Mathilde Raverdeau and Kingston H. G. Mills This information is current as of March 26, 2014.

References Subscriptions Permissions Email Alerts

J Immunol 2014; 192:2953-2958; ; doi: 10.4049/jimmunol.1303245 http://www.jimmunol.org/content/192/7/2953

This article cites 92 articles, 48 of which you can access for free at: http://www.jimmunol.org/content/192/7/2953.full#ref-list-1 Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscriptions Submit copyright permission requests at: http://www.aai.org/ji/copyright.html Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/cgi/alerts/etoc

The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 9650 Rockville Pike, Bethesda, MD 20814-3994. Copyright © 2014 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

Modulation of T Cell and Innate Immune Responses by Retinoic Acid

The

Brief Reviews

Journal of

Immunology

Modulation of T Cell and Innate Immune Responses by Retinoic Acid Retinoic acid (RA) is produced by a number of cell types, including macrophages and dendritic cells, which express retinal dehydrogenases that convert vitamin A to its main biologically active metabolite, all-trans RA. All-trans RA binds to its nuclear retinoic acid receptors that are expressed in lymphoid cells and act as transcription factors to regulate cell homing and differentiation. RA production by CD103+ dendritic cells and alveolar macrophages functions with TGF-b to promote conversion of naive T cells into Foxp3+ regulatory T cells and, thereby, maintain mucosal tolerance. Furthermore, RA inhibits the differentiation of naive T cells into Th17 cells. However, Th1 and Th17 responses are constrained during vitamin A deficiency and in nuclear RA receptor a–defective mice. Furthermore, RA promotes effector T cell responses during infection or autoimmune diseases. Thus, RA plays a role in immune homeostasis in the steady-state but activates pathogenic T cells in conditions of inflammation. The Journal of Immunology, 2014, 192: 2953–2958.

V

itamin A or retinol (ROL) is essential for pre- and postnatal development, eyesight, and reproduction, and it plays a crucial role in the maintenance of the immune system (1–4). Exclusively provided through the diet, the different forms of ROL are absorbed by enterocytes and are predominantly stored in the liver (5, 6). In cells, ROL is either stored or hydrolyzed into retinal by ubiquitous alcohol dehydrogenases. A second and irreversible hydrolysis reaction allows the formation of retinoic acid (RA), the main biologically active metabolite of ROL (7). This limiting step is catalyzed by cellspecific retinaldehyde dehydrogenases (RALDH1, RALDH2, and RALDH3) and defines the range of action of RA (8). RALDH enzymes produce two isoforms of RA: all-trans RA (atRA) and 9-cis RA (9cisRA) (9). Both atRA and 9cisRA are autocrine or paracrine ligands of the nuclear RA receptors (RARa, RARb, and RARg), whereas the nuclear rexinoid receptors (RXRa, RXRb, and RXRg) only bind to 9cisRA (10, 11). Nevertheless, 9cisRA has not been detected in vivo, with the Immune Regulation Research Group and Immunology Research Centre, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland Received for publication December 4, 2013. Accepted for publication January 13, 2014.

exception of the pancreas, in which it has been implicated in glucose metabolism (12). It is commonly understood that RARs act as heterodimers with RXRs and that the activity of the RXR is dependent on the activation of their ligand-inducible RAR partners (13). However, more recent studies (14–17) described intra- and extranuclear pathways driven by RARs independently of RXRs. Nevertheless, the majority of known functions of RA in immunity can be attributed to the canonical RAR/RXR pathway, driven by atRA produced by RALDH2 and acting through RARa. In this brief review, we highlight the recent key findings on the effects of vitamin A on T cells and innate immune responses. RA in innate immunity, including its influence on APCs

A number of studies demonstrated that atRA has an important modulating role in innate immunity, with the most recent reports showing that RA has a central function in the differentiation of dendritic cells (DCs), the key APCs for activating naive T cells. Indeed, DCs express the three isotypes of RAR, especially RARa, and are able to respond directly to RA (18). It was shown that RA regulates the development and homeostasis of splenic CD11b+CD8a2CD42Esamhigh DCs and the developmentally related intestinal CD11b+CD103+ DCs but not other splenic or gut DC populations (19, 20). These two DC subtypes are specialized in MHC class II–restricted Ag presentation to CD4+ T cells both in vitro and in vivo and in the differentiation of IL-17–producing CD4+ T cells (Th17 cells) in vivo, respectively (21, 22). However the mechanism by which RA regulates the fate of these DCs remains to be determined. One hypothesis proposed by several groups is that RA influences the Notch-2 pathway; Notch-2 signaling is required for the differentiation of CD11b+CD8a2CD42Esamhigh and CD11b+CD103+ DCs and is influenced by RA during development (22–25). In addition to its impact on commitment of specific DC populations, RA influences the function of DCs. For example, atRA can modulate monocyte-derived DCs toward a mucosaltype DC, which secretes TGF-b and IL-6 and has the capacity to induce gut-homing receptors on T cells (26). atRA also enhances the migratory properties of DCs, which is crucial for their Ag-presenting function during infection. atRA induces Abbreviations used in this article: atRA, all-trans retinoic acid; DC, dendritic cell; ILC, innate lymphoid cell; iTreg, inducible regulatory T cell; MLN, mesenteric lymph node; MMP, metalloproteinase; RA, retinoic acid; RALDH, retinaldehyde dehydrogenase; RAR, retinoic acid receptor; ROL, retinol; RXR, rexinoid receptor; TIMP, tissue inhibitor of matrix metalloproteinase; Treg, regulatory T cell; VAD, vitamin A deficiency.

This work was supported by Science Foundation Ireland. Address correspondence and reprint requests to Dr. Kingston H. G. Mills, Immune Regulation Research Group and Immunology Research Centre, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland. E-mail address: [email protected] www.jimmunol.org/cgi/doi/10.4049/jimmunol.1303245

Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

Mathilde Raverdeau and Kingston H. G. Mills

2954

BRIEF REVIEWS: RETINOIC ACID MODULATION OF IMMUNE RESPONSES

proinflammatory cytokines IL-6, IL-12p70, and TNF-a through a SOCS3-dependent mechanism (18, 45). In contrast, PGE2 appears to be a central negative regulator of RALDH expression in DCs, possibly through the induction of cAMP, a known repressor of RALDH (44). Importantly, recent studies (48, 49) showed a decrease in RALDH expression by intestinal and MLN DCs during infection with Trichuris muris and during colitis induced by T cell transfer in mice. These findings point to a crucial role for the inflammatory milieu in the regulation of RA production during inflammation. However, it is important to note that the influence of RA on the function of DCs in a proinflammatory context depends on the cytokines involved in the inflammation. For example, DePaolo et al. (50) demonstrated that IL-15 acts with RA on DCs to induce the phosphorylation of JNK, leading to the release of the proinflammatory cytokines IL-12p70 and IL-23. Influence of RA on regulatory T cells and effector/pathogenic T cells

A study by Iwata et al. (34) was one of the first to describe a role for RA in the biology of T cells; they found that RA enhances the expression of the gut-homing molecules a4b7 and CCR9 on CD4+ T cells, thus promoting preferential migration into the intestinal lamina propria and GALT. This function is supported by the intestinal RA-producing CD103+ DCs through RARa and RARb but not RARg, whereas the activation of RXR enhances the effect but appears to be dispensable (34, 51). In fact, the RA/RARa pathway directly controls the expression of the integrin subunit a4 (52). Moreover, expression of the integrins a4b7 and a4b1 (also known as VLA-4 and implicated in the migration of T cells into tissues) is greatly decreased on the surface of T cells during a state of vitamin A deficiency (VAD), leading to defective migratory properties of these cells (52). The observation that RA influences T cell migration was followed 3 y later by a major breakthrough on the function of RA in the maintenance of intestinal homeostasis, with the discovery that local RA produced by DCs and macrophages in the intestine and GALT promotes conversion of naive CD4+ T cells into induced regulatory T cells (iTregs) (37, 38, 40, 53). Moreover, RA confers gut-homing properties on the regulatory T cells (Tregs) by increasing their expression of a4b7 and CCR9 (40, 54), partly through a BATF-dependent mechanism (55). In both mice and humans, RA cooperates with TGF-b to promote conversion of naive CD4+ T cells into Foxp3+ iTregs (37, 53, 56), and this effect is mediated, at least in part, through RARa (53, 57). Interestingly, in a mouse model of Crohn’s disease, excessive stocks of vitamin A induced the conventional CCR9+a4b7+Foxp3+ population of Tregs, whereas a state of VAD generated CD103+CCR7+Foxp3+ Tregs; both Treg types helped to control intestinal inflammation (58). The mechanism by which RA promotes iTregs is unclear, and there are a number of possibilities. RA appears to determine whether naive CD4+ T cells differentiate into iTregs or Th17 cells, both of which require TGF-b. atRA inhibits expression of IL-6Ra on naive T cells, which, together with TGF-b, promotes differentiation of naive T cells into Th17 cells; IL-6 has the reciprocal effect of inhibiting the expression of RARa (41, 59, 60). Moreover atRA acts through RARa in activated CD4+ T cells to promote the expression of Foxp3, independently of the cytokine IL-2, the TGF-b–signaling molecules SMAD3

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

expression of the matrix metalloproteinases (MMP) MMP-9 and MMP-14 but not MMP-2, and it decreases the production of their inhibitors (tissue inhibitor of matrix metalloproteinase; TIMP) TIMP-1, TIMP-2, and TIMP-3, as well as the surface expression of the adhesion molecule CD11a on DCs in vitro (27, 28). MMP-9 and MMP-14 are involved in DC migration (29–31), and it was demonstrated that atRA promotes the migration of DCs in vivo toward draining lymph nodes in a murine model of cancer (27). This property of atRA is mediated through RARa, because its repression leads to a reduction in MMP-9 expression, whereas expression of CD11a on DCs and adherence of the CD11c+ DCs are both increased (28, 31). The enhancement of MMP-9 expression by atRA is likely to be mediated directly through the binding of RARa to the regulatory sequences of Mmp9 (31), although further studies are required to confirm this hypothesis. In a proinflammatory context, atRA also has a direct influence on the Ag-presenting capacity of DCs. The positive or negative influence of atRA seems to be dependent on the proinflammatory context and/or the type of DC. During the development of experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis, atRA influences the APC function of splenic DCs by decreasing surface expression of MHC class II and the costimulatory molecules CD80 and CD86 (32). It was also shown that atRA can activate skin DCs called Langerhans cells; in the presence of the proinflammatory cytokines TNF-a and IL-1-b, atRA upregulates MHC class II and CD86 on Langerhans cells and induces binding of NF-kB to its response element on DNA (33). In addition to its influence on DC differentiation, RA is directly produced by DCs; these cells express the RALDH enzymes, in particular RALDH2 (34). The production of RA by DCs was first described for intestinal CD103+ DCs, located mainly in the lamina propria, but also in the associated Peyer’s patches and mesenteric lymph nodes (MLNs) (34–37). Interestingly, the effect of atRA on DCs probably results from the autocrine action of RA produced by DCs themselves (34–37). Nevertheless, there are other sources of RA in the gut, such as intestinal macrophages, which express both RALDH1 and RALDH2 (38). The imprinting of intestinal CD103+ DCs to produce RA is not directly influenced by dietary vitamin A, but instead seems to rely on the stores of ROL and occurs directly in the small intestine (39). A number of studies (37, 39–41) demonstrated the importance of RA production by CD103+ DCs in the gut and its associated lymphoid tissues (GALT). However, RA is also produced in other DC populations, particularly in DCs located at the environmental interfaces, such as the skin, the lungs, and the corresponding draining lymph nodes (42, 43). At the molecular level, RA signaling in DCs is enhanced by RA itself (44, 45), as well as by the cytokines GM-CSF (35, 44), IL-4 (35, 45, 46), IL-13 (44), the MAPK p38a (45, 47), and TLR activation (18, 35, 46). Manicassamy et al. (18) showed that specific activation of the TLR2-signaling pathway induces strong expression of RALDH2 in splenic DCs and, as a consequence, SOCS3 expression, which suppresses the production of proinflammatory cytokines, including IL-23. Moreover, IL-4 enhances expression of RALDH2 and RARb at the transcriptional level, thus creating a positive-control loop on the RA pathway within inflammatory DCs (45). Interestingly, treatment of inflammatory DCs with IL-4 and atRA suppresses production of

The Journal of Immunology

of Foxp3+ Tregs and mediate respiratory tolerance via the production of RA and TGF-b (71). A complementary study (72) showed that lung-resident tissue macrophages coexpress TGF-b and RALDH and have regulatory functions. Furthermore, in a model of experimental allergic asthma, treatment with atRA attenuates airways inflammation by inhibiting Th2 and Th17 responses (73). Despite its role in promoting the differentiation of Tregs required to maintain mucosal homeostasis, recent studies established a central role for RA in the activation of effector CD4+ T cells during inflammation (Fig. 1). Thus, in a proinflammatory context, the RA-signaling pathway in CD4+ T cells is enhanced at the site of inflammation (74). CD4+ T cell effector function and migration to the site of inflammation were inhibited by conditional ablation of RA signaling in T cells (74). Hall et al. (75) showed that the RA–RARa axis is essential for the production of the proinflammatory cytokines IFN-g and IL-17A by Th1 and Th17 cells in response to infection. Moreover, in a proinflammatory context involving cytokines, such as IL-15, RA acts through DCs to decrease conversion of naive T cells into Tregs and to enhance Th1 cell polarization (50). RA also has direct effects on CD8+ T cells; Allie et al. (76) found that the absence of functional RARa resulted in a lack of effector CD8+ T cells, whereas the population of central memory CD8+ T cells appeared to be increased. Importantly, both splenic and MLN DCs are able to enhance the RA pathway in CD8+ T cells (77), and both MLN and intestinal CD103+ DCs induced the RARa-dependent expression of the guthoming molecules on activated CD8+ T cells (77–79). Impact of RA on gd T cells, NK cells, and innate lymphoid cells

In addition to its effect on T cells and DCs, RA modulates innate lymphoid cells (ILCs) and gd T cells at the interface of

X

FIGURE 1. Immunomodulatory effects of RA. In the steady-state, RA produced by CD103+ DCs and alveolar macrophages helps to maintain immune homeostasis and mucosal tolerance by working with TGF-b to promote expansion of Tregs from naive T cells, while inhibiting Th1 and Th17 cells. In addition, RA constrains the inhibitory effect of CD4+CD44high T cells on Tregs. However, in conditions of inflammation (e.g., during autoimmunity or infection), when the inflammatory cytokines IL-1b, IL-12, IL-18, and IL-23 are produced, RA promotes IFN-g production by Th1 cells, IL-17 production by Th17 cells, and IL-22 production by gd T cells. During inflammation, RA also works with IL-15 to decrease conversion of naive T cells into Tregs.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

and SMAD4, and the transcription factors STAT3 and STAT5 (59, 61, 62). Furthermore, RA enhances the expression of arginase 1 in DCs in vitro, an enzyme known to promote the production of Tregs (63). In contrast, atRA decreases T cell expression of RORgt, a transcription factor that is crucial for the differentiation of Th17 cells (60). It was reported recently that RA induces expression of the micro-RNA miR-10a in iTreg, which suppresses the negative effect of Bcl-6 and Ncor-2 on the stability of the iTreg population (64). RA also induces iTreg conversion indirectly through inhibiting a population of CD4+CD44high memory T cells, which block the differentiation of naive T cells into Tregs, by secreting the cytokines IL-4, IL-21, and IFN-g (59, 65, 66). In the absence of RARa-mediated modulation of CD4+CD44high memory T cells, the effect of RA on iTreg conversion is significantly decreased (59, 65). Interestingly, in the GALT, but not in lymph nodes and spleen, activation of CD44high T cells in the absence of TGF-b leads to the expression of the RA-degrading enzyme CYP26B1, which, in turn, inhibits the expression of CCR9 on T cells (67). Surprisingly, although enhancing the proportion of Tregs, both human and murine studies (68, 69) found that RA downregulates the expression of IL-10, an antiinflammatory cytokine often produced by Treg during inflammation. Because RA-producing DCs are found in organs at the environmental interfaces, it is important to note that in the skin a subset of DCs that, unlike RA-producing intestinal DCs, do not express CD103 but are CD11b+CD24+, express RALDH2, and produce RA (42). Moreover, these cells are able to induce Foxp3+ iTregs from naive CD4+ T cells (42). In the liver, hepatic stellate cells produce RA and are able to induce Foxp3+ Tregs in the presence of DCs and TGF-b1 (70). In the lungs, alveolar CD11c+F4/80+ macrophages promote the induction

2955

2956

BRIEF REVIEWS: RETINOIC ACID MODULATION OF IMMUNE RESPONSES

Conclusions

Although the benefits of dietary vitamin A supplementation in controlling child mortality in developing countries is still controversial (88), there is evidence both from human and mouse studies that VAD is associated with defective immune responses to infection or following vaccination (75, 89). These studies led to the conclusion that vitamin A or its metabolite RA plays a role in driving adaptive immunity. Ironically, the early studies (37, 41) on the immunomodulatory functions of RA demonstrated a role, with TGF-b, in promoting the conversion of naive T cells to iTregs. RA produced by gut-resident CD103+ DCs helps to maintain intestinal tolerance by promoting expansion of Tregs in vitro and inhibiting Th17 cells both in vitro and in vivo (41). However, further investigation revealed that, in the context of inflammation, during infection or in autoimmunity, RA has the opposite effect of promoting effector T cell responses (75), explaining the original observations about VAD. Thus, the immunomodulatory effects of RA on the immune system is both tissue and context dependent and, although VAD may curtail effector T cell responses during infection or following vaccination, inhibition of RA binding to its receptor can reduce iTregs, which has the indirect effect of enhancing effector T cell responses during cancer (90). In terms of therapeutic application of the immunomodulatory effects of RA or its inhibitors, we demonstrated that

treatment with atRA is protective against intestinal inflammation induced by gut microbes through enhancement of IL-22 production by gd T cells and group 3 ILCs (80). Furthermore, atRA is protective against experimental autoimmune encephalomyelitis (91), in which autoimmune inflammation is mediated by Th1 and Th17 cells. Conversely, treatment of children with vitamin A can enhance the efficacy of infectious disease vaccines (92). Furthermore, we demonstrated that treatment of mice with an RARa inhibitor can enhance the protective efficacy of a DC vaccine against B16 tumors, suppressing the induction of Tregs and promoting Th1 responses (90). Therefore, modulating RA signaling by dietary supplementation with vitamin A, treatment with agonists, or inhibitors of RAR has potential for enhancing natural and vaccine-induced immunity against infections or cancer, as well as in the treatment of immunemediated diseases in humans.

Disclosures

K.H.G.M. is a cofounder and shareholder in Opsona Therapeutics and TriMod Therapeutics, university spin-out companies from Trinity College involved in the development of immunotherapeutics.

References

1. Warkany, J., and E. Schraffenberger. 1946. Congenital malformations induced in rats by maternal vitamin A deficiency; defects of the eye. Arch. Ophthal. 35: 150–169. 2. Wolbach, S. B., and P. R. Howe. 1925. Tissue Changes Following Deprivation of Fat-Soluble a Vitamin. J. Exp. Med. 42: 753–777. 3. Dowling, J. E., and G. Wald. 1958. Vitamin a Deficiency and Night Blindness. Proc. Natl. Acad. Sci. USA 44: 648–661. 4. Ross, A. C., and C. B. Stephensen. 1996. Vitamin A and retinoids in antiviral responses. FASEB J. 10: 979–985. 5. Harrison, E. H. 2012. Mechanisms involved in the intestinal absorption of dietary vitamin A and provitamin A carotenoids. Biochim. Biophys. Acta 1821: 70–77. 6. Blomhoff, R. 1994. Transport and metabolism of vitamin A. Nutr. Rev. 52: S13–S23. 7. Dowling, J. E., and G. Wald. 1960. The Biological Function of Vitamin A Acid. Proc. Natl. Acad. Sci. USA 46: 587–608. 8. Duester, G., F. A. Mic, and A. Molotkov. 2003. Cytosolic retinoid dehydrogenases govern ubiquitous metabolism of retinol to retinaldehyde followed by tissue-specific metabolism to retinoic acid. Chem. Biol. Interact. 143-144: 201–210. 9. Lin, M., and J. L. Napoli. 2000. cDNA cloning and expression of a human aldehyde dehydrogenase (ALDH) active with 9-cis-retinal and identification of a rat ortholog, ALDH12. J. Biol. Chem. 275: 40106–40112. 10. Durand, B., M. Saunders, P. Leroy, M. Leid, and P. Chambon. 1992. All-trans and 9-cis retinoic acid induction of CRABPII transcription is mediated by RAR-RXR heterodimers bound to DR1 and DR2 repeated motifs. Cell 71: 73–85. 11. Heyman, R. A., D. J. Mangelsdorf, J. A. Dyck, R. B. Stein, G. Eichele, R. M. Evans, and C. Thaller. 1992. 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell 68: 397–406. 12. Kane, M. A., A. E. Folias, A. Pingitore, M. Perri, K. M. Obrochta, C. R. Krois, E. Cione, J. Y. Ryu, and J. L. Napoli. 2010. Identification of 9-cis-retinoic acid as a pancreas-specific autacoid that attenuates glucose-stimulated insulin secretion. Proc. Natl. Acad. Sci. USA 107: 21884–21889. 13. Germain, P., J. Iyer, C. Zechel, and H. Gronemeyer. 2002. Co-regulator recruitment and the mechanism of retinoic acid receptor synergy. Nature 415: 187–192. 14. Dedieu, S., and P. Lefebvre. 2006. Retinoids interfere with the AP1 signalling pathway in human breast cancer cells. Cell. Signal. 18: 889–898. 15. Piskunov, A., and C. Rochette-Egly. 2012. A retinoic acid receptor RARa pool present in membrane lipid rafts forms complexes with G protein aQ to activate p38MAPK. Oncogene 31: 3333–3345. 16. Vernet, N., C. Dennefeld, F. Guillou, P. Chambon, N. B. Ghyselinck, and M. Mark. 2006. Prepubertal testis development relies on retinoic acid but not rexinoid receptors in Sertoli cells. EMBO J. 25: 5816–5825. 17. Chen, N., B. Onisko, and J. L. Napoli. 2008. The nuclear transcription factor RARalpha associates with neuronal RNA granules and suppresses translation. J. Biol. Chem. 283: 20841–20847. 18. Manicassamy, S., R. Ravindran, J. Deng, H. Oluoch, T. L. Denning, S. P. Kasturi, K. M. Rosenthal, B. D. Evavold, and B. Pulendran. 2009. Toll-like receptor 2dependent induction of vitamin A-metabolizing enzymes in dendritic cells promotes T regulatory responses and inhibits autoimmunity. Nat. Med. 15: 401–409. 19. Klebanoff, C. A., S. P. Spencer, P. Torabi-Parizi, J. R. Grainger, R. Roychoudhuri, Y. Ji, M. Sukumar, P. Muranski, C. D. Scott, J. A. Hall, et al. 2013. Retinoic acid controls the homeostasis of pre-cDC-derived splenic and intestinal dendritic cells. J. Exp. Med. 210: 1961–1976. 20. Beijer, M. R., R. Molenaar, G. Goverse, R. E. Mebius, G. Kraal, and J. M. den Haan. 2013. A crucial role for retinoic acid in the development of Notch-dependent murine splenic CD82 CD42 and CD4+ dendritic cells. Eur. J. Immunol. 43: 1608–1616.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

innate and adaptive immunity. Consistent with the effect of CD4+ T cells, we found that RA inhibits IL-17A production by gd T cells but, interestingly, enhances the production of IL-22 by gd T cells and group 3 ILCs stimulated with IL-1b and IL-23 (80). Furthermore, in vivo administration of RA promotes recovery from intestinal inflammation induced by dextran sulfate sodium or following infection with Citrobacter rodentium, and this is associated with enhanced production of IL-22 and the antimicrobial peptides REG3b and REG3g (80). Consistent with the protective effects of atRA, Chenery et al. (81) showed that mice lacking the RA-degrading enzyme CYP26B1 are protected against T cell–induced intestinal inflammation. In a model of autoimmune disease, experimental autoimmune uveitis, administration of atRA is associated with a decrease in expression of the activation markers CD25 and CD69 on pathogenic gd T cells. Furthermore, because atRA reduces IL-17A production by gd T cells (80) and because IL17A and IL-21 produced by gd T cells can activate Th17 cells (82), RA may decrease the ability of gd T cells to activate autoreactive Th17 cells (83). This provides further evidence that RA can have direct and indirect effects on CD4+ effector T cells. Studies in humans (84) showed that the RA–RARa pathway induces expression of the gut-homing integrin a4b7 and its ligand addressin (MAdCAM-1) on circulating Vd2+ gd T cells, while decreasing their production of the skin-homing receptor cutaneous lymphocyte Ag. There is also evidence that RA can influence the function of NK cells. RA enhances expression of the lipid Ag-presenting molecules CD1d on DCs, thereby promoting activation of NKT cells (85). Moreover, the number of circulating NK and NKT cells in humans is positively regulated by the level of the ROL stores (69). In tumor cells, RA also increases the expression of the RA early inducible genes RAE-1 and MICA/B, which activate NK cells through binding to NKG2D (86, 87).

The Journal of Immunology

47. Huang, G., Y. Wang, and H. Chi. 2013. Control of T cell fates and immune tolerance by p38a signaling in mucosal CD103+ dendritic cells. J. Immunol. 191: 650–659. 48. Hurst, R. J., and K. J. Else. 2013. The retinoic acid-producing capacity of gut dendritic cells and macrophages is reduced during persistent T. muris infection. Parasite Immunol. 35: 229–233. 49. Laffont, S., K. R. Siddiqui, and F. Powrie. 2010. Intestinal inflammation abrogates the tolerogenic properties of MLN CD103+ dendritic cells. Eur. J. Immunol. 40: 1877–1883. 50. DePaolo, R. W., V. Abadie, F. Tang, H. Fehlner-Peach, J. A. Hall, W. Wang, E. V. Marietta, D. D. Kasarda, T. A. Waldmann, J. A. Murray, et al. 2011. Coadjuvant effects of retinoic acid and IL-15 induce inflammatory immunity to dietary antigens. Nature 471: 220–224. 51. Takeuchi, H., A. Yokota, Y. Ohoka, H. Kagechika, C. Kato, S. Y. Song, and M. Iwata. 2010. Efficient induction of CCR9 on T cells requires coactivation of retinoic acid receptors and retinoid X receptors (RXRs): exaggerated T cell homing to the intestine by RXR activation with organotins. J. Immunol. 185: 5289–5299. 52. Kang, S. G., J. Park, J. Y. Cho, B. Ulrich, and C. H. Kim. 2011. Complementary roles of retinoic acid and TGF-b1 in coordinated expression of mucosal integrins by T cells. Mucosal Immunol. 4: 66–82. 53. Kang, S. G., H. W. Lim, O. M. Andrisani, H. E. Broxmeyer, and C. H. Kim. 2007. Vitamin A metabolites induce gut-homing FoxP3+ regulatory T cells. J. Immunol. 179: 3724–3733. 54. Siewert, C., A. Menning, J. Dudda, K. Siegmund, U. Lauer, S. Floess, D. J. Campbell, A. Hamann, and J. Huehn. 2007. Induction of organ-selective CD4+ regulatory T cell homing. Eur. J. Immunol. 37: 978–989. 55. Wang, C., S. Thangamani, M. Kim, B. H. Gu, J. H. Lee, E. J. Taparowsky, and C. H. Kim. 2013. BATF is required for normal expression of gut-homing receptors by T helper cells in response to retinoic acid. J. Exp. Med. 210: 475–489. 56. Sun, C. M., J. A. Hall, R. B. Blank, N. Bouladoux, M. Oukka, J. R. Mora, and Y. Belkaid. 2007. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp. Med. 204: 1775–1785. 57. Schambach, F., M. Schupp, M. A. Lazar, and S. L. Reiner. 2007. Activation of retinoic acid receptor-alpha favours regulatory T cell induction at the expense of IL17-secreting T helper cell differentiation. Eur. J. Immunol. 37: 2396–2399. 58. Kang, S. G., C. Wang, S. Matsumoto, and C. H. Kim. 2009. High and low vitamin A therapies induce distinct FoxP3+ T-cell subsets and effectively control intestinal inflammation. Gastroenterology 137: 1391–1402.e1–6. 59. Nolting, J., C. Daniel, S. Reuter, C. Stuelten, P. Li, H. Sucov, B. G. Kim, J. J. Letterio, K. Kretschmer, H. J. Kim, and H. von Boehmer. 2009. Retinoic acid can enhance conversion of naive into regulatory T cells independently of secreted cytokines. J. Exp. Med. 206: 2131–2139. 60. Zhou, X., N. Kong, J. Wang, H. Fan, H. Zou, D. Horwitz, D. Brand, Z. Liu, and S. G. Zheng. 2010. Cutting edge: all-trans retinoic acid sustains the stability and function of natural regulatory T cells in an inflammatory milieu. J. Immunol. 185: 2675–2679. 61. Mucida, D., K. Pino-Lagos, G. Kim, E. Nowak, M. J. Benson, M. Kronenberg, R. J. Noelle, and H. Cheroutre. 2009. Retinoic acid can directly promote TGFbeta-mediated Foxp3(+) Treg cell conversion of naive T cells. Immunity 30: 471– 472, author reply 472–473. 62. Elias, K. M., A. Laurence, T. S. Davidson, G. Stephens, Y. Kanno, E. M. Shevach, and J. J. O’Shea. 2008. Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat-5 independent signaling pathway. Blood 111: 1013–1020. 63. Chang, J., S. Thangamani, M. H. Kim, B. Ulrich, S. M. Morris, Jr., and C. H. Kim. 2013. Retinoic acid promotes the development of Arg1-expressing dendritic cells for the regulation of T-cell differentiation. Eur. J. Immunol. 43: 967–978. 64. Takahashi, H., T. Kanno, S. Nakayamada, K. Hirahara, G. Sciume`, S. A. Muljo, S. Kuchen, R. Casellas, L. Wei, Y. Kanno, and J. J. O’Shea. 2012. TGF-b and retinoic acid induce the microRNA miR-10a, which targets Bcl-6 and constrains the plasticity of helper T cells. Nat. Immunol. 13: 587–595. 65. Hill, J. A., J. A. Hall, C. M. Sun, Q. Cai, N. Ghyselinck, P. Chambon, Y. Belkaid, D. Mathis, and C. Benoist. 2008. Retinoic acid enhances Foxp3 induction indirectly by relieving inhibition from CD4+CD44hi cells. Immunity 29: 758–770. 66. Takeuchi, H., A. Yokota-Nakatsuma, Y. Ohoka, H. Kagechika, C. Kato, S. Y. Song, and M. Iwata. 2013. Retinoid X receptor agonists modulate Foxp3+ regulatory T cell and Th17 cell differentiation with differential dependence on retinoic acid receptor activation. J. Immunol. 191: 3725–3733. 67. Takeuchi, H., A. Yokota, Y. Ohoka, and M. Iwata. 2011. Cyp26b1 regulates retinoic acid-dependent signals in T cells and its expression is inhibited by transforming growth factor-b. PLoS ONE 6: e16089. 68. Maynard, C. L., R. D. Hatton, W. S. Helms, J. R. Oliver, C. B. Stephensen, and C. T. Weaver. 2009. Contrasting roles for all-trans retinoic acid in TGF-betamediated induction of Foxp3 and Il10 genes in developing regulatory T cells. J. Exp. Med. 206: 343–357. 69. Ahmad, S. M., M. J. Haskell, R. Raqib, and C. B. Stephensen. 2009. Markers of innate immune function are associated with vitamin a stores in men. J. Nutr. 139: 377–385. 70. Dunham, R. M., M. Thapa, V. M. Velazquez, E. J. Elrod, T. L. Denning, B. Pulendran, and A. Grakoui. 2013. Hepatic stellate cells preferentially induce Foxp3+ regulatory T cells by production of retinoic acid. J. Immunol. 190: 2009–2016. 71. Coleman, M. M., D. Ruane, B. Moran, P. J. Dunne, J. Keane, and K. H. Mills. 2013. Alveolar macrophages contribute to respiratory tolerance by inducing FoxP3 expression in naive T cells. Am. J. Respir. Cell Mol. Biol. 48: 773–780. 72. Soroosh, P., T. A. Doherty, W. Duan, A. K. Mehta, H. Choi, Y. F. Adams, Z. Mikulski, N. Khorram, P. Rosenthal, D. H. Broide, and M. Croft. 2013. Lungresident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J. Exp. Med. 210: 775–788.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

21. Persson, E. K., H. Uronen-Hansson, M. Semmrich, A. Rivollier, K. Ha¨gerbrand, J. Marsal, S. Gudjonsson, U. Ha˚kansson, B. Reizis, K. Kotarsky, and W. W. Agace. 2013. IRF4 transcription-factor-dependent CD103(+)CD11b(+) dendritic cells drive mucosal T helper 17 cell differentiation. Immunity 38: 958–969. 22. Lewis, K. L., M. L. Caton, M. Bogunovic, M. Greter, L. T. Grajkowska, D. Ng, A. Klinakis, I. F. Charo, S. Jung, J. L. Gommerman, et al. 2011. Notch2 receptor signaling controls functional differentiation of dendritic cells in the spleen and intestine. Immunity 35: 780–791. 23. Wingert, R. A., R. Selleck, J. Yu, H. D. Song, Z. Chen, A. Song, Y. Zhou, B. Thisse, C. Thisse, A. P. McMahon, and A. J. Davidson. 2007. The cdx genes and retinoic acid control the positioning and segmentation of the zebrafish pronephros. PLoS Genet. 3: 1922–1938. 24. Mitsiadis, T. A., M. Lardelli, U. Lendahl, and I. Thesleff. 1995. Expression of Notch 1, 2 and 3 is regulated by epithelial-mesenchymal interactions and retinoic acid in the developing mouse tooth and associated with determination of ameloblast cell fate. J. Cell Biol. 130: 407–418. 25. Ying, M., S. Wang, Y. Sang, P. Sun, B. Lal, C. R. Goodwin, H. Guerrero-Cazares, A. Quinones-Hinojosa, J. Laterra, and S. Xia. 2011. Regulation of glioblastoma stem cells by retinoic acid: role for Notch pathway inhibition. Oncogene 30: 3454– 3467. 26. Saurer, L., K. C. McCullough, and A. Summerfield. 2007. In vitro induction of mucosa-type dendritic cells by all-trans retinoic acid. J. Immunol. 179: 3504–3514. 27. Darmanin, S., J. Chen, S. Zhao, H. Cui, R. Shirkoohi, N. Kubo, Y. Kuge, N. Tamaki, K. Nakagawa, J. Hamada, et al. 2007. All-trans retinoic acid enhances murine dendritic cell migration to draining lymph nodes via the balance of matrix metalloproteinases and their inhibitors. J. Immunol. 179: 4616–4625. 28. Lackey, D. E., S. L. Ashley, A. L. Davis, and K. A. Hoag. 2008. Retinoic acid decreases adherence of murine myeloid dendritic cells and increases production of matrix metalloproteinase-9. J. Nutr. 138: 1512–1519. 29. Ratzinger, G., P. Stoitzner, S. Ebner, M. B. Lutz, G. T. Layton, C. Rainer, R. M. Senior, J. M. Shipley, P. Fritsch, G. Schuler, and N. Romani. 2002. Matrix metalloproteinases 9 and 2 are necessary for the migration of Langerhans cells and dermal dendritic cells from human and murine skin. J. Immunol. 168: 4361–4371. 30. Yang, M. X., X. Qu, B. H. Kong, Q. L. Lam, Q. Q. Shao, B. P. Deng, K. H. Ko, and L. Lu. 2006. Membrane type 1-matrix metalloproteinase is involved in the migration of human monocyte-derived dendritic cells. Immunol. Cell Biol. 84: 557–562. 31. Lackey, D. E., and K. A. Hoag. 2010. Vitamin A upregulates matrix metalloproteinase9 activity by murine myeloid dendritic cells through a nonclassical transcriptional mechanism. J. Nutr. 140: 1502–1508. 32. Zhan, X. X., Y. Liu, J. F. Yang, G. Y. Wang, L. Mu, T. S. Zhang, X. L. Xie, J. H. Wang, Y. M. Liu, Q. F. Kong, et al. 2013. All-trans-retinoic acid ameliorates experimental allergic encephalomyelitis by affecting dendritic cell and monocyte development. Immunology 138: 333–345. 33. Geissmann, F., P. Revy, N. Brousse, Y. Lepelletier, C. Folli, A. Durandy, P. Chambon, and M. Dy. 2003. Retinoids regulate survival and antigen presentation by immature dendritic cells. J. Exp. Med. 198: 623–634. 34. Iwata, M., A. Hirakiyama, Y. Eshima, H. Kagechika, C. Kato, and S. Y. Song. 2004. Retinoic acid imprints gut-homing specificity on T cells. Immunity 21: 527–538. 35. Yokota, A., H. Takeuchi, N. Maeda, Y. Ohoka, C. Kato, S. Y. Song, and M. Iwata. 2009. GM-CSF and IL-4 synergistically trigger dendritic cells to acquire retinoic acid-producing capacity. Int. Immunol. 21: 361–377. 36. Hammerschmidt, S. I., M. Ahrendt, U. Bode, B. Wahl, E. Kremmer, R. Fo¨rster, and O. Pabst. 2008. Stromal mesenteric lymph node cells are essential for the generation of gut-homing T cells in vivo. J. Exp. Med. 205: 2483–2490. 37. Coombes, J. L., K. R. Siddiqui, C. V. Arancibia-Ca´rcamo, J. Hall, C. M. Sun, Y. Belkaid, and F. Powrie. 2007. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic aciddependent mechanism. J. Exp. Med. 204: 1757–1764. 38. Denning, T. L., Y. C. Wang, S. R. Patel, I. R. Williams, and B. Pulendran. 2007. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat. Immunol. 8: 1086–1094. 39. Jaensson-Gyllenba¨ck, E., K. Kotarsky, F. Zapata, E. K. Persson, T. E. Gundersen, R. Blomhoff, and W. W. Agace. 2011. Bile retinoids imprint intestinal CD103+ dendritic cells with the ability to generate gut-tropic T cells. Mucosal Immunol. 4: 438–447. 40. Benson, M. J., K. Pino-Lagos, M. Rosemblatt, and R. J. Noelle. 2007. All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation. J. Exp. Med. 204: 1765–1774. 41. Mucida, D., Y. Park, G. Kim, O. Turovskaya, I. Scott, M. Kronenberg, and H. Cheroutre. 2007. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 317: 256–260. 42. Guilliams, M., K. Crozat, S. Henri, S. Tamoutounour, P. Grenot, E. Devilard, B. de Bovis, L. Alexopoulou, M. Dalod, and B. Malissen. 2010. Skin-draining lymph nodes contain dermis-derived CD103(2) dendritic cells that constitutively produce retinoic acid and induce Foxp3(+) regulatory T cells. Blood 115: 1958–1968. 43. Ruane, D., L. Brane, B. S. Reis, C. Cheong, J. Poles, Y. Do, H. Zhu, K. Velinzon, J. H. Choi, N. Studt, et al. 2013. Lung dendritic cells induce migration of protective T cells to the gastrointestinal tract. J. Exp. Med. 210: 1871–1888. 44. Stock, A., S. Booth, and V. Cerundolo. 2011. Prostaglandin E2 suppresses the differentiation of retinoic acid-producing dendritic cells in mice and humans. J. Exp. Med. 208: 761–773. 45. Zhu, B., T. Buttrick, R. Bassil, C. Zhu, M. Olah, C. Wu, S. Xiao, W. Orent, W. Elyaman, and S. J. Khoury. 2013. IL-4 and retinoic acid synergistically induce regulatory dendritic cells expressing Aldh1a2. J. Immunol. 191: 3139–3151. 46. Elgueta, R., F. E. Sepulveda, F. Vilches, L. Vargas, J. R. Mora, M. R. Bono, and M. Rosemblatt. 2008. Imprinting of CCR9 on CD4 T cells requires IL-4 signaling on mesenteric lymph node dendritic cells. J. Immunol. 180: 6501–6507.

2957

2958

BRIEF REVIEWS: RETINOIC ACID MODULATION OF IMMUNE RESPONSES 83. Liang, D., A. Zuo, H. Shao, W. K. Born, R. L. O’Brien, H. J. Kaplan, and D. Sun. 2013. Retinoic acid inhibits CD25+ dendritic cell expansion and gd T-cell activation in experimental autoimmune uveitis. Invest. Ophthalmol. Vis. Sci. 54: 3493– 3503. 84. McCarthy, N. E., Z. Bashir, A. Vossenka¨mper, C. R. Hedin, E. M. Giles, S. Bhattacharjee, S. G. Brown, T. J. Sanders, K. Whelan, T. T. MacDonald, et al. 2013. Proinflammatory Vd2+ T cells populate the human intestinal mucosa and enhance IFN-g production by colonic ab T cells. J. Immunol. 191: 2752–2763. 85. Szatmari, I., A. Pap, R. R€ uhl, J. X. Ma, P. A. Illarionov, G. S. Besra, E. Rajnavolgyi, B. Dezso, and L. Nagy. 2006. PPARgamma controls CD1d expression by turning on retinoic acid synthesis in developing human dendritic cells. J. Exp. Med. 203: 2351–2362. 86. Cerwenka, A., A. B. Bakker, T. McClanahan, J. Wagner, J. Wu, J. H. Phillips, and L. L. Lanier. 2000. Retinoic acid early inducible genes define a ligand family for the activating NKG2D receptor in mice. Immunity 12: 721–727. 87. Jinushi, M., T. Takehara, T. Tatsumi, T. Kanto, V. Groh, T. Spies, R. Kimura, T. Miyagi, K. Mochizuki, Y. Sasaki, and N. Hayashi. 2003. Expression and role of MICA and MICB in human hepatocellular carcinomas and their regulation by retinoic acid. Int. J. Cancer 104: 354–361. 88. Awasthi, S., R. Peto, S. Read, S. Clark, V. Pande, and D. Bundy; DEVTA (Deworming and Enhanced Vitamin A) team. 2013. Vitamin A supplementation every 6 months with retinol in 1 million pre-school children in north India: DEVTA, a cluster-randomised trial. Lancet 381: 1469–1477. 89. Ahmad, S. M., M. J. Haskell, R. Raqib, and C. B. Stephensen. 2009. Vitamin A status is associated with T-cell responses in Bangladeshi men. Br. J. Nutr. 102: 797– 802. 90. Galvin, K. C., L. Dyck, N. A. Marshall, A. M. Stefanska, K. P. Walsh, B. Moran, S. C. Higgins, L. S. Dungan, and K. H. Mills. 2013. Blocking retinoic acid receptora enhances the efficacy of a dendritic cell vaccine against tumours by suppressing the induction of regulatory T cells. Cancer Immunol. Immunother. 62: 1273–1282. 91. Racke, M. K., D. Burnett, S. H. Pak, P. S. Albert, B. Cannella, C. S. Raine, D. E. McFarlin, and D. E. Scott. 1995. Retinoid treatment of experimental allergic encephalomyelitis. IL-4 production correlates with improved disease course. J. Immunol. 154: 450–458. 92. Sudfeld, C. R., A. M. Navar, and N. A. Halsey. 2010. Effectiveness of measles vaccination and vitamin A treatment. Int. J. Epidemiol. 39(Suppl. 1): i48–i55.

Downloaded from http://www.jimmunol.org/ at UNIVERSITAETS-UND LANDESBIBLIOTHEK DUESSELDORF on March 26, 2014

73. Wu, J., Y. Zhang, Q. Liu, W. Zhong, and Z. Xia. 2013. All-trans retinoic acid attenuates airway inflammation by inhibiting Th2 and Th17 response in experimental allergic asthma. BMC Immunol. 14: 28. 74. Pino-Lagos, K., Y. Guo, C. Brown, M. P. Alexander, R. Elgueta, K. A. Bennett, V. De Vries, E. Nowak, R. Blomhoff, S. Sockanathan, et al. 2011. A retinoic aciddependent checkpoint in the development of CD4+ T cell-mediated immunity. J. Exp. Med. 208: 1767–1775. 75. Hall, J. A., J. L. Cannons, J. R. Grainger, L. M. Dos Santos, T. W. Hand, S. Naik, E. A. Wohlfert, D. B. Chou, G. Oldenhove, M. Robinson, et al. 2011. Essential role for retinoic acid in the promotion of CD4(+) T cell effector responses via retinoic acid receptor alpha. Immunity 34: 435–447. 76. Allie, S. R., W. Zhang, C. Y. Tsai, R. J. Noelle, and E. J. Usherwood. 2013. Critical role for all-trans retinoic acid for optimal effector and effector memory CD8 T cell differentiation. J. Immunol. 190: 2178–2187. 77. Svensson, M., B. Johansson-Lindbom, F. Zapata, E. Jaensson, L. M. Austenaa, R. Blomhoff, and W. W. Agace. 2008. Retinoic acid receptor signaling levels and antigen dose regulate gut homing receptor expression on CD8+ T cells. Mucosal Immunol. 1: 38–48. 78. Eksteen, B., J. R. Mora, E. L. Haughton, N. C. Henderson, L. Lee-Turner, E. J. Villablanca, S. M. Curbishley, A. I. Aspinall, U. H. von Andrian, and D. H. Adams. 2009. Gut homing receptors on CD8 T cells are retinoic acid dependent and not maintained by liver dendritic or stellate cells. Gastroenterology 137: 320–329. 79. Tan, X., J. L. Sande, J. S. Pufnock, J. N. Blattman, and P. D. Greenberg. 2011. Retinoic acid as a vaccine adjuvant enhances CD8+ T cell response and mucosal protection from viral challenge. J. Virol. 85: 8316–8327. 80. Mielke, L. A., S. A. Jones, M. Raverdeau, R. Higgs, A. Stefanska, J. R. Groom, A. Misiak, L. S. Dungan, C. E. Sutton, G. Streubel, et al. 2013. Retinoic acid expression associates with enhanced IL-22 production by gd T cells and innate lymphoid cells and attenuation of intestinal inflammation. J. Exp. Med. 210: 1117–1124. 81. Chenery, A., K. Burrows, F. Antignano, T. M. Underhill, M. Petkovich, and C. Zaph. 2013. The retinoic acid-metabolizing enzyme Cyp26b1 regulates CD4 T cell differentiation and function. PLoS ONE 8: e72308. 82. Sutton, C. E., S. J. Lalor, C. M. Sweeney, C. F. Brereton, E. C. Lavelle, and K. H. Mills. 2009. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity 31: 331–341.

Modulation of T cell and innate immune responses by retinoic Acid.

Retinoic acid (RA) is produced by a number of cell types, including macrophages and dendritic cells, which express retinal dehydrogenases that convert...
1MB Sizes 3 Downloads 3 Views