Chronic Immune Activation in Systemic Lupus Erythematosus and the Autoimmune PTPN22 Trp620 Risk Allele Drive the Expansion of FOXP3+ Regulatory T Cells and PD-1 Expression

Ricardo C Ferreira, Xaquin Castro Dopico, João J Oliveira, Daniel B Rainbow, Jennie H Yang, Dominik Trzupek, Sarah A Todd, Mhairi McNeill, Maristella Steri, Valeria Orrù, Edoardo Fiorillo, Daniel J M Crouch, Marcin L Pekalski, Francesco Cucca, Tim I Tree, Tim J Vyse, Linda S Wicker, John A Todd, Ricardo C Ferreira, Xaquin Castro Dopico, João J Oliveira, Daniel B Rainbow, Jennie H Yang, Dominik Trzupek, Sarah A Todd, Mhairi McNeill, Maristella Steri, Valeria Orrù, Edoardo Fiorillo, Daniel J M Crouch, Marcin L Pekalski, Francesco Cucca, Tim I Tree, Tim J Vyse, Linda S Wicker, John A Todd

Abstract

In systemic lupus erythematosus (SLE), perturbed immunoregulation underpins a pathogenic imbalance between regulatory and effector CD4+ T-cell activity. However, to date, the characterization of the CD4+ regulatory T cell (Treg) compartment in SLE has yielded conflicting results. Here we show that patients have an increased frequency of CD4+FOXP3+ cells in circulation owing to a specific expansion of thymically-derived FOXP3+HELIOS+ Tregs with a demethylated FOXP3 Treg-specific demethylated region. We found that the Treg expansion was strongly associated with markers of recent immune activation, including PD-1, plasma concentrations of IL-2 and the type I interferon biomarker soluble SIGLEC-1. Since the expression of the negative T-cell signaling molecule PTPN22 is increased and a marker of poor prognosis in SLE, we tested the influence of its missense risk allele Trp620 (rs2476601C>T) on Treg frequency. Trp620 was reproducibly associated with increased frequencies of thymically-derived Tregs in blood, and increased PD-1 expression on both Tregs and effector T cells (Teffs). Our results support the hypothesis that FOXP3+ Tregs are increased in SLE patients as a consequence of a compensatory mechanism in an attempt to regulate pathogenic autoreactive Teff activity. We suggest that restoration of IL-2-mediated homeostatic regulation of FOXP3+ Tregs by IL-2 administration could prevent disease flares rather than treating at the height of a disease flare. Moreover, stimulation of PD-1 with specific agonists, perhaps in combination with low-dose IL-2, could be an effective therapeutic strategy in autoimmune disease and in other immune disorders.

Keywords: FOXP3; PD-1; PTPN22 Arg620Trp; autoimmunity; immunotherapy; regulatory T cells (Tregs); systemic lupus erythematosus (SLE); type I interferon.

Copyright © 2019 Ferreira, Castro Dopico, Oliveira, Rainbow, Yang, Trzupek, Todd, McNeill, Steri, Orrù, Fiorillo, Crouch, Pekalski, Cucca, Tree, Vyse, Wicker and Todd.

Figures

Figure 1
Figure 1
Regulatory CD4+ T cell (Treg) frequency is increased in the circulation of SLE patients. (A) Gating strategy for the delineation of CD4+ regulatory T cells (Tregs). (B,C) Scatter plots depict the frequency (geometric mean ± 95% CI) of CD127lowCD25hi Tregs (B), and CD127lowCD25hi FOXP3+ Tregs (C) in PBMCs isolated from SLE patients (depicted in red) and matching healthy volunteers (depicted in blue). (D) Gating strategy for the delineation of the CD45RA+ naïve and CD45RA− memory CD4+ T-cell compartments. (E,F) Scatter plots depict the frequency (geometric mean ± 95% CI) of memory CD45RA−(E), and naïve CD45RA+(F) CD127lowCD25hi FOXP3+ Tregs. Data were obtained from a discovery clinic-attending cohort (cohort 1) consisting of 34 SLE patients and 24 healthy volunteers, and from a population-based replication cohort (cohort 2) consisting of 41 SLE patients and 112 healthy volunteers. P-values were calculated using two-tailed Student's t-tests comparing the frequency of the assessed immune subsets in patients and controls. SLE, systemic lupus erythematosus; HC, healthy control; ns, not significant.
Figure 2
Figure 2
Frequency of FOXP3+ T cells in SLE patients is increased in both CD25low and CD25hi CD4+ T-cell subsets. (A) gating strategy for the delineation of the CD25lowFOXP3+ Treg subset. (B,C) Scatter plots depict the frequency (geometric mean ± 95% CI) of CD25lowFOXP3+ Tregs in SLE patients (red) and matching healthy volunteers (blue), defined as the frequency of FOXP3+ cells among CD127lowCD25low T cells (B) or as the frequency of CD127lowCD25lowFOXP3+ T cells among total CD4+ T cells (C). (D) Scatter plot depicts the frequency (geometric mean ± 95% CI) of total CD4+FOXP3+ Tregs in SLE patients (red) and matching healthy volunteers (blue), defined as the frequency of FOXP3+ cells among total CD4+ T cells. Data were obtained from a discovery clinic-attending cohort (cohort 1) consisting of 34 SLE patients and 24 healthy volunteers, and from a population-based replication cohort (cohort 2) consisting of 41 SLE patients and 112 healthy volunteers. P-values were calculated using two-tailed Student's t-tests comparing the frequency of the assessed immune subsets in patients and controls. SLE, systemic lupus erythematosus; HC, healthy control.
Figure 3
Figure 3
Expanded FOXP3+ Tregs in SLE patients are predominantly HELIOS+. (A,B) Gating strategy for the delineation of the CD45RA− CD25low(A) and CD25hi(B) Treg subsets stratified by the expression of the canonical Treg transcription factors FOXP3 and HELIOS. (C,D) Scatter plots depict the frequency (geometric mean ± 95% CI) of CD25low FOXP3+HELIOS+(C), and CD25hi FOXP3+HELIOS+(D) Tregs in SLE patients (red) and matched healthy volunteers (blue). (E,F) Scatter plots depict the frequency (geometric mean ± 95% CI) of CD25low FOXP3+HELIOS−(E), and CD25hi FOXP3+HELIOS−(F) Tregs in SLE patients and matched healthy volunteers. Data were obtained from a discovery clinic-attending cohort (cohort 1) consisting of 34 SLE patients and 24 healthy volunteers, and from a population-based replication cohort (cohort 2) consisting of 41 SLE patients and 112 healthy volunteers. P-values were calculated using two-tailed Student's t-tests comparing the frequency of the assessed immune subsets in patients and controls. SLE, systemic lupus erythematosus; HC, healthy control; ns, not significant.
Figure 4
Figure 4
Single-cell RNA-sequencing reveals phenotypic similarities between CD25lowFOXP3+ T cells and conventional CD25hiFOXP3+ Tregs. (A) Uniform manifold approximation projection (UMAP) plot depicting clustering of all captured CD4+ single cells using the combined proteomics and transcriptomics data. Data were extracted from a targeted single-cell RNA-sequencing dataset (ref. 29), and depicts the analysis of pre-sorted CD127lowCD25hi (N = 7,711) and CD127lowCD25low (N = 7,115) T cells from one SLE patient and two control donors, including one type 1 diabetes (T1D) patient and one healthy donor. (B) Expression levels of the CD45RA (black to green) and CD45RO (black to red) isoforms using oligo-conjugated antibodies. (C) Expression of the canonical Treg transcription factors FOXP3 and HELIOS in the identified resting CD4+ T-cell clusters. (D) Detection of FOXP3+ cells (as assessed by the expression of >=1 copy of FOXP3 in each single cell) in either the pre-sorted CD127lowCD25hi (blue) or CD127lowCD25low (red) populations. Relative frequencies of FOXP3+ cells in SLE and control donors is indicated in the figure. (E) Frequency of FOXP3+ cells from either the CD127lowCD25hi or CD127lowCD25low populations in each identified regulatory T cell (Treg) or effector T cell (Teff) cluster. Clusters were ordered according to their relative positioning along the naïve to memory (Mem) differentiation axis. (F,G) Volcano plots depict the differential expression of the assessed genes at the mRNA (N = 397 genes) and protein (N = 24) level between: (i) CD127lowCD25low FOXP3+ vs. CD127lowCD25low FOXP3− T cells (F); and (ii) CD127lowCD25hi FOXP3+ Tregs vs. CD127lowCD25low FOXP3− T cells (G).
Figure 5
Figure 5
Memory FOXP3+HELIOS+ Tregs from SLE patients display hallmarks of recent immune activation. (A–D) Representative histograms and summary scatter plots depict the frequency (geometric mean ± 95% CI) of: (i) PD-1+(A); (ii) CD25 mean fluorescence intensity (B); (iii) TIGIT−(C); and (iv) Ki-67+(D) cells within CD45RA− CD25hi FOXP3+HELIOS+ Tregs. Data were obtained from a discovery clinic-attending cohort (cohort 1) consisting of 34 SLE patients (depicted in red) and 24 healthy volunteers (depicted in blue) and from a population-based replication cohort (cohort 2) consisting of 41 SLE patients and 112 healthy volunteers. Samples from cohorts 1 and 2 were processed within a period of 11 months of each other. For each parameter an illustrative histogram is provided as a reference depicting the expression of the marker in the CD45RA+ FOXP3+HELIOS+ naïve Treg population (depicted in gray). P-values were calculated using two-tailed Student's t-tests comparing the frequency of the assessed immune subsets in patients and controls. SLE, systemic lupus erythematosus; HC, healthy control; ns, not significant.
Figure 6
Figure 6
FOXP3+ Treg expansion is a marker of recent autoimmune reaction. (A,B) Correlation between the frequency of CD25lowFOXP3+ Tregs in blood and the circulating concentration of the plasma type I interferon (IFN) marker soluble SIGLEC-1 (sSIGLEC-1). Data shown depict the correlation in SLE patients (red) and matched healthy volunteers (black) in either the discovery clinic-attending cohort—cohort 1 (A) or the replication population-based cohort—cohort 2 (B). (C,D) Data shown depict the correlation between the total CD4+FOXP3+ CD45RA− memory Tregs and the plasma concentration of sSIGLEC-1 in cohort 1 (C) and cohort 2 (D). (E,F) Data shown depict the correlation between the frequency of CD25lowFOXP3+(E) or total FOXP3+(F) CD45RA− memory Tregs and the plasma concentration of IL-2 measured in the 41 SLE patients from cohort 2. The correlation coefficients (r) and the respective P-values from the linear regression in SLE patients and healthy controls are shown in the plots.
Figure 7
Figure 7
The autoimmunity missense PTPN22 Trp620 risk allele is associated with increased frequency of CD127lowCD25+ Tregs in circulation. (A) Gating strategy depicting the delineation of total CD127lowCD25hi Tregs in blood. Data were obtained by surface immunostaining of freshly isolated whole blood from 486 healthy subjects selected by genotype from the Cambridge BioResource. (B–D) Scatter plots depict the frequency (geometric mean ± 95% CI) of: (i) CD127lowCD25hi Tregs (B); (ii) CD45RA− CD127lowCD25hi memory Tregs (C); and (iii) CD45RA+ CD127lowCD25hi naive Tregs (D) in PTPN22 Arg620/Arg620 (N = 362; red), Arg620/Trp620 (N = 85; blue), and Trp620/Trp620 (N = 39; green) donors. P-values were calculated by linear regression analysis. ns, not significant.
Figure 8
Figure 8
T cells from PTPN22 Trp620/Trp620 donors display an activated phenotype and increased proliferative capacity. (A,B) Scatter plots depict the frequency (geometric mean ± 95% CI) of PD-1+ cells within: (i) CD45RA− CD25hi FOXP3+HELIOS+ Tregs (A); and (ii) CD45RA− CD25int/low Teffs (B) from 152 healthy volunteers recruited from the Cambridge BioResource. Data were stratified according to the genotype of the autoimmune-associated PTPN22 Arg620Trp variant. (C) Treg suppressive capacity was assessed in a subset of 27 common Arg620 homozygous and 24 age- and sex-matched rare Trp620 homozygous donors by measuring the proliferation of autologous CD45RA− CD25int/low T effector cells (mTeffs) in co-culture after in vitro activation for 6 days with anti-CD2/3/28 beads, at a ratio of either 1:1 or 1:0.5 mTeff:Treg (D) Increased proliferation of mTeffs, in the absence of Tregs, from a given donor correlates with reduced suppression of mTeff proliferation when autologous Tregs were present at a ratio of 1 mTeff: 0.5 Treg. (E) Proliferative capacity of CD45RA− memory Teffs and CD45RA+ naïve Teffs was assessed after 6 days in vitro stimulation with anti-CD2/3/28 beads in the absence of Tregs in culture. P-values were calculated using two-tailed student's t-tests comparing the frequency of the assessed immune subsets between the PTPN22 Arg620/Arg620 and Trp620/Trp620 genotype groups. ns, not significant; mTeffs, CD45RA− CD25int/low T effector cells; CPM, proliferation readings—[3H]thymidine incorporation rate (fmoles/g).

References

    1. Tsokos GC. Systemic lupus erythematosus. N Engl J Med. (2011) 365:2110–21. 10.1056/NEJMra1100359
    1. Kaul A, Gordon C, Crow MK, Touma Z, Urowitz MB, van Vollenhoven R, et al. . Systemic lupus erythematosus. Nat Rev Dis Prim. (2016) 2:16039. 10.1038/nrdp.2016.39
    1. Kammer GM. Altered regulation of IL-2 production in systemic lupus erythematosus: an evolving paradigm. J Clin Invest. (2005) 115:836–40. 10.1172/JCI24791
    1. Lieberman LA, Tsokos GC. The IL-2 defect in systemic lupus erythematosus disease has an expansive effect on host immunity. J Biomed Biotechnol. (2010) 2010:740619. 10.1155/2010/740619
    1. Langefeld CD, Ainsworth HC, Graham DSC, Kelly JA, Comeau ME, Marion MC, et al. . Transancestral mapping and genetic load in systemic lupus erythematosus. Nat Commun. (2017) 8:16021. 10.1038/ncomms16021
    1. Onengut-Gumuscu S, Chen W-M, Burren O, Cooper NJ, Quinlan AR, Mychaleckyj JC, et al. . Fine mapping of type 1 diabetes susceptibility loci and evidence for colocalization of causal variants with lymphoid gene enhancers. Nat Genet. (2015) 47:381–6. 10.1038/ng.3245
    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) 30:899–911. 10.1016/j.immuni.2009.03.019
    1. Kim O, Klaus T. Regulatory T cells in systemic lupus erythematosus. Eur J Immunol. (2014) 45:344–55. 10.1002/eji.201344280
    1. La Cava A. Tregs in SLE: an update. Curr Rheumatol Rep. (2018) 20:6. 10.1007/s11926-018-0714-8
    1. Wing JB, Tanaka A, Sakaguchi S. Human FOXP3+ regulatory T cell heterogeneity and function in autoimmunity and cancer. Immunity. (2019) 50:302–16. 10.1016/j.immuni.2019.01.020
    1. Ferreira RC, Simons HZ, Thompson WS, Rainbow DB, Yang X, Cutler AJ, et al. . Cells with Treg-specific FOXP3 demethylation but low CD25 are prevalent in autoimmunity. J Autoimmun. (2017) 84:75–86. 10.1016/j.jaut.2017.07.009
    1. Valencia X, Yarboro C, Illei G, Lipsky PE. Deficient CD4+CD25high T regulatory cell function in patients with active systemic lupus erythematosus. J Immunol. (2007) 178:2579–88. 10.4049/jimmunol.178.4.2579
    1. Bonelli M, Savitskaya A, Steiner C-W, Rath E, Smolen JS, Scheinecker C. Phenotypic and functional analysis of CD4+CD25–Foxp3+ T cells in patients with systemic lupus erythematosus. J Immunol. (2009) 182:1689–95. 10.4049/jimmunol.182.3.1689
    1. Suen J-L, Li H-T, Jong Y-J, Chiang B-L, Yen J-H. Altered homeostasis of CD4+ FoxP3+ regulatory T-cell subpopulations in systemic lupus erythematosus. Immunology. (2009) 127:196–205. 10.1111/j.1365-2567.2008.02937.x
    1. Schneider A, Long SA, Cerosaletti K, Ni CT, Samuels P, Kita M, et al. In active relapsing-remitting multiple sclerosis, effector T cell resistance to adaptive Tregs Involves IL-6–Mediated Signaling. Sci Transl Med. (2013) 5:170ra15 10.1126/scitranslmed.3004970
    1. Stanford SM, Bottini N. PTPN22: the archetypal non-HLA autoimmunity gene. Nat Rev Rheumatol. (2014) 10:602–11. 10.1038/nrrheum.2014.109
    1. McKinney EF, Lyons PA, Carr EJ, Hollis JL, Jayne DRW, Willcocks LC, et al. . A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med. (2010) 16:586–91. 10.1038/nm.2130
    1. Ivashkiv LB. PTPN22 in autoimmunity: different cell and different way. Immunity. (2013) 39:91–3. 10.1016/j.immuni.2013.07.007
    1. Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, Mikulski Z, et al. . The autoimmunity-associated gene PTPN22 potentiates toll-like receptor-driven, type 1 interferon-dependent immunity. Immunity. (2013) 39:111–22. 10.1016/j.immuni.2013.06.013
    1. Vang T, Congia M, Macis MD, Musumeci L, Orrú V, Zavattari P, et al. . Autoimmune-associated lymphoid tyrosine phosphatase is a gain-of-function variant. Nat Genet. (2005) 37:1317–9. 10.1038/ng1673
    1. Zhang J, Zahir N, Jiang Q, Miliotis H, Heyraud S, Meng X, et al. . The autoimmune disease–associated PTPN22 variant promotes calpain-mediated Lyp/Pep degradation associated with lymphocyte and dendritic cell hyperresponsiveness. Nat Genet. (2011) 43:902–7. 10.1038/ng.904
    1. Humrich JY, von Spee-Mayer C, Siegert E, Alexander T, Hiepe F, Radbruch A, et al. . Rapid induction of clinical remission by low-dose interleukin-2 in a patient with refractory SLE. Ann Rheum Dis. (2015) 74:791–2. 10.1136/annrheumdis-2014-206506
    1. von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis. (2016) 75:1407–15. 10.1136/annrheumdis-2015-207776
    1. He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, et al. . Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med. (2016) 22:991–3. 10.1038/nm.4148
    1. Thompson WS, Pekalski ML, Simons HZ, Smyth DJ, Castro-Dopico X, Guo H, et al. . Multi-parametric flow cytometric and genetic investigation of the peripheral B cell compartment in human type 1 diabetes. Clin Exp Immunol. (2014) 177:571–85. 10.1111/cei.12362
    1. Rainbow DB, Yang X, Burren O, Pekalski ML, Smyth DJ, Klarqvist MDR, et al. . Epigenetic analysis of regulatory T cells using multiplex bisulfite sequencing. Eur J Immunol. (2015) 45:3200–3. 10.1002/eji.201545646
    1. Oliveira J, Karrar S, Rainbow D, Pinder C, Clarke P, Rubio Garcia A, et al. . The plasma biomarker soluble SIGLEC-1 is associated with the type I interferon transcriptional signature, ethnic background and renal disease in systemic lupus erythematosus. Arthritis Res Ther. (2018) 20:152. 10.1186/s13075-018-1649-1
    1. Ferreira RC, Rainbow DB, Rubio García A, Pekalski ML, Porter L, Oliveira JJ, et al. . Human IL-6RhiTIGIT– CD4+CD127lowCD25+ T cells display potent in vitro suppressive capacity and display a distinct Th17 profile. Clin Immunol. (2017) 179:25–39. 10.1016/j.clim.2017.03.002
    1. Trzupek D, Dunstan M, Cutler AJ, Lee M, Godfrey L, Aschenbrenner D, et al. Simultaneous mRNA and protein quantification at the single-cell level delineates trajectories of CD4+ T-cell differentiation. bioRxiv. (2019) 706275 10.1101/706275
    1. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, et al. . Comprehensive integration of single-cell data. Cell. (2019) 177:1888–902.e21. 10.1016/j.cell.2019.05.031
    1. Finak G, McDavid A, Yajima M, Deng J, Gersuk V, Shalek AK, et al. . MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. (2015) 16:278. 10.1186/s13059-015-0844-5
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, et al. . PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. (2007) 81:559–75. 10.1086/519795
    1. Kim H-J, Barnitz RA, Kreslavsky T, Brown FD, Moffett H, Lemieux ME, et al. . Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science. (2015) 350:334–9. 10.1126/science.aad0616
    1. Võsa U, Claringbould A, Westra H-J, Bonder MJ, Deelen P, Zeng B, et al. Unraveling the polygenic architecture of complex traits using blood eQTL metaanalysis. bioRxiv. (2018) 447367 10.1101/447367
    1. Ferreira RC, Rainbow DB, Rubio García A, Pekalski ML, Porter L, Oliveira JJ, et al. . In-depth immunophenotyping of IL-6R on the human peripheral regulatory T cell (Treg) compartment. Data Br. (2017) 12:676–91. 10.1016/j.dib.2017.04.043
    1. Alexander T, Sattler A, Templin L, Kohler S, Groß C, Meisel A, et al. . Foxp3+ Helios+ regulatory T cells are expanded in active systemic lupus erythematosus. Ann Rheum Dis. (2013) 72:1549–58. 10.1136/annrheumdis-2012-202216
    1. Golding A, Hasni S, Illei G, Shevach EM. The percentage of FoxP3+Helios+ Treg cells correlates positively with disease activity in systemic lupus erythematosus. Arthritis Rheum. (2013) 65:2898–906. 10.1002/art.38119
    1. Todd JA, Evangelou M, Cutler AJ, Pekalski ML, Walker NM, Stevens HE, et al. . Regulatory T cell responses in participants with type 1 diabetes after a single dose of interleukin-2: a non-randomised, open label, adaptive dose-finding trial. PLoS Med. (2016) 13:e1002139. 10.1371/journal.pmed.1002139
    1. Liu W, Putnam AL, Xu-yu Z, Szot GL, Lee MR, Zhu S, et al. . CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+T reg cells. J Exp Med. (2006) 203:1701–11. 10.1084/jem.20060772
    1. Srivastava S, Koch MA, Pepper M, Campbell DJ. Type I interferons directly inhibit regulatory T cells to allow optimal antiviral T cell responses during acute LCMV infection. J Exp Med. (2014) 211:961–74. 10.1084/jem.20131556
    1. Bacher N, Raker V, Hofmann C, Graulich E, Schwenk M, Baumgrass R, et al. . Interferon-α suppresses cAMP to disarm human regulatory T cells. Cancer Res. (2013) 73:5647–56. 10.1158/0008-5472.CAN-12-3788
    1. Yan B, Ye S, Chen G, Kuang M, Shen N, Chen S. Dysfunctional CD4+,CD25+ regulatory T cells in untreated active systemic lupus erythematosus secondary to interferon-α-producing antigen-presenting cells. Arthritis Rheum. (2008) 58:801–12. 10.1002/art.23268
    1. Golding A, Rosen A, Petri M, Akhter E, Andrade F. Interferon-alpha regulates the dynamic balance between human activated regulatory and effector T cells: implications for antiviral and autoimmune responses. Immunology. (2010) 131:107–17. 10.1111/j.1365-2567.2010.03280.x
    1. Crow MK. Type I interferon in organ-targeted autoimmune and inflammatory diseases. Arthritis Res Ther. (2010) 12:S5. 10.1186/ar2886
    1. Feng H, Yin J, Han Y-P, Zhou X-Y, Chen S, Yang L, et al. . Sustained changes of Treg and Th17 cells during interferon-α therapy in patients with chronic hepatitis B. Viral Immunol. (2015) 28:412–7. 10.1089/vim.2015.0024
    1. Gómez-Martín D, Díaz-Zamudio M, Crispín JC, Alcocer-Varela J. Interleukin 2 and systemic lupus erythematosus: beyond the transcriptional regulatory net abnormalities. Autoimmun Rev. (2009) 9:34–9. 10.1016/j.autrev.2009.02.035
    1. Alcocer-Varela J, Alarcón-Segovia D. Longitudinal study on the production of and cellular response to interleukin-2 in patients with systemic lupus erythematocus. Rheumatol Int. (1995) 15:57–63. 10.1007/BF00262709
    1. Huang YP, Miescher PA, Zubler RH. The interleukin 2 secretion defect in vitro in systemic lupus erythematosus is reversible in rested cultured T cells. J Immunol. (1986) 137:3515–20.
    1. Giang S, La Cava A. Regulatory T cells in SLE: biology and use in treatment. Curr Rheumatol Rep. (2016) 18:67. 10.1007/s11926-016-0616-6
    1. Mizui M, Tsokos GC. Targeting regulatory T cells to treat patients with systemic lupus erythematosus. Front Immunol. (2018) 9:786. 10.3389/fimmu.2018.00786
    1. Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, et al. . Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med. (2011) 365:2067–77. 10.1056/NEJMoa1105143
    1. Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K, Brohawn P, et al. . Anifrolumab, an anti–interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol. (2016) 69:376–86. 10.1002/art.39962
    1. Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei GG, et al. . Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: a randomised, double-blind, placebo-controlled study. Ann Rheum Dis. (2016) 75:1909–16. 10.1136/annrheumdis-2015-208562
    1. Paluch C, Santos AM, Anzilotti C, Cornall RJ, Davis SJ. Immune checkpoints as therapeutic targets in autoimmunity. Front Immunol. (2018) 9:2306. 10.3389/fimmu.2018.02306
    1. Asano T, Meguri Y, Yoshioka T, Kishi Y, Iwamoto M, Nakamura M, et al. PD-1 modulates regulatory T cell homeostasis during low-dose IL-2 therapy. Blood. (2017) 129:2186–97. 10.1182/blood-2016-09-741629
    1. Stathopoulou C, Gangaplara A, Mallett G, Flomerfelt FA, Liniany LP, Knight D, et al. . PD-1 inhibitory receptor downregulates asparaginyl endopeptidase and maintains Foxp3 transcription factor stability in induced regulatory T cells. Immunity. (2018) 49:247–63.e7. 10.1016/j.immuni.2018.05.006

Source: PubMed

3
Subskrybuj