]. Regulatory T cells (Tregs) were also found in the atherosclerotic plaque. Several subsets of Tregs, which are responsible for maintenance of immunological tolerance and suppressing immune overactivity of effector T cells [3], diminish atherosclerosis development by down-regulation of activated T cell responses [4–6]. These Tregs subsets secrete two major anti-inflammatory cytokines, interleukin-10 (IL-10) and transforming growth factor (TGF-␤) capable to reduce proatherogenic inflammatory response in atheroslerosis [7]. Indeed, the balance between effector T cells and Tregs is sufficient to control of atheroslerosis development and progression. In this review, we characterize Tregs subtypes, their induction, function and role in atheroslerosis. We also consider approaches

24

0165-2478/$ – see front matter © 2013 Published by Elsevier B.V. http://dx.doi.org/10.1016/j.imlet.2013.01.014

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

2 43 44 45

aimed to restore/stimulate Tregs function in atherosclerotic patients and improve dysbalance between Tregs and effector T cells by activating an endogenous immune response.

46

2. Tregs subsets and their function

47

2.1. Natural CD4+ CD25+ Foxp3+ Tregs

81

There are several subpopulations of Tregs including naturally occurring CD4+ CD25+ Foxp3+ Tregs and inducible Tregs subsets [3]. Natural CD4+ CD25+ Tregs, which are produced in the thymus, are the key players in suppressing self-reactive immune responses and mantaining dominant self-tolerance [8]. They constitutevely express CD25, an ␣-subunit of the trimeric IL-2 receptor, at high levels thereby producing the complete high-affinity IL2R␣␤␥ complex that is capable to respond to physiologically low concentrations of IL-2 in vivo. This cytokine is critical in the generation and maintainance of CD4+ CD25+ Tregs [9]. In CD4+ CD25+ Tregs, IL-2 induces expression of Foxp3, a transcription factor that is crucial for their development and function [10] and is currently the most reliable marker for them [11,12]. Foxp3 acts to control the core module of Tregs suppressive function by regulating expression of a number of key molecules such as CTLA-4 and CD25 [13]. Natural CD4+ CD25+ Foxp3+ Tregs can suppress a variety of immune cells including CD4+ and CD8+ T cells, B cells, natural killer (NK) cells, NKT cells, monocytes, and dendritic cells (DCs) (Fig. 1). A main function of natural CD4+ CD25+ Tregs is suppression of activation of naïve T cells, but they can also inhibit activated effector T cells and memory CD4+ and CD8+ cells [14]. Natural CD4+ CD25+ Foxp3+ Tregs inhibit immune responses through cell-to-cell contact by suppressing T-cell receptor (TCR)induced proliferation and IL-2 transcription in target T cells [15] or CTLA-4-mediated down-regulation of CD80/CD86 expression in DCs [16]. In addition, natural CD4+ CD25+ Foxp3+ Tregs can exhibit in vitro potent granzyme B-dependent, partially perforinindependent cytotoxic cells that are capable of specifically killing antigen-presenting B cells [17]. Upon CD3/CD46 activation, Grossman et al. [18] also reported the ability of human natural CD4+ CD25+ Foxp3+ Tregs to express the serine protease granzyme A and kill CD4+ T cells and other target cells in a perforin-dependent manner.

82

2.2. Tregs type 1

83

A subpopulation of inducible Tregs called Tregs type 1 (Tr1) was first described by Groux et al. [19] who induced these cells from T-cell receptor–transgenic mice by repeated stimulation of naïve T cells with ovalbumin and IL-10. This CD4+ T cell subset exhibits a unique cytokine profile distinct from that of T helper (Th)0, Th1, or Th2 cells. These Tr1 cells primarily produce IL-10 and TGF-␤ and some IL-5 and interferon-␥ (IFN-␥) with little or no IL-2 or IL-4 and proliferate poorly after polyclonal T-cell receptormediated activation. Tr1 cells express markers LAG3, CD49b, ICOS, PD-1, and LAP [20]. Functional studies on Tr1 cells have indicated that Tr1 cells have immunosuppressive properties and have been shown to prevent the development of Th1-mediated autoimmune diseases [19]. Compared to natural CD4+ CD25+ Foxp3+ Tregs, Tr1 cells do not express Foxp3 [21]. IL-10-producing Tr1 cells are capable to suppress a variety of immune cells including DCs and Th17 cells [22]. Treg-derived IL-10 is important for control of inflammation at environmental interfaces but seems to be dispensable for control of systemic autoimmunity [23]. Along that line, IL-10- or IL-10 receptor-deficient mice do not develop autoimmunity, but are susceptible to colitis in the presence of triggering flora [24].

48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80

84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101

2.3. Th3 cells

102

Th3 cells are another subset of inducible Tregs. These cells, which are primary producers of TGF-␤, may be induced on periphery by TGF-␤. Th3 induction may be enhanced by IL-4 and IL-10 [25]. Carrier et al. [26] developed TGF-␤-transgenic mice in which TGF-␤ was linked to the IL-2 promoter and T cells transiently overexpressed TGF-␤ upon TCR stimulation but produce little or no IL-2, IL-4, IL-10, IL-13, or IFN-␥. Th3 cells derived from these mice were capable to induce Foxp3 expression in both CD25+ and CD25− T cell populations [26] and rescue IL-2-deficient mice from autoimmunity due to the induction of CD25− Tregs in the periphery [27]. Indeed, Th3 cells may play a critical role in inducing and maintaining peripheral tolerance by driving differentiation of Foxp3+ Tregs in the periphery by secretion TGF-␤. TGF-␤-derived Foxp3+ CD25+/− Th3 Tregs represent a different cell lineage from thymic-derived CD25+ Tregs in the periphery. TGF-␤-deficient mice develop T cell-mediated autoimmunity within several weeks after birth [28]. A similar phenotype is observed in mice lacking TGF-␤ responsiveness specifically in T cells [28]. These mice showed enhanced Th1 and Th2 responses and immunopathology including colitis; however, these mice were also resistant to the induction of experimental autoimmune encephalitis likely due to impaired Th17 induction [29]. Indeed, the role of TGF-␤ in Treg-mediated suppression might depend very much on the type of effector cell and the site of the immune response, and TGF-␤ may even promote proinflammatory Th17 responses. Tregs can produce high amounts of membrane-bound and soluble TGF-␤, and blocking TGF-␤ partially abrogated suppression of T cell proliferation in vitro suggesting that Treg-produced TGF-␤ controls autoimmunity [30]. Treg-produced TGF-␤ can induce apoptosis of lymphoid cells including self-reactive effector T cells through cleavage of Bcl-xL with activated caspase-1- like protease [31]. Activated CD4+ T cells induced by DCs are particularly sensitive to TGF-␤ [32].

103

2.4. Foxp3+ Tregs

135

Compared to natural CD4+ CD25+ Foxp3+ Tregs, which arise from the thymus and are released into peripheral tissues after thymic-positive selection, inducible Foxp3+ Tregs are generated via peripheral conversion (after antigen-specific stimulation) from mature, naïve CD4+ T cells or from “rescued” self-reactive effector T lymphocytes [33]. Foxp3+ Tregs can be induced in periphery from CD4+ CD25− naïve T cells by IL-2 and TGF-␤ [34,35]. In synergy with TGF-␤, retinoic acid generated in functionally specialized mucosal DCs can induce Foxp3 expression in CD4+ naïve T cells [36]. Compared to natural CD4+ CD25+ Foxp3+ Tregs whose CpG island within the Foxp3 locus is demethylated, it was reported to be methylated in TGF-␤-induced Foxp3+ Tregs [37]. It seems that the demethylation status is a prerequisite for stable Foxp3 expression and suppressive activity. As a consequence, methylation profile of the Foxp3 promoter would facilitate the distinction of truly committed Tregs [38]. In contrast to natural CD4+ CD25+ Foxp3+ Tregs, TGF-␤-induced Foxp3+ Tregs rapidly lose both Foxp3 expression and suppression activity [39]. IL-4-producing Th2 cells seem to be a major cause for the disappearance of Foxp3+ Tregs during long culture. IL-4 is an important suppressor of Foxp3 induction, which down-regulates Foxp3 expression in a STAT6-dependent manner [39]. IL-4 activates the Th2 transcription factor Gata3 that blocks Foxp3 transcription by direct binding to the FoxP3 promoter [40]. Foxp3 expression is efficiently suppressed also by transcription factor PU.1, which is transiently induced during Th2 differentiation [41]. In addition to its effect on Tregs, TGF-␤ also induces the differentiation of Th17 cells in the presence of a pro-inflammatory cytokine, IL-6 [42]. In a sharp contrast to Tregs, which actively

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134

136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

3

Fig. 1. Interactions between denritic cells (DCs) and T cells. DCs may present possible atherosclerosis self-antigens (possibly derived from heat shock protein-60 and oxidized low density lipoprotein) to naïve T cells in the context of costimulatory molecules like CD40, OX40L, CD80, and CD86 eliciting T cell differentiation and proliferation. Although a T helper (Th)1-biased response is documented in atherosclerosis, there is also a significant body of evidence suggesting a possible role for Th2 cells in mature lesions. Recently discovered Th17 also play proinflammatory and proatherogenic role. Regulatory T cells (Tregs) that could be induced from naïve T cells by tolerogenic DCs are able to suppress function of all types of effector T cells and immunostimulatory properties of mature DCs. Treg cells have antiatherogenic effects and play a protective role against atherosclerosis, mainly by inhibiting proinflammatory Th1 and Th17 cells trough cell-to-cell contacts and secreting anti-inflammatory cytokines interleukin (IL)-10 and transforming growth factor (TGF)-␤.

165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196

suppress immune responses, Th17 cells are involved in promoting autoimmune and inflammatory responses. Intriguingly, retinoic acid inhibits Th17 differentiation [42] by reducing the expression of ROR␥t, a master transcription factor for Th17 cells [43]. TGF-␤-induced Foxp3+ Tregs suppress Th17 differentiation by antagonizing ROR␥t function [44]. Interestingly, Foxp3+ Tregs producing IL-17 were found in humans [45]. Upon T-cell receptor stimulation in the presence of IL-1␤, IL-2, IL-21, and IL-23, peripheral CD4+ Fox3+ Tregs can differentiate to IL-17 producers. IL-17-secreting Tregs express Foxp3 and ROR␥t transcription factors. The biological significance of T cells that display the function of Treg and the opposing function of Th17 is unclear. In normal conditions, these cells can potentially contribute to the antimicrobial innate immune defense while controlling inflammation and autoimmunity at the same time, particularly at mucosal sites. IL-17-producing Tregs could be implicated in controling Th17/Treg balance, which is altered in cardiovascular pathology including acute coronary syndrome [46] and atheroslerosis [47]. In the presence of IL-6 alone without exogenous addition of TGF␤, natural CD4+ CD25+ Foxp3+ Tregs can differentiate themselves into IL-17 producers [48,49]. In contrast, TGF-␤ and IL-2-induced Foxp3+ Tregs are resistant to IL-6-dependent conversion to Th17 cells [50]. Thus, natural and TGF-␤-induced Foxp3+ Tregs are not mirror images of each other. These two subsets differ in their principal antigen specificities and in the T-cell receptor signal strength and co-stimulatory requirements needed for their generation [51]. Indeed, natural Foxp3+ Tregs may have undesirable effects through their ability to differentiate into pathogenic Th17 in the presence of IL-6 and/or IL-23 at sites of inflammation. Induced Foxp3+ Tregs thereby might retain suppressive function at inflammatory sites.

3. Protective role of Tregs in cardiovascular pathology

197

3.1. Interactions between Tregs and DCs in atherosclerosis

198

Two rodent models of atherosclerosis, apolipoprotein (apo)Edeficient and LDL receptor (LDLR)-deficient mice, were widely used to assess molecular mechanisms of atherogenesis including T-cell mediated proinflammatory responses. Monocytes migrating through the intima from the early atherosclerosis stages may differentiate into macrophages or DCs affected by inflammatory cytokines and chemokines. DCs and DC precursors may also migrate from bloodstream and accumulate in the plaque on chemokines and the adhesion molecule-dependent pathway [52]. Significantly increased levels of DCs were found in atherosclerotic lesions of apoE-deficient mice [53]. DCs are a heterogeneous population of bone marrow-derived immune cells that specialize in capturing, processing and presenting antigens to T lymphocytes in order to induce and control immunity. DCs, after entering the vascular tissue, screen the environment for potential antibodies. In atherosclerotic plaque, immature DCs can capture and process self-antigens derived from dying cells or from neutrophil extracellular traps such as heat shock proteins (HSP)60/65 [54], self-antigenic protein-DNA complexes [55], and others. Oxidized LDL (oxLDL) contribute to induction of a proinflammatory cytokine profile in human DCs leading to DCmaturation and -differentiation [56]. After capturing an antigen, DCs differentiate into mature DCs capable to induce activation and differentiation naïve T cells towards the proinflammatory phenotype [57,58]. Tregs contribute to the maturation, activation, and function of DCs [52]. Tregs inhibit the antigen-presenting function of proatherogenic DCs through CTLA-4-dependent suppression of

199

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226

G Model IMLET 5317 1–13 4 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

their CD80 or CD86 expression [59]. CTLA-4, an important coinhibitory molecule, is consitutively expressed in natural CD4+ CD25+ Tregs. CTLA-4 binds to B7 family molecules such as B7-1 (CD80) and B7-2 (CD86) abundantly expressed on the surface of macrophages and DCs and conveys a negative signal to DCs reducing expression of both costimulatory molecules [60]. By interaction with CD80 or CD86, Treg-derived CTLA-4 may induce indoleamine 2,3-dioxygenase (IDO) expression on DC. This enzyme converts tryptophane to kynurenine, a potent immunosuppresive metabolite capable to induce de novo formation of Tregs from naïve T cells in the local environment [61]. Through cell-to-cell contacts with mature DCs, Tregs may also inhibit the antigen-presenting function via the mechanism of trogocytosis (e.g., transfer of membrane fragments and cell surface proteins between cells) of CD80 and CD86 by Tregs [62]. This process, which is mediated by CTLA-4, CD28, and programmed death ligand-1 (PDL-1), includes acquisition of CD80 and CD86 from the surface of DCs and leads to enhanced ability of Tregs to suppress naïve CD4+ T-cell proliferation. CTLA-4 is a key factor in the downregulation of costimulatory molecules CD80 and CD86 expressed on antigen-expressing mature DCs, since Tregs from CTLA-4-deficient mice were not able to inhibit CD80/CD86 expression [63]. Indeed, CTLA-4 engagement of CD80 and CD86 on DCs present a critical mechanism of tolerance induction in vivo [64]. Thus, CTLA-4 can be considered as a potential therapeutic target for atheroslerotic disease in the development of strategies aimed to enhance Treg-mediated suppression function or increase downregulation of effector T cell function [65]. Lymphocyte function-associated antigen 1 (LFA-1), a T-cell adhesion molecule (CD11a/CD18), is necessary for cell-to-cell contact between a Treg and an antigen-presenting DC [59]. Tregs exert the CD80/86-down-regulating effect even in the presence of strong DC-maturating stimuli such as granulocyte-macrophage colonystimulating factor (GM-CSF), tumor necrosis factor (TNF)-␣, IFN-␥, type I IFN, and lipopolysaccharide. Indeed, Tregs may successfully prevent proinflammatory stimuli from antigen-presenting DCs to naïve T cells by binding to immature DCs with subsequent LFA1/CTLA-4-dependent suppression of CD80/CD86 expression in DCs. DCs can in turn regulate Tregs by their suppression or induction. Weber et al. [66] reported a proatherogenic population of CCL17-produced monocyte-derived CD11c+ DCs, which were able to supress Tregs. Recently, Choi et al. [67] found in aortic tissue a subtype of monocyte-derived DCs (CD11chigh MHCIIhigh CD11b− CD103− ) playing the atheroprotective role and capable to induce Tregs in the atheroslerotic plaque. Tolerogenic DCs that basically have immature phenotype (CD86− CD80− CD40− MHCII− ) may induce Tregs through cell-tocell contact with naïve T cells and by secreting cytokines such TGF-␤ and IL-10. In the presence of IL-10, peripheral Tregs differentiate into Tregs, and IL-10 production by tolerogenic DCs contributes to the induction of IL-10-producing Tr1. DCs and Tregs can also interact via chemokines CCL17 and CCL22 and their receptors, chemokine receptor (CCR) 4 and 8 [68]. These receptors are expressed on Tregs and may bind their ligands CCL17 and CCL22 secreted by DCs. Enchancement of CCL22 on tolerogenic DCs may induce and recruit Tregs at inflammatory sites such as ateroclerotic plaque. Stimulatory effect of CCL22/CCR4 on induction of Tregs may be mediated by IDO, since mice deficient for CCR4 exhibited markedly reduced expression of this enzyme in mesenteric lymph nodal DCs [69]. These findings indicate that reciprocal interactions between the DCs and Tregs via both B7/CTLA-4 and CCL22/CCR4 mediated by the immunoregulatory enzyme IDO may play a key role in the pathogenesis of inflammatory disease including the induction of inflammation in atherosclerosis. Indeed, induction of tolerogenic DCs in order to enhance or improve atheroprotective

effects of Tregs may be considered as a plausible therapeutic strategy for preventing atherosclerosis.

293

3.2. Influence of Tregs on Th1/Th2 immune responses in atheroslerosis

295

The majority of pathogenic T cells in atherosclerosis are of the Th1 profile producing high levels of IFN-␥ [70]. IFN-␥ displays pleiotropic proatherogenic effects including activation of monocytes/macrophages and DCs, inhibiting vascular muscle cells proliferation, down-regulating extracellular matrix (ECM) collagen production and up-regulating matrix metalloproteinases [71]. The proatherogenic activity of IFN-␥ was clearly shown in apoEdeficient mice deficient for this cytokine or its receptor [72] and by exogenous administration of IFN-␥ to the apoE-deficient mice [73]. The proatherogenic role of IL-12 and IL-18 was demonstrated in the apoE-deficient atherosclerosis mouse model [74]. IL-12 produced by DCs, monocytes and macrophages, is crucial for differentiation of naïve T cells into Th1 cells by stimulating IFN-␥ production and down-regulating Th2 type cytokines (IL-4 and IL-5) in T cells. In synergy with IL-12, IL-18 up-regulates expression of IFN-␥ [75]. Th2 cells secrete IL-4, IL-5, IL-10, and IL-13 and provide help for antibody production by B cells. Th2 cells are rarely detected within the atherosclerotic lesions. However, in hyperlipidemic conditions, Th1/Th2 balance may be switched towards Th2-bias [76]. The role of the Th2 pathway in the development of atherosclerosis seems to be multifaceted depending on the stage and/or site of the lesion, as well as on the experimental model [77]. In atheroslerosisresistant mice strains such as C57BL/6 and BALB/c fed on fat diet, IL-4-mediated Th2-bias has been shown to protect against early fatty streak development [78]. In more permissive model such as apoE- or LDLR-deficient mice, IL-4 shows the proatherogenic role [79] that could be even more exacerbated by prolonged hypercholesterolemia [80]. As shown in hypercholesterolemic apoE−/− mice immunized with oxLDL, other Th2-type cytokines such as IL5 and IL-33 exert antiatherogenic function through the induction of humoral immunity associated with the production of high levels of IgM-type anti-oxLDL antibodies [81,82]. Along with Th1/Th2 switch, hypercholesterolemia may also stimulate Th3 immune effector responses by inducing TGF-␤ levels, increasing amounts of TGF-␤1+ CD4+ Th3 regulatory cells in lesions, and elevating Th3dependent IgG2b antibodies to oxLDL [83]. As mentioned above, natural CD4+ CD25+ Foxp3+ Tregs are able to supress a variety of immune cells including Th1 and Th2 lymphocytes. Ait-Oufella et al. [84] showed that endogenous natural CD4+ CD25+ Foxp3+ Tregs may be powerful inhibitors of atheroslerosis in LDLR-deficient mice. Interaction between the costimulatory molecules CD80/CD86 and CD28 is known to be crucial for the generation and homeostasis of Tregs [85]. Using the irradiation/bone marrow transplantation model, Ait-Oufella et al. [84] reported that Treg deficiency due to reconstitution with CD80−/− /CD86−/− and CD28−/− bone marrow led to enhanced atheroslerotic plaque development in mice. Transplantation of CD28−/− splenocytes to apoE−/− Rag2−/− mice deficient for lymphocytes resulted in accelareted atheroslerosis compared to the transplantation of normal splenocytes. Adoptive transfer of natural CD4+ CD25+ Foxp3+ Tregs greatly reduced atheroslerosis in apoEdeficient mice [86]. However, these studies did not provide precise evidence for the atheroprotective role of natural CD4+ CD25+ Foxp3+ Tregs since deficiency for CD80−/− /CD86−/− and CD28−/− may influence function of not only Tregs but also other effector T cells. A protective role of Tr1 cells against atheroslerosis via suppression of Th1-mediated inflammation was first showed in apoE-deficient mice co-immunized with ovalbumin in complete Freund’s adjuvant and Tr1 cells [4]. The immunization with Tr1

297

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

294

296

298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

384

cells resulted in significant reduction in atherosclerotic lesion size, decrease in the production of IFN-␥, and increase in IL-10 production. Overexpression of IL-10 by activated T lymphocytes reduced atherosclerosis in LDLR−/− mice likely due to the anti-inflammatory effect of Tr1-like cells [87]. The anti-atherogenic role of Tr1 cells was further supported by Ait-Oufella and co-authors [88] who promoted endogenous adaptive Tr1 cell response in apoE-deficient mice by treatment with measles virus nucleoprotein, a vaccine with anti-inflammatory properties. The Tr1-mediated response markedly inhibited progression of atherosclerosis and accumulation of T lymphocytes and macrophages in lesions. Interestingly, Tregs were shown to inhibit formation of new plaques on the early atheroslerosis stages whereas their influence on pre-existing lesions is limited by reducing plaque size and inducing a ‘stable’ plaque phenotype [89]. Plaque stabilization shown in atheroslerosis-prone murine strains, is mediated by CD40–CD40L interactions [90]. The CD40–CD40L dyad is known to interact with other co-stimulatory molecules, to activate antigen-presenting cells and to contribute to T-cell priming and B-cell isotype switching. Disrupting the CD40-CD40L co-stimulatory pathway reduces atherosclerosis and induces a stable atherosclerotic plaque phenotype that is low in inflammation and high in fibrosis [91]. Since CD40-CD40L interactions directly promote development of proatherogenic Th1 cells, Tregs-mediated suppression of these cells should decrease CD40–CD40L interactions within the plaque and in turn stabilize the plaque. Inhibition of CD40 expression on effector T cells, DCs and B cells may increase Tregs population and suppress a humoral immune response in atherosclerosis [92].

385

3.3. Th17/Treg balance

386

Th17 cells, a recently identified Th cell population, express a lineage-specific factor RORC (ROR␥t in mice) and represent a distinct lineage of CD4+ T cells [93]. TGF-␤ and inflammatory cytokines such as IL-6, IL-21, IL-1␤, and IL-23 are required in the generation of Th17 cells. Th17 cells play a sharp proinflammatory role by secretion of inflammatory cytokine IL-17 [94]. In the immune response, Th17 cells are critical for the clearance of extracellular pathogens including Candida and Klebsiella [95]. However, under certain conditions, Th17 cells become a leader in orchestrating inflammatory and autoimmune pathology. By producing two IL-17 isoforms (IL17A and IL-17F) and other effector molecules such as IL-21, IL-22, GM-CSF, and CCL20, Th17 cells are known to contribute to the pathogenesis of several inflammatory and autoimmune diseases [96]. The pathogenic role of Th17 cells in atherosclerosis was demonstrated in ApoE-deficient mice [47,97,98]. In atherosclerosis and atherosclerotic vascular complications such as acute coronary syndrome, the inflammatory response is accompanied with significant increase in Th17 cell population and elevated production of IL-17 [99,100], which is correlated with the severity and progression of vascular disease [101]. IL-17 is involved in cardiovascular pathology by inducing the mitochondrial-dependent apoptosis of vascular endothelial cells [102] and cooperating with IFN-␥ in the induction of proinflammatory responses in vascular smooth muscle cells (VSMCs) [103]. The mechanism by which Th17-derived IL17A promotes activation of VSMCS may involve NAD(P)H-oxidase dependent generation of reactive oxygen species [104] that in turn induces a proinflammatory signaling [105]. In atherosclerotic disease, the vascular ECM is subjected to the inflammation-driven degradation and remodeling, with induction of collagen II, collagen IV, malondialdehyde-modified laminin, and fragments of other ECM proteins as self-antigens [106]. IL-17 plays a primary role in mediating cellular autoimmunity against ECM structural components in atherosclerotic arteries of both humans and mice [107,108]. In addition, Th17-derived IL-17 promotes activation and

357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383

387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420

5

expansion of IL-17A/IL17-F producing neutrophils [99] and IL-17Esecreting B cells [109] in advanced and complicated plaques further enhancing atherosclerotic progression. Assessment of the role of IL-17A and its receptor IL-17A in apoE-deficient mice showed that the IL-17A/IL-17RA axis increases aortic arch inflammation during atherogenesis through the induction of aortic chemokines, and the acceleration of neutrophil and monocyte recruitment to this site [110]. Blockage of IL-17A with anti-IL-17A antibodies attenuated development of atherosclerosis in apoE-deficient mice [111,112]. Altogether, these findings clearly suggest for the proatherogenic role of Th17 and IL-17. Studies in patients with coronary atherosclerosis reported Th17/Treg functional imbalance associated with significant increase in peripheral Th17 number, Th17-related cytokines (IL-17, IL-6, and IL-23) and transcription factor (ROR␥t) levels and obvious decrease in Treg number, Treg-related cytokines (IL-10 and TGF-␤1) and transcription factor (Foxp3) levels [46,113]. Indeed, restoring the Th17/Treg balance may be a promising therapeutic approach in the treatment of atherosclerosis and particularly atherosclerotic complications such as acute coronary syndrome and chronic heart failure. A possible way to improve the Th17/Treg bias is to stimulate TGF-␤, which is known to orchestrate differentiation of both Th17 and Tregs in a concentration-dependent manner [114]. At low concentrations, TGF-␤ synergizes with IL-6 and IL-21 to promote IL-23 receptor expression favoring Th17 cell differentiation. High concentrations of TGF-␤ repress IL23R expression and favor Foxp3+ Treg cells. ROR␥t and Foxp3 are co-expressed in naïve CD4+ T cells exposed to TGF-␤ and in a subset of T cells found in the murine small intestinal lamina propria [44]. TGF-␤-induced Foxp3 inhibits ROR␥t function such as ROR␥t-induced expression of IL-17A mRNA through their interaction and thereby induces polarization into inducible Tregs [115]. Along with the subset of IL-17 producing inducible peripheral Foxp3+ Tregs, a subpopulation of IL-17-producing natural CD4+ CD25+ Foxp3+ Tregs was recently discovered [116]. These cells express high amounts of inducible costimulator (ICOS) and have a unique cytokine secretion profile (IL-10, IL-17, IL-35, TGF-␤, and IFN-␥) distinguishable from all other FoxP3+ Tregs. In vitro, ICOS+ Tregs can be induced by 2,4-dinitrofluorobenzene immunization of cultured CD4+ CD25+ Tregs. These cell share features of Th17/Th1 cells and Tregs. ICOS+ Tregs were shown to possess a highly suppressive activity to CD8+ T-cell effector responses in a haptenspecific manner in vivo [116]. By secreting IL-35, ICOS-positive Tregs were reported to inhibit Th17-mediated IL-17 production and suppress airway hyperresponsiveness in the mouse model of allergic airways disease [117]. The population of ICOS+ Tregs was found to be decreased in patients with myocardial infarction [118]. Since ICOS+ Tregs have capacity to down-regulate Th17mediated proinflammatory responses, this Tregs subset is likely to have the atheroprotective role. Immunosuppressive properties of IL-17-producing inducible and natural Tregs should be further assessed in order to explore their promising therapeutic potential in the future.

421

4. Approaches to enhance Treg-mediated immune regulation in atheroslerosis

475

Studies in animal models showed the significance of T cell co-stimulatory and co-inhibitory pathways in the regulation of inflammation in atherosclerosis. These pathways are critical in the control of function of naïve and effector T cells and in the generation and function of T regs. Due to the proinflammatory microenvironment, Tregs function may be markedly down-regulated or impaired and Tregs numbers may be reduced in atheroslerosis [119]. Indeed,

477

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474

476

478 479 480 481 482 483

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

6

486

strategies focused on the improvement and/or enhancement of Tregs function and increasing their amount could be considered as a novel therapeutic approach against atherosclerosis.

487

4.1. Induction of mucosal tolerance

488

A promising strategy to induce anti-inflammatory effector T cells and Tregs is the induction of oral tolerance, e.g. the specific suppression of cellular and/or humoral immune reactivity to an antigen by prior administration of the antigen by the oral (nasal) route [120]. Antigen-specific immunomodulation by vaccination is an attractive approach to prevent or treat chronic inflammatory diseases including atheroslerosis. By mobilizing protective immune responses in an antigen-specific manner, side effects due to hampered host defense against infections may be avoided. The mucosal tolerance (with antiatherogenic consequences) may be induced by a variety of agents such as monoclonal antibodies and immunogenic peptides listed in Table 1. To induce an antigen-specific response, investigators used self-antigens or peptides derived from self-antigens usually presented in the atheroslerotic plaque such as oxLDL [121], apoB [122,123], HSP60/65 [124–126], and beta 2-glycoprotein I [127]. Conjugation of a peptide to the subunit B of cholera toxin promotes uptake of an antigen via the nasal and oral mucosa and induction of protective immunity [128]. Natural human LDL is definded as a population of lipoproteins that form lipoprotein particles of 18–28 nm in diameter and can be isolated by ultracentrifugation within a density range of 1.019–1.063 kg/L [129]. In the central lipophilic core, an LDL particle contains up to 170 triglycerides and up to 1600 cholesteryl esther molecules. The core is surrounded by a monolayer comprising ∼700 phospholipid molecules and 600 molecules of free cholesterol. A large molecule of apolipoprotein B-100 (apoB-100; 4356 amino acids) is embedded to the outer layer. A total of 2700 molecules of free fatty acids are distributed in the LDL particle, and a half of those are polyunsaturated fatty acids (PUFAs) [130]. Compared to other lipoprotein classes, LDL are enriched (up to 50%) with cholesterol, and increased plasma concentrations of LDL-cholesterol are widely recognized as an established cardiovascular risk factor. Reaching LDL-cholesterol level of less than 100 mg/dL is recommended for lipid-lowering therapy to prevent a high risk of a cardiovascular disease [131]. Oxidation of LDL accumulated in the arterial wall is occurred on early stages of atherosclerosis. There are many possible mechanisms of LDL oxidation including enzymatic action of membrane-associated NAD(P)H oxidase of activated neutrophils, monocytes, and macrophages and 15-lipoxygenase [132] and influence of various reactive oxygen species and radicals such as superoxide anion [133], thiyl radicals and superoxide [134], peroxynitrite [135], and by-products of the myeloperoxidase reaction (hypochlorous acid and tyrosyl radical) [136]. Transition metal ions such as Cu2+ and Fe3+ capable to catalyze LDL oxidation may be used for oxLDL generation in cell-free systems. LDL oxidized by a cellfree system is physiochemically and biologically indistinguishable from LDL oxidized by a cellular system [137]. Oxidation of LDL is a free radical-mediated process resulting in numerous structural changes, all of which depend on a common initiating event, the peroxidation of PUFAs in LDL. Due to the presence of conjugated double bonds, PUFAs are very sensitive to oxidation and could be subsequently oxidized to peroxy radicals and hydroperoxides [129]. Cholesterol in LDL can be oxidized to oxysterols such as 7-ketocholesterol [138]. In LDL oxidation, the propagation phase is followed by a decomposition or degradation phase, during which there is cleavage of double bonds resulting in the formation of aldehydes. The major aldehydes produced include malondialdehyde (MDA), malondialdehyde-acetaldehyde (MAA),

484 485

489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547

4-hydroxynonenal, and hexanal, which can cross-link with amino groups on apoB-l00 [139,140]. Changes in the protein moiety also occur during the oxidation of LDL. After oxidation, there is an increase in the negative charge on the LDL particle, possibly due to the derivatization of positively charged amino groups through the formation of a Schiff base with aldehydes. Also, after oxidation, apo B-100 undergoes oxidative scission leading to fragmentation [141]. MAA-modified proteins were found in the atherosclerotic plaque. MAA-adducted proteins are involved in the inflammatory reaction that occurs in atherosclerosis [142]. Different classes of modified LDL were shown to differ in their capacity to induce oral tolerance in animal models of atheroslerosis. If the oral administration of oxLDL was able to induce the oral tolerance in LDLR-deficient mice, MDA-LDL did not [117]. The atheroprotective effect of oral administration of oxLDL may be partially explained by the ability of oxLDL to induce tolerogenic DCs that could in turn induce Tregs and inhibit proinflammatory responces [143]. The difference between oxLDL and MDA-LDL in inducing oral tolerance may result from the binding complement anaphylatoxin C3a to MAA-LDL and MDA-LDL [144]. MAA- and MDA-LDL and proteins containing MDA adducts or MAA adducts may bind C3a in vivo and contribute to inflammatory processes involving activation of the complement system in atherosclerosis [142].

548

4.2. Induction of Tregs by anti-CD3 antibody

573

Monoclonal antibodies such as anti-IL-2 [145] or anti-CD3 antibodies [6] suitable for induction of mucosal tolerance could possess immunomodulatory properties. The IL-2/anti-IL-2 antibody complex formed with IL-2 and anti-IL-2 neutralizing monoclonal antibody is able to selectively stimulate natural CD4+ CD25+ Foxp3+ Tregs and greatly attenuate progression of atherosclerosis in apoEdeficient mice [145]. Preclinical studies including nasal or oral administration of anti-CD3 antibodies showed their high efficacy in induction of Tregs and suppression of autoimmune and inflammatory pathology in mice [146,147]. Based on the results achieved in animal experiments, a humanized anti-CD3 monoclonal antibody was orally administered to healthy human individuals [148]. At present, this antibody (otelixizumab) is clinically tested at Phase III for the prevention of type 1 diabetes [149]. The therapy with this antibody is typically safe and biologically active since it induces many favorable immunologic effects such as suppressed Th1 and Th17 responses, induced immunosuppressive Tregs, increased production of anti-inflammatory cytokines (IL-10 and TGF-␤), and decreased DC-mediated proinflammatory cytokine (IL-6 and IL-23) secretion [82]. Indeed, oral administration of anti-CD3 antibody may be considered as a promising strategy to induce Tregs-mediated anti-inflammatory response in order to reduce atheroslerosis development and progression. In atherosclerosis mice, oral administration of anti-CD3 antibody resulted in obvious beneficial effects including reduced plaque development when administered before a high-cholesterol diet and markedly decreased lesion progression in animals with already established atherosclerosis [150]. Treatment with antiCD3 antibody was shown to induce CD4+ CD25+/− Foxp3+ Tregs (Th3) and CD4+ CD25− LAP+ Tregs, likely in gut-associated lymphoid tissues [151]. The latter Tregs subclass expresses surface latencyassociated peptide (LAP), the amino-terminal domain of the TGF-␤ precursor peptide, which remains noncovalently associated with the TGF-␤ peptide after cleavage forming the latent TGF-␤ complex [152]. It seems that CD4+ CD25− LAP+ Tregs may prevent atherosclerosis [6] but have a limited capacity to attenuate the established

574

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572

575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

7

Table 1 Examples of therapeutics used for stimulation of anti-inflammatory immunosuppressive function of Tregs and increasing their population in atherosclerotic vascular disease. Agent and treatment regiment

Experimental model

Tregs population

Outcome

Reference

Statin (simvastatin): 50 g/kg daily for 6 weeks

ApoE-deficient mice

Foxp3+ Tregs

[162]

ApoB-CTB fusion protein (peptide p210 3136–3155 fused with the cholera toxin B subunit): 15 ␮g intranasally twice a week for 12 weeks HSP60; HSP60 peptide 253–268: orally 4 times in 8 days before starting fat-rich diet

ApoE-deficient mice

Tr1

Increase in Tregs numbers in plaques; 2–2.5-fold increase in expression of Foxp3, IL-10, and TGF-␤ mRNA in plaques; 1.5–2-fold increase in protein levels of Foxp3, IL-10, and TGF-␤ in plaques; 2.5–3.5-fold decrease in circulating blood levels of IFN-␥ and IL-17 Reduction in aortic lesion size by 35%; 2-fold increase in amounts of IL-10- producing Tr1 cells; 11-fold increase in expression of IL-10 mRNA in plaques

LDLR-deficient mice

CD4+ CD25+ Foxp3+ Tregs

oxLDL: 100 ␮g by intraperitoneal injection before starting fat-rich diet

LDLR-deficient mice

CD4+ CD25+ Foxp3+ Tregs

FTY720 (sphingosine-1-phosphate receptor antagonist): 5 mg/kg orally to mice at 4 weeks of age fed on high-cholesterol diet Anti-CD3 monoclonal antibody: 5 ␮g intragastrically daily for 5 days

ApoE-deficient mice

Foxp3+ Tregs

ApoE-deficient mice

CD25+ Foxp3+ Tregs

621

atherosclerosis [145]. However, up-regulated secretion of TGF-␤ by CD4+ CD25− LAP+ Tregs may induce other TGF-␤-dependent Tregs subsets in other lympoid organs. In anti-CD3-treated mice, a significant increase in amounts of induced Foxp3+ Tregs was observed in mesenteric lymph nodes [6] and spleen [151]. Induced Tregs may migrate from lymphoid organs to atherosclerotic plaques and dampen local immune responses resulting in the attenuation of atherosclerotic lesion formation in already developed atherosclerosis. Alternately, CD4+ CD25+/− Foxp3+ Tregs may be directly induced by anti-CD3 antibody [153].

622

4.3. Induction of tolerogenic DCs by 1,25-dihydroxyvitamin D3

623

Tolerogenic DCs may be induced by oral administration of vitamin D, or its hormonally active form calcitriol (1,25dihydroxyvitamin D3 ). This hormone plays a central role in calcium metabolism and bone formation but also exerts immunomodulatory (immunosuppressive) function. Calcitriol exhibits antiinflammatory properties by inhibiting Th17 cell differentiation through the down-regulation of IL-6 and IL-23 [154] and inhibiting differentiation and maturation of DCs [155]. Therefore, calcitrioltreated immature DCs became tolerogenic and can promote Foxp3+ Tregs, Tr1, and Th3 cells via production of IL-10 and TGF-␤ [156]. Finally, 1,25-dihydroxyvitamin D3 can directly modulate differentiation of Foxp3+ Tregs [157,158]. In a recent double-blind, placebo-controlled trial, Bock et al. [159] showed that intake of calcitriol at high dose (140,000 IU monthly) for 3 months significantly increased the frequency of Tregs in healthy volunteers. The atheroprotective effect of 1,25-dihydroxyvitamin D3 was demonstrated in apoE-deficient mice treated daily with 200 ng of calcitriol for 12 weeks [160]. Atheroslerotic lesion formation was significantly attenuated, with decreasing monocyte/macrophage accumulation in lesions and reducing numbers of mature

612 613 614 615 616 617 618 619 620

624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642

[122]

Reduction in aortic lesion size by 27%; reduction in lesion size in the carotid arteries by 80%; 1.3–1.5-fold increase in amounts of CD4+ CD25+ Foxp3+ Tregs in blood, spleen, and mesenteric lymph nodes by day 14; 2–3-fold increase in production of TGF-␤ and IL-10 in plaques by day 14 Reduction in initiation rate of atherosclerosis by 30–71%; Reduction in progression rate of atherosclerosis by 45%; 1.8-fold increase in amounts of CD4+ CD25+ Foxp3+ Tregs in spleen, and mesenteric lymph nodes by day 14; 3.2-fold increase in production of TGF-␤ in plaques by day 14 Reduction in aortic lesion size by 40% (4 weeks post-administration); 1.6–2.3-fold increase in amounts of Foxp3+ Tregs and LAP+ Tregs; 4.5-fold increase in production of TGF-␤ in plaques

[126]

Reduction in aortic lesion size by 33% (10 weeks post-administration); 1.2-fold increase in amounts of CD25+ Foxp3+ Tregs in spleen and mesenteric lymph nodes; 2.2-fold increase in production of TGF-␤ in plaques 4-fold decrease in production of IFN-␥ in plaques

[6]

[121]

[170]

CD11c+ CD86+ DCs in the mesenteric lymph nodes, spleen, and intestine. In the plaque, Foxp3+ Tregs counts were elevated and their immunosuppressive capacity was increased in association with stimulated IL-10 production and inhibited IL-12 expression. Calcitriol increased mRNA levels of CCL22 and its receptor CCR4 in DCs suggesting that tolerogenic DCs release Tregs-attracting chemokines and draw Tregs to bind to CCR4 on the surface of DCs [161].

643 644 645 646 647 648 649 650

4.4. Statins

651

Tregs and DCs can be quantitatively and qualitatively modulated by small molecule drugs possessing immunomodulatory features. For example, statins (lipid-lowering agents) exert pleiotropic effects on atherosclerosis including the modulation of inflammatory responses. In atorvastatin-treated apoE-deficient mice, population of Foxp3+ Tregs was increased and Tregs +Th2/Th1 + Th17 balance was improved [162]. A short-term (4week) therapy with atorvastatin at daily dose of 20 mg significantly inhibited the immunostimulatory function of DCs in patients with abdominal aortic aneurism [162]. In hyperlipidemic subjects treated with simvastatin or pravastatin, numbers of natural CD4+ CD25+ Foxp3+ Tregs were increased [163]. Atorvastatin was shown to be capable to induce CD4+ CD25+ Foxp3+ Tregs from human peripheral CD4+ CD25− Foxp3− T cells and enhance their functional suppressive properties. A stimulatory effect of atorvastatin on induction of Tregs, Foxp3 expression, and TGF-␤ production was observed in patients with ST-segment elevation myocardial infarction [164]. Simvastatin promoted an accumulation of Tregs, subsequently decreased Th1 and Th17 response and modulated the Th1/Th2 balance toward a Th2 phenotype in the atherosclerotic plaques,

652

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

8

674

and this result may be an additional mechanism of simvastatin in stabilizing vulnerable plaques [165].

675

4.5. Lysosphingolipid sphingosine 1-phosphate analogs

676

699

The biologically active lysosphingolipid sphingosine 1phosphate (S1P) is an important lipid mediator generated from phospholipids on cell activation [166]. A large body of evidence has documented the pleiotropic effects of S1P in various cell types and organs including the immune system. S1P was demonstrated to interfere with proliferation, migration, and cytokine secretion by lymphocytes and to prevent their recirculation from lymphoid compartments to peripheral sites of antigen presentation [167]. FTY720, a synthetic S1P analog, possesses immunosuppressory properties via inhibiting T lymphocyte proliferation, downregulating secretion of proinflammatory cytokines IL-6, IFN-␥, and TNF-␣2, and stimulating the anti-inflammatory path of activation of macrophages towards the M2 phenotype [168]. FTY720 was shown to inhibit atheroslerosis in LDLR-null [168] and apoEdeficient mice [169]. Oral administration of this sphingolipid analog to apoE-deficient mice was shown to result in significant stimulation of CD4+ CD25− LAP+ and CD4+ CD25+/− Foxp3+ Tregs and suppression of Th1 immune response [170]. Recently, FTY720 (Fingolimod) was approved as a new therapeutic drug for multiple sclerosis in more than 50 countries [171]. Hopefully, due to the marked capacity of Fingolimod to inhibit the early development of atherosclerosis in mice via induction of a Tregs response and inhibition of effector T responses, this sphingolipid analog will be clinically tested soon in patients with coronary artery disease.

700

4.6. Steroids

701

Glucocorticoids such as hydrocortisone and their synthetic analogs such as dexamethasone, methylprednisolone, and pregnisone possess potent immunosuppressive and anti-inflammatory properties. Treatment with intranasal steroid was shown to inhibit nasal inflammation and induce increased amounts of Foxp3+ Tregs and anti-inflammatory cytokine (IL-10) in nasal polyps [172]. Methylprednisolone treatment resulted in significant increase in a population of highly suppressive DRhigh+ CD45RA− Tregs in transplanted patients [173]. Similarly, dexamethasonetreated monocytes differentiate into tolerogenic IL-10-producing DCs capable to induce IL-10-producing Tregs and restore immune tolerance [174,175]. In contrast, aldosterone, a mineralocorticoid hormone, may support inflammation, fibrosis, and vascular remodeling, e.g. proatherogenic pathogenic mechanisms. As a component of the renin–angiotensin–aldosterone system, aldosterone at high concentrations may contribute to the vascular damage through the induction of hypertension, oxidative stress, and vascular inflammation [176]. Human VSMCs exposed to aldosterone present an increase in type I and III collagen, IL-16, and CTLA-4 expression, molecules associated with fibrosis, inflammation, and vascular calcification [177]. Adoptive transfer of natural CD4+ CD25+ Foxp3+ Tregs leads to suppression of aortic and renal vascular inflammation, reduction of macrophage and T-cell infiltration into the vascular wall and other improvements in mice with aldosteronemediated vascular injury [178]. These findings may suggest for potential beneficial effects of treatment of coronary artery disease with glucocorticoids. However, these hormones are usually used in therapy of severe proinflammatory diseases such as eczema, psoriasis, asthma, and arthritis or as immunosuppressive agents in transplantation due to the systemic inhibiting effect on the immune system. Glucocorticoids are unlikely to be efficient for treatment of chronic diseases with persisting low-dose inflammation such as atherosclerosis

673

677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698

702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734

since long-term administration of these steroids significantly increases risk of side effects including weight gain, mood swings, sleep disruptions, dizziness, glaucoma, lowering bone denstiry, and an overall weakened immune system [179]. In addition, treatment of apoE-deficient mice with dexamethasone was found to increase hyperlipidemia through mechanisms related to the increase in the synthesis of triglyceride-rich lipoproteins [180].

735

4.7. Non-steroid anti-inflammatory drugs (NSAIDs)

742

Most of NSAIDs are non-selective inhibitors of both forms of cyclooxygenase (COX), COX-1 and COX-2, an enzyme involved in the biosynthesis of prostanoids. The pharmacological inhibition of COX provides relief from the symptoms of inflammation and pain. Aspirin, a broadly used NSAID, may mediate immune regulation in a wider manner including mechanisms, which are not limited by the only COX inhibition. For example, aspirin is able to induce tolerogenic DCs from immature DCs through the suppression of NF␬B-dependent signaling, reduced expression of co-stimulatory molecules (CD80, CD86, ICOS), and induction of immunoglobulinlike transcript-3, a B-cell immune receptor preventing activation of B lymphocytes [181]. Indeed, tolerogenic DCs can then induce differentiation of naïve T cells to CD4+ CD25− Foxp3+ Tregs and support expansion of natural CD4+ CD25+ Foxp3+ Tregs [182]. New aspirin derivatives such as ASC14 (a hydrogen sulfide-releasing aspirin) may possess advanced anti-inflammatory properties in preventing atherosclerosis and thrombosis via several pathways including suppression of a fraktalkine-dependent macrophage activation and migration [183], blocking platelet aggregation, and down-regulating fibrinogen receptor activation [184]. Aspirin continues therefore to be of interest due to its wide effects on immune regulation including the capacity to induce tolerogenic DCs at therapeutic concentrations in humans. Other NSAID classes may also possess anti-atherosclerotic properties through COX and acetyl-CoA carboxylase inhibition, lipid-lowering effects, antioxidant activity, and suppressing inflammation [185,186]. Compared to non-specific COX inhibitors, coxibs (rofecoxib, celecoxib, and valdecoxib) that are selectively block COX-2 activity are advantageous in therapy of arthritis and other painful inflammatory syndromes but become associated with increased risk myocardial infarction and stroke as shown in many studies. COX-2-specific inhibitors enhance Th1 type immune response associated with increased production of inflammatory cytokines, unstable plaque phenotype, and acute atherosclerosis complications [187]. By disrupting the physiological balance between thromboxane and prostacyclin, coxibs increase atherosclerosis, thrombogenesis, and the risk of cardiovascular complications. In addition, through uncoupling mitochondrial oxidative phosphorylation, coxibs may induce production of reactive oxygen species and increase cardiovascular risk [188]. COX-2-specific inhibitors suppress IDO production and Tregs, and this may be beneficial for disruption of tumor-induced tolerance but not for prevention of atherogenesis [189].

743

4.8. Disease-modifying anti-rheumatic drugs (DMARDs)

786

DMARDs is a category of unrelated drugs defined due to their ability to dampen the development of rheumatoid arthritis (RA) and other autoimmune proinflammatory diseases. These include tetracycline antibiotics (doxycycline, minocycline), DNA synthesis blockers (methotrexate, azathioprine, leflunomide), TNF-␣-specific monoclonal antibodies (ifliximab, adalimumab, golimumab), non-antibody inhibitors of IL-1 and TNF-␣ (chloroquine, hydroxycloroquine, sulfasalazine) and some other agents. In many studies, methotrexate (MTX), a folate metabolism inhibitor, showed a potent atheroprotective effect by stimulating reverse

787

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

736 737 738 739 740 741

744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785

788 789 790 791 792 793 794 795 796

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828

cholesterol transport, cholesterol oxidation [190], down-regulation of proinflammatory genes (TNF-␣, IL-1␤, CXCL-2, VAP-1, etc.), and up-regulation of anti-inflammatory cytokine TNF-␤1 [191]. Compared to other DMARDs such as sulfasalazine and leflunomide, MTX did not inhibit suppressive function of CD4+ CD25+ Tregs and Foxp3 expression [192]. In a murine model of collageninduced arthritis, co-treatment with MTX and etanercept resulted in marked suppression of proinflammatory T cell subsets and their production of proinflammatory cytokines and restoring natural CD4+ CD25+ Foxp3+ Tregs numbers in the thymus and spleen [193].TNF-␣ is a pleiotropic cytokine, which can have proinflammatory or immunosuppressive effects depending on the context, duration of exposure and disease state. In concert with IL-2, TNF-␣ selectively activates Tregs resulting in proliferation, up-regulation of FoxP3 expression and increases in their suppressive activity [194]. However, in rheumatoid arthritis and other proinflammatory environments, this cytokine down-regulates the function of both natural CD4+ CD25+ Tregs and TGF␤1-induced CD4+ CD25+ T regs [195]. Accordingly, specific inhibition of TNF-␣ with monoclonal antibodies restores Tregs immunosuppressive function and increases numbers of natural CD4+ CD25+ Foxp3+ Tregs in patients with autoimmune and inflammatory pathology [196–198]. The higher mortality rate among rheumatoid arthritis patients in comparison with the general population is largely attributable to cardiovascular disease, particularly coronary atherosclerosis. Overall, the immunosuppressive capacity of TNF-␣ inhibitory antibodies, MTX, and other DMARDs has beneficial effects on the cardiovascular system of patients affected with rheumatoid arthritis [199–201]. Longer exposure to low doses of corticosteroids increases cardiovascular risk while treatment with anti-TNF agents, despite some inconsistency, delays and even reverses the progression of endothelial dysfunction and atherosclerosis [199].

829

5. Conclusion

830

In addition to Tregs, tolerogenic DCs have a critical role in the regulation of T cell response in atherosclerosis. Modulation of intestinal immune system by inducing both Tregs and tolerogenic DCs might be a therapeutic approach for the prevention of atherosclerosis. Oral immune modulation by drugs and therapeutic agents possessing immunoregulatory activities is the simplest way to induce gut-associated immune tolerance. Because the intestinal immune system must protect our body from invading pathogens and food antigens, intestinal DCs are crucial for determining inflammatory or tolerogenic immune responses. The presence of the specific CD103+ CD11b+ DCs subset in lamina propria capable to capture antigens and induce Tregs through the production of TGF-␤ and retinoic acid [202,203] is a key component in the induction of intestinal immune tolerance. On the other hand, there are other subpopulations of mucosal DCs that are resided in the gut-associated lymphoid tissue and that are able to induce inflammatory response [204]. Thus, the careful analysis of the mechanisms of how different subtypes of intestinal DCs respond to various antigens and determining the response toward either immunity or tolerance should contribute to the efficiency of the tolerance-inducing therapy. To facilitate assessment of a role of each DC subtype including aortic DC populations, advanced approaches may be used such as generating mice with selectively labeled DC subsets by a reporter gene such as green fluorescent protein or obtaining mice strains each lacking a specific DC subset. These murine models may set the stage for future work on, for example, the mechanistic function of convenient DCs and monocyte-derived DCs in the aorta, both during steady state and during the development of atherosclerosis.

831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859

9

Such a strategy became to use. For example, Koltsova et al. [58] reported the development of apoE-deficient mice with CD11− DCs specifically labeled with yellow fluorescent protein. Using this model, a live-cell imaging of dynamic interactions between DCs and T cell in atherosclerotic aorta was obtained. Using an antimouse plasmacytoid dendritic cell antigen 1 (PDCA-1) antibody, Macritchie et al. [205] specifically depleted proatherogenic plasmacytoid DCs in apoE-deficient mice resulted in significant reduction of atherosclerosis progression and more stable plaque phenotype. Similar advanced approaches can be implemented for evaluating a role of each Treg subset in atherosclerosis. Regarding Tregs, a special attention should be paid for further characterization and studying immune regulatory function (in the atherosclerosis context) of recently discovered subtypes of Tregs such as IL17-producing inducible Foxp3+ Tregs and IL-17-producing natural CD4+ CD25+ Foxp3+ Tregs, since these Tregs can share features of Th2/Th17 and Th1/Treg cells. The oral administration of immunomodulatory molecules seems to be the optimal way of inducing a physiologically relevant Tregs-dependent antiatherogenic response because intravenous injection of immune modulators such as anti-CD3 antibody and superagonist anti-CD28 antibody may activate excessive immune responses [206]. The newest potential strategy of enhancing endogenous immune responses is the direct transplantation of autologous Tregs expanded ex vivo. At present, this cell therapy approach is under consideration to be implemented for treatment of autoimmune or graft-versus-host disease (clinical trials NCT01624077, NCT00651716, and NCT00675831). If Tregs-based cell therapy will be beneficial for treatment of such inflammatory diseases, it becomes possible to use this approach for atherosclerosis treatment in humans. However, multiple issues including biosafety, biocompatibility, homing ability, and functional activity should be investigated before the clinical approval of this technique.

860

Conflict of interests

893

The authors have no conflicts of interest to declare.

861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892

894

Acknowledgement

895

The work is supported by the Russian Ministry of Education and Sciences.

896

References

898

[1] Gerrity RG, Naito HK, Richardson M, Schwartz CJ. Dietary induced atherogenesis in swine. Morphology of the intima in prelesion stages. Am J Pathol 1979;95:775–92. [2] Hansson GK, Libby P. The immune response in atherosclerosis: a doubleedged sword. Nat Rev Immunol 2006;6:508–19. [3] Peterson RA. Regulatory T-cells: diverse phenotypes integral to immune homeostasis and suppression. Toxicol Pathol 2012;40:186–204. [4] Mallat Z, Gojova A, Brun V, Esposito B, Fournier N, Cottrez F, et al. Induction of a regulatory T cell type 1 response reduces the development of atherosclerosis in apolipoprotein E-knockout mice. Circulation 2003;108:1232–7. [5] Gotsman I, Grabie N, Gupta R, Dacosta R, MacConmara M, Lederer J, et al. Impaired regulatory T-cell response and enhanced atherosclerosis in the absence of inducible costimulatory molecule. Circulation 2006;114:2047–55. [6] Sasaki N, Yamashita T, Takeda M, Shinohara M, Nakajima K, Tawa H, et al. Oral anti-CD3 antibody treatment induces regulatory T cells and inhibits the development of atherosclerosis in mice. Circulation 2009;120:1996–2005. [7] Mallat Z, Tedgui A. Cytokines as regulators of atherosclerosis in murine models. Curr Drug Targets 2007;8:1264–72. [8] Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic selftolerance maintained by activated T cells expressing IL-2 receptor ␣-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 1995;155:1151–64. [9] Shevach EM, DiPaolo RA, Andersson J, Zhao DM, Stephens GL, Thornton AM. The lifestyle of naturally occurring CD4+ CD25+ Foxp3+ regulatory T cells. Immunol Rev 2006;212:60–73. [10] Zorn E, Nelson EA, Mohseni M, Porcheray F, Kim H, Litsa D, et al. IL-2 regulates FOXP3 expression in human CD4+ CD25+ regulatory T cells through a

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

897

899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925

G Model IMLET 5317 1–13 10 926 927 928

[11]

929 930

[12]

931 932

[13]

933 934

[14]

935 936 937

[15]

938 939 940

[16]

941 942 943

[17]

944 945 946

[18]

947 948 949

[19]

950 951 952

[20]

953 954 955

[21]

956 957 958

[22]

959 960 961

[23]

962 963 964

[24]

965 966 967

[25]

968 969

[26]

970 971 972

[27]

973 974 975

[28]

976 977 978

[29]

979 980 981

[30]

982 983 984 985

[31]

986 987 988

[32]

989 990 991 992

[33]

993 994 995

[34]

996 997 998

[35]

999 1000 1001 1002

[36]

1003 1004 1005 1006

[37]

1007 1008 1009 1010 1011

[38]

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006;108:1571–9. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role of Scurfin in CD4+ CD25+ regulatory T cells. Nat Immunol 2003;4:337–42. Miyara M, Sakaguchi S. Human FoxP3+ CD4+ regulatory T cells: their knowns and unknowns. Immunol Cell Biol 2011;89:346–51. Wing JB, Sakaguchi S. Multiple treg suppressive modules and their adaptability. Front Immunol 2012;3:178. Suvas S, Kumaraguru U, Pack CD, Lee S, Rouse BT. CD4+ CD25+ T cells regulate virus- specific primary and memory CD8+ T cell responses. J Exp Med 2003;198:889–901. Thornton AM, Shevach EM. CD4+ CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med 1998;188:287–96. Cederbom L, Hall H, Ivars F. CD4+ CD25+ regulatory T cells down-regulate co- stimulatory molecules on antigen-presenting cells. Eur J Immunol 2000;30:1538–43. Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contactmediated suppression by CD4+ CD25+ regulatory cells involves a granzyme Bdependent, perforin-independent mechanism. J Immunol 2005;174:1783–6. Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Human Ley TJ. T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 2004;21:589–601. Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, et al. A CD4+ T-cell subset inhibits antigen-specific T cell responses and prevents colitis. Nature 1997;389:737–42. Roncarolo MG, Gregori S, Lucarelli B, Ciceri F, Bacchetta R. Clinical tolerance in allogeneic hematopoietic stem cell transplantation. Immunol Rev 2011;241:145–63. Roncarolo MG, Gregori S, Battaglia M, Bacchetta R, Fleischhauer K, Levings MK. Interleukin-10-secreting type 1 regulatory T cells in rodents and humans. Immunol Rev 2006;212:28–50. Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, et al. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 2011;34:566–78. Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 2008;28:546–58. Spencer SD, DiMarco F, Hooley J, Pitts-Meek S, Bauer M, Ryan AM, et al. The orphan receptor CRF2-4 is an essential subunit of the interleukin 10 receptor. J Exp Med 1998;187:571–8. Weiner HL. Oral tolerance: immune mechanisms and the generation of Th3type TGF-␤-secreting regulatory cells. Microbes Infect 2001;3:947–54. Carrier Y, Juan J, Kuchroo VK, Weiner HL. Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-␤ T cell-transgenic mice. J Immunol 2007;178:179–85. Carrier Y, Juan J, Kuchroo VK, Weiner HL. Th3 cells in peripheral tolerance. II. TGF-␤-transgenic Th3 cells rescue IL-2-deficient mice from autoimmunity. J Immunol 2007;178:172–8. Li MO, Sanjabi S, Flavell RA. Transforming growth factor-␤ controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006;25:455–71. Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-␤1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 2007;26:579–91. Nakamura K, Kitani A, Fuss I, Pedersen A, Harada N, Nawata H, et al. TGF-␤1 plays an important role in the mechanism of CD4+ CD25+ regulatory T cell activity in both humans and mice. J Immunol 2004;172: 834–42. Chung EJ, Choi SH, Shim YH, Bang YJ, Hur KC, Kim CW. Transforming growth factor-␤ induces apoptosis in activated murine T cells through the activation of caspase 1-like protease. Cell Immunol 2000;204:46–54. Lin CM, Wang FH, Lee PK. Activated human CD4+ T cells induced by dendritic cell stimulation are most sensitive to transforming growth factor-␤: implications for dendritic cell immunization against cancer. Clin Immunol 2002;102:96–105. Workman CJ, Szymczak-Workman AL, Collison LW, Pillai MR, Vignali DAA. The development and function of regulatory T cells. Cell Mol Life Sci 2009;66:2603–22. Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, et al. Conversion of peripheral CD4+ CD25− naive T cells to CD4+ CD25+ regulatory T cells by TGF-␤ induction of transcription factor Foxp3. J Exp Med 2003;198:1875–86. Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-␤ induces a regulatory phenotype in CD4+ CD25− T cells through Foxp3 induction and down-regulation of Smad7. J Immunol 2004;172:5149–53. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-␤ and retinoic acid-dependent mechanism. J Exp Med 2007;204:1757–64. Lal G, Zhang N, van der Touw W, Ding Y, Ju W, Bottinger EP, et al. Epigenetic regulation of Foxp3 expression in regulatory T cells by DNA methylation. J Immunol 2009;182:259–73. Janson PC, Winerdal ME, Marits P, Thorn M, Ohlsson R, Winqvist O. FOXP3 promoter demethylation reveals the committed Treg population in humans. PLoS ONE 2008;3:e1612.

[39] Takaki H, Ichiyama K, Koga K, Chinen T, Takaesu G, Sugiyama Y, et al. STAT6 Inhibits TGF-␤1-mediated Foxp3 induction through direct binding to the Foxp3 promoter, which is reverted by retinoic acid receptor. J Biol Chem 2008;283:14955–62. [40] Mantel PY, Kuipers H, Boyman O, Rhyner C, Ouaked N, Rückert B, et al. GATA3-driven Th2 responses inhibit TGF-␤1-induced FOXP3 expression and the formation of regulatory T cells. PLoS Biol 2007;5:e329. [41] Hadjur S, Bruno L, Hertweck A, Cobb BS, Taylor B, Fisher AG, et al. IL4 blockade of inducible regulatory T cell differentiation: the role of Th2 cells, Gata3 and PU.1. Immunol Lett 2009;122:37–43. [42] Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol 2007;25:821–52. [43] Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006;126:1121–33. [44] Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, et al. TGF-␤-induced Foxp3 inhibits Th17 cell differentiation by antagonizing ROR␥t function. Nature 2008;453:236–40. [45] Voo KS, Wang YH, Santori FR, Boggiano C, Wang YH, Arima K, et al. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci U S A 2009;106:4793–8. [46] Cheng X, Yu X, Ding YJ, Fu QQ, Xie JJ, Tang TT, et al. The Th17/Treg imbalance in patients with acute coronary syndrome. Clin Immunol 2008;127:89–97. [47] Xie JJ, Wang J, Tang TT, Chen J, Gao XL, Yuan J, et al. The Th17/Treg functional imbalance during atherogenesis in ApoE−/− mice. Cytokine 2010;49:185–93. [48] Xu L, Kitani A, Fuss I, Strober W. Cutting edge: regulatory T cells induce CD4+ CD25− Foxp3− T cells or are self-induced to become Th17 cells in the absence of exogenous TGF-␤. J Immunol 2007;178:6725–9. [49] Kitani A, Xu L. Regulatory T cells and the induction of IL-17. Mucosal Immunol 2008;1(Suppl. 1):S43–6. [50] Zheng SG, Wang J, Horwitz DA. Cutting edge: Foxp3+ CD4+ CD25+ regulatory T cells induced by IL-2 and TGF-␤ are resistant to Th17 conversion by IL-6. J Immunol 2008;180:7112–6. [51] Horwitz DA, Zheng SG, Gray JD. Natural and TGF-␤-induced Foxp3+ CD4+ CD25+ regulatory T cells are not mirror images of each other. Trends Immunol 2008;29:429–35. [52] Tacke F, Alvarez D, Kaplan TJ, Jakubzick C, Spanbroek R, Llodra J, et al. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques. J Clin Invest 2007;117:185–94. [53] Bobryshev YV, Babaev VR, Lord RS, Watanabe T. Ultrastructural identification of cells with dendritic cell appearance in atherosclerotic aorta of apolipoprotein E deficient mice. J Submicrosc Cytol Pathol 1999;31:527–31. [54] Benagiano M, D’Elios MM, Amedei A, Azzurri A, van der Zee R, Ciervo A, et al. Human 60-kDa heat shock protein is a target autoantigen of T cells derived from atherosclerotic plaques. J Immunol 2005;174:6509. [55] Döring Y, Manthey HD, Drechsler M, Lievens D, Megens RT, Soehnlein O, et al. Auto-antigenic protein-DNA complexes stimulate plasmacytoid dendritic cells to promoteatherosclerosis. Circulation 2012;125:1673–83. [56] Nickel T, Schmauss D, Hanssen H, Sicic Z, Krebs B, Jankl S, et al. oxLDL uptake by dendritic cells induces upregulation of scavenger-receptors, maturation and differentiation. Atherosclerosis 2009;205:442–50. [57] Döring Y, Zernecke A. Plasmacytoid dendritic cells in atherosclerosis. Front Physiol 2012;3:230. [58] Koltsova EK, Garcia Z, Chodaczek G, Landau M, McArdle S, Scott SR, et al. Dynamic T cell-APC interactions sustain chronic inflammation in atherosclerosis. J Clin Invest 2012;122:3114–26. [59] Onishi Y, Fehervari Z, Yamaguchi T, Sakaguchi S. Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. Proc Natl Acad Sci U S A 2008;105:10113–8. [60] Bour-Jordan H, Esensten JH, Martinez-Llordella M, Penaranda C, Stumpf M, Bluestone JA. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol Rev 2011;241:180–205. [61] Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, et al. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 2003;4:1206–12. [62] Gu P, Gao JF, D’Souza CA, Kowalczyk A, Chou KY, Zhang L. Trogocytosis of CD80 and CD86 by induced regulatory T cells. Cell Mol Immunol 2012;9:136–46. [63] Gao JF, McIntyre MS, Juvet SC, Diao J, Li X, Vanama RB, et al. Regulation of antigen-expressing dendritic cells by double negative regulatory T cells. Eur J Immunol 2011;41:2699–708. [64] Oderup C, Cederbom L, Makowska A, Cilio CM, Ivars F. Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+ CD25+ regulatory T-cell-mediated suppression. Immunology 2006;118:240–9. [65] Gotsman I, Sharpe AH, Lichtman AH. T-cell costimulation and coinhibition in atherosclerosis. Circ Res 2008;103:1220–31. [66] Weber C, Meiler S, Döring Y, Koch M, Drechsler M, Megens RT, et al. CCL17expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice. J Clin Invest 2011;121:2898–910. [67] Choi JH, Cheong C, Dandamudi DB, Park CG, Rodriguez A, Mehandru S, et al. Flt3 signaling-dependent dendritic cells protect against atherosclerosis. Immunity 2011;35:819–31. [68] Iellem A, Mariani M, Lang R, Recalde H, Panina-Bordignon P, Sinigaglia F, et al. Unique chemotactic response profile and specific expression of chemokine

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

1098 1099 1100

[69]

1101 1102 1103 1104

[70]

1105 1106 1107

[71]

1108 1109

[72]

1110 1111

[73]

1112 1113 1114

[74]

1115 1116

[75]

1117 1118 1119

[76]

1120 1121 1122

[77]

1123 1124

[78]

1125 1126 1127

[79]

1128 1129 1130

[80]

1131 1132 1133

[81]

1134 1135

[82]

1136 1137

[83]

1138 1139 1140 1141

[84]

1142 1143 1144

[85]

1145 1146

[86]

1147 1148 1149

[87]

1150 1151 1152 1153

[88]

1154 1155 1156

[89]

1157 1158 1159

[90]

1160 1161 1162

[91]

1163 1164 1165

[92]

1166 1167

[93]

1168 1169 1170

[94]

1171 1172 1173

[95]

1174 1175

[96]

1176 1177 1178

[97]

1179 1180 1181 1182 1183

[98]

receptors CCR4 and CCR8 by CD4+ CD25+ regulatory T cells. J Exp Med 2001;194:847–53. Onodera T, Jang MH, Guo Z, Yamasaki M, Hirata T, Bai Z, et al. Constitutive expression of IDO by dendritic cells of mesenteric lymph nodes: functional involvement of the CTLA-4/B7 and CCL22/CCR4 interactions. J Immunol 2009;183:5608–14. Laurat E, Poirier B, Tupin E, Caligiuri G, Hansson GK, Bariéty J, et al. In vivo downregulation of T helper cell 1 immune responses reduces atherogenesis in apolipoprotein E-knockout mice. Circulation 2001;104:197–202. McLaren JE, Ramji DP. Interferon ␥: a master regulator of atherosclerosis. Cytokine Growth Factor Rev 2009;20:125–35. Gupta S, Pablo AM, Jiang XC, Wang N, Tall AR, Schindler C. IFN-␥ potentiates atherosclerosis in apoE knock-out mice. J Clin Invest 1997;99:2752–61. Whitman SC, Ravisankar P, Elam H, Daugherty A. Exogenous interferon␥ enhances atherosclerosis in apolipoprotein E−/− mice. Am J Pathol 2000;157:1819–24. Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res 2008;79:360–76. Ranjbaran H, Sokol SI, Gallo A, Eid RE, Iakimov AO, D’Alessio A, et al. An inflammatory pathway of IFN-␥ production in coronary atherosclerosis. J Immunol 2007;178:592–604. Zhou X, Paulsson G, Stemme S, Hansson GK. Hypercholesterolemia is associated with a T helper Th1/Th2 switch of the autoimmune response in atherosclerotic apo E-knockout mice. J Clin Invest 1998;101:1717–25. Robertson AK, Hansson GK. T cells in atherogenesis: for better or for worse. Arterioscler Thromb Vasc Biol 2006;26:2421–32. Huber SA, Sakkinen P, David C, Newell MK, Tracy RP. T helper-cell phenotype regulates atherosclerosis in mice under conditions of mild hypercholesterolemia. Circulation 2001;103:2610–6. King VL, Szilvassy SJ, Daugherty A. Interleukin-4 deficiency decreases atherosclerotic lesion formation in a site-specific manner in female LDL receptor−/− mice. Arterioscler Thromb Vasc Biol 2002;22:456–61. Davenport P, Tipping PG. The role of interleukin-4 and interleukin-12 in the progression of atherosclerosis in apolipoprotein E-deficient mice. Am J Pathol 2003;163:1117–25. Binder CJ, Shaw PX, Chang MK, Boullier A, Hartvigsen K, Hörkkö S, et al. The role of natural antibodies in atherogenesis. J Lipid Res 2005;46:1353–63. Miller SA, St Onge E. Otelixizumab: a novel agent for the prevention of type 1 diabetes mellitus. Expert Opin Biol Ther 2011;11:1525–32. Zhou X, Johnston TP, Johansson D, Parini P, Funa K, Svensson J, et al. Hypercholesterolemia leads to elevated TGF-␤1 activity and T helper 3dependent autoimmune responses in atherosclerotic mice. Atheroslerosis 2009;204:381–7. Ait-Oufella H, Salomon BL, Potteaux S, Robertson AK, Gourdy P, Zoll J, et al. Natural regulatory T cells control the development of atherosclerosis in mice. Nat Med 2006;12:178–80. Wang S, Chen L. T lymphocyte co-signaling pathways of the B7-CD28 family. Cell Mol Immunol 2004;1:37–42. Mor A, Planer D, Luboshits G, Afek A, Metzger S, Chajek-Shaul T, et al. Role of naturally occurring CD4+CD25+ regulatory T cells in experimental atherosclerosis. Arterioscler Thromb Vasc Biol 2007;27:893–900. Pinderski LJ, Fischbein MP, Subbanagounder G, Fishbein MC, Kubo N, Cheroutre H, et al. Overexpression of interleukin-10 by activated T lymphocytes inhibits atherosclerosis in LDL receptor-deficient mice by altering lymphocyte and macrophage phenotypes. Circ Res 2002;90:1064–71. Ait-Oufella H, Horvat B, Kerdiles Y, Herbin O, Gourdy P, Khallou-Laschet J, et al. Measles virus nucleoprotein induces a regulatory immune response and reduces atherosclerosis in mice. Circulation 2007;116:1707–13. Foks AC, Frodermann V, ter Borg M, Habets KL, Bot I, Zhao Y, et al. Differential effects of regulatory T cells on the initiation and regression of atherosclerosis. Atherosclerosis 2011;218:53. Lievens D, Eijgelaar WJ, Biessen EA, Daemen MJ, Lutgens E. The multifunctionality of CD40L and its receptor CD40 in atherosclerosis. Thromb Haemost 2009;102:206–14. Mach F, Schönbeck U, Sukhova GK, Atkinson E, Libby P. Reduction of atherosclerosis in mice by inhibition of CD40 signalling. Nature 1998;394:200–3. Suzuki M, Zheng X, Zhang X, Ichim TE, Sun H, Kubo N, et al. Inhibition of allergic responses by CD40 gene silencing. Allergy 2009;64:387–97. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005;6:1123–32. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, et al. A distinct lineage of CD4T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 2005;6:1133–41. Hernández-Santos N, Gaffen SL. Th17 cells in immunity to Candida albicans. Cell Host Microbe 2012;11:425–35. Maddur MS, Miossec P, Kaveri SV, Bayry J. Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol 2012;181:8–18. Pejnovic N, Vratimos A, Lee SH, Popadic D, Takeda K, Akira S, et al. Increased atherosclerotic lesions and Th17 in interleukin-18 deficient apolipoprotein E-knockout mice fed high-fat diet. Mol Immunol 2009;47:37–45. Gao Q, Jiang Y, Ma T, et al. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J Immunol 2010;185:5820.

11

[99] Wang Z, Lee J, Zhang Y, Wang H, Liu X, Shang F, et al. Increased Th17 cells in coronary artery disease are associated with neutrophilic inflammation. Scand Cardiovasc J 2011;45:54–61. [100] Zhao Z, Wu Y, Cheng M, Ji Y, Yang X, Liu P, et al. Activation of Th17/Th1 and Th1, but not Th17, is associated with the acute cardiac event in patients with acute coronary syndrome. Atherosclerosis 2011;217:518–24. [101] Liu Z, Lu F, Pan H, Zhao Y, Wang S, Sun S, et al. Correlation of peripheral Th17 cells and Th17-associated cytokines to the severity of carotid artery plaque and its clinical implication. Atherosclerosis 2012;221:232–41. [102] Zhu F, Wang Q, Guo C, Wang X, Cao X, Shi Y, et al. IL-17 induces apoptosis of vascular endothelial cells: a potential mechanism for human acute coronary syndrome. Clin Immunol 2011;141:152–60. [103] Eid RE, Rao DA, Zhou J, Lo SF, Ranjbaran H, Gallo A, et al. Interleukin-17 and interferon-␥ are produced concomitantly by human coronary arteryinfiltrating T cells and act synergistically on vascular smooth muscle cells. Circulation 2009;119:1424–32. [104] Pietrowski E, Bender B, Huppert J, White R, Luhmann HJ, Kuhlmann CR. Pro-inflammatory effects of interleukin-17A on vascular smooth muscle cells involve NAD(P)H- oxidase derived reactive oxygen species. J Vasc Res 2011;48:52–8. [105] Lee HS, Son SM, Kim YK, Hong KW, Kim CD. NAD(P)H oxidase participates in the signaling events in high glucose-induced proliferation of vascular smooth muscle cells. Life Sci 2003;72:2719–30. [106] Dunér P, To F, Berg K, Björkbacka H, Engelbertsen D, Fredrikson GN, et al. Immune responses against aldehyde-modified laminin accelerate atherosclerosis in Apoe−/− mice. Atherosclerosis 2010;212:457–65. [107] Dart ML, Jankowska-Gan E, Huang G, Roenneburg DA, Keller MR, Torrealba JR, et al. Interleukin-17-dependent autoimmunity to collagen type V in atherosclerosis. Circ Res 2010;107:1106–16. [108] Postigo J, Genre F, Iglesias M, Fernández-Rey M, Buelta L, Carlos RodríguezRey J, et al. Exacerbation of type II collagen-induced arthritis in apolipoprotein E-deficient mice in association with the expansion of Th1 and Th17 cells. Arthritis Rheum 2011;63:971–80. [109] de Boer OJ, van der Meer JJ, Teeling P, van der Loos CM, Idu MM, van Maldegem F, et al. Differential expression of interleukin-17 family cytokines in intact and complicated human atherosclerotic plaques. J Pathol 2010;220:499–508. [110] Butcher MJ, Gjurich BN, Phillips T, Galkina EV. The IL-17A/IL-17RA axis plays a proatherogenic role via the regulation of aortic myeloid cell recruitment. Circ Res 2012;110:675–87. [111] Erbel C, Chen L, Bea F, Wangler S, Celik S, Lasitschka F, et al. Inhibition of IL17A attenuates atherosclerotic lesion development in apoE-deficient mice. J Immunol 2009;183:8167–75. [112] Smith E, Prasad KM, Butcher M, Dobrian A, Kolls JK, Ley K, et al. Blockade of interleukin-17A results in reduced atherosclerosis in apolipoprotein E-deficient mice. Circulation 2010;121:1746–55. [113] Li N, Bian H, Zhang J, Li X, Ji X, Zhang Y. The Th17/Treg imbalance exists in patients with heart failure with normal ejection fraction and heart failure with reduced ejection fraction. Clin Chim Acta 2010;411:1963–8. [114] Ivanov II, Zhou L, Littman DR. Transcriptional regulation of Th17 differentiation. Semin Immunol 2007;19:409–17. [115] Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, Nakaya M, et al. Foxp3 inhibits ROR␥t-mediated IL-17A mRNA transcription through direct interaction with ROR␥t. J Biol Chem 2008;283:17003–8. [116] Vocanson M, Rozieres A, Hennino A, Poyet G, Gaillard V, Renaudineau S, et al. Inducible costimulator (ICOS) is a marker for highly suppressive antigenspecific T cells sharing features of TH17/TH1 and regulatory T cells. J Allergy Clin Immunol 126:280–9, 289.e1–7. Q2 [117] Whitehead GS, Wilson RH, Nakano K, Burch LH, Nakano H, Cook DN. IL-35 production by inducible costimulator (ICOS)-positive regulatory T cells reverses established IL-17-dependent allergic airways disease. J Allergy Clin Immunol 2010;129, 207-15.e1–207-15.e. [118] Ghourbani Gazar S, Andalib A, Hashemi M, Rezaei A. CD4+ Foxp3+ Treg and its ICOS+ subsets in patients with myocardial infarction. Iran J Immunol 2012;9:53–60. [119] Mor A, Luboshits G, Planer D, Keren G, George J. Altered status of CD4+ CD25+ regulatory T cells in patients with acute coronary syndromes. Eur Heart J 2006;27:2530–7. [120] Pabst O, Mowat AM. Oral tolerance to food protein. Mucosal Immunol 2012;5:232–9. [121] van Puijvelde GH, Hauer AD, de Vos P, van den Heuvel R, van Herwijnen MJ, van der Zee R, et al. Induction of oral tolerance to oxidized low-density lipoprotein ameliorates atherosclerosis. Circulation 2006;114:1968–76. [122] Klingenberg R, Lebens M, Hermansson A, Fredrikson GN, Strodthoff D, Rudling M, et al. Intranasal immunization with an apolipoprotein B-100 fusion protein induces antigen-specific regulatory T cells and reduces atherosclerosis. Arterioscler Thromb Vasc Biol 2010;30:946–52. [123] Wigren M, Kolbus D, Dunér P, Ljungcrantz I, Söderberg I, Björkbacka H, et al. Evidence for a role of regulatory T cells in mediating the atheroprotective effect of apolipoprotein B peptide vaccine. J Intern Med 2011;269:546–56. [124] Harats D, Yacov N, Gilburd B, Shoenfeld Y, George J. Oral tolerance with heat shock protein 65 attenuates mycobacterium tuberculosis-induced and highfat-diet-driven atherosclerotic lesions. J Am Coll Cardiol 2002;40:1333–8. [125] 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;106:1708–15.

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219 1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269

G Model IMLET 5317 1–13 12 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305 1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

[126] van Puijvelde GH, van Es T, van Wanrooij EJ, Habets KL, de Vos P, van der Zee R, et al. Induction of oral tolerance to HSP60 or an HSP60-peptide activates T cell regulation and reduces atherosclerosis. Arterioscler Thromb Vasc Biol 2007;27:2677–83. [127] George J, Yacov N, Breitbart E, Bangio L, Shaish A, Gilburd B, et al. Suppression of early atherosclerosis in LDL-receptor deficient mice by oral tolerance with ␤2-glycoprotein I. Cardiovasc Res 2004;62:603–9. [128] Sun JB, Raghavan S, Sjoling A, Lundin S, Holmgren J. Oral tolerance induction with antigen conjugated to cholera toxin B subunit generates both Foxp3+ CD25+ and Foxp3− CD25− CD4+ regulatory T cells. J Immunol 2006;177:7634–44. [129] Esterbauer H, Gebicki J, Puhi H, Jurgens G. The role of lipid peroxidation and antioxidants in the oxidative modification of LDL. Free Radic Biol Med 1992;13:341–90. [130] Jonas A, Phillips MC. Lipoprotein structure. In: Vance DE, Vance JE, editors. Biochemistry of Lipids, Lipoproteins and Membranes. 5th ed. New York: Elsevier Science; 2008. p. 485–506. [131] Brunzell JD, Davidson M, Furberg CD, Goldberg RB, Howard BV, Stein JH, et al. Lipoprotein management in patients with cardiometabolic risk: consensus conference report from the American Diabetes Association and the American College of Cardiology Foundation. J Am Coll Cardiol 2008;51:1512–24. [132] Aviram M, Rosenblat M, Etzioni A, Levy R. Activation of NADPH oxidase required for macrophage-mediated oxidation of low-density lipoprotein. Metabolism 1996;45:1069–79. [133] Heinecke JW, Baker H, Rosen H, Chait A. Superoxide mediated modification of low density lipoprotein by human arterial smooth muscle cells. J Clin Invest 1986;77:757–62. [134] Heinecke JW, Kawamura M, Suzuki L, Chait A. Oxidation of low density lipoprotein by thiols: superoxide dependent and independent mechanisms. J Lipid Res 1993;34:2051–61. [135] Hogg N, Darley-Usmar DM, Graham A, Moncada S. Peroxynitrite and atherosclerosis. Biochem Soc Trans 1993;21:358–62. [136] Heinecke JW. Cellular mechanisms for the oxidative modification of lipoproteins: implications for atherogenesis. Coronary Artery Dis 1994;5:205–10. [137] Steinberg D, Parthasarathy S, Carew TE, Khoo JC, Witztum JL. Beyond cholesterol: modifications of low density lipoprotein that increase its atherogenicity. N EngI J Med 1989;320:915–24. [138] Kritharides L, Jessup W, Gifford J, Dean RT. A method for defining stages of LDL oxidation by separation of cholesterol and cholesterol ester oxidation products using high pressure liquid chromatography. Anal Biochem 1993;213:79–89. [139] Esterbauer H, Jürgens G, Quehenberger O, Koller E. Autoxidation of human low density lipoprotein: loss of polyunsaturated fatty acids and vitamin E and generation of aldehydes. J Lipid Res 1987;28:495–509. [140] Hoff HF, O’Neil J, Chisolm 3rd GM, Cole TB, Quehenberger O, Esterbauer H, et al. Modification of low density lipoprotein with 4-hydroxynonenal induces uptake by macrophages. Arteriosclerosis 1989;9:538–49. [141] Parthasarathy S, Rankin SM. Role of oxidized low density lipoprotein in atherogenesis. Prog Lipid Res 1992;31:127–43. [142] Hill GE, Miller JA, Baxter BT, Klassen LW, Duryee MJ, Tuma DJ, et al. Association of malondialdehyde-acetaldehyde (MAA) adducted proteins with atherosclerotic-induced vascular inflammatory injury. Atherosclerosis 1998;141:107–16. [143] Habets KL, van Puijvelde GH, van Duivenvoorde LM, van Wanrooij EJ, de Vos P, Tervaert JW, et al. Vaccination using oxidized low-density lipoproteinpulsed dendritic cells reduces atherosclerosis in LDL receptor-deficient mice. Cardiovasc Res 2010;85:622–30. [144] Veneskoski M, Turunen SP, Kummu O, Nissinen A, Rannikko S, Levonen AL, et al. Specific recognition of malondialdehyde and malondialdehyde acetaldehyde adducts on oxidized LDL and apoptotic cells by complement anaphylatoxin C3a. Free Radic Biol Med 2011;51:834–43. [145] Dinh TN, Kyaw TS, Kanellakis P, To K, Tipping P, Toh BH, et al. Cytokine therapy with interleukin-2/anti-interleukin-2 monoclonal antibody complexes expands CD4+ CD25+ Foxp3+ regulatory T cells and attenuates development and progression of atherosclerosis. Circulation 2012;126:1256–66. [146] Ochi H, Abraham M, Ishikawa H, Frenkel D, Yang K, Basso AS, et al. Oral CD3-specific antibody suppresses autoimmune encephalomyelitis by inducing CD4+ CD25− LAP+ T cells. Nat Med 2006;12:627–35. [147] 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;56:2103–9. [148] Ilan Y, Zigmond E, Lalazar G, Dembinsky A, Ben Ya’acov A, Hemed N, et al. Oral administration of OKT3 monoclonal antibody to human subjects induces a dose-dependent immunologic effect in T cells and dendritic cells. J Clin Immunol 2010;30:167–77. [149] Reichert JM. Antibody-based therapeutics to watch in 2011. Mabs 2011;3:76–99. [150] Steffens S, Burger F, Pelli G, Dean Y, Elson G, Kosco-Vilbois M, et al. Short-term treatment with anti-CD3 antibody reduces the development and progression of atherosclerosis in mice. Circulation 2006;114:1977–84. [151] 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;181:6038–50. [152] Chen ML, Yan BS, Bando Y, Kuchroo VK, Weiner HL. Latency-associated peptide identifies a novel CD4+ CD25+ regulatory T cell subset with

[153] [154]

[155]

[156]

[157]

[158]

[159]

[160]

[161]

[162]

[163]

[164]

[165]

[166] [167] [168]

[169]

[170]

[171]

[172]

[173]

[174]

[175]

[176]

[177]

TGF-␤-mediated function and enhanced suppression of experimental autoimmune encephalomyelitis. J Immunol 2008;180:7327–37. Zhang L, Zhao Y. The regulation of Foxp3 expression in regulatory CD4+ CD25+ T cells: multiple pathways on the road. J Cell Physiol 2007;211:590–7. Daniel C, Sartory NA, Zahn N, Radeke HH, Stein JM. Immune modulatory treatment of trinitrobenzene sulfonic acid colitis with calcitriol is associated with a change of a T helper (Th) 1/Th17 to a Th2 and regulatory T cell profile. J Pharmacol Exp Ther 324:23-33. Q3 Penna G, Adorini L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol 2000;164:2405–11. Unger WW, Laban S, Kleijwegt FS, van der Slik AR, Roep BO. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1. Eur J Immunol 2009;39:3147–59. Penna G, Roncari A, Amuchastegui S, Daniel KC, Berti E, Colonna M, et al. Expression of the inhibitory receptor ILT3 on dendritic cells is dispensable for induction of CD4+ Foxp3+ regulatory T cells by 1,25-dihydroxyvitamin D3. Blood 2005;106:3490–7. Gorman S, Kuritzky LA, Judge MA, Dixon KM, McGlade JP, Mason RS, et al. Topically applied 1,25-dihydroxyvitamin D3 enhances the suppressive activity of CD4+ CD25+ cells in the draining lymph nodes. J Immunol 2007;179:6273–83. Bock G, Prietl B, Mader JK, Höller E, Wolf M, Pilz S, et al. The effect of vitamin D supplementation on peripheral regulatory T cells and ␤ cell function in healthy humans: a randomized controlled trial. Diabetes Metab Res Rev 2011;27:942–5. Takeda M, Yamashita T, Sasaki N, Nakajima K, Kita T, Shinohara M, et al. Oral administration of an active form of vitamin D3 (calcitriol) decreases atherosclerosis in mice by inducing regulatory T cells and immature dendritic cells with tolerogenic functions. Arterioscler Thromb Vasc Biol 2010;30:2495–503. Penna G, Amuchastegui S, Giarratana N, Daniel KC, Vulcano M, Sozzani S, et al. 1,25-Dihydroxyvitamin D3 selectively modulates tolerogenic properties in myeloid but not plasmacytoid dendritic cells. J Immunol 2007;178:145–53. Meng X, Zhang K, Li J, Dong M, Yang J, An G, et al. Statins induce the accumulation of regulatory T cells in atherosclerotic plaque. Mol Med 2012;18:598–605. Kajimoto K, Miyauchi K, Kasai T, Shimada K, Kojima Y, Shimada A, et al. Shortterm 20-mg atorvastatin therapy reduces key inflammatory factors including c-Jun N-terminal kinase and dendritic cells and matrix metalloproteinase expression in human abdominal aortic aneurysmal wall. Atherosclerosis 2009;206:505–11. Mausner-Fainberg K, Luboshits G, Mor A, Maysel-Auslender S, Rubinstein A, Keren G, et al. The effect of HMG-CoA reductase inhibitors on naturally occurring CD4+ CD25+ T cells. Atheroslerosis 2008;197:829–39. Zhang D, Wang S, Guan Y, Wang L, Xie W, Li N, et al. Effect of oral atorvastatin on CD4+ CD25+ regulatory T cells, FoxP3 expression, and prognosis in patients with ST-segment elevated myocardial infarction before primary percutaneous coronary intervention. J Cardiovasc Pharmacol 2011;57:536–41. Spiegel M, Milstien S. Sphingosine-1-phosphate: an enigmatic signalling lipid. Nat Rev Mol Cell Biol 2003;4:397–407. Rosen H, Goetzl EJ. Sphingosine 1-phosphate and its receptors: an autocrine and paracrine network. Nat Rev Immunol 2005;5:560–70. Nofer JR, Bot M, Brodde M, Taylor PJ, Salm P, Brinkmann V, et al. FTY720, a synthetic sphingosine 1 phosphate analogue, inhibits development of atherosclerosis in low-density lipoprotein receptor-deficient mice. Circulation 2007;115:501–8. Keul P, Tölle M, Lucke S, von Wnuck Lipinski K, Heusch G, Schuchardt M, et al. The sphingosine-1-phosphate analogue FTY720 reduces atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2007; 27:607. Huang K, Li SQ, Wang WJ, Liu LS, Jiang YG, Feng PN, et al. Oral FTY720 administration induces immune tolerance and inhibits early development of atherosclerosis in apolipoprotein E-deficient mice. Int J Immunopathol Pharmacol 2012;25:397–406. Chiba K, Adachi K. Discovery of fingolimod, the sphingosine 1-phosphate receptor modulator and its application for the therapy of multiple sclerosis. Future Med Chem 2012;4:771–81. Li HB, Cai KM, Liu Z, Xia JH, Zhang Y, Xu R, et al. Foxp3+ T regulatory cells (Tregs) are increased in nasal polyps (NP) after treatment with intranasal steroid. Clin Immunol 2008;129:394–400. Seissler N, Schmitt E, Hug F, Sommerer C, Zeier M, Schaier M, et al. Methylprednisolone treatment increases the proportion of the highly suppressive HLA-DR+ -Treg-cells in transplanted patients. Transpl Immunol 2012;27:157–61. Roelen DL, van den Boogaardt DE, van Miert PP, Koekkoek K, Offringa R, Claas FH. Differentially modulated dendritic cells induce regulatory T cells with different characteristics. Transpl Immunol 2008;19:220–8. Unger WW, Laban S, Kleijwegt FS, van der Slik AR, Roep BO. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1. Eur J Immunol 2009;39:3147–59. Waanders F, de Vries LV, van Goor H, Hillebrands JL, Laverman GD, Bakker SJ, et al. Aldosterone, from (patho)physiology to treatment in cardiovascular and renal damage. Curr Vasc Pharmacol 2011;9:594–605. Jaffe IZ, Mendelsohn ME. Angiotensin II and aldosterone regulate gene transcription via functional mineralocortocoid receptors in human coronary artery smooth muscle cells. Circ Res 2005;96:643–50.

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

1356 1357 1358 1359 1360 1361 1362 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391 1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441

G Model IMLET 5317 1–13

ARTICLE IN PRESS D.A. Chistiakov et al. / Immunology Letters xxx (2013) xxx–xxx

1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477 1478 1479 1480 1481 1482 1483 1484

[178] Kasal DA, Barhoumi T, Li MW, Yamamoto N, Zdanovich E, Rehman A, et al. T regulatory lymphocytes prevent aldosterone-induced vascular injury. Hypertension 2012;59:324–30. [179] Longui CA. Glucocorticoid therapy: minimizing side effects. J Pediatr (Rio J) 2007;83(5 Suppl.):S163–77. [180] Tous M, Ribas V, Escolà-Gil JC, Blanco-Vaca F, Calpe-Berdiel L, Coll B, et al. Manipulation of inflammation modulates hyperlipidemia in apolipoprotein E-deficient mice: a possible role for interleukin-6. Cytokine 2006;34:224–32. [181] Buckland M, Lombardi G. Aspirin and the induction of tolerance by dendritic cells. Handb Exp Pharmacol 2009;(188):197–213. [182] Buckland M, Jago CB, Fazekasova H, Scott K, Tan PH, George AJ, Lechler R, Lombardi G. Aspirin-treated human DCs up-regulate ILT-3 and induce hyporesponsiveness and regulatory activity in responder T cells. Am J Transplant 2006;6:2046–59. [183] Zhang H, Guo C, Zhang A, Fan Y, Gu T, Wu D, Sparatore A, Wang C. Effect of S-aspirin, a novel hydrogen-sulfide-releasing aspirin (ACS14), on atherosclerosis in apoE-deficient mice. Eur J Pharmacol 2012;697:106–16. [184] Pircher J, Fochler F, Czermak T, Mannell H, Kraemer BF, Wörnle M, Sparatore A, Del Soldato P, Pohl U, Krötz F. Hydrogen sulfide releasing aspirin derivative ACS14 exerts strong antithrombotic effects in vitro and in vivo. Arherioscler Thromb Vasc Biol 2012;32:2884–91. [185] Burleigh ME, Babaev VR, Patel MB, Crews BC, Remmel RP, Morrow JD, et al. Inhibition of cyclooxygenase with indomethacin phenethylamide reduces atherosclerosisin apoE-null mice. Biochem Pharmacol 2005;70:334–42. [186] Dabhi JK, Solanki JK, Mehta A. Antiatherosclerotic activity of ibuprofen, a nonselective COX inhibitor—an animal study. Indian J Exp Biol 2008;46:476–81. [187] Padol IT, Hunt RH. Association of myocardial infarctions with COX-2 inhibition may be related to immunomodulation towards a Th1 response resulting in atheromatous plaque instability: an evidence-based interpretation. Rheumatology (Oxford) 2010;49:837–43. [188] Fosslien E. Cardiovascular complications of non-steroidal anti-inflammatory drugs. Ann Clin Lab Sci 2005;35:347–85. [189] Lee SY, Choi HK, Lee KJ, Jung JY, Hur GY, Jung KH, et al. The immune tolerance of cancer is mediated by IDO that is inhibited by COX-2 inhibitors through regulatory T cells. J Immunother 2009;32:22–8. [190] Coomes E, Chan ES, Reiss AB. Methotrexate in atherogenesis and cholesterol metabolism. Cholesterol 2011;2011:503028. [191] Bulgarelli A, Martins Dias AA, Caramelli B, Maranhão RC. Treatment with methotrexate inhibits atherogenesis in cholesterol-fed rabbits. J Cardiovasc Pharmacol 2012;59:308–14. [192] Oh JS, Kim YG, Lee SG, So MW, Choi SW, Lee CK, et al. The effect of various disease-modifying anti-rheumatic drugs on the suppressive function of CD4+CD25+ regulatory T cells. Rheumatol Int, in press. http://dx.doi.org/10.1007/s00296-012-2365-9

13

[193] Huang B, Wang QT, Song SS, Wu YJ, Ma YK, Zhang LL. Combined use of etanercept and MTX restores CD4+ /CD8+ ratio and Tregs in spleen and thymus in collagen-induced arthritis. Inflamm Res 2012;61:1229–39. [194] Chen X, Oppenheim JJ. TNF-␣: an activator of CD4+ FoxP3+ TNFR2+ regulatory T cells. Curr Dir Autoimmun 2010;11:119–34. [195] Valencia X, Stephens G, Goldbach-Mansky R, Wilson M, Shevach EM, Lipsky PE. TNF downmodulates the function of human CD4+ CD25hi T-regulatory cells. Blood 2006;108:253–61. [196] Ricciardelli I, Lindley KJ, Londei M, Quaratino S. Anti tumour necrosis-alpha therapy increases the number of FOXP3 regulatory T cells in children affected by Crohn’s disease. Immunology 2008;125:178–83. [197] Boschetti G, Nancey S, Sardi F, Roblin X, Flourié B, Kaiserlian D. Therapy with anti-TNF␣ antibody enhances number and function of Foxp3+ regulatory T cells in inflammatory bowel diseases. Inflamm Bowel Dis 2011;17:160–70. [198] Calleja S, Cordero-Coma M, Rodriguez E, Llorente M, Franco M, Ruiz de Morales JG. Adalimumab specifically induces CD3+ CD4+ CD25high Foxp3+ CD127− T-regulatory cells and decreases vascular endothelial growth factor plasma levels in refractory immunomediated uveitis: a non-randomized pilot intervention study. Eye (Lond) 2012;26:468–77. [199] Atzeni F, Turiel M, Caporali R, Cavagna L, Tomasoni L, Sitia S, et al. The effect of pharmacological therapy on the cardiovascular system of patients with systemic rheumatic diseases. Autoimmun Rev 2010;9:835–9. [200] Marks JL, Edwards CJ. Protective effect of methotrexate in patients with rheumatoid arthritis and cardiovascular comorbidity. Ther Adv Musculoskelet Dis 2012;4:149–57. [201] Tam LS, Shang Q, Li EK, Wang S, Li RJ, Lee KL, et al. Infliximab is associated with improvement in arterial stiffness in patients with early rheumatoid arthritis—a randomized trial. J Rheumatol 2012;39:2267–75. [202] Coombes JL, Powrie F. Dendritic cells in intestinal immune regulation. Nat Rev Immunol 2008;8:435–46. [203] Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med 2007;204:1775–85. [204] Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, et al. ATP drives lamina propria TH 17 cell differentiation. Nature 2008;455:808–12. [205] Macritchie N, Grassia G, Sabir SR, Maddaluno M, Welsh P, Sattar N, et al. Plasmacytoid dendritic cells play a key role in promoting atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 2012;32:2569–79. [206] Eastwood D, Findlay L, Poole S, Bird C, Wadhwa M, Moore M, et al. Monoclonal antibody TGN1412 trial failure explained by species differences in CD28 expression on CD4+ effector memory T-cells. Br J Pharmacol 2010;161:512–26.

Please cite this article in press as: Chistiakov DA, et al. Regulatory T cells in atherosclerosis and strategies to induce the endogenous atheroprotective immune response. Immunol Lett (2013), http://dx.doi.org/10.1016/j.imlet.2013.01.014

1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528