Chapter 5
T Cells in Autoimmune Diseases Amir Sharabi1,2, George C. Tsokos1 1Division
of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; 2Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
INTRODUCTION T cells develop in the thymus from bone marrow–derived hematopoietic cell precursor, and during their development they acquire their T cell receptor (TCR) that is educated not to recognize self-antigens. During this selection process, the T cells gain and lose the expression of CD4 and CD8 co-receptors that eventually result in the generation of conventional T cells and T regulatory (Treg) cells that can be sent out to the periphery [1]. However, some autoreactive T cells do leak into the periphery as T cell selection in the thymus is not protected totally from mistakes. Furthermore, in circumstances of deletion of genes that are critical for T cell development in the thymus, such as phosphatase and tensin homolog (PTEN), a potent negative regulator of PI3K signaling [2], and zeta-chain-associated protein kinase 70 (ZAP-70) that is critical for the depletion of autoreactive T cells [3], autoimmune diseases will ultimately develop. The thymic autoreactive T cells, which escape to the periphery, will be later controlled by peripheral tolerance mechanisms able to prevent autoreactive T cells from inducing pathogenic immune responses in target organs. The breakdown of peripheral T cell tolerance to self-antigens enables autoreactive T cells to become active and mediate the effector immune responses through inflammation and support to autoantibody-producing B cells.
GENETIC PREDISPOSITION AND T CELL GENETIC VARIANTS IN AUTOIMMUNE DISEASES Genome-wide association studies have shown that the susceptibility for developing autoimmune diseases is rather polygenic with multiple genetic variants [4]. The highest genetic risk derives from MHC loci, which indicates a key role for antigen recognition by T cells for them to become pathogenic. Additional genetic variants include CD25 (IL-2 receptor, α chain), IL-2, CTLA-4, serine/threonine protein phosphatase 2A (PP2A), and tyrosine protein phosphatase [5,6]. Alternative splicing of T cell genes gives rise to several splice gene variants that are pathogenic. Alternative splicing of the CD3 zeta mRNA in systemic lupus erythematosus (SLE) T cells results in an unstable variant, which contributes to abnormalities with TCR signaling [7]. Furthermore, CD44v3 and CD44v6 are common splice isoforms of the CD44 gene in SLE T cells, which have been shown to correlate with the extent of disease activity, the presence of lupus nephritis, and with positivity for anti-dsDNA antibodies [8]. CD44 variant isoforms are important for the homing capacity of T cells to target organs. In addition, the products of alternative spliced CREM gene, cAMP responsive element modulator alpha (CREMα) and cAMP early repressor I (ICER), represent transcriptional repressors of certain functional repressions. Lupus-prone B6.lpr mice deficient of ICER/CREM are protected from systemic and organ specific autoimmunity [9].
T CELLS IN THE PATHOGENESIS OF AUTOIMMUNE DISEASES There are different subsets of T cells that are defined by their TCR composition (e.g., αβ chains or γδ chains), the expression of co-receptors, namely CD4 for T helper (Th) cells and CD8 for T cytotoxic (Tc) cells, and the expression of master genes, transcription factors, and cytokines. Most of T cell subsets have a role in the pathogenesis of autoimmune diseases (Fig. 5.1), either because of the expression and production of pathogenic cytokines or because of impaired or excessive function.
Th17 Cells CD4 T cells that express the transcription factor ROR-γt produce on stimulation the proinflammatory cytokines IL-17, IL-21, IL-22, IFN-γ, and Granulocyte-monocyte colony-stimulating factor (GM-CSF), all of which promote the development of autoimmune diseases in mice and humans [10,11]. In target organs they generate ectopic lymphoid follicles [12], wherein B cells are activated and become autoreactive [13,14]. Mosaic of Autoimmunity. https://doi.org/10.1016/B978-0-12-814307-0.00005-0 Copyright © 2019 Elsevier Inc. All rights reserved.
29
30 SECTION | II Cellular and Molecular Mechanisms
FIGURE 5.1 T cell subsets playing role in autoimmune disease.
Double-Negative T Cells These cells are αβ TCR T cells that do not express CD4 and CD8. Their origin is not clear; however, because they share genes with those of CD8 T cells, it is suggested that at least some of them originate from autoreactive CD8+ T cells that were stimulated continuously [15–17]. In mice, the repression of CD8 locus is triggered by engagement of these cells with autoantigens [18]. In patients with SLE, CREMα mediates CD8 repression through binding to the CD8 locus and the recruitment of HDAC1 and DNMT1, which turn the chromatin inaccessible [19]. In several autoimmune diseases, doublenegative (DN) T cells are expanded and may comprise 1%–5% of peripheral T cells. They produce IL-17 and can help B cells to produce autoantibody. IL-17–producing DN T cells accumulate in the kidney and probably other tissues [20–23].
γδ T Cells These cells represent a minor T cell population that has adaptive and innate-like characteristics important for mucosal immunity [24]. They can present antigen, secrete inflammatory cytokines, promote antibody production, and inhibit Treg cells, thereby playing a pathogenic role in autoimmune diseases [25]. Stress molecules such as those of the HSP60 family augment γδ T cell activity. γδ T cells affect dendritic cells to produce IL-17 resulting in inflammatory cell infiltration in the kidney, and depletion of γδ T cells can diminish this infiltration [26–28].
T Follicular Helper Cells These are CD4 T cells that express the transcription factor Bcl6 and the chemokine receptor CXCR5, which enable them to migrate into germinal centers. Within the germinal centers of lymph nodes and kidneys, T follicular helper (Tfh) cells help the activation of B cells and lead to differentiation of long-lived plasma cells. This process yields the production of high-affinity autoantibodies as it leads to immunoglobulin class switching [29–32]. Tfh cells are frequent in autoimmune
T Cells in Autoimmune Diseases Chapter | 5 31
diseases, and their pathogenic role in SLE has been demonstrated in murine models [31]. In patients with SLE, the number of Tfh cells correlates with both the numbers of plasmablasts and the titers of anti-dsDNA autoantibodies. Interestingly, a subset of Tfh cells in SLE patients was shown to secrete IL-17 and promote B cell activation in the kidney leading to the development of nephritis [29].
Th1 Cells These are CD4 T cells that express the αβ TCR and IFN-γ as their signature cytokine. These are the pathogenic cells that mediate the development of diffuse proliferative lupus nephritis and crescentic glomerulonephritis, anti–GBM-induced glomerulonephritis, and antineutrophil cytoplasmic antibody (ANCA)–associated vasculitis [33–35].
Th2 Cells These are CD4 T cells that express the αβ TCR and produce IL-4 as their signature cytokine. IL-4 activates signal transducer and activator of transcription 6 (STAT6) and leads to the expression of their principle transcription factor, GATA. They also secrete IL-5, IL-6, IL-9, IL-13, and IL-17E that support the humoral response. Th2 cells mediate the development of membranous lupus nephritis in MRL/lpr mice whose Th1 response is genetically deleted [37]. It is noteworthy that in addition to the effects of Th cells on B cells, BAFF and APRIL are B cell survival and growth factors that contribute to the pathogenicity of B cells, and patients with autoimmune diseases produce high levels of these factors [36].
Th9 Cells Th9 cells secrete IL-9 and require TGF-β and IL-4 for their induction [38,39]. They express Foxp3 and IL-4 but at much lower levels than Tregs and Th2 cells, respectively. TGF-β promotes the development of Th9 cells in both mice and humans through the induction of the ETS-transcription factor PU.1 [40,41]. PU.1 binds to the IL-9 promoter and promotes Th9 subset polarization. IL-4 activates STAT6, which stimulates an increase of IL-9 in Th9 cells. MRL/lpr mice with established lupus have expanded Th9 cells in their spleens and kidneys, which correlate with the production of anti-dsDNA antibodies [42]. In addition, experimental autoimmune encephalomyelitis (EAE) could be induced by Th9 cells and IL-9, and the inflammation in the central nervous system was shown to be independent of Th1 and Th17 cells [43].
CD8 T Cells In addition to their cytotoxic role they have against intracellular pathogens and tumors, CD8 T cells are known for their ability to regulate autoimmune and allergic diseases [44]. In experimental autoimmune glomerulonephritis, depletion of CD8+ T cells reduced the severity of disease [45]. Furthermore, in SLE and ANCA-associated vasculitis, an altered gene expression profile in CD8+ T cells was shown to correlate with a negative disease outcome [46]. Also, patients with ANCAassociated nephritis and lupus nephritis have a higher urinary CD8 to CD4 T cell ratio [47].
CD4 Treg Cells Among the CD4 and CD8 T cells, the most studied Treg cell population resides within the CD4 T cells that express the αβ TCR and the master gene Foxp3. Those CD4 Treg cells develop in the thymus and have typical characteristics that enable them to suppress the immune response [48]. CD4 Treg cells also play a role in tissue repair [49,50]. Defects in several signaling molecules and pathways were shown to hamper Treg cell function, including PP2A, mechanistic target of rapamycin (mTOR) C1, PI3 phosphatase, PTEN, and calcium/calmodulin-dependent protein kinase IV (CaMKIV) [49]. In autoimmune diseases, both decreased numbers and compromised function have been implicated [51,52].
CD8 Treg Cells These are CD8 T cells of multiple subsets that have suppressive capabilities. Their number, function, or both are impaired in patients with autoimmune diseases and in murine models of autoimmunity [53]. Frequent subsets of CD8 Treg cells express low levels of CD28. CD8 knockout mice that were induced with EAE improved their outcome following the adoptive transfer of CD8+CD28low T cells from wild-type animals [54]. Furthermore, in murine SLE, treatment with tolerogenic peptides ameliorated the disease manifestations through the induction of CD8 and CD4 Treg cells [55,56], and CD8 Treg cells were essential for the optimal function of CD4 Treg cells [56,57], suggesting a cross talk between CD8 and CD4 Treg cells.
32 SECTION | II Cellular and Molecular Mechanisms
MOLECULAR PATHWAYS IN PATHOGENIC AUTOREACTIVE T CELLS Central pathways in T cell development in the thymus, which result in the development of autoimmune diseases, are beyond the scope of this manuscript, but mostly related to interference with the process of educating the TCR recognition of selfantigens and the deletion of autoreactive T cells [1–3]. The main molecular pathways in the periphery that participate in the induction and activation of autoreactive T cells are described here (Fig. 5.2).
CD3 Zeta This T cell surface molecule is part of the CD3 complex, which transduces the signaling from TCR following antigen recognition. Single-nucleotide polymorphisms in the CD3 zeta locus affect its expression, and it is downregulated in several autoimmune diseases [58–60]. Consequently, decreased expression of CD3 zeta can lead to the migration and accumulation of peripheral T cells in tissues where they produce IFN-γ and IL-17 and trigger inflammation [61–63]. T cells with decreased CD3 zeta expression have a higher capacity to infiltrate tissues partly because they overexpress the adhesion molecule CD44 [64]. T cells from patients with SLE also express elevated levels of CD44 and specifically v6 variant of CD44, which has been associated with lupus nephritis [8].
Rho-Associated Protein Kinase This is a serine/threonine kinase that phosphorylates ezrin/radixin/moesin (ERM) protein complex for the regulation of cell migration [65], and the CD44–Rho-associated protein kinase (ROCK)–ERM axis is activated in patients with autoimmune diseases, including SLE [66–68]. Inflammatory conditions for mesangial cells and podocytes also result in ROCK activation [69]. Rock activation can also result in the development of Th17 cells [68].
Protein Phosphatase 2A This is a serine/threonine phosphatase composed of three subunits (each with different isoforms), which is involved in pivotal cellular reactions and responses, including transcription, and cell migration and survival [70]. PP2A-beta subunit plays a role in the survival of autoreactive T cells as it controls T cell apoptosis following IL-2 deprivation, and T cells from patients with SLE have decreased levels of PP2A-beta subunit [71]. In Treg cells, however, it is required for their development through the suppression of mTORC1 [72]. Interestingly, mice whose T cells overexpress PP2A do not develop autoimmune syndrome spontaneously, yet on challenge with anti-GBM antibody, they develop glomerulonephritis that is responsive to treatment with anti–IL-17 antibody [73].
FIGURE 5.2 Molecular pathways in T cells related to autoimmune disease.
T Cells in Autoimmune Diseases Chapter | 5 33
CREM This is a family of transcription factors that become activated following intracellular increase in cAMP. Two isoforms, CREMα and ICER, play a role in T cell differentiation, epigenetic-regulated suppression of IL-2, and regulation of CD8 expression by CREMα [9,19,74–78]. CREMα is increased in SLE T cells and represses IL-2 transcription through at least three pathways: (1) binding to CREB-binding site to withhold the transcription initiation machinery, (2) the recruitment of HDAC1 and DNMT-3a to impose epigenetic closure of the IL-2 locus, and (3) binding and repressing c-fos to reduce AP-1 formation required for IL-2 transcription [74,75]. CREMα together with ICER result in demethylation and acetylation of the IL-17 promoter, processes that upregulate IL-17 transcription [9,74,75].
Calcium/Calmodulin-Dependent Protein Kinase IV This is a serine/threonine kinase that regulates gene expression through transcription factors associated with activation and development of T cell subsets. In SLE patients and lupus-prone mice, the expression of CaMKIV is increased in T cells following their activation [77,78]. This results in decreased IL-2 production and compromised Treg cell function [77]. Specifically, in SLE T cells, CaMKIV enhances the binding of CREMα to the IL-2 promoter and reduces its activity [78]. Depletion of CaMKIV in T cells from both mice and human with SLE recovers IL-2 production and Treg cell differentiation [77]. CaMKIV also promotes Th17 cell differentiation through the Akt/mTOR pathway [79] and the recruitment of these cells into tissues, including the kidneys [80], where it affects the function of mesangial cells [81] and podocytes [82].
Mechanistic Target of Rapamycin Pathway mTOR is a serine/threonine kinase that regulates T cell activation, proliferation, and survival, and it is frequently activated in autoimmune diseases [83]. There are two major mTOR complexes, namely mTORC1 and mTORC2, and the former plays a role in Th1 and Th17 cell differentiation, whereas the latter is essential for Th2 differentiation [84]. In SLE, the mTORC1 is activated by PI3K/Akt pathway, CaMKIV, and ROCK [68,79,85], which impairs Treg cells and increases the development and tissue recruitment of Th1 and Th17 cells to target organs [86].
Signal Transducer and Activator of Transcription This is a family of signaling molecules downstream the cytokine receptors that dictate the differentiation and function of immune cells, including T cells. Following the engagement of a cytokine receptor, STAT molecules are phosphorylated by janus kinases and become activated. For instance, STAT4 promotes Th1 cell differentiation in response to IL-12; STAT6 promotes Th2 cell differentiation in response to IL-4; STAT3 promotes Th17 or Tfh cells in response to IL-6, IL-21, IL-23, and TGF-β; and STAT5 promotes Treg cells in response to IL-2 and TGF-β. These differentiation events of T cells are regulated by mTOR pathway [87]. SLE T cells express high levels of STAT3 [88], and STAT3-induced IL-17 production and Tfh cell differentiation are required for infiltration and accumulation of immune complexes in murine lupus nephritis [89,90].
EPIGENETICS AND AUTOIMMUNITY Epigenetic pathways enable cells to make functional adjustments through the expression or repression of genes in response to a variety of stimuli ranging from cell surface molecules to signaling molecules. Methylation of DNA CpGs and histone modifications are common epigenetic mechanisms, and gene expression is increased when DNA is hypomethylated [91]. In SLE, genes that are hypomethylated and play a pathogenic role include CD70, CD11A, CD40L, IL-4, IL-10, IL-13, and IL-17 [91,92]. Furthermore, PP2A gene is hypomethylated in SLE T cells [93], and increased activity of PP2A results in ERK/MEK dephosphorylation and decreased DNMT1 activity with a resultant hypomethylation of the pathogenic molecules, CD70 and CD11a [94].
CONCLUDING REMARKS The study of T cells in patients with systemic autoimmunity and lupus-prone mice has generated novel insights into the pathogenesis of the disease and more importantly in the expression of organ damage. Study of T cell subsets has demonstrated the poor function of Treg cells and the poor control of the autoimmune and the inflammatory responses. Recognition of the expansion of DN T cells has provided insight into their ability to help B cells to produce anti-dsDNA antibodies and to produce the proinflammatory cytokine IL-17 in the periphery and when they infiltrate the kidney. Study of the CD8
34 SECTION | II Cellular and Molecular Mechanisms
cell subset and their poor cytotoxic function has provided a better understanding of the inability of SLE patients to fend off infections. More importantly, detailed studies of the biochemistry of T cell signaling events and gene transcription processes have unveiled a number of plausible treatment targets and biomarkers of disease activity. For example, a ROCK inhibitor is in Phase II trial, and mTOR inhibitors have been considered enthusiastically along with Syk and CaMKIV inhibitors. Low-dose IL-2 has been extolled in uncontrolled reports, whereas an anti-p40 (IL-23) monoclonal antibody has completed a successful Phase II trial. In parallel, a number of surface molecules (CD44v3 or C6) or gene expression profiles have been shown to have biomarker value. Yet, T cells in patients with SLE and other autoimmune diseases present enormous complexity, and continuous intense studies will reveal not only novel targets and biomarkers but also potential to develop tools toward personalized medicine.
REFERENCES [1] Klein L, Kyewski B, Allen PM, Hogquist KA. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat Rev Immunol 2014;14:377–91. [2] Liu X, Karnell JL, Yin B, Zhang R, Zhang J, Li P, Choi Y, Maltzman JS, Pear WS, Bassing CH, Turka LA. Distinct roles for PTEN in prevention of T cell lymphoma and autoimmunity in mice. J Clin Investig 2010;120:2497–507. [3] Negishi I, Motoyama N, Nakayama K, Nakayama K, Senju S, Hatakeyama S, Zhang Q, Chan AC, Loh DY. Essential role for ZAP-70 in both positive and negative selection of thymocytes. Nature 1995;376:435–8. [4] Cotsapas C, Hafler DA. Immune-mediated disease genetics: the shared basis of pathogenesis. Trends Immunol 2013;34:22–6. [5] Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 2007;447:661–78. [6] Tan W, Sunahori K, Zhao J, Deng Y, Kaufman KM, Kelly JA, Langefeld CD, Williams AH, Comeau ME, Ziegler JT, et al. BIOLUPUS Network; GENLES Network. Association of PPP2CA polymorphisms with systemic lupus erythematosus susceptibility in multiple ethnic groups. Arthritis Rheum 2011;63:2755–63. [7] Nambiar MP, et al. T cell signaling abnormalities in systemic lupus erythematosus are associated with increased mutations/polymorphisms and splice variants of T cell receptor zeta chain messenger RNA. Arthritis Rheum 2001;44:1336–50. [8] Crispin JC, et al. Expression of CD44 variant isoforms CD44v3 and CD44v6 is increased on T cells from patients with systemic lupus erythematosus and is correlated with disease activity. Arthritis Rheum 2010;62:1431–7. [9] Yoshida N, et al. ICER is requisite for Th17 differentiation. Nat Commun 2016;7:12993. [10] Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med 2009;361:888–98. [11] Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014;14:585–600. [12] Deteix C, et al. Intragraft Th17 infiltrate promotes lymphoid neogenesis and hastens clinical chronic rejection. J Immunol 2010;184:5344–51. [13] Mitsdoerffer M, et al. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci USA 2010;107:14292–7. [14] Schaffert H, et al. IL-17-producing CD4+ T cells contribute to the loss of B-cell tolerance in experimental autoimmune myasthenia gravis. Eur J Immunol 2015;45:1339–47. [15] Thomson CW, Lee PL, Zhang L. Double-negative regulatory T cells: non-conventional regulators. Immunol Res 2006;35:163–78. [16] Rodríguez-Rodríguez N, et al. Programmed cell death 1 and Helios distinguish TCR-αβ+ double-negative (CD4-CD8-) T cells that derive from selfreactive CD8 T cells. J Immunol 2015;194:4207–14. [17] Crispín JC, Tsokos GC. Human TCR-alpha beta+ CD4- CD8- T cells can derive from CD8+ T cells and display an inflammatory effector phenotype. J Immunol 2009;183:4675–81. [18] Rodríguez-Rodríguez N, et al. Pro-inflammatory self-reactive T cells are found within murine TCR-αβ+ CD4− CD8− PD-1+ cells. Eur J Immunol 2016;46:1383–91. [19] Hedrich CM, Crispín JC, Rauen T, Ioannidis C, Koga T, Rodriguez Rodriguez N, Apostolidis SA, Kyttaris VC, Tsokos GC. cAMP responsive element modulator (CREM) α mediates chromatin remodeling of CD8 during the generation of CD3+ CD4- CD8- T cells. J Biol Chem 2014;289:2361–70. [20] Crispín JC, et al. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol 2008;181:8761–6. [21] Oliveira JB, et al. Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop. Blood 2010;116:e35–40. [22] Alunno A, et al. CD4−CD8− T-cells in primary Sjögren’s syndrome: association with the extent of glandular involvement. J Autoimmun 2014;51:38–43. [23] Tarbox JA, et al. Elevated double negative T cells in pediatric autoimmunity. J Clin Immunol 2014;34:594–9. [24] Lalor SJ, McLoughlin RM. Memory γδ T cells-newly appreciated protagonists in infection and immunity. Trends Immunol 2016;37:690–702. [25] Paul S, et al. Role of gamma-delta (γδ) T cells in autoimmunity. J Leukoc Biol 2015;97:259–71. [26] Turner JE, et al. IL-17A production by renal γδ T cells promotes kidney injury in crescentic GN. J Am Soc Nephrol 2012;23:1486–95. [27] Peng X, et al. IL-17A produced by both γδ T and Th17 cells promotes renal fibrosis via RANTES-mediated leukocyte infiltration after renal obstruction. J Pathol 2015;235:79–89. [28] Yin S, et al. Hyperactivation and in situ recruitment of inflammatory Vδ2 T cells contributes to disease pathogenesis in systemic lupus erythematosus. Sci Rep 2014;5:14432.
T Cells in Autoimmune Diseases Chapter | 5 35
[29] Liarski VM, Kaverina N, Chang A, Brandt D, Yanez D, Talasnik L, Carlesso G, Herbst R, Utset TO, Labno C, Peng Y, Jiang Y, Giger ML, Clark MR. Cell distance mapping identifies functional T follicular helper cells in inflamed human renal tissue. Sci Transl Med 2014;6:230ra46. [30] Craft JE. Follicular helper T cells in immunity and systemic autoimmunity. Nat Rev Rheumatol 2012;8:337–47. [31] Ueno H. T follicular helper cells in human autoimmunity. Curr Opin Immunol 2016;43:24–31. [32] Morita R, et al. Human blood CXCR5+CD4+ T cells are counterparts of T follicular cells and contain specific subsets that differentially support antibody secretion. Immunity 2011;34:108–21. [33] Hünemörder S, et al. TH1 and TH17 cells promote crescent formation in experimental autoimmune glomerulonephritis. J Pathol 2015;237:62–71. [34] Steinmetz OM, et al. CXCR3 mediates renal Th1 and Th17 immune response in murine lupus nephritis. J Immunol 2009;183:4693–704. [35] Paust HJ, et al. CXCR3+ regulatory T cells control TH1 responses in crescentic GN. J Am Soc Nephrol 2016;27:1933–42. [36] Shimizu S, et al. Membranous glomerulonephritis development with Th2-type immune deviations in MRL/lpr mice deficient for IL-27 receptor (WSX-1). J Immunol 2005;175:7185–92. [37] Hiepe F, Radbruch A. Plasma cells as an innovative target in autoimmune disease with renal manifestations. Nat Rev Nephrol 2016;12:232–40. [38] Veldhoen M, Uyttenhove C, van Snick J, Helmby H, Westendorf A, Buer J, Martin B, Wilhelm C, Stockinger B. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat Immunol 2008;9:1341–6. [39] Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, Mitsdoerffer M, Strom TB, Elyaman W, Ho IC, Khoury S, Oukka M, Kuchroo VK. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+IL-10+ Foxp3(-) effector T cells. Nat Immunol 2008;9:1347–55. [40] Chang HC, Sehra S, Goswami R, Yao W, Yu Q, Stritesky GL, Jabeen R, McKinley C, Ahyi AN, Han L, Nguyen ET, Robertson MJ, Perumal NB, Tepper RS, Nutt SL, Kaplan MH. The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation. Nat Immunol 2010;11:527–34. [41] Jabeen R, Kaplan MH. The symphony of the ninth: the development and function of Th9 cells. Curr Opin Immunol 2012;24:303–7. [42] Yang J, Li Q, Yang X, Li M. Interleukin-9 is associated with elevated anti-double-stranded DNA antibodies in lupus-prone mice. Mol Med 2015;21:364–70. [43] Jäger A, Dardalhon V, Sobel RA, Bettelli E, Kuchroo VK. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J Immunol 2009;183:7169–77. [44] Kim HJ, Cantor H. Regulation of self-tolerance by Qa-1-restricted CD8(+) regulatory T cells. Semin Immunol 2011;23:446–52. [45] Reynolds J, Norgan VA, Bhambra U, Smith J, Cook HT, Pusey CD. Anti-CD8 monoclonal antibody therapy is effective in the prevention and treatment of experimental autoimmune glomerulonephritis. J Am Soc Nephrol 2002;13:359–69. [46] McKinney EF, Lyons PA, Carr EJ, Hollis JL, Jayne DR, Willcocks LC, Koukoulaki M, Brazma A, Jovanovic V, Kemeny DM, Pollard AJ, Macary PA, Chaudhry AN, Smith KG. A CD8+ T cell transcription signature predicts prognosis in autoimmune disease. Nat Med 2010;16:586–91. [47] Kopetschke K, et al. The cellular signature of urinary immune cells in Lupus nephritis: new insights into potential biomarkers. Arthritis Res Ther 2015;17:94. [48] Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012;30:531–64. [49] Kasper IR, Apostolidis SA, Sharabi A, Tsokos GC. Empowering regulatory T cells in autoimmunity. Trends Mol Med 2016;22:784–97. [50] Arpaia N, et al. A distinct function of regulatory T cells in tissue protection. Cell 2015;162:1078–89. [51] Ferretti C, La Cava A. Adaptive immune regulation in autoimmune diabetes. Autoimmun Rev 2016;15:236–41. [52] Ghali JR, Wang YM, Holdsworth SR, Kitching AR. Regulatory T cells in immune-mediated renal disease. Nephrology (Carlton) 2016;21:86–96. [53] Costantino CM, Baecher-Allan CM, Hafler DA. Human regulatory T cells and autoimmunity. Eur J Immunol 2008;38:921–4. [54] Najafian N, Chitnis T, Salama AD, Zhu B, Benou C, Yuan X, Clarkson MR, Sayegh MH, Khoury SJ. Regulatory functions of CD8+CD28- T cells in an autoimmune disease model. J Clin Investig 2003;112:1037–48. [55] Hahn BH, Singh RP, La Cava A, Ebling FM. Tolerogenic treatment of lupus mice with consensus peptide induces Foxp3-expressing, apoptosisresistant, TGFbeta-secreting CD8+ T cell suppressors. J Immunol 2005;175:7728–37. [56] Sharabi A, Mozes E. The suppression of murine lupus by a tolerogenic peptide involves foxp3-expressing CD8 cells that are required for the optimal induction and function of foxp3-expressing CD4 cells. J Immunol September 1, 2008;181(5):3243–51. [57] Arazi A, Sharabi A, Zinger H, Mozes E, Neumann AU. In vivo dynamical interactions between CD4 Tregs, CD8 Tregs and CD4+ CD25- cells in mice. PLoS One 2009;4:e8447. [58] Moulton VR, Tsokos GC. T cell signaling abnormalities contribute to aberrant immune cell function and autoimmunity. J Clin Investig 2015;125:2220–7. [59] Li P, et al. TCR-CD3ζ gene polymorphisms and expression profile in rheumatoid arthritis. Autoimmunity 2016;49:466–71. [60] Zayed H. Genetic epidemiology of type 1 diabetes in the 22 Arab countries. Curr Diab Rep 2016;16:37. [61] Zhang Z, et al. TCRzetadim lymphocytes define populations of circulating effector cells that migrate to inflamed tissues. Blood 2007;109:4328–35. [62] Yoshimoto K, Setoyama Y, Tsuzaka K, Abe T, Takeuchi T. Reduced expression of TCR zeta is involved in the abnormal production of cytokines by peripheral T cells of patients with systemic lupus erythematosus. J Biomed Biotechnol 2010;2010:509021. [63] Ferraccioli G, Zizzo G. The potential role of Th17 in mediating the transition from acute to chronic autoimmune inflammation: rheumatoid arthritis as a model. Discov Med 2011;11:413–24. [64] Deng GM, Beltran J, Chen C, Terhorst C, Tsokos GC. T Cell CD3ζ deficiency enables multiorgan tissue inflammation. J Immunol 2013;191:3563–7. [65] Loirand G. Rho kinases in health and disease: from basic science to translational research. Pharmacol Rev 2015;67:1074–95. [66] Nishikimi T, Matsuoka H. Molecular mechanisms and therapeutic strategies of chronic renal injury: renoprotective effect of rho-kinase inhibitor in hypertensive glomerulosclerosis. J Pharmacol Sci 2006;100:22–8.
36 SECTION | II Cellular and Molecular Mechanisms
[67] Komers R. Rho kinase inhibition in diabetic kidney disease. Br J Clin Pharmacol 2013;76:551–9. [68] Isgro J, et al. Enhanced rho-associated protein kinase activation in patients with systemic lupus erythematosus. Arthritis Rheum 2013;65:1592–602. [69] Hayashi K, et al. Molecular mechanisms and therapeutic strategies of chronic renal injury: role of rho-kinase in the development of renal injury. J Pharmacol Sci 2006;100:29–33. [70] Sharabi A, Kasper IR, Tsokos GC. The serine/threonine protein phosphatase 2A controls autoimmunity. Clin Immunol 2017;17:S1521–6616. [71] Crispín JC, Apostolidis SA, Finnell MI, Tsokos GC. Induction of PP2A Bβ, a regulator of IL-2 deprivation-induced T-cell apoptosis, is deficient in systemic lupus erythematosus. Proc Natl Acad Sci USA 2011;108:12443–8. [72] Apostolidis SA, et al. Phosphatase PP2A is requisite for the function of regulatory T cells. Nat Immunol 2016;17:556–64. [73] Crispín JC, et al. Cutting edge: protein phosphatase 2A confers susceptibility to autoimmune disease through an IL-17-dependent mechanism. J Immunol 2012;188:3567–71. [74] Rauen T, Hedrich CM, Tenbrock K, Tsokos GC. cAMP responsive element modulator: a critical regulator of cytokine production. Trends Mol Med 2013;19:262–9. [75] Hedrich CM, et al. cAMP response element modulator α controls IL2 and IL17A expression during CD4 lineage commitment and subset distribution in lupus. Proc Natl Acad Sci USA 2012;109:16606–11. [76] Hedrich CM, et al. cAMP-responsive element modulator α (CREMα) trans-represses the transmembrane glycoprotein CD8 and contributes to the generation of CD3+CD4-CD8- T cells in health and disease. J Biol Chem 2013;288:31880–7. [77] Koga T, Ichinose K, Mizui M, Crispín JC, Tsokos GC. Calcium/calmodulin-dependent protein kinase IV suppresses IL-2 production and regulatory T cell activity in lupus. J Immunol 2012;189:3490–6. [78] Juang YT, Wang Y, Solomou EE, Li Y, Mawrin C, Tenbrock K, Kyttaris VC, Tsokos GC. Systemic lupus erythematosus serum IgG increases CREM binding to the IL-2 promoter and suppresses IL-2 production through CaMKIV. J Clin Investig 2005;115:996–1005. [79] Koga T, et al. CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance. J Clin Investig 2014;124:2234–45. [80] Koga T, et al. CaMK4 facilitates the recruitment of IL-17-producing cells to target organs through the CCR6/CCL20 axis in Th17-driven inflammatory diseases. Arthritis Rheumatol 2016;68:1981–8. [81] Ichinose K, Rauen T, Juang YT, Kis-Toth K, Mizui M, Koga T, Tsokos GC. Cutting edge: calcium/Calmodulin-dependent protein kinase type IV is essential for mesangial cell proliferation and lupus nephritis. J Immunol 2011;187:5500–4. [82] Ichinose K, Ushigusa T, Nishino A, Nakashima Y, Suzuki T, Horai Y, Koga T, Kawashiri SY, Iwamoto N, Tamai M, Arima K, Nakamura H, Obata Y, Yamamoto K, Origuchi T, Nishino T, Kawakami A, Tsokos GC. Lupus nephritis IgG induction of calcium/calmodulin-dependent protein kinase IV expression in podocytes and alteration of their function. Arthritis Rheumatol 2016;68:944–52. [83] Perl A. Activation of mTOR (mechanistic target of rapamycin) in rheumatic diseases. Nat Rev Rheumatol 2016;12:169–82. [84] Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol 2011;12:295–303. [85] Suárez-Fueyo A, Barber DF, Martínez-Ara J, Zea-Mendoza AC, Carrera AC. Enhanced phosphoinositide 3-kinase δ activity is a frequent event in systemic lupus erythematosus that confers resistance to activation-induced T cell death. J Immunol 2011;187:2376–85. [86] Kshirsagar S, et al. Akt-dependent enhanced migratory capacity of Th17 cells from children with lupus nephritis. J Immunol 2014;193:4895–903. [87] Saleiro D, Platanias LC. Intersection of mTOR and STAT signaling in immunity. Trends Immunol 2015;36:21–9. [88] Harada T, et al. Increased expression of STAT3 in SLE T cells contributes to enhanced chemokine-mediated cell migration. Autoimmunity 2007;40:1–8. [89] Amarilyo G, Lourenço EV, Shi FD, La Cava A. IL-17 promotes murine lupus. J Immunol 2014;193:540–3. [90] Yiu G, et al. Development of Th17-associated interstitial kidney inflammation in lupus-prone mice lacking the gene encoding STAT-1. Arthritis Rheumatol 2016;68:1233–44. [91] Hedrich CM, Crispin JC, Tsokos GC. Epigenetic regulation of cytokine expression in systemic lupus erythematosus with special focus on T cells. Autoimmunity 2014;47:234–41. [92] Patel DR, Richardson BC. Dissecting complex epigenetic alterations in human lupus. Arthritis Res Ther 2013;15:201. [93] Sunahori K, Juang YT, Tsokos GC. Methylation status of CpG islands flanking a cAMP response element motif on the protein phosphatase 2Ac alpha promoter determines CREB binding and activity. J Immunol 2009;182:1500–8. [94] Sunahori K, Nagpal K, Hedrich CM, Mizui M, Fitzgerald LM, Tsokos GC. The catalytic subunit of protein phosphatase 2A (PP2Ac) promotes DNA hypomethylation by suppressing the phosphorylated mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)/ phosphorylated ERK/DNMT1 protein pathway in T-cells from controls and systemic lupus erythematosus patients. J Biol Chem 2013;288:21936–44.