Peripherally induced tregs - role in immune homeostasis and autoimmunity

Mahesh Yadav, Stephen Stephan, Jeffrey A Bluestone, Mahesh Yadav, Stephen Stephan, Jeffrey A Bluestone

Abstract

Thymically derived Foxp3(+) regulatory T cells (tTregs) constitute a unique T cell lineage that is essential for maintaining immune tolerance to self and immune homeostasis. However, Foxp3 can also be turned on in conventional T cells as a consequence of antigen exposure in the periphery, under both non-inflammatory and inflammatory conditions. These so-called peripheral Tregs (pTregs) participate in the control of immunity at sites of inflammation, especially at the mucosal surfaces. Although numerous studies have assessed in vitro generated Tregs (termed induced or iTregs), these cells most often do not recapitulate the functional or phenotypic characteristics of in vivo generated pTregs. Thus, there are still many unanswered questions regarding the T cell receptor (TCR) repertoire and function of pTregs as well as conditions under which they are generated in vivo, and the degree to which these characteristics identify specialized features of pTregs versus features that are shared with tTregs. In this review, we summarize the current state of our understanding of pTregs and their relationship to the tTreg subset. We describe the recent discovery of unique cell surface markers and transcription factors (including Neuropilin-1 and Helios) that can be used to distinguish tTreg and pTreg subsets in vivo. Additionally, we discuss how the improved ability to distinguish these subsets provided new insights into the biology of tTregs versus pTregs and suggested differences in their function and TCR repertoire, consistent with a unique role of pTregs in certain inflammatory settings. Finally, these recent advances will be used to speculate on the role of individual Treg subsets in both tolerance and autoimmunity.

Keywords: Helios; autoimmunity; immune tolerance; neuropilin-1; regulatory T cell.

Figures

Figure 1
Figure 1
Model depicting the generation and function of tTregs and pTregs. Nrp-1hi tTregs are generated in the thymus and are important in maintaining immune homeostasis and controlling autoimmune responses. During the course of an immune response, Nrp-1lo pTregs are generated in response to Ag presentation by specialized APCs and control effector T cells (Teff) at the site of inflammation. pTregs help in controlling inflammation locally and may be more effective than tTregs at suppressing Teff due to overlapping antigen specificity.
Figure 2
Figure 2
Stability of Treg subsets in MBP.TCR.Tg 1B3 mice using lineage reporter system. The MBP.TCR.Tg mouse when crossed onto RAG−/− background lacks tTregs but generates pTregs in the periphery highlighted by red box (GFP+YFP− subset). The FACS plots depicting expression of GFP and YFP by CD4+ T cells from LNs of 3- to 4-week-old MBP.TCR.Tg.RAG−/−.Foxp3-Cre × R26-YFP or MBP.TCR.Tg.RAG+/−. Foxp3-Cre × R26-YFP mice are shown. In the current gating strategy, GFP+YFP+ population represents the stable Treg subset whereas GFP−YFP+ gate represents unstable Tregs, which previously expressed Foxp3. Cells gated on CD4+ T cells are shown and numbers around the outlined areas indicate percent. Graph on bottom shows the frequency of GFP−YFP+ among YFP+ cells with each symbol representing an individual mouse and bars representing mean values for each group.
Figure 3
Figure 3
Distinct TCR repertoire of pTregs and tTregs. (A) Venn diagram showing distribution of unique and overlapping pTreg, tTreg and Tconv CDR3 sequences. Nrp-1hi tTregs, Nrp-1lo pTregs and CD4+Foxp3− Tconv cells were sorted from MBP.TCR.Tg (1B3)-Foxp3.GFP mice. cDNA was amplified with Vα2-specific primers and amplicons were subcloned and sequenced to determine CDR3 sequences. (B) Frequency of unique CDR3 sequences (identified by peptide number along horizontal axis) in Nrp-1hi tTregs (black bars; top graph), Foxp3− Tconv cells (white bars; top graph) and Nrp-1lo pTregs (gray bars; bottom graph) sorted from 1B3 mice. Data from one representative mouse is presented here.

References

    1. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet (2001) 27:20–110.1038/83713
    1. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet (2001) 27:68–7310.1038/83784
    1. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet (2001) 27:18–2010.1038/83707
    1. Bluestone JA, Abbas AK. Natural versus adaptive regulatory T cells. Nat Rev Immunol (2003) 3:253–710.1038/nri1032
    1. Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol (2013) 14:307–810.1038/ni.2554
    1. 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-beta induction of transcription factor Foxp3. J Exp Med (2003) 198:1875–8610.1084/jem.20030152
    1. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature (2006) 441:235–810.1038/nature04753
    1. Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-beta induces a regulatory phenotype in CD4+CD25− T cells through Foxp3 induction and down-regulation of Smad7. J Immunol (2004) 172:5149–53
    1. Kretschmer K, Apostolou I, Hawiger D, Khazaie K, Nussenzweig MC, Von Boehmer H. Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol (2005) 6:1219–2710.1038/ni1265
    1. Knoechel B, Lohr J, Kahn E, Bluestone JA, Abbas AK. Sequential development of interleukin 2-dependent effector and regulatory T cells in response to endogenous systemic antigen. J Exp Med (2005) 202:1375–8610.1084/jem.20050855
    1. Bilate AM, Lafaille JJ. Induced CD4+Foxp3+ regulatory T cells in immune tolerance. Annu Rev Immunol (2012) 30:733–5810.1146/annurev-immunol-020711-075043
    1. Bonomo A, Kehn PJ, Shevach EM. Post-thymectomy autoimmunity: abnormal T-cell homeostasis. Immunol Today (1995) 16:61–710.1016/0167-5699(95)80089-1
    1. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol (1995) 155:1151–64
    1. Asano M, Toda M, Sakaguchi N, Sakaguchi S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J Exp Med (1996) 184:387–9610.1084/jem.184.2.387
    1. Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B, et al. A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity (2011) 35:109–2210.1016/j.immuni.2011.03.029
    1. Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, et al. Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature (2012) 482:395–910.1038/nature10772
    1. Samstein RM, Josefowicz SZ, Arvey A, Treuting PM, Rudensky AY. Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict. Cell (2012) 150:29–3810.1016/j.cell.2012.05.031
    1. Feuerer M, Jiang W, Holler PD, Satpathy A, Campbell C, Bogue M, et al. Enhanced thymic selection of FoxP3+ regulatory T cells in the NOD mouse model of autoimmune diabetes. Proc Natl Acad Sci U S A (2007) 104:18181–610.1073/pnas.0708899104
    1. Hill JA, Feuerer M, Tash K, Haxhinasto S, Perez J, Melamed R, et al. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity (2007) 27:786–80010.1016/j.immuni.2007.09.010
    1. Feuerer M, Hill JA, Kretschmer K, Von Boehmer H, Mathis D, Benoist C. Genomic definition of multiple ex vivo regulatory T cell subphenotypes. Proc Natl Acad Sci U S A (2010) 107:5919–2410.1073/pnas.1002006107
    1. Kim JM. Molecular mechanisms of regulatory T cell development and suppressive function. Prog Mol Biol Transl Sci (2010) 92:279–31410.1016/S1877-1173(10)92011-5
    1. Curotto de Lafaille MA, Lino AC, Kutchukhidze N, Lafaille JJ. CD25− T cells generate CD25+Foxp3+ regulatory T cells by peripheral expansion. J Immunol (2004) 173:7259–68
    1. Apostolou I, von Boehmer H. In vivo instruction of suppressor commitment in naive T cells. J Exp Med (2004) 199:1401–810.1084/jem.20040249
    1. Curotto de Lafaille MA, Kutchukhidze N, Shen S, Ding Y, Yee H, Lafaille JJ. Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation. Immunity (2008) 29:114–2610.1016/j.immuni.2008.05.010
    1. Haribhai D, Lin W, Edwards B, Ziegelbauer J, Salzman NH, Carlson MR, et al. A central role for induced regulatory T cells in tolerance induction in experimental colitis. J Immunol (2009) 182:3461–810.4049/jimmunol.0802535
    1. Wan YY, Flavell RA. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci U S A (2005) 102:5126–3110.1073/pnas.0501701102
    1. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol (2010) 184:3433–4110.4049/jimmunol.0904028
    1. Akimova T, Beier UH, Wang L, Levine MH, Hancock WW. Helios expression is a marker of T cell activation and proliferation. PLoS ONE (2011) 6:e24226.10.1371/journal.pone.0024226
    1. Gottschalk RA, Corse E, Allison JP. Expression of Helios in peripherally induced foxp3+ regulatory T cells. J Immunol (2012) 188:976–8010.4049/jimmunol.1102964
    1. Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-Bucktrout S, et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med (2012) 209(1713–1722):S1711–910.1084/jem.20120822
    1. Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, et al. Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3+ T reg cells. J Exp Med (2012) 209(1723–1742):S1721.10.1084/jem.20120914
    1. Kim JK, Samaranayake M, Pradhan S. Epigenetic mechanisms in mammals. Cell Mol Life Sci (2009) 66:596–61210.1007/s00018-008-8432-4
    1. Musri MM, Parrizas M. Epigenetic regulation of adipogenesis. Curr Opin Clin Nutr Metab Care (2012) 15:342–910.1097/MCO.0b013e3283546fba
    1. Huehn J, Polansky JK, Hamann A. Epigenetic control of FOXP3 expression: the key to a stable regulatory T-cell lineage? Nat Rev Immunol (2009) 9:83–910.1038/nri2474
    1. Polansky JK, Kretschmer K, Freyer J, Floess S, Garbe A, Baron U, et al. DNA methylation controls Foxp3 gene expression. Eur J Immunol (2008) 38:1654–6310.1002/eji.200838105
    1. Miyao T, Floess S, Setoguchi R, Luche H, Fehling HJ, Waldmann H, et al. Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity (2012) 36:262–7510.1016/j.immuni.2011.12.012
    1. Ohkura N, Hamaguchi M, Morikawa H, Sugimura K, Tanaka A, Ito Y, et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity (2012) 37:785–9910.1016/j.immuni.2012.09.010
    1. Cipolletta D, Feuerer M, Li A, Kamei N, Lee J, Shoelson SE, et al. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature (2012) 486:549–5310.1038/nature11132
    1. Cipolletta D, Kolodin D, Benoist C, Mathis D. Tissular T(regs): a unique population of adipose-tissue-resident Foxp3+CD4+ T cells that impacts organismal metabolism. Semin Immunol (2011) 23:431–710.1016/j.smim.2011.06.002
    1. Feuerer M, Herrero L, Cipolletta D, Naaz A, Wong J, Nayer A, et al. Lean, but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters. Nat Med (2009) 15:930–910.1038/nm.2002
    1. Soroosh P, Doherty TA, Duan W, Mehta AK, Choi H, Adams YF, et al. Lung-resident tissue macrophages generate Foxp3+ regulatory T cells and promote airway tolerance. J Exp Med (2013) 210:775–8810.1084/jem.20121849
    1. Thorstenson KM, Khoruts A. Generation of anergic and potentially immunoregulatory CD25+CD4 T cells in vivo after induction of peripheral tolerance with intravenous or oral antigen. J Immunol (2001) 167:188–95
    1. Tone Y, Furuuchi K, Kojima Y, Tykocinski ML, Greene MI, Tone M. Smad3 and NFAT cooperate to induce Foxp3 expression through its enhancer. Nat Immunol (2008) 9:194–20210.1038/ni1549
    1. Kim JM, Rudensky A. The role of the transcription factor Foxp3 in the development of regulatory T cells. Immunol Rev (2006) 212:86–9810.1111/j.0105-2896.2006.00426.x
    1. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. 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 (2007) 204:1765–7410.1084/jem.20070719
    1. Gottschalk RA, Corse E, Allison JP. TCR ligand density and affinity determine peripheral induction of Foxp3 in vivo. J Exp Med (2010) 207:1701–1110.1084/jem.20091999
    1. Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity (2006) 25:455–7110.1016/j.immuni.2006.07.011
    1. 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) 204:1775–8510.1084/jem.20070602
    1. Mucida D, Kutchukhidze N, Erazo A, Russo M, Lafaille JJ, Curotto de Lafaille MA. Oral tolerance in the absence of naturally occurring Tregs. J Clin Invest (2005) 115:1923–3310.1172/JCI24487
    1. Zheng SG, Wang JH, Stohl W, Kim KS, Gray JD, Horwitz DA. TGF-beta requires CTLA-4 early after T cell activation to induce FoxP3 and generate adaptive CD4+CD25+ regulatory cells. J Immunol (2006) 176:3321–9
    1. Verhagen J, Genolet R, Britton GJ, Stevenson BJ, Sabatos-Peyton CA, Dyson J, et al. CTLA-4 controls the thymic development of both conventional and regulatory T cells through modulation of the TCR repertoire. Proc Natl Acad Sci U S A (2013) 110:E221–3010.1073/pnas.1208573110
    1. Salomon B, Bluestone JA. Complexities of CD28/B7: CTLA-4 costimulatory pathways in autoimmunity and transplantation. Annu Rev Immunol (2001) 19:225–5210.1146/annurev.immunol.19.1.225
    1. Tai X, Cowan M, Feigenbaum L, Singer A. CD28 costimulation of developing thymocytes induces Foxp3 expression and regulatory T cell differentiation independently of interleukin 2. Nat Immunol (2005) 6:152–6210.1038/ni1160
    1. Collette Y, Benziane A, Razanajaona D, Olive D. Distinct regulation of T-cell death by CD28 depending on both its aggregation and T-cell receptor triggering: a role for Fas-FasL. Blood (1998) 92:1350–63
    1. Yamazaki S, Bonito AJ, Spisek R, Dhodapkar M, Inaba K, Steinman RM. Dendritic cells are specialized accessory cells along with TGF− for the differentiation of Foxp3+ CD4+ regulatory T cells from peripheral Foxp3 precursors. Blood (2007) 110:4293–30210.1182/blood-2007-05-088831
    1. Belkaid Y, Oldenhove G. Tuning microenvironments: induction of regulatory T cells by dendritic cells. Immunity (2008) 29:362–7110.1016/j.immuni.2008.08.005
    1. Darrasse-Jeze G, Deroubaix S, Mouquet H, Victora GD, Eisenreich T, Yao KH, et al. Feedback control of regulatory T cell homeostasis by dendritic cells in vivo. J Exp Med (2009) 206:1853–6210.1084/jem.20090746
    1. Mahnke K, Knop J, Enk AH. Induction of tolerogenic DCs: ‘you are what you eat’. Trends Immunol (2003) 24:646–5110.1016/j.it.2003.09.012
    1. Mahnke K, Qian Y, Knop J, Enk AH. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood (2003) 101:4862–910.1182/blood-2002-10-3229
    1. 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-beta and retinoic acid-dependent mechanism. J Exp Med (2007) 204:1757–6410.1084/jem.20070590
    1. Idoyaga J, Fiorese C, Zbytnuik L, Lubkin A, Miller J, Malissen B, et al. Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Invest (2013) 123:844–54
    1. Lombardi V, Speak AO, Kerzerho J, Szely N, Akbari O. CD8alpha(+)beta(−) and CD8alpha(+)beta(+) plasmacytoid dendritic cells induce Foxp3(+) regulatory T cells and prevent the induction of airway hyper-reactivity. Mucosal Immunol (2012) 5:432–4310.1038/mi.2012.20
    1. Gallegos AM, Bevan MJ. Central tolerance to tissue-specific antigens mediated by direct and indirect antigen presentation. J Exp Med (2004) 200:1039–4910.1084/jem.20041457
    1. Watanabe N, Wang YH, Lee HK, Ito T, Cao W, Liu YJ. Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus. Nature (2005) 436:1181–510.1038/nature03886
    1. Aschenbrenner K, D’Cruz LM, Vollmann EH, Hinterberger M, Emmerich J, Swee LK, et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nat Immunol (2007) 8:351–810.1038/ni1444
    1. Coquet JM, Ribot JC, Babala N, Middendorp S, Van Der Horst G, Xiao Y, et al. Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27-CD70 pathway. J Exp Med (2013) 210:715–2810.1084/jem.20112061
    1. Gardner JM, Devoss JJ, Friedman RS, Wong DJ, Tan YX, Zhou X, et al. Deletional tolerance mediated by extrathymic Aire-expressing cells. Science (2008) 321:843–710.1126/science.1159407
    1. Mottet C, Uhlig HH, Powrie F. Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J Immunol (2003) 170:3939–43
    1. Huter EN, Punkosdy GA, Glass DD, Cheng LI, Ward JM, Shevach EM. TGF-beta-induced Foxp3+ regulatory T cells rescue scurfy mice. Eur J Immunol (2008) 38:1814–2110.1002/eji.200838346
    1. DiPaolo RJ, Brinster C, Davidson TS, Andersson J, Glass D, Shevach EM. Autoantigen-specific TGFbeta-induced Foxp3+ regulatory T cells prevent autoimmunity by inhibiting dendritic cells from activating autoreactive T cells. J Immunol (2007) 179:4685–93
    1. Nguyen TL, Sullivan NL, Ebel M, Teague RM, Dipaolo RJ. Antigen-specific TGF-beta-induced regulatory T cells secrete chemokines, regulate T cell trafficking, and suppress ongoing autoimmunity. J Immunol (2011) 187:1745–5310.4049/jimmunol.1004112
    1. Chaudhry A, Rudensky AY. Control of inflammation by integration of environmental cues by regulatory T cells. J Clin Invest (2013) 123:939–4410.1172/JCI57175
    1. Cretney E, Kallies A, Nutt SL. Differentiation and function of Foxp3(+) effector regulatory T cells. Trends Immunol (2013) 34:74–8010.1016/j.it.2012.11.002
    1. Zheng Y, Chaudhry A, Kas A, Deroos P, Kim JM, Chu TT, et al. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature (2009) 458:351–610.1038/nature07674
    1. Chaudhry A, Rudra D, Treuting P, Samstein RM, Liang Y, Kas A, et al. CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science (2009) 326:986–9110.1126/science.1172702
    1. Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol (2009) 10:595–60210.1038/ni.1731
    1. Hsu P, Santner-Nanan B, Dahlstrom JE, Fadia M, Chandra A, Peek M, et al. Altered decidual DC-SIGN+ antigen-presenting cells and impaired regulatory T-cell induction in preeclampsia. Am J Pathol (2012) 181:2149–6010.1016/j.ajpath.2012.08.032
    1. Cerdeira AS, Kopcow HD, Karumanchi SA. Regulatory T cells in preeclampsia: some answers, more questions? Am J Pathol (2012) 181:1900–210.1016/j.ajpath.2012.09.020
    1. Rowe JH, Ertelt JM, Xin L, Way SS. Pregnancy imprints regulatory memory that sustains anergy to fetal antigen. Nature (2012) 490:102–610.1038/nature11462
    1. Glinka Y, Prud’Homme GJ. Neuropilin-1 is a receptor for transforming growth factor beta-1, activates its latent form, and promotes regulatory T cell activity. J Leukoc Biol (2008) 84:302–1010.1189/jlb.0208090
    1. Sarris M, Andersen KG, Randow F, Mayr L, Betz AG. Neuropilin-1 expression on regulatory T cells enhances their interactions with dendritic cells during antigen recognition. Immunity (2008) 28:402–1310.1016/j.immuni.2008.01.012
    1. Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martinez-Llordella M, Ashby M, et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol (2009) 10:1000–710.1038/ni.1774
    1. Zhou L, Chong MM, Littman DR. Plasticity of CD4+ T cell lineage differentiation. Immunity (2009) 30:646–5510.1016/j.immuni.2009.05.001
    1. Zhou X, Bailey-Bucktrout S, Jeker LT, Bluestone JA. Plasticity of CD4(+) FoxP3(+) T cells. Curr Opin Immunol (2009) 21:281–510.1016/j.coi.2009.05.007
    1. Bailey-Bucktrout SL, Bluestone JA. Regulatory T cells: stability revisited. Trends Immunol (2011) 32:301–610.1016/j.it.2011.04.002
    1. Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY. An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nat Immunol (2006) 7:401–1010.1038/ni1318
    1. Pacholczyk R, Ignatowicz H, Kraj P, Ignatowicz L. Origin and T cell receptor diversity of Foxp3+CD4+CD25+ T cells. Immunity (2006) 25:249–5910.1016/j.immuni.2006.05.016
    1. Fazilleau N, Bachelez H, Gougeon ML, Viguier M. Cutting edge: size and diversity of CD4+CD25high Foxp3+ regulatory T cell repertoire in humans: evidence for similarities and partial overlapping with CD4+CD25− T cells. J Immunol (2007) 179:3412–6
    1. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. Peripheral education of the immune system by colonic commensal microbiota. Nature (2011) 478:250–410.1038/nature10434
    1. Zhang L, Nakayama M, Eisenbarth GS. Insulin as an autoantigen in NOD/human diabetes. Curr Opin Immunol (2008) 20:111–810.1016/j.coi.2007.11.005
    1. Levisetti MG, Suri A, Petzold SJ, Unanue ER. The insulin-specific T cells of nonobese diabetic mice recognize a weak MHC-binding segment in more than one form. J Immunol (2007) 178:6051–7
    1. Levisetti MG, Lewis DM, Suri A, Unanue ER. Weak proinsulin peptide-major histocompatibility complexes are targeted in autoimmune diabetes in mice. Diabetes (2008) 57:1852–6010.2337/db08-0068
    1. Stadinski BD, Zhang L, Crawford F, Marrack P, Eisenbarth GS, Kappler JW. Diabetogenic T cells recognize insulin bound to IAg7 in an unexpected, weakly binding register. Proc Natl Acad Sci U S A (2010) 107:10978–8310.1073/pnas.1006545107
    1. Crawford F, Stadinski B, Jin N, Michels A, Nakayama M, Pratt P, et al. Specificity and detection of insulin-reactive CD4+ T cells in type 1 diabetes in the nonobese diabetic (NOD) mouse. Proc Natl Acad Sci U S A (2011) 108:16729–3410.1073/pnas.1113954108
    1. Mohan JF, Petzold SJ, Unanue ER. Register shifting of an insulin peptide-MHC complex allows diabetogenic T cells to escape thymic deletion. J Exp Med (2011) 208:2375–8310.1084/jem.20111502
    1. Mohan JF, Levisetti MG, Calderon B, Herzog JW, Petzold SJ, Unanue ER. Unique autoreactive T cells recognize insulin peptides generated within the islets of Langerhans in autoimmune diabetes. Nat Immunol (2010) 11:350–410.1038/ni.1850
    1. Daniel C, Weigmann B, Bronson R, Von Boehmer H. Prevention of type 1 diabetes in mice by tolerogenic vaccination with a strong agonist insulin mimetope. J Exp Med (2011) 208:1501–1010.1084/jem.20110574
    1. Strauch UG, Obermeier F, Grunwald N, Gurster S, Dunger N, Schultz M, et al. Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis. Gut (2005) 54:1546–5210.1136/gut.2004.059451
    1. Ostman S, Rask C, Wold AE, Hultkrantz S, Telemo E. Impaired regulatory T cell function in germ-free mice. Eur J Immunol (2006) 36:2336–4610.1002/eji.200535244
    1. Ishikawa H, Tanaka K, Maeda Y, Aiba Y, Hata A, Tsuji NM, et al. Effect of intestinal microbiota on the induction of regulatory CD25+ CD4+ T cells. Clin Exp Immunol (2008) 153:127–3510.1111/j.1365-2249.2008.03668.x
    1. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science (2011) 331:337–4110.1126/science.1198469
    1. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell (2009) 139:485–9810.1016/j.cell.2009.09.033
    1. Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A (2010) 107:12204–910.1073/pnas.0909122107
    1. Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science (2011) 332:974–710.1126/science.1206095
    1. Farache J, Koren I, Milo I, Gurevich I, Kim KW, Zigmond E, et al. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity (2013) 38:581–9510.1016/j.immuni.2013.01.009
    1. Farache J, Zigmond E, Shakhar G, Jung S. Contributions of dendritic cells and macrophages to intestinal homeostasis and immune defense. Immunol Cell Biol (2013) 91:232–910.1038/icb.2012.79
    1. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol (2007) 8:1086–9410.1038/ni1511
    1. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity (2009) 30:899–91110.1016/j.immuni.2009.03.019
    1. Battaglia A, Buzzonetti A, Monego G, Peri L, Ferrandina G, Fanfani F, et al. Neuropilin-1 expression identifies a subset of regulatory T cells in human lymph nodes that is modulated by preoperative chemoradiation therapy in cervical cancer. Immunology (2008) 123:129–3810.1111/j.1365-2567.2007.02737.x
    1. Milpied P, Renand A, Bruneau J, Mendes-Da-Cruz DA, Jacquelin S, Asnafi V, et al. Neuropilin-1 is not a marker of human Foxp3+ Treg. Eur J Immunol (2009) 39:1466–7110.1002/eji.200839040
    1. Walker MR, Kasprowicz DJ, Gersuk VH, Benard A, Van Landeghen M, Buckner JH, et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+. J Clin Invest (2003) 112:1437–4310.1172/JCI200319441
    1. Morgan ME, Van Bilsen JH, Bakker AM, Heemskerk B, Schilham MW, Hartgers FC, et al. Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol (2005) 66:13–2010.1016/j.humimm.2004.05.016
    1. Roncador G, Brown PJ, Maestre L, Hue S, Martinez-Torrecuadrada JL, Ling KL, et al. Analysis of FOXP3 protein expression in human CD4+CD25+ regulatory T cells at the single-cell level. Eur J Immunol (2005) 35:1681–9110.1002/eji.200526189
    1. Pillai V, Ortega SB, Wang CK, Karandikar NJ. Transient regulatory T-cells: a state attained by all activated human T-cells. Clin Immunol (2007) 123:18–2910.1016/j.clim.2006.10.014
    1. Gavin MA, Torgerson TR, Houston E, Deroos P, Ho WY, Stray-Pedersen A, et al. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc Natl Acad Sci U S A (2006) 103:6659–6410.1073/pnas.0509484103
    1. Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, Bacchetta R, et al. Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol (2007) 19:345–5410.1093/intimm/dxm014
    1. Wang J, Ioan-Facsinay A, Van Der Voort EI, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol (2007) 37:129–3810.1002/eji.200636435
    1. Tran DQ, Ramsey H, Shevach EM. Induction of FOXP3 expression in naive human CD4+FOXP3− T cells by T cell receptor stimulation is TGF{beta}-dependent but does not confer a regulatory phenotype. Blood (2007) 110:2983–9010.1182/blood-2007-06-094656
    1. McClymont SA, Putnam AL, Lee MR, Esensten JH, Liu W, Hulme MA, et al. Plasticity of human regulatory T cells in healthy subjects and patients with type 1 diabetes. J Immunol (2011) 186:3918–2610.4049/jimmunol.1003099
    1. Duhen T, Duhen R, Lanzavecchia A, Sallusto F, Campbell DJ. Functionally distinct subsets of human Foxp3+ Treg cells that phenotypically mirror effector Th cells. Blood (2012) 119:4430–4010.1182/blood-2011-11-392324
    1. Himmel ME, MacDonald KG, Garcia RV, Steiner TS, Levings MK. Helios+ and Helios− cells coexist within the natural Foxp3+ T regulatory cell subset in humans. J Immunol (2013) 190:2001–810.4049/jimmunol.1201379
    1. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med (2004) 199:971–910.1084/jem.20031579
    1. Beriou G, Costantino CM, Ashley CW, Yang L, Kuchroo VK, Baecher-Allan C, et al. IL-17-producing human peripheral regulatory T cells retain suppressive function. Blood (2009) 113:4240–910.1182/blood-2008-10-183251
    1. Long SA, Cerosaletti K, Bollyky PL, Tatum M, Shilling H, Zhang S, et al. Defects in IL-2R signaling contribute to diminished maintenance of FOXP3 expression in CD4(+)CD25(+) regulatory T-cells of type 1 diabetic subjects. Diabetes (2010) 59:407–1510.2337/db09-0694
    1. Dominguez-Villar M, Baecher-Allan CM, Hafler DA. Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat Med (2011) 17:673–510.1038/nm.2389

Source: PubMed

3
구독하다