Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2017 Sep 6;314(5):F679–F698. doi: 10.1152/ajprenal.00236.2017

Regulatory T cells in acute and chronic kidney diseases

Rahul Sharma 1,, Gilbert R Kinsey 1
PMCID: PMC6031912  PMID: 28877881

Abstract

Foxp3-expressing CD4+ regulatory T cells (Tregs) make up one subset of the helper T cells (Th) and are one of the major mechanisms of peripheral tolerance. Tregs prevent abnormal activation of the immune system throughout the lifespan, thus protecting from autoimmune and inflammatory diseases. Recent studies have elucidated the role of Tregs beyond autoimmunity. Tregs play important functions in controlling not only innate and adaptive immune cell activation, but also regulate nonimmune cell function during insults and injury. Inflammation contributes to a multitude of acute and chronic diseases affecting the kidneys. This review examines the role of Tregs in pathogenesis of renal inflammatory diseases and explores the approaches for enhancing Tregs for prevention and therapy of renal inflammation.

Keywords: AKI, cytokine, inflammation, IRI, nephrotoxicity, Tregs

INTRODUCTION

The immune system is a double-edged sword, which on one hand performs immunosurveillance to protect from pathogenic invaders such as viruses, bacteria, fungi, etc., as well as internal threats such as cancers and neoplasm, but on the other hand is involved in clearance and recycling of dying cells. The immune system is armed with checkpoints, which ensure that once the pathogen has been eliminated, the immune system returns to a quiescent basal state. The immune system exerts these functions while distinguishing what is self and what is not. The “self” includes the harmless commensal organisms, which do not activate the immune system and are “tolerated.”

The two components of the immune system, namely innate and adaptive immunity, use diverse mechanisms to identify threats. The innate immune cells such as the phagocytes (primarily granulocytes, macrophages, and dendritic cells) identify conserved pathogen associated molecular patterns or danger associated molecular patterns (DAMPs) to identify the threat (144, 157). The adaptive immune cells on the other hand are educated in primary lymphoid organs to differentiate self and foreign antigens through a mechanism categorized as central tolerance, where any developing T and B cells die through apoptosis if their T-cell receptors (TCRs) or B-cell receptors (BCRs) react strongly to self-antigens (185, 245). Only B cells whose BCR does not react to self-antigen or T cells whose TCR reacts moderately to the self-antigen presented on the major histocompatibility complex (MHC) or human leukocyte antigen survive and are exported to the periphery. T cells responding to the antigen on MHC I give rise to CD8+ cytotoxic T cells, whereas T cells antigen recognition on MHC II generates CD4+ helper T cells (Th).

RECOGNITION AND IDENTITY OF REGULATORY T CELLS

A subset of Th cells, which react strongly to self-antigen-MHC II complex, does not get eliminated and is rather positively selected to be exported to periphery and regulate the activation of other immune cells. These cells express the forkhead box p3 (Foxp3) transcription factor and have been christened regulatory T cells (Tregs) (92, 170, 217). Cells with similar regulatory properties were first recognized in mice decades ago, when it was found that removal of thymus between days 2 and 4 after birth results in autoimmune diseases, and replenishing the thymocytes in these day-3 thymectomized mice prevented the development of autoimmunity, suggesting that certain T cells emigrate from the thymus of postnatal animals, which have the ability to “suppress” autoimmunity (160, 166).

Later work from the laboratories of Sakaguchi, Shevach, and others identified the suppressor cells in the thymus as well as periphery to express high levels of interleukin (IL)-2 high-affinity receptor (IL-2Rα or CD25) as CD4+CD25hi cells (196, 237). Subsequently, in the early 2000s Foxp3 was identified as a lineage-defining factor and a marker to confidently identify Tregs, based on studies on mutations in the Foxp3 gene in mice (scurfy) and humans [immune dysregulation, polyendocrinopathy, enteropathy, and X-linked (IPEX) syndrome] (14, 254). Absence of functional Foxp3 induces a multiorgan inflammatory syndrome and death in infancy. Early replenishment with CD4+CD25+ cells, the majority of which also express Foxp3, from normal mice prevented the mortality and autoimmune syndrome, indicating that the CD4+CD25+ Tregs possess in their repertoire, the ability to suppress multiorgan inflammation (21, 210, 223). Fontenot et al. (57) and Williams and Rudensky (255) generated mice with targeted deletion of Foxp3 and Hori et al. (77) made Foxp3 overexpressing mice and confirmed these findings. Owing to their specificity to self-antigen and constant exposure to self-antigen, Foxp3+ Tregs express the properties of activated antigen-experienced cells including high expression of CD44 and CD25 (IL-2Rα) (54, 59, 181, 195).

CD25 is not merely a surface marker for Tregs, but the survival and function of Tregs is also critically dependent on IL-2 (4, 63, 237, 242). Similar to the deficiency of Foxp3, lack of IL-2/IL-2R causes multiorgan inflammatory disease and death in infancy (4, 153, 259). CD25 expression is also upregulated on activated non-Treg cells, although not to the same extent as on Tregs, however, making it harder to distinguish Tregs from activated T cells. Fluorescent reporters for Foxp3 expression have been generated in mice, thus enabling sophisticated studies (58, 74). Such approaches are not possible in human studies. However, inclusion of IL-7Rα (CD127) has helped distinguish Tregs from activated T cells, such that CD4+CD25hiCD127lo cells are widely accepted as Tregs with more than 95% of these cells expressing Foxp3 (205).

Attempts have also been made to distinguish the thymic-derived Tregs (tTregs) from peripherally-induced Tregs (pTregs). Thornton et al. (236) postulated the expression of Helios transcription factor to differentiates tTregs from pTregs, such that the proportion of Helios+ Tregs is higher in thymus than periphery, with the proportion of Helios+ Tregs declining in the periphery with age. Helios was also found to regulate the fitness of CD44+CD62Llo effector Tregs. Although there was no overt pathology of Treg-specific deletion of Helios, such Tregs had impaired ability to regulate activation of T cells and germinal center (GC) responses (204). Other cell surface markers have been documented to differentiate the tTregs and pTregs including the “T cell immunoreceptor with Ig and ITIM domain” (TIGIT), FcR-like 3 (FCRL3), Neuropilin-1 (Nrp1), etc. (17, 265, 272), with some controversy (221, 228).

SUBSETS OF TREGs

The Foxp3 Tregs generated during T-cell selection in thymus are commonly known as thymus-derived Tregs (tTregs) or natural Tregs (nTregs). Tregs not only regulate immune response to self-antigen but also play an important role in maintaining tolerance to commensal organisms, food, and air-borne antigens as well as the fetus, which essentially is a semi-allograft (6, 80, 91). Foxp3+ Tregs are also generated from naïve T cells during antigenic response to nonself or neoantigens in the presence of transforming growth factor (TGF)-β and IL-2 and are called induced Tregs (iTregs), peripherally-derived Tregs (pTregs), or adaptive Tregs (aTregs) in the literature (31, 116, 122). The majority of iTregs reside at the surfaces that are frequently exposed to the environment, such as skin, mucosa, or placenta. Other subsets of Tregs have been defined that do not express Foxp3, yet are immunosuppressive and produce IL-10, TGF-β, or IL-35 and are termed Tr1, Th3, or Tr35, respectively (25, 36, 37, 71, 169). Regulatory cells other than those belonging to Th lineage have also been described. These include CD4CD8 double-negative Tregs, Qa-1-restricted CD8 Tregs, CD28+CD8+ Tregs, regulatory NK cells, regulatory B cells, etc. (150, 191, 218, 234, 247, 249). For the purpose of this review, we will focus our discussion on the CD4+Foxp3+ Tregs only.

ESSENTIAL REQUIREMENTS FOR TREG GENERATION AND FUNCTION

Although several pathways have been identified for the generation, survival, and function of Tregs (Fig. 1), TCR and IL-2 signaling remain as the minimum requirements for generation of Tregs in the thymus. We discuss these and other mechanisms below.

Fig. 1.

Fig. 1.

Regulators and regulatory mechanisms of Tregs. The CD4+Foxp3+ Tregs are primarily defined by the expression of Foxp3 transcription factor and high-affinity IL-2 receptor (IL-2Rα or CD25). The most important factors for thymic Tregs, which also express Helios transcription factor, include TCR/CD28 signaling and IL-2. In addition, several other mechanisms are utilized for the differentiation, maintenance, and functions of Tregs. TGF-β, which is one of the suppression mechanisms used by Tregs, along with IL-2, converts naïve T cells to Tregs during T-cell receptor (TCR) stimulation. IL-33 is the newest factor that can promote differentiation, proliferation, and function of Tregs. Other major mechanisms are highlighted. Tregs can suppress target cells through IL-2 consumption, adenosine, soluble mediators, costimulatory molecules, and direct cytotoxicity. Trafficking and adhesion molecules also play important roles in Treg function. Costimulatory molecules: CD28; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; TIGIT, T-cell immunoreceptor with Ig and ITIM domains; GARP’, glycoprotein A repetitions predominant; GITR, glucocorticoid-induced tumor necrosis factor-related receptor; PD-1, programmed death 1. Cytokines: IL-2, interleukin-2; tripartite IL-2 receptor IL-2Rα, CD25, IL-2Rβ, and IL-2Rγ; ST2, IL-33 receptor; interleukin (IL)-2 receptor α chain IL-33, interleukin-33; 2A2AR; TGF-β, transforming growth factor-β; TGF-βR, TGFβ receptor. Soluble mediators of suppression: IL-10, interleukin-10; IL-35, interleukin-35. Cytotoxicity: granzyme B; FasL, Fas ligand. Adhesion molecules: ItgαVβ8, integrin αVβ8 (also involved in TGF-β activation); CD103, integrin αE; Nrp1, neuropilin-1. Adenosine metabolism: CD39 and CD73, ectonucleotidases; A2A adenosine receptor.

T-Cell Receptor

The first identification that antigen specificity is essential for generation of Tregs came from TCR-transgenic mice (6, 91, 248), with specificity to a non-self-antigen (e.g., ovalbumin). In these mice, the majority of T cells express a rearranged TCR for ovalbumin (OVA). However, Foxp3-expressing Tregs in such mice were only found in the minor population of cells not bearing the transgenic TCR. On a recombination (Rag1)- deficient background the endogenous TCR was not expressed, resulting in a complete lack of Tregs. When the cognate antigen (ovalbumin) was introduced as a second transgene, large number of CD4+ T cells were deleted, but a sizable proportion of the remaining OVA-specific T cells became Tregs. Interestingly, in an OTII-TCR-transgenic Foxp3 mutant mice (scurfy), it was primarily the nontransgenic TCR-bearing cells that expanded in the target organs and contributed to the pathology (211). Similarly, T cells engineered to express tTreg-derived TCRs expanded more in the lymphopenic hosts, indicating their greater self-reactivity (123).

Persistent TCR signaling is not only important for generation of Tregs in thymus, but sustained expression of TCR is necessary for maintaining their suppressive activity (203). By using a sophisticated double-transgenic mouse strain bearing Foxp3eGFP-Cre-ERT2 mated to loxp-flanked TCRα chain, TracFL/WT, or TracFL/FL mice, TCR expression can be deleted in the peripheral Tregs by tamoxifen treatment, and it was shown that although expression of Foxp3 was not affected, a specific subset of CD44hiCD62Llo effector-like Tregs was lost (131). This also correlated with developing fulminant autoimmunity in less than 2 wk of TCR deletion, indicating that sustained expression of TCR is critical for maintenance of Treg suppressive activity. Interestingly, in these TCR-deleted Tregs that expressed normal levels of CD25 and other Tregs associated markers (CD39, CD73, GITR), the responsiveness to IL-2 was maintained, thus suggesting that CD25 expression on Tregs is independent of continued TCR expression. However, it must be noted that Tregs once activated by one antigen can suppress proliferation of not only T-cell activation to a different antigen, but also in a xenogeneic manner (184).

IL-2/IL-2R

IL-2 was discovered more than three decades ago and was labeled as T-cell growth factor. However, studies with mice deficient in IL-2 or IL-2Rα (CD25) have shown that IL-2 is critical for the survival and maintenance of Tregs (33, 40, 58, 79) and may be dispensable for Th1, but not Th2 cells (212, 214). Mice deficient in IL-2 or IL-2R have a reduced proportion of Tregs, especially in the periphery (4, 63). As a result, the IL-2/IL-2R-deficient mice develop a lympho-proliferative disease and die young (208, 274). Interestingly, absence of CD25 expression results in Treg deficiency, which leads to polyclonal activation of T-cells and increased production of IL-2. The reduced consumption (likely by Tregs) and increased production of IL-2 leads to accumulation of very high levels of IL-2 (serum levels exceeding 1 mg/ml), which drives an expansion of memory phenotype CD8+ T cells (216). The mortality, IL-2 levels, and CD8 T-cell proliferation could be normalized by adoptive transfer of CD4+CD25+ Tregs from wild-type mice. STAT5 regulates Foxp3 expression in mice and humans (164, 260). Ablation of STAT5, which is downstream to IL-2 signaling also results in loss of Tregs in thymus as well as the periphery, with phenotypes similar to the IL-2/IL-2R-deficient mice (22, 268). Activated Stat5 binds to two different regulatory elements in the Foxp3 gene (see below) and has an important role in its expression, in addition to being a paracrine proliferative signal for Tregs. During thymopoiesis, CD4+CD25+Foxp3 precursors of Tregs could be identified, which begin to express Foxp3 in response to IL-2 in vitro and in vivo (141, 215, 268). IL-2 not only regulates Tregs directly as a proliferation/maintenance factor and by increasing the expression of Foxp3 directly, but also regulates the responsiveness of Tregs to other stimuli through CD103 (213) and ST2 (see below).

Costimulation

Whereas the interaction of TCR with MHC-antigen complex provides “signal 1,” adequate T-cell activation depends on “signal 2” provided by the interaction of costimulatory molecules (B7-1 and 2, also known as CD80 and CD86) on the antigen-presenting cells (APCs) with their counterpart on the T cells (CD28). These signals are also important for Treg differentiation, since Treg selection is based on the strength of the activation signal. The proportion of Tregs was strongly reduced in mice with genetic deficiency of CD28 or CD80/CD86 (197, 229, 232, 248). Although CD28 signaling in conventional T cells is critical for IL-2 production, interaction of the COOH-terminal cytoplasmic tail of CD28 with “lck” plays an intrinsic role in inducing the expression of Foxp3, glucocorticoid-induced TNFR-related (GITR), and CTLA-4 on antigen-experienced double-positive thymocytes (229). Several transcription factors downstream to the TCR/CD28 signaling bind to the promoter as well as intronic regulatory regions of the Foxp3 gene, as further discussed below. CTLA-4 is another costimulatory factor expressed late on T cells after multiple rounds of stimulation that, similar to CD28, interacts with CD80/CD86 on the APCs (24, 117). Unlike the activation signal relayed by CD28, CTLA-4 is believed to send inhibitory signal in the T cell. Since Tregs are always activated in response to the ubiquitous self-antigen (186, 231), they also express CTLA-4, and in Tregs, CTLA-4 ligation to CD80/CD86 sends a signal to downmodulate the activation of APCs (187, 233). Global or Treg-specific ablation of CTLA-4 resulted in greater number of Tregs; however, it impaired the suppressive activity of Tregs and induced activation and proliferation of DC and inflammatory conditions (61, 178, 256). CTLA-4 mutations have been reported to have a high degree of correlation with several autoimmune diseases, including Type 1 diabetes and multiple sclerosis (30, 53, 199).

Programmed death 1 (PD-1), expressed on T cells and PD-L1/PD-L2 (B7-H1 and B7-H2), expressed on the APC as well as nonimmune cells is another pair of costimulatory receptors, which are expressed late during immune reactions [reviewed in (97, 125)]. Although known as a marker of exhaustion and relaying negative signals to T cells (119), PD-1 plays an important role in the generation of iTregs. Interaction of PD-1/PD-L1 synergizes with TGF-β pathway for induction of iTregs, and mice lacking PD-L1/PD-L2 are impaired for induction of iTregs (60). We have found that Tregs from PD-1-deficient mice are also impaired in their ability to inhibit the activation of innate immune cells (105). However, in the context of cancer biology, PD-1 expression on tumor-infiltrating Tregs is a marker of Treg exhaustion, and PD-1Hi cells isolated from tumors have lower suppressive activity (147). The TNF receptor family member GITR is also a costimuatory molecule expressed on Tregs (159, 219), and treatment of mice with Fc-GITR-L increased Treg numbers (138). GITR along with the other TNFR family members OX-40, ICOS, and TNFR2 were proposed to regulate the responsiveness of tTreg to IL-2 (189). However, ligation of these receptors also induces proliferation and activation of effector T cells and has been shown to inhibit Treg activity (224).

IL-33/ST2 Pathway

While studying the role of IL-2 in CD4+ T cells from the scurfy mice, we found that ST2 (IL1RL1) was one of the most highly responsive cell surface molecules regulated by IL-2 (212). ST2 is the receptor for IL-33, an alarmin expressed in the nucleus of several cell types, which is known to be released on cellular damage (27, 139). Since tTregs are critically dependent on IL-2, they may express ST2 as well. Indeed, we found that FACS-sorted nTregs but not iTregs expressed ST2 mRNA and that a major proportion of Tregs in a naïve mouse expressed ST2 (226). Furthermore, similar to IL-2, IL-33 alone could induce expansion of Tregs systemically. Importantly, IL-2 and IL-33 synergized in vivo to expand Tregs much more potently than either cytokine alone. Similar observations were made by Schiering et al. (201), which showed that ST2-expressing Tregs are enriched in the intestine especially among the Helios+ subset. IL-33 treatment promoted proliferation of tTregs and TGFβ-mediated differentiation of iTregs in vitro. They also showed that ST2+ Tregs had higher expression of CD103, OX40, and Foxp3. IL-33 treatment in vitro also increased the recruitment of Gata3 and Pol II at the promoter region of the Foxp3 gene. In another recent study, IL-33 treatment increased the production of IL-2 by DC, which contributed to expansion of CD4+ST2+Foxp3+ Tregs (156).

A novel hybrid cytokine (IL233) bearing the activities of IL-2 and IL-33 cytokine domain in a single molecule offered improved synergy to increase Tregs as compared with the co-administration of IL-2 and IL-33 (226). Since IL-33 is released on cellular damage, it may constitute a signal for recruitment of Tregs to prevent inflammation and activation of auto-reactive T cells in response to neoantigens released during cellular damage. Interestingly, helminth infestations induce release of IL-33 from intestinal epithelial cells (85), and helminths have also been shown to increase Treg levels. Helminths also protect from autoimmune Type 1 diabetes and multiple sclerosis in mouse models (55). Clinical trials are ongoing for helminth-induced immunomodulation therapy in relapsing-remitting multiple sclerosis (NCT00645749, NCT01413243). Expansion of Tregs in response to IL-33 may be one of the mechanisms in the therapeutic use of helminths for inflammatory diseases.

TGF-β Pathway

TGF-β is an immunoregulatory cytokine, which signals through a heterodimeric receptor composed of TGF-βRI and TGF-βRII subnits, is a pleiotropic cytokine and inhibits the activation of multiple immune cell types. A role of TGF-β was proposed in Treg biology when it was found that TGF-β1-deficient mice die early with multiorgan inflammatory disease (146). Expression of a dominant-negative form of TGF-βRII or ablation of TGF-βRII on T cells also recapitulated the inflammatory phenotype (154). However, when TGF-βRI was deleted late in antigen-experienced cells, it did not affect Tregs and mice were spared from autoimmunity (146). It was then realized that TGF-β may be dispensable for generation of tTregs but is critical for the induction of pTregs (222). Further studies as discussed below also demonstrated that Smad3, which is downstream to TGF-β signaling, does not bind Foxp3 promoter or the conserved tTreg-specific regulatory region in the Foxp3 gene, rather it binds the pTreg-associated elements (202, 238). Indeed, naïve CD4+ T cells (Tn) were shown to be converted to Foxp3+ Tregs by TCR stimulation in the presence of TGF-β1 and IL-2 (40). During antigenic stimulation, TGF-β1 can also convert naïve T cells to the regulatory Th3 cells, which do not express Foxp3 but themselves produce TGF-β to regulate immune activation and Foxp3+ Tregs (25). Other TGF-β pathway-related molecules have been found to contribute to Treg maintenance and function. These include the latent TGF-β-binding proteins (LTBPs) termed GARP/LRRC32 (44, 239), the integrins αVβ8 (45), and αE(CD103) (72). Interestingly, IL-2 was shown to regulate CD103 expression on helper T cells and contribute to the migration and recruitment of T cells and Tregs to skin as well as gut and lung mucosal surfaces and lung (213).

Adenosine Metabolism and Tregs

Tregs express at least three cell surface receptors that are involved in modulation of extracellular ATP and its role in immune activation that include CD39, CD73, and adenosine 2a receptor (A2AR) (41). Extracellular ATP released during injury is a potent inflammatory stimulus. CD39 and CD73 expressed on Treg cell surface are involved in dephosphorylation of ATP or ADP to AMP (CD39) and subsequently to adenosine (by CD73). Tregs deficient for CD73 and thus lacking in their ability to produce adenosine were also impaired in their ability to protect in inflammatory disorders (105). By binding to A2ARs, adenosine inhibits effector T-cell activation, while it also induces differentiation and activation of iTregs (171). A2AR activation upregulated PD-1 and IL-10 expression on Tregs and promoted their in vivo protective activity in the mainly innate inflammation model of kidney ischemia-reperfusion injury (105).

Other Regulators of Treg Maintenance and Function

Several other regulators of tolerance have now been found to contribute to the generation, maintenance, and function of the various overlapping and nonoverlapping subsets of Tregs. The vast majority of them were initially identified among pTregs in gut-associated lymphoid tissue and have now been extended for other organs. These will be mentioned here, with references provided for further details. The prominent among them are all-trans retinoic acid (ATRA) (15, 38, 227), Vitamin D3 (96), and indoleamine-pyrrole 2,3-dioxygenase (IDO) (207). Exposure of gut and dermal DC to ATRA and Vitamin D3, respectively, increases the expression of IDO, which promote the differentiation and proliferation of Tregs. Short-chain fatty acids (especially butyrate), produced by the gut Clostridium bacteria were also shown to promote differentiation, proliferation, and activation of Tregs (7, 64). The AKT/mTOR pathway is a negative regulator of iTreg differentiation, and the mTOR inhibitor rapamycin increased Treg generation and has been proposed for therapeutic use in inflammatory conditions (75, 198, 270).

TRANSCRIPTIONAL AND EPIGENETIC REGULATION OF TREGs

The realization that Foxp3 plays a central role in Treg lineage and function prompted studies on the regulation of its expression. Although elegant studies have identified several factors for the maintenance and function of Tregs as discussed below, the exact event, which determines Foxp3 expression in thymus is still elusive.

Transcriptional and Epigenetic Regulation of Foxp3

The Foxp3 gene has four distinct regulatory regions (Fig. 2), the promoter region and three conserved noncoding sequence elements (CNS1, CNS2, and CNS3) (277). A unique demethylation pattern is observed at these regulatory regions of freshly isolated Tregs but not on the conventional T cells.

Fig. 2.

Fig. 2.

Transcriptional regulation of Foxp3. Foxp3 transcription is controlled by binding of a diverse set of transcription factors to distinct regulatory elements in the Foxp3 gene, namely promoter, conserved noncoding sequence (CNS) 1, CNS2, and CNS3. Regulators downstream to TCR/CD28 signaling bind at all the elements. IL-2 signaling recruits activated STAT5 to the promoter and CNS2, while TGF-β-mediated Smad and other peripheral (induced) Tregs related transcriptional factors (RAR, Foxo) bind to CNS1. Foxp3 is an autoregulator and binds to CNS2 as a trinary complex with Cbf-β and RUNX1. Demethylation of CpG islands in CNS2 regulate tTreg stability, and CNS1 may regulate iTreg differentiation, whereas CNS3 may regulate precursors to Tregs. The upstream signals and transcription factors are color coded. Upstream pathways inducing expression of the downstream transcription factors are depicted in the same color. AP1, activator protein 1; c-Rel and p65, members of NF-κB transcription factor family; CREB, cyclic-AMP-responsive element-binding protein; NFAT, nuclear factor of activated T cells; STAT5, signal transducer and activator of transcription; ST2, receptor for IL-33; Gata3, GATA binding protein 3, Pol II, RNA polymerase II; SMAD3, mothers against decapentaplegic homolog; Cbf-β, core-binding factor, β subunit; Foxp3, forkhead box P3; RUNX, runt-related transcription factor 1; Nr4a, nuclear receptor 4a; Foxo, forkhead box O; RAR, retinoic acid receptor. [From Huehn and Beyer (84).]

Promoter.

During T-cell development the CpG motifs in the Foxp3 promoter are completely demethylated in tTregs but are methylated in conventional CD4+ T cells, which is even higher on T-cell activation (88, 101, 272). The promoter region gets demethylated during iTreg induction similar to tTregs. Multiple transcription factors that are known to be required for tTreg-cell differentiation bind to the Foxp3 promoter region (reviewed in Ref. 84). These include cAMP-responsive element-binding protein (CREB), nuclear factor of activated T cells (NFAT), activator protein 1 (AP1), signal transducer and activator of transcription 5 (STAT5), nuclear receptor 4a (Nr4a), c-Rel, and p65, and this is consistent with the role of TCR, CD28, and IL-2 signals in the upregulation of Foxp3 expression (Fig. 2). Recently, IL-33 was also shown to induce the recruitment of Gata3 and Pol II to the Foxp3 promoter region and enhance its expression (201).

CNS1.

The CNS1 element, which is in the first intron, does not have any CpG motifs and is regulated primarily by histone modifications (277). Consistent with the role of TGF-β in the differentiation of iTregs and not the tTregs, Smad2/3 only binds in the CNS1 regions, because deletion of CNS1 only affected the differentiation of iTregs but not tTregs (238). Other transcription factors bound to CNS1 region are NFAT and AP1 (277), indicating the requirement of TCR/CD28 stimulation for iTreg differentiation as well as by retinoic acid receptor (RAR) (277), as has been verified by the role of ATRA in pTreg differentiation in the gastrointestinal system (Fig. 2).

CNS2.

The CNS2 region is what really set the tTregs apart from all other Treg and non-Treg T cells, and is a bona fide epigenetic marker for tTreg-lineage. CNS2, also located in the first intron of the Foxp3 gene, is fully demethylated in tTregs and thus is also called Treg-specific demethylated region, TSDR (56, 182, 277). Demethylation status of CpG island in CNS2 is critical for the stability of Treg lineage. This region is methylated in the double-positive thymocytes as well as in single-positive non-Treg CD4+ T cells. The TGF-β-mediated differentiation of iTregs also does not induce demethylation of the CNS2 region, eluding to the instability of Foxp3 expression in these cells. Activated CD4+ conventional T cells in humans transiently express Foxp3, yet the CNS2 regions in these cells remain methylated, and the cells gradually lose Foxp3 expression (272). Interestingly, deletion of CNS2 region did not induce overt autoimmunity or a complete loss of Foxp3 expression in CNS2-deleted mice, although the frequency of Foxp3+ cells and the intensity of Foxp3 expression was reduced as compared with CNS2-sufficient Tregs (52, 135). Methylation of CNS2 is likely an active mechanism to inhibit Foxp3 expression in non-Treg cells as deletion or chemical inhibition of DNA methyltransferase 1 induced expression of Foxp3 on NK and CD8 T cells (93).

The CNS2 region is bound by several transcription factors including Foxp3 itself, indicating the role of Foxp3 and CNS2 in lineage commitment and stability of Foxp3 expression. The other transcription factors bound to CNS2 include c-Rel, p65, NFAT, CREB (TCR/CD28 related), STAT5 (IL-2 signaling), and Runx1-Cbf-β. Runx1-Cbf-β complex binds to demethylated CNS2 region and recruits Foxp3 to the CNS2 element, and deletion of Cbf-β in Tregs led to a loss of Foxp3 expression stability (109, 193).

CNS3.

Studies on chromatin remodeling of the Foxp3 locus shows that the CNS3 regions are the first to open during thymopoiesis and stay open in Tregs as well as all CD4+ T cells (277). It is primarily bound by NFκB family transcription factors c-Rel and p65 transcription factors and is believed to mark Treg precursors.

Recently, Foxo1 and Foxo3 were also shown to bind to Foxp3 regulatory regions, and deletion of Foxo proteins in CD4 T cells led to lower proportion and activity of Foxp3+ Tregs. Binding sites for Foxo proteins, which are downstream to the AKT/PI-3K pathway, were also discovered in CNS1 and CNS3 regions, but not CNS2, indicating their role in iTregs (98, 173, 174). The complexity of this issue is further illustrated by the finding that deletion of both Foxo1 and Foxo3 by using CD4-Cre resulted in lower proportion and numbers of Foxp3+ cells, whereas deletion of Foxo1 by using a Foxp3-Cre resulted in greater numbers of Foxp3+ cells; however, their activity was impaired, and in either case the mice were susceptible to multiorgan inflammatory disease (98, 174).

Transcriptional Regulation by Foxp3

Foxp3 itself is a winged-helix DNA binding protein and a unique transcriptional factor that serves both as a transcriptional activator and transcriptional repressor. Deletion of the DNA binding domain of Foxp3 or fusing GFP to the COOH terminus of Foxp3 rendering it inactive helped elucidate the role of Foxp3 as a transcription factor (67, 140). Tregs from these mice retained some Treg-associated programs in terms of CD25, CTLA-4, and GITR expression. These Tregs also retained their anergic status and the inability to produce IL-2. However, now these cells started to express IL-4 and IL-17 under stimulatory conditions (255). Foxp3 can interact with the TH17 transcription factors RORγt and Rorα as well as the interferon regulated factor (IRF)-4 (276, 279). These interactions may be involved in repression of the RORγ and IRF-4-related inflammatory program. In addition, Foxp3 was found to interact with NFAT, AP1, and Runx/Cbf-β, and these interactions might play a role in regulation of the Foxp3-mediated Treg program (see Transcriptional and Epigenetic Regulation of Foxp3 above). Although, Gata3 binds to the Foxp3 promoter, coexpression of Gata3 or T-bet with Foxp3 was shown to enable Tregs to restrict the respective Th2 and Th1 inflammatory responses more efficiently (113, 257).

MicroRNA and Tregs

In addition to the above-mentioned transcriptional programs regulating Foxp3 and regulated by Foxp3, microRNA (miRNA) also have important functions in Treg biology by regulating Foxp3, as well as being regulated by Foxp3. Deletion of two RNA processing enzymes Dicer and Drosha in Tregs induced fatal multiorgan inflammatory disease, similar to the scurfy mice (35, 143). Ablating the miRNA generative capacity of Tregs impaired the peripheral homeostasis and suppressive capacity of Tregs; however, the expression of other Treg-associated markers was maintained (280). Different miRNA species regulate specific components of Treg function, e.g., miRNA-155 controls responsiveness of Tregs to IL-2 (149), whereas miR-142-3p regulates the suppressive function (82). MiR-125a-5p increased the resistance to IL-6-mediated decline of Foxp3 expression (132), and miRNA-146 controls STAT1 signaling in Tregs and their ability to restrict Th1 responses (261). In clinical studies, miR-141 and miR-200a were associated with MS and were proposed to regulate the Treg/Th17 balance (165).

TREG SUPPRESSION MECHANISMS

As discussed above, the suppressor function and lineage differentiation of Tregs are differentially regulated. Several molecular mechanisms have been delineated over the years that are used by Tregs to inhibit activation of the immune system (Fig. 1). Tregs suppress the activation of cells of the innate and adaptive immune system, including T cells, B cells, NK cells, DC, and macrophages, as well as granulocytes. In Foxp3-deficient scurfy mice, T-cell activation can be observed as early as the first week after birth, which rapidly amplifies to almost all the immune cells and production of inflammatory cytokines (210). It is worthwhile to note that in the OTII-transgenic scurfy mice, primarily the T-cell-bearing OVA-specific TCR did not expand and the nontransgenic TCR-bearing cells rapidly expanded and infiltrated the target organs, indicating a specific loss of regulation of autoimmunity (275). Continued expression of TCR was shown to be critical for maintaining the suppressive function of Tregs (131).

IL-2 Consumption

One of the earliest mechanisms proposed for Treg suppressor activity was consumption of IL-2 and outcompeting activated T cells for IL-2, owing to the constitutive high-level expression of CD25 on Tregs (12, 235). In CD25-deficient mice, not only Treg proportions were compromised, IL-2 consumption was also impaired, leading to accumulation of abnormally high circulating IL-2 levels, which induced inflammatory pathology as well abnormal expansion of memory phenotype CD8 T cells (216). Adoptive transfer of CD4+CD25+ T cells from normal mice normalized the IL-2 and CD8+ T cell levels. A recent elegant study showed that IL-2 consumption and downstream STAT5 signaling was important to maintain the suppressor function of Tregs (33). Furthermore, IL-2 capture by Tregs was critical for suppressing CD8+ T-cell activation, but was dispensable for controlling the CD4+ T cells.

Soluble Mediators

TGF-β1 and IL-10 are immunoregulatory cytokines that are produced by many cell types, including Foxp3+ Tregs as well as Tr1 and Th3 cells, and constitute soluble mediators of Treg suppression activity (133, 134, 158, 192). Tregs deficient in these cytokines have impaired ability to regulate immune activation. Other soluble mediators include IL-35, which can induce infectious tolerance by converting naïve CD4+ T cells into IL-35-expressing Tr35 cells (36, 37). As mentioned above, Tregs also have the ability to convert pro-inflammatory extracellular ATP to anti-inflammatory adenosine through CD39 and CD73 (41, 105, 171). Adenosine can inhibit the activation of innate and adaptive cells in a paracrine manner and also stimulate Tregs to express PD-1 in an autocrine manner through A2AR signaling.

Contact-Dependent Mechanisms

An early mechanism proposed for Treg-mediated suppression was identified by the common appearance of CTLA-4 in various genetic screens for autoimmune diseases. Deletion of CTLA-4 impaired the suppressive function of Tregs, without affecting the Treg differentiation (11, 278). CTLA-4 ligation to CD80/CD86 on APCs is critical for maintaining immune tolerance, and the lack of CTLA-4 resulted in higher expression of CD80 and CD86, which provided greater costimulatory signals to the effector T cells (278). We believe that during homeostasis, the MHC II molecules are occupied by self-antigen, prompting Tregs to stay in association with DC and lowering their activation status through CTLA-4 and CD80/CD86 interactions (209). During infections, the self-antigen on MHC II will be replaced with the foreign peptide, which may cause Tregs to dislodge. Furthermore, signaling through DAMPs will upregulate MHC II for display of the pathogen’s antigens on their surface and CD80/CD86 for enhanced costimulatory potential. This will allow the immune reaction to go forward to eliminate the pathogen. However, once the danger signal is gone and the pathogen is cleared, the MHC II of DC will be again loaded with self-antigen, thus allowing Tregs to resume the association with DC for enforcing self-tolerance. Other costimulatory molecules that are used by Tregs to increase their interactions with DC include Lag3 and TIGIT that play a role in the suppressive function of Tregs (83, 269).

Cytotoxicity

Tregs can also directly kill activated immune cells through granzyme B as a soluble mediator. These studies, originally reported in murine models, were also demonstrated using human adaptive Tregs that can induce cytolysis by using granzyme B (69, 273). Another cytolytic mechanism employed by Tregs to kill CD8+, but not CD4+ T cells, is through Fas-FasL in a cell-cell contact-dependent manner (225). The latter studies were performed in vitro, but do prompt revisiting the relevance of autoinflammatory disease in Fas or FasL mutant mice.

THERAPEUTIC IMPLICATIONS OF TREGs IN INFLAMMATORY RENAL DISEASE

In most, if not all, studies on inflammatory diseases, including acute and chronic renal disease, depletion of Tregs worsens the disease, whereas Treg supplementation offers robust protection (43, 108, 226). Inflammatory diseases of the kidney are a major medical burden. Kidneys are heavily perfused organs and responsible for filtration of blood. The differential distribution of blood flow and partial pressure of oxygen between the renal cortex (50 mmHg) and medulla (10–20 mmHg) makes the medulla especially susceptible to hypoxia (20, 50, 73). Kidneys get up to 25% of cardiac output and constitute only 1% of the body mass, leading to renal accumulation of substances that interfere with cellular metabolism, making them vulnerable (180, 243). Renal manifestations are often secondary to other ailments and contribute to mortality and morbidity. Diabetic kidney disease, either due to Type 1 or Type 2 diabetes, is a major cause of morbidity in the United States, with the incidence on the rise (43). Autoimmune disease such as lupus (SLE), which is marked with high levels of circulating autoantibodies, manifests in half of patients as immune complex deposits in the kidneys and renal failure in up to 10% of patients (5). A sizable number of patients undergoing chemotherapy suffer from nephrotoxicity-associated renal damage, which adds not only to the health burden, but also limits the administration of potent chemotherapeutic drugs. Several other renal disorders such as IgA nephropathy, membranous glomerulopathy, focal segmental glomerulonephritis, etc., are worsened by the immune system. A large proportion of these patients never recover and proceed to end-stage renal disease (ESRD), with transplantation as the only resort. Renal transplantation along with alloreactivity is also plagued by ischemia-reperfusion injury (IRI), which is further amplified by the inflammatory immune response. Last but not least, acute kidney injury (AKI) occurs in a larger proportion of all hospitalized patients and up to 50% of ICU patients. Although a sizable proportion of these patients recover, 5–20% of them progress to ESRD (26). Inflammation plays a major role in the pathogenesis. Thus strategies that can intervene with inflammation should prove to be beneficial. Tregs have proven to be one of the most potent enforcers of peripheral tolerance and can prevent undue activation of the immune system. Below we will discuss the role of Tregs in the major inflammatory renal diseases with the exception of transplantation, the literature on which is vast and deserves a separate review article by itself.

Ischemic Kidney Injury and Tregs

Prevention of AKI by Treg supplementation.

Inflammation plays a major role in the pathogenesis of AKI (9, 87). To test the role of Tregs in AKI, we targeted Tregs by using an antibody against CD25, which depleted ~50% of Tregs and performed IRI (107). Depletion of Tregs before IRI worsened the inflammation, acute tubular necrosis, and renal function assessed by plasma creatinine. In another approach, Rag1-deficient (knockout) mice reconstituted with lymph node cells from Treg-deficient scurfy mice and subjected to IRI had worse renal function, injury, and inflammation as compared with Rag1 knockout (KO) mice that received lymph node cells from Treg-replete wild-type mice (107). Importantly, in the same study, supplementation of scurfy mouse lymph node cells with CD4+CD25+ cells from wild-type mice inhibited the greater AKI associated with scurfy lymph node cell transfer to Rag1 KO mice. Adoptive transfer of freshly isolated wild-type Tregs to lymphocyte-deficient Rag1 KO mice directly demonstrated that the protective effect of Tregs in kidney IRI does not require adaptive immune cells. Expression of the immunosuppressive cytokine IL-10 was critical for the Treg-mediated protection as adoptive transfer of Tregs from IL-10-deficient mice was unable to protect in renal IRI (107). Mechanistically, Tregs effectively prevent the accumulation of neutrophils and mononuclear phagocytic cells in the kidney during reperfusion, by pathways that are not completely understood to date. Interestingly, Treg depletion with PC61 in a cecal ligation puncture of sepsis-induced AKI improved survival and renal outcomes, suggesting that Tregs protect mainly from sterile renal inflammation (129).

The use of Tregs or Treg-enhancing strategies have been investigated to evaluate protection after the onset of injury in AKI models. Gandolfo et al. (65) found increased trafficking of Tregs in the kidneys when measured 3 and 10 days post-IRI. Depletion of Tregs with an anti-CD25 (PC61) antibody starting 1 day after IRI resulted in continued injury and inflammation in the kidneys, leading to worse renal function and mortality. Adoptive transfer of Tregs 1 day after IRI surgery reduced Th1 cells as measured at day 3 and improved recovery measured day 10 post-IRI.

Preconditioning.

Ischemic preconditioning has been shown to protect from IRI and is believed to be mediated, in part, through increased recruitment of Tregs to the postischemic kidneys (106). Indeed, depletion of Tregs with PC61 after the preconditioning ischemia and before the second ischemic episode reduced the preconditioning-mediated protection from IRI. In another study, reconstitution with Tregs from donor preconditioned mice restored the protection; however, the efficacy of the preconditioned Tregs to restore the protection was not compared with those isolated from nonpreconditioned mice (34). Galectin-9 is a β-galactoside binding protein, which is a urate transporter in the kidneys, is also expressed in myeloid cells, and is known for its role in dendritic cell maturation and inducing apoptosis of promoting iTreg differentiation (151). Combination of galectin-9 pretreatment with ischemic preconditioning offered better protection from IRI than ischemic preconditioning alone (271). The protection correlated with a decrease in IFNγ-producing CD4+ T cells and an increase in the proportion of Foxp3+ Tregs in peripheral blood, spleen, and kidneys. Galectin-9 treatment by itself without ischemic preconditioning, however, did not protect from IRI.

Liang et al. (137) also investigated preconditioning in the setting of IRI, demonstrating that indeed preconditioning-mediated protection form IRI involves Tregs, as antibody depletion of Tregs took away the protection. In the same study, administration of a superagonistic antibody against CD28 before IRI was also shown to increase Tregs and protect from IRI, with the protection being reversed on Treg depletion. However, there is a caution associated with the CD28 superagonists, because a clinical trial employing TGN1412, a humanized CD28 superagonist antibody, had severe deleterious consequences at a dose 500 times lower than what was considered safe in animal studies (78, 86).

Heat shock preconditioning upregulated heat shock protein 70 (Hsp70) in mice along with an increase in the Tregs (102). Adoptive transfer of T cells from heat-preconditioned mice protected T-cell-deficient nu/nu mice from renal IRI. This protection was lost if Tregs were depleted before the T-cell adoptive transfer and restored when Tregs were replenished. Inhibition of Hsp70 expression by quercetin treatment of the heat-preconditioned mice attenuated the Treg expansion and renoprotection. Furthermore, Tregs from Hsp70-deficient heat-preconditioned mice only offered partial protection in this study.

IL-2 and IL-33.

The role of IL-2 in Treg homeostasis is well established. A complex of IL-2 and anti-IL-2 antibody was shown to induce robust proliferation of Tregs, CD8+ T, NK, and NK-T cells (19). Pretreatment of mice with IL-2 and anti-IL-2 complexes expanded spleen and kidney Tregs and offered protection in renal IRI (103). Furthermore, administration of IL-2/anti-IL2 antibody complex 1 day after IRI for 3 days improved the recovery of kidneys with reduced apoptosis and increased proliferation (PCNA+) of renal tubular cells (103).

We have found that IL-2 regulates the expression of IL-33 receptor, ST2 (212). The COOH-terminal half of IL-33 resembles the IL-1 family of cytokines and signals through the common IL1RAcP through MyD88/TRAF/NFκB pathway, and was originally thought to be involved with the Th2 response. However, we found that a major proportion of Tregs express ST2 and that Tregs increase in response to IL-33 stimulation, as also shown in other studies (201). Since IL-2 upregulates ST2 and increases the responsiveness of Tregs to IL-33, we treated mice with a mixture of IL-2 and IL-33, which induced a robust and rapid increase in Treg levels systemically and protected mice from IRI much more efficiently than IL-2 or IL-33 administered alone (226). Furthermore, a novel hybrid cytokine (termed IL233), containing IL-2 and IL-33 in a single molecule, was more efficient than the mixture of IL-2 and IL-33 at equimolar ratios to increase Tregs as measured in the spleen and blood. This may likely be due to better targeting of IL-33 to Tregs, owing to using IL-2 as a handle. Pretreatment with the IL233 hybrid cytokine also protected mice from IRI at a one-third lower molar dose as compared with the IL-2 and IL-33 mixture. Importantly, IL233 treatment increased the intrarenal level of Tregs before IRI, which was even more pronounced after renal IRI, indicating the active participation of Tregs in protection from ischemic kidney injury. In addition, IL233 administration after IRI surgery also completely protected mice from loss of renal function and mortality for 28 days when the experiment was terminated, whereas all the control mice were moribund in 3 days. When analyzed at day 28 after the IRI, the IL233 mice were indistinguishable from uninjured mice, without any renal fibrosis or any other structural abnormalities (226).

The spleen is a reservoir for blood as well as immune cells. Our data in this study showed that ischemic injury induces mobilization of Tregs from the spleen, and if the mice were pretreated with IL233, the mobilization was even more robust. Correspondingly, more Tregs were present in the kidneys of IL233-treated mice postischemia as compared with saline-treated controls. CD4+CD25+ Tregs isolated from IL233-treated mice not only had greater ability to suppress proliferation of conventional T cells in vitro, but they were also able to prevent IRI more efficiently than Tregs isolated from vehicle (saline)-treated mice. In agreement with the improved mobilization of Tregs by IL233 treatment, the Tregs isolated from IL233-treated mice trafficked to the kidneys in greater numbers, whereas the Tregs from the saline-treated mice accumulated at greater numbers in the spleen (M. E. Stremska, V. Sabapathy, and R. Sharma, unpublished observations). IL233 also increases the number of group 2 innate lymphoid cells, ILC2 in the spleen and kidneys, and adoptive transfer of ex vivo-expanded ILC2 also protected from IRI (226). We believe that IL-33 being an alarmin is designed by nature to negate the threats posed by cellular damage and serves as a signal for Tregs (and Th2 and ILC2) to restrict Th1-mediated inflammation. While our study was ongoing, others have also found a protective role for IL-33 in intestinal inflammatory diseases by increasing Treg levels (201).

Sphingosine.

Sphingosine 1 phosphate analog FTY720 (fingolimod) has been shown to be beneficial in inflammatory disease (32). In a mouse model of IRI, FTY720 administration post-IRI increased the levels of CD11b+CD11c+ DC in the spleens and kidneys, which coincided with a greater expression of Foxp3 mRNA in these organs, suggesting that Tregs may also participate in the FTY720-mediated protection from IRI (104). Pretreament with another natural sphingosine analog sphingosine N,N-dimethylsphingosine (DMS) also protected mice from bilateral IRI with the protection correlating with greater infiltration of total CD4+ and Foxp3+ cell in the kidneys before IRI (120). Inhibiting Tregs by blocking CD25 or CTLA-4 negated the DMS mediated protection. However, in this study DMS administration after IRI did not offer any protection.

Adenosine and other nucleotides.

Interactions of adenosine with the A2AR offer protection from inflammatory disease, and Tregs are involved in generation of adenosine through cell surface expression of CD39 and CD73 (41). An autocrine role of the adenosine pathway was identified in Tregs that contributes to the protective function of Tregs in IRI (105). Adoptive transfer of CD73 or A2AR-deficient Tregs was unable to protect in IRI model, indicating an intrinsic role of adenosine generation and utilization in Treg protective ability. Treatment of CD73 KO Tregs with A2AR agonist ATL1222 before the adoptive transfer restored their protective potential. Furthermore, ex vivo pharmacological stimulation of A2AR on Tregs promoted their protective ability such that as little as 40,000 Tregs significantly inhibited IRI-associated renal dysfunction and injury.

Extracellular ATP is considered pro-inflammatory through binding to the purinergic P2X and P2Y receptors and blocking of P2X7 receptor, which is highly expressed in the kidneys was beneficial in inflammatory renal conditions (28). In a recent study, Koo et al. (114) investigated the use of periodate-oxidized ATP (oATP), a P2X7 receptor (P2X7R) antagonist (121), in the settings of IRI. Mice treated with oATP for 7 consecutive days until the day of IRI were protected from renal dysfunction, tubular injury, and inflammation (114). Accordingly, the P2X7-deficient mice were protected from IRI. Interestingly, this protection was associated with an increase in Tregs in the kidneys, and the protection was abrogated on Treg depletion. Furthermore, adoptive transfer of P2X7-sufficient, but not P2X7-deficient, Tregs showed protective response to oATP treatment in immunodeficient Rag1 KO mice from IRI; however, the P2X7 KO Tregs were more protective than the WT Tregs (114). Treatment of mice with oATP also protected mice from renal dysfunction and fibrosis when administered after IRI (114).

PD-1 and PD-1 ligands.

Microarray comparison of Tregs from A2AR-sufficient and deficient Tregs showed that PD-1 expression on Tregs is linked with A2AR stimulation (105). The protection offered by Treg-A2AR stimulation was abrogated if the Tregs were treated with a blocking antibody to PD-1. Subsequently, Jaworska et al. (89) showed that the expression of PD-1 on Tregs and its ligands PD-L1 and PD-L2 in recipient mice are important for protection from renal IRI.

Other anti-inflammatory mechanisms in IRI.

Mesenchymal stem cells (MSC) have long been proposed to have anti-inflammatory properties in addition to their tissue regenerative capacity. The efficacy of MSC to intervene in renal IRI has been investigated (81). MSC infused 6 h after IRI surgery protected from renal inflammation and injury. Importantly, a greater number of Tregs accumulated in the spleen and kidneys of MSC-infused mice. This protection was abrogated if the mice were splenectomized or if Tregs were depleted using an anti-CD25 antibody.

Mycophenolate mofetil (MMF) is often used post-renal transplant to prevent inflammation, and it was found that in an animal model of IRI, MMF could alter the recruitment of Tregs to the kidneys along with CD4+ and CD8+ T cells and may affect the contribution of Tregs to renal repair (66).

Nephrotoxic Renal Injury and Tregs

Different classes of nephrotoxins induce diverse renal pathology in humans and mice. The most widely studied nephrotoxic injuries include cisplatin, Adriamycin (doxorubicin), gentamycin, aristolochic acid, and folic acid. However, the role of Tregs has mostly been studied in the former two models only. Whereas cisplatin primarily induces tubular damage, Adriamycin can induce both tubular and glomerular injury and is often proposed as a model of focal segmental glomerulosclerosis (130). Irrespective of the involvement of renal segments, inflammation plays a major role in the amplification of the injury and pathogenesis. The role of Tregs in preventing inflammation and injury and therapeutic use of Tregs or Treg-enhancing strategies have been explored in renal manifestations of these conditions as discussed below.

Tregs in cisplatin-induced renal injury.

Lee et al. (127) were the first to demonstrate that CD4+CD25+ Tregs protect from cisplatin-induced renal dysfunction and damage. Depletion of Tregs in WT mice exacerbated the cisplatin-induced injury, whereas adoptive transfer of Tregs either in the T-cell deficient nu/nu mice or intact Balb/c mice reduced the serum creatinine and BUN levels along with reduced renal damage, reduced renal levels of TNFα and IL-1β, as well as reduced mortality associated with cisplatin. Kim et al. (99) first showed that bee venom increased Treg proportions and protects from cisplatin-induced renal injury and later demonstrated that a constituent of bee venom, phospholipase A2 (PLA2), modulated Treg numbers and protected mice in a model of cisplatin-induced AKI (100). PLA2 increased Treg proportions in vitro and also induced greater accumulation of IL-10 and IL-10-producing Tregs and DC in the kidneys after cisplatin treatment. The PLA2-mediated renoprotection was abolished after anti-CD25 antibody depletion of Tregs. Importantly, bee venom-associated increase in Tregs and renoprotection did not affect the antitumor activity in tumor-bearing mice. Among other regulators of Treg function in prevention of AKI, it was recently shown that TLR9-deficient Tregs were not able to inhibit cisplatin-induced AKI, although the level and in vitro suppressive function of Tregs in TLR9 KO mice was intact (3). The authors postulated reduced expression of adhesion molecules CD11a and CD44 on Tregs from TLR9-KO mice to be responsible for the lack of protective ability.

As mentioned above, MSCs have protective properties in the renal IRI model (81). Recently, the effect of MSC administration was evaluated in cisplatin-induced AKI (177). Treatment with umbilical cord blood-derived MSCs 1 day after cisplatin administration attenuated intrarenal MCP-1 and IL-6 and showed higher expression of IL-10 and VEGF. This was postulated to be due to an increase in Tregs in the kidneys, because depletion of Tregs after MSC treatment abrogated the protection. However, treatment of mice on day 3 after cisplatin did not increase Tregs or protect renal function, indicating that MSCs do not promote renal repair in cisplatin nephrotoxicity (177).

Pretreatment of mice with the IL233 hybrid cytokine, which increases Tregs rapidly, was also able to rescue mice from nephrotoxic injury induced by cisplatin in C57Bl/6J mice, as observed with lower plasma creatinine and BUN levels and preserved renal architecture. This protection was preceded with an increase in circulating Treg levels (226). Interestingly, in a study by Akcay et al. (2), IL-33 was shown to exacerbate injury and inflammation in the cisplatin model and increased recruitment of CD4+ T cells to the kidneys in a CXCL1-dependent manner. IL-33 levels were increased in the blood and kidneys following cisplatin administration. Furthermore, coinjection with cisplatin and IL-33 worsened the injury as compared with cisplatin alone. However, the effects of IL-33 treatment on Tregs was not studied. The difference observed between this study and our studies could be that we pretreated mice with IL233 hybrid cytokine before administering cisplatin, and IL-33 was injected as a fusion to IL-2, thus targeting the majority of IL-33 activity to Tregs, which offered protection.

Tregs in Adriamycin nephropathy.

Foxp3 is the lineage-defining transcription factor for Tregs. Retroviral transduction of CD4+ T cells by Fontenot et al. (57) and Hori et al. (77) imparted Treg-like phenotype and regulatory function. A similar strategy was adopted by Wang et al. (252) in one of the earliest studies to show that adoptive transfer of T cells with a retroviral transduction with Foxp3 gene, 1 wk after induction of Adriamycin, protected the mice from renal dysfunction and injury. Subsequently, the same group showed that adoptive transfer of Tregs isolated from wild-type BALB/c mice protected syngeneic SCID mice from Adriamycin (152); however, the Treg-mediated protection is dependent on stimulation with self-antigen, as Tregs isolated from DO11.10 TCR-Tg mice, in which most T cells have specificity to ovalbumin, were unable to protect. Furthermore, the Treg-adoptive transfer-mediated protection was attenuated if the mice were treated with a neutralizing antibody to TGF-β. TGF-β and IL-10 induce distinct categories of immunomodulatory macrophages (M2c) compared with M2a macrophages, which are induced by IL-4 and IL-13 (148). M2c macrophages were more potent in inducing Tregs and protecting mice from the glomerulosclerosis, tubular atrophy, interstitial expansion, and proteinuria induced by Adriamycin, as compared with M2a macrophage. The advantage of M2c over M2a was lost after Treg depletion. In line with the protective role of adenosine in protection from AKI (105, 172), CD25+ Tregs from mice transgenic for human CD39 (hCD39-Tg) offered greater protection in the Adriamycin model as compared with either wild-type CD25+ Tregs or CD25 T cells from hCD39-Tg mice (251).

In our own studies, pretreatment with the IL-2 and IL-33 containing hybrid cytokine (IL233), which offered protection from ischemic and cisplatin injury, also potently protected BALB/c mice from Adriamycin (226). IL233 treatment as late as 2 wk post-Adriamycin infusion reversed ongoing nephropathy and protected kidneys from renal dysfunction, inflammation, injury, and fibrosis, indicating that enhancing Tregs with IL233 late in nephrotoxic injury models may be a viable strategy to limit ongoing injury and promote repair (V. Sabapathy, N. Cheru, M. Saleh, and R. Sharma, unpublished observations).

Tregs in Glomerulopathies

Autoimmune diseases such as systemic lupus erythematosus (SLE or lupus), Goodpasture’s syndrome, IgA nephropathy (idiopathic), and others often have glomerular manifestations. Defects in Foxp3+ Tregs have been indicated in these conditions, especially in lupus glomerulonephritis (GN) as discussed below.

Tregs in Lupus Glomerulonephritis

Lupus GN and Treg deficiency.

SLE is a systemic autoimmune disease manifested as increased production of autoantibodies, immune complex deposition, and multiorgan inflammation (39, 240). Kidneys are affected in over 50% of lupus patients, leading to glomerulonephritis (29), with a fifth of these patients proceeding to ESRD (253). Defects in IL-2 production by T cells from lupus patients were found to contribute to autoimmunity by Lippe et al. (142) in patients and mouse models. IL-2 deficiency interferes with Treg homeostasis and activation-induced cell death, and promotes differentiation of T follicular helper (TfH) (10, 90, 168) and TH17 cells (16, 124) (241). Clinical and experimental studies show that Treg deficiency correlates with incidence and severity of lupus (reviewed in Ref. 118). We will review the studies for lupus GN below.

Xing et al. (263) found that in a cohort of 60 lupus patients the proportion of CD4+CD25+Foxp3+ Tregs, as well as the level of Foxp3 expression per cell (measured as the mean fluorescence intensity of Foxp3), was lower in SLE patients as compared with healthy controls. These values were further reduced in patients with active lupus GN. This was accompanied by a corresponding increase of Th17 cells in SLE patients that was further increased in SLE patients with GN, indicating that an imbalance in Treg/Th17 ratios contributed to renal pathogenesis lupus patients. A similar Treg/Th17 imbalance was observed in another study with a cohort of 42 patients with new-onset lupus nephritis (262). Additionally, lower serum levels and higher urinary levels of TGF-β1, also correlated with higher lupus scores and increased nephritis in this cohort, indicating a diagnostic value of urinary TGF-β1 with disease activity.

Another independent study comparing 17 patients with lupus and 20 patients with lupus GN showed greater Th17 and reduced CD4+CD25+Foxp3+ Treg cells in patients with nephritis compared with SLE patients without nephritis (23). A recent study suggested that reduced numbers of regulatory CD4+CD25+Foxp3+ cells as a biomarker of the activity of SLE, particularly of renal involvement. In a pilot histological study, kidney biopsy samples showed lower proportions of Foxp3+ cells in patients with active proliferative class IV nephritis as compared with that of that of ANCA glomerulonephritis, acute tubulointerstitial nephritis, and nephroangiosclerosis (1). Coexpression of IL-17 in Foxp3+ cells (called Treg17) was proposed to impart Tregs with increased ability to suppress Th17 responses (110). By using a pristane-induced lupus GN model, abrogation of STAT3 expression specifically in Tregs resulted in higher disease score, which correlated with reduced expression of CC chemokine receptor 6 (CCR6) in Tregs, suggesting that STAT3-mediated CCR6 expression may enable Treg17 to prevent Th17-mediated pathogenesis. In contrast, a recent study demonstrated that Tregs coexpressing Foxp3 and the Th17 lineage-specific transcription factor Rorγt inhibited the anti-inflammatory Th2 responses in the pristane-induced lupus nephritis model (112). A Foxp3-creRORCfl/fl -mediated ablation of IL-17 production in Tregs ameliorated the vasculitis and nephritis phenotypes.

Treg-based therapeutic approaches in Lupus GN.

Using a model of d3-thymectomy-induced accelerated GN in NZM2328 mice, Bagavant et al. (8) showed that adoptive transfer of freshly isolated Tregs prevented production of autoantibodies and inflammation in prostate, thyroid, and lacrimal glands; however, this did not inhibit GN. Scalapino et al. (200) showed that ex vivo-expanded CD4+CD25+CD62LHi cells reduced the incidence and progression of GN in NZB/W F1 mice, suggesting that increasing Tregs may be a viable therapeutic approach for lupus GN.

Other strategies that were found to be beneficial in lupus GN were accompanied with an increase in Tregs. In clinical trials, depletion of B cells with rituximab correlated with increased levels of Tregs (244). Although not demonstrated directly, treatment with abatacept (CTLA4-Ig) in combination with MMF and glucocorticoids improved autoantibody levels, complement and urinary protein levels (62), and was hypothesized to function through increasing Treg abundance (220). In preclinical studies, a tolerogenic regimen with a low-dose infusion of nucleosomal peptide in lupus-prone SNF1 mice was effective at reducing production of inflammatory mediators and restricted leukocyte infiltration in kidneys (95). Importantly, the treatment induced differentiation of CD4+CD25+ and CD8+ Tregs, which produced TGF-β1 for mediating suppression. Adoptive transfer of the Tregs expanded by the nucleosomal peptide also suppressed lupus-associated autoimmune responses in recipients. Long-term oral treatment of lupus-prone NZB/WF1 mice with curcumin attenuated production of autoantibodies, immune complex deposition, renal inflammation, and proteinuria (126). This protection was abrogated by treatment with PC61, suggesting that the interaction of curcumin with Tregs participated in the protection.

IL-2 treatment to expand Tregs for treatment of autoimmune diseases has been investigated experimentally (70, 128, 145). Low-dose IL-2 therapy improved the outcomes in Graft versus host disease (GVHD) and Hepatitis C virus (HCV) associated vasculitis (155, 194). Treatment with low-dose IL-2, although beneficial, may also induce deleterious effects at high concentrations (13, 49, 175). In two recent clinical trial, low-dose IL-2 therapy corrected T-cell defects by expanding Tregs, inhibiting TfH and TH17 cells, and also lowered the SELENA-SLEDAI disease index; however, data for GN was lacking (76, 246). Mizui et al. (162) showed that treatment of MRL/lpr mice with recombinant AAV-expressing IL-2 increased Tregs and reduced inflammation. However, this effect was attributed to an increase in CD8+ T cells and NK cells. In another experimental study, treatment of NZB/WF1 mice with an IL-2/anti-IL-2 antibody complex increased Tregs in spleen and kidneys and reduced autoantibody levels (267). This was accompanied with reduced renal immune complex deposition, lower inflammation in the kidneys, lower proteinuria scores, and decreased mortality. Importantly, the treatment was more effective than the MMF and steroid combination therapy.

Lupus-prone NZM2328 mice have a kinetic loss in the ability to produce IL-2, which led to an age-dependent decrease in total Tregs, as well as ST2-expressing Tregs (R. Sharma, M. Stremska, C. Dai, S. Mohammad, S. M. Fu, unpublished observations). A synergistic treatment with IL-2 and IL-33, especially in the form of IL233 hybrid cytokine, which bears the activity of IL-2 and IL-33 in a single molecule and was found to protect mice from AKI (see IL-2 and IL-33 above), also inhibited the onset of lupus GN in an accelerated model using NZM2328 mice. A one-time induction therapy with daily injection for 5 days, especially with the IL233 hybrid cytokine, increased Tregs in the renal lymph nodes for several weeks and inhibited the production of IFNγ and TNFα. Importantly, IL233 administration after the onset of proteinuria, induced persistent remission lasting for months in >85% of mice. IL233 treatment also protected MRL/lpr mice from proteinuria and mortality accompanied by an increase in Tregs and attenuation of pro-inflammatory cytokines.

Tregs and Nephrotoxic Serum Nephritis

The nephrotoxic serum nephritis (NTS) model is commonly used to study rapidly progressive glomerulonephritis, which culminates in glomerulosclerosis (94). It is induced by injection of antibodies raised either in rabbits or sheep that are directed against the renal glomerular basement membrane (GBM). The disease is characterized by rapid inflammation and infiltration of leukocytes in the kidneys. In the very first report (258) for the therapeutic use of Tregs for GN, adoptive transfer of 1 million CD4+CD25+ Tregs before induction of anti-GBM GN reduced inflammation and protected from renal damage. By using eGFP marked Tregs, it was found that the transferred Tregs trafficked to the lymph nodes. This trafficking was important for the protection because in another independent study Tregs deficient for CCR7 were unable to protect mice (47). In this model, IL-9 production by Tregs was important for recruitment of mast cells to the lymph nodes, which was required for the protection, as adoptive transfer of Tregs from IL-9-deficient mice did not protect from NTS, and mast cell-deficient mice were also found to be more susceptible, thus indicating a yet unidentified role of a Treg and mast cell nexus in protection from GN (48). Although not shown, mast cells or other myeloid cells may participate in providing costimulation for Treg-mediated protection because in an unrelated study treatment of rats with a super-agonistic antibody to CD28 (JJ316) robustly increased Tregs and protected rats from NTS (230). A heightened Th1 response due to Treg depletion plays a pathogenic role in NTS (179), and increased expression of the Th1-specific transcription factor T-Bet in Tregs made them better suppressors of Th1-driven GN, partly by improving the trafficking of Tregs to Th1-target regions in a CXCR3-dependent manner (167). However, Tregs coexpressing the Th17-specific factor RORγt contributed to NTS pathogenicity (111).

Studies with adoptive transfer of Tregs, although promising, should be carried out carefully, because during polyclonal expansion of Tregs using anti-CD3/CD28 stimulation with TGFβ and IL-2, there is a possibility of generating inducible Tregs, which in a recent study were shown to be ineffective in protecting from experimental sheep anti-GBM-induced GN and failed to suppress Th1 responses (68).

Tregs and Diabetic Kidney Disease

Diabetic kidney disease has become the largest contributor to ESRD and mortality (176, 266). Defects in IL-2 and Tregs are much appreciated in autoimmune T1D (183, 206), with clinical trials ongoing for Treg-adoptive therapy and Treg enhancement strategies (18). Recent studies show that inflammation contributes to ESRD in Type 2 diabetes (T2D) also (51, 115, 163), which is primarily considered a metabolic syndrome. In a clinical study, compared with healthy controls, circulating levels of Tregs were significantly lower in T2D patients with ongoing micro- and macroalbuminuria, and significant inverse correlations were noted between the disease course (measured as urinary albumin excretion rate) and circulating Treg levels (264). In experimental studies, Treg depletion with anti-CD25 antibody worsened proteinuria in diabetic mice, whereas adoptive transfer of Tregs was found to be protective (46). Treatment of genetically obese diabetic mice with the IL-2 and IL-33-containing IL233 hybrid cytokine (similar to the AKI and lupus GN studies) rapidly and robustly increased Foxp3+ Treg in lymphoid organs and adipose tissue, accompanied with greater accumulation of M2 macrophages. Importantly, the IL233 cytokine treatment induced normoglycemia, improved glucose tolerance, and protected mice from renal dysfunction (R. Sharma, S. Mohammad, M. Stremska, unpublished observations). In addition, the IL233 treatment also increased the recruitment of ILC2 to the adipose tissue, which may participate in regulating the metabolic syndrome, indicating that Treg modulation by IL-2 and IL-33 can be an attractive strategy to not only restrict inflammation but also regulate metabolism in a Treg-dependent manner. Indeed, the discovery that metabolism of Tregs is different—in terms of preference toward fatty acid oxidation as a source of energy as compared with preferred glycolytic mechanisms by inflammatory T cells—has evolved our thinking in the interplay of Tregs and metabolism (161).

CONCLUSION AND FUTURE IMPLICATIONS

The immune system is designed to constantly patrol the body to look for pathogens and other danger signals. We have begun to appreciate the role of the immune system in regulation of not only threats from pathogens but also regulation of cues that are macro- or microenvironmental. Although several mechanisms have been proposed for loss of tolerance during insult and injury, the onset of autoimmunity is still a black box. Nevertheless, Tregs play an important role, as evidenced above in several inflammatory diseases, including those in the kidney. In addition to their anti-inflammatory potential, Tregs may also serve as an indicator of successful immunomodulatory therapy, for example, in patients with membranous nephropathy, where at baseline Tregs were low and on rituximab therapy the Treg levels increased and were indicative of improvement of disease score, whereas in the nonresponders Treg levels were unchanged as compared with pretreatment baseline (190). Multiple anti-inflammatory mechanisms have been identified for renal and other inflammatory diseases, all of which converge on the central role of Tregs. Inflammatory pathways have the ability to adapt and change their course if one pathway is restricted. Therefore, multiple pathways need to be simultaneously evoked to offer optimal protection in these conditions. In our recent studies mentioned above, use of two pleiotropic pathways, an adaptive (IL-2) and an innate (IL-33), have shown remarkable synergy in strengthening the survival and function of Tregs and other renoprotective cell types, including group 2 innate lymphoid cells (188, 250). It is important to employ the benefits of IL-33 to our advantage, as has been demonstrated by multiple recent studies. By itself, IL-33 may have deleterious effects (42, 136), but directing IL-33 activity preferentially to Tregs, with the help of IL-2, may be a promising clinical approach to therapeutic Treg enhancement (Fig. 3). IL-33 and other mediators of cellular damage such as adenosine, sphingosine, and heat shock proteins are natural signals of cellular damage, which have the potential to activate inflammatory immune responses through release of intracellular DAMPs and neoantigens. These signals, therefore, also serve to activate Tregs to simultaneously limit collateral damage due to a runaway inflammatory response. Although techniques for manufacturing Tregs and Treg-adoptive therapy have progressed significantly with clinical trials ongoing in multiple disorders, many challenges remain, and strategies to improve endogenous Tregs are likely to offer significant advantages. One caveat is in conditions where endogenous Tregs are genetically defective, such as IPEX, where a combined approach of Treg-adoptive transfer and Treg-enhancing strategies may need to be employed for best protection.

Fig. 3.

Fig. 3.

Treg-enhancing strategies to protect from renal inflammatory diseases. In addition to the adoptive transfer, several other approaches have been used to promote the number and function of endogenous Tregs. These include small molecules such as fingolimod (FTY720), mycophenolate mofetil (MMF), adenosine receptor (A2AR) agonists, oxidized ATP (oATP), constituent of bee venom (PLA2); cell transfusions such as mesenchymal stem cells (MSCs), tolerogenic dendritic cells (DC); macromolecules including, rituximab, CD28 superagonists, low-dose IL-2, IL-2/anti-IL-2 complex and the recently described IL233 fusion cytokine, which bears IL-2 and IL-33 activities in one molecule. IL-2 upregulates the ST2 (IL-33 receptor) on Tregs enabling IL-2 and IL-33 to synergize and promote activation, proliferation, and recruitment of Tregs to protect from renal inflammatory diseases induced by ischemia-reperfusion, nephrotoxicity, lupus nephritis, and diabetic nephropathy, all of which contribute to end-stage renal disease (ESRD). Please see the text for details.

GRANTS

Support for this study was provided by National Institutes of Health Grants 1R01-DK-105833 [to R. Sharma (contact PI) and Shu Man Fu], 2R01-AI-116725 (subcontract from Fred Perrino, Wake Forest University, to R. Sharma), 1R01-DK-104963 (to G. R. Kinsey), 1R21- DK-112105 (to G. R. Kinsey), a UVA AstraZeneca Research Alliance award (to R. Sharma), and the LaunchPad Diabetes Fund (to R. Sharma). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or other funding agencies.

DISCLOSURES

A patent (US9840545 B2) for IL233 hybrid cytokine was awarded on December 12, 2017.

AUTHOR CONTRIBUTIONS

R.S. conceived and designed research; R.S. performed experiments; R.S. analyzed data; R.S. and G.R.K. interpreted results of experiments; R.S. and G.R.K. prepared figures; R.S. and G.R.K. drafted manuscript; R.S. and G.R.K. edited and revised manuscript; R.S. and G.R.K. approved final version of manuscript.

ACKNOWLEDGMENTS

R. Sharma and G. R. Kinsey thank the Center for Immunity, Inflammation and Regenerative Medicine, Division of Nephrology, Department of Medicine.

REFERENCES

  • 1.Afeltra A, Gigante A, Margiotta DP, Taffon C, Cianci R, Barbano B, Liberatori M, Amoroso A, Rossi Fanelli F. The involvement of T regulatory lymphocytes in a cohort of lupus nephritis patients: a pilot study. Intern Emerg Med 10: 677–683, 2015. doi: 10.1007/s11739-015-1212-x. [DOI] [PubMed] [Google Scholar]
  • 2.Akcay A, Nguyen Q, He Z, Turkmen K, Won Lee D, Hernando AA, Altmann C, Toker A, Pacic A, Ljubanovic DG, Jani A, Faubel S, Edelstein CL. IL-33 exacerbates acute kidney injury. J Am Soc Nephrol 22: 2057–2067, 2011. doi: 10.1681/ASN.2010091011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Alikhan MA, Summers SA, Gan PY, Chan AJ, Khouri MB, Ooi JD, Ghali JR, Odobasic D, Hickey MJ, Kitching AR, Holdsworth SR. Endogenous toll-like receptor 9 regulates AKI by promoting regulatory T cell recruitment. J Am Soc Nephrol 27: 706–714, 2016. doi: 10.1681/ASN.2014090927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Almeida AR, Legrand N, Papiernik M, Freitas AA. Homeostasis of peripheral CD4+ T cells: IL-2Rα and IL-2 shape a population of regulatory cells that controls CD4+ T cell numbers. J Immunol 169: 4850–4860, 2002. doi: 10.4049/jimmunol.169.9.4850. [DOI] [PubMed] [Google Scholar]
  • 5.Anders HJ, Rovin B. A pathophysiology-based approach to the diagnosis and treatment of lupus nephritis. Kidney Int 90: 493–501, 2016. doi: 10.1016/j.kint.2016.05.017. [DOI] [PubMed] [Google Scholar]
  • 6.Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of regulatory T cells with known specificity for antigen. Nat Immunol 3: 756–763, 2002. doi: 10.1038/ni816. [DOI] [PubMed] [Google Scholar]
  • 7.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S, Ivanov II, Umesaki Y, Itoh K, Honda K. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331: 337–341, 2011. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bagavant H, Tung KS. Failure of CD25+ T cells from lupus-prone mice to suppress lupus glomerulonephritis and sialoadenitis. J Immunol 175: 944–950, 2005. doi: 10.4049/jimmunol.175.2.944. [DOI] [PubMed] [Google Scholar]
  • 9.Bajwa A, Kinsey GR, Okusa MD. Immune mechanisms and novel pharmacological therapies of acute kidney injury. Curr Drug Targets 10: 1196–1204, 2009. doi: 10.2174/138945009789753174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ballesteros-Tato A, León B, Graf BA, Moquin A, Adams PS, Lund FE, Randall TD. Interleukin-2 inhibits germinal center formation by limiting T follicular helper cell differentiation. Immunity 36: 847–856, 2012. doi: 10.1016/j.immuni.2012.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Barnes MJ, Griseri T, Johnson AM, Young W, Powrie F, Izcue A. CTLA-4 promotes Foxp3 induction and regulatory T cell accumulation in the intestinal lamina propria. Mucosal Immunol 6: 324–334, 2013. doi: 10.1038/mi.2012.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Barthlott T, Moncrieffe H, Veldhoen M, Atkins CJ, Christensen J, O’Garra A, Stockinger B. CD25+ CD4+ T cells compete with naive CD4+ T cells for IL-2 and exploit it for the induction of IL-10 production. Int Immunol 17: 279–288, 2005. doi: 10.1093/intimm/dxh207. [DOI] [PubMed] [Google Scholar]
  • 13.Becker MO, Brückner C, Scherer HU, Wassermann N, Humrich JY, Hanitsch LG, Schneider U, Kawald A, Hanke K, Burmester GR, Riemekasten G. The monoclonal anti-CD25 antibody basiliximab for the treatment of progressive systemic sclerosis: an open-label study. Ann Rheum Dis 70: 1340–1341, 2011. doi: 10.1136/ard.2010.137935. [DOI] [PubMed] [Google Scholar]
  • 14.Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, Kelly TE, Saulsbury FT, Chance PF, Ochs HD. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 27: 20–21, 2001. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 15.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 204: 1765–1774, 2007. doi: 10.1084/jem.20070719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441: 235–238, 2006. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 17.Bin Dhuban K, d’Hennezel E, Nashi E, Bar-Or A, Rieder S, Shevach EM, Nagata S, Piccirillo CA. Coexpression of TIGIT and FCRL3 identifies Helios+ human memory regulatory T cells. J Immunol 194: 3687–3696, 2015. doi: 10.4049/jimmunol.1401803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, Herold KC, Lares A, Lee MR, Li K, Liu W, Long SA, Masiello LM, Nguyen V, Putnam AL, Rieck M, Sayre PH, Tang Q. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 7: 315ra189, 2015. doi: 10.1126/scitranslmed.aad4134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 311: 1924–1927, 2006. doi: 10.1126/science.1122927. [DOI] [PubMed] [Google Scholar]
  • 20.Brezis M, Rosen S. Hypoxia of the renal medulla—its implications for disease. N Engl J Med 332: 647–655, 1995. doi: 10.1056/NEJM199503093321006. [DOI] [PubMed] [Google Scholar]
  • 21.Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27: 68–73, 2001. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 22.Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA. IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J Immunol 178: 280–290, 2007. doi: 10.4049/jimmunol.178.1.280. [DOI] [PubMed] [Google Scholar]
  • 23.Cai XY, Luo M, Lin XJ, Xu YL, Tang C, Ye JH, Li WN, He ZX, Yu N. [Expression and significance of Th17 and Treg cells in peripheral blood of patients with systemic lupus erythematosus]. Zhonghua Yi Xue Za Zhi 92: 460–463, 2012. [DOI] [PubMed] [Google Scholar]
  • 24.Carreno BM, Collins M. The B7 family of ligands and its receptors: new pathways for costimulation and inhibition of immune responses. Annu Rev Immunol 20: 29–53, 2002. doi: 10.1146/annurev.immunol.20.091101.091806. [DOI] [PubMed] [Google Scholar]
  • 25.Carrier Y, Yuan J, Kuchroo VK, Weiner HL. Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-β T cell-transgenic mice. J Immunol 178: 179–185, 2007. doi: 10.4049/jimmunol.178.1.179. [DOI] [PubMed] [Google Scholar]
  • 26.Case J, Khan S, Khalid R, Khan A. Epidemiology of acute kidney injury in the intensive care unit. Crit Care Res Pract 2013: 479730, 2013. doi: 10.1155/2013/479730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cayrol C, Girard JP. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol 31: 31–37, 2014. doi: 10.1016/j.coi.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 28.Cekic C, Linden J. Purinergic regulation of the immune system. Nat Rev Immunol 16: 177–192, 2016. doi: 10.1038/nri.2016.4. [DOI] [PubMed] [Google Scholar]
  • 29.Cervera R, Khamashta MA, Font J, Sebastiani GD, Gil A, Lavilla P, Mejía JC, Aydintug AO, Chwalinska-Sadowska H, de Ramón E, Fernández-Nebro A, Galeazzi M, Valen M, Mathieu A, Houssiau F, Caro N, Alba P, Ramos-Casals M, Ingelmo M, Hughes GR; European Working Party on Systemic Lupus Erythematosus . Morbidity and mortality in systemic lupus erythematosus during a 10-year period: a comparison of early and late manifestations in a cohort of 1,000 patients. Medicine (Baltimore) 82: 299–308, 2003. doi: 10.1097/01.md.0000091181.93122.55. [DOI] [PubMed] [Google Scholar]
  • 30.Chang TT, Kuchroo VK, Sharpe AH. Role of the B7-CD28/CTLA-4 pathway in autoimmune disease. Curr Dir Autoimmun 5: 113–130, 2002. doi: 10.1159/000060550. [DOI] [PubMed] [Google Scholar]
  • 31.Chen W, Jin W, Hardegen N, Lei KJ, Li L, Marinos N, McGrady G, Wahl SM. Conversion of peripheral CD4+CD25 naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3. J Exp Med 198: 1875–1886, 2003. doi: 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chi H. Sphingosine-1-phosphate and immune regulation: trafficking and beyond. Trends Pharmacol Sci 32: 16–24, 2011. doi: 10.1016/j.tips.2010.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, Gasteiger G, Feng Y, Fontenot JD, Rudensky AY. An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17: 1322–1333, 2016. doi: 10.1038/ni.3540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cho WY, Choi HM, Lee SY, Kim MG, Kim HK, Jo SK. The role of Tregs and CD11c+ macrophages/dendritic cells in ischemic preconditioning of the kidney. Kidney Int 78: 981–992, 2010. doi: 10.1038/ki.2010.266. [DOI] [PubMed] [Google Scholar]
  • 35.Chong MM, Rasmussen JP, Rudensky AY, Littman DR. The RNAseIII enzyme Drosha is critical in T cells for preventing lethal inflammatory disease. J Exp Med 205: 2005–2017, 2008. doi: 10.1084/jem.20081219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Collison LW, Chaturvedi V, Henderson AL, Giacomin PR, Guy C, Bankoti J, Finkelstein D, Forbes K, Workman CJ, Brown SA, Rehg JE, Jones ML, Ni HT, Artis D, Turk MJ, Vignali DA. IL-35-mediated induction of a potent regulatory T cell population. Nat Immunol 11: 1093–1101, 2010. doi: 10.1038/ni.1952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Collison LW, Workman CJ, Kuo TT, Boyd K, Wang Y, Vignali KM, Cross R, Sehy D, Blumberg RS, Vignali DA. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 450: 566–569, 2007. doi: 10.1038/nature06306. [DOI] [PubMed] [Google Scholar]
  • 38.Coombes JL, Siddiqui KR, Arancibia-Cárcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-β and retinoic acid-dependent mechanism. J Exp Med 204: 1757–1764, 2007. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Crow MK. Developments in the clinical understanding of lupus. Arthritis Res Ther 11: 245, 2009. doi: 10.1186/ar2762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Davidson TS, DiPaolo RJ, Andersson J, Shevach EM. Cutting Edge: IL-2 is essential for TGF-β-mediated induction of Foxp3+ T regulatory cells. J Immunol 178: 4022–4026, 2007. doi: 10.4049/jimmunol.178.7.4022. [DOI] [PubMed] [Google Scholar]
  • 41.Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 204: 1257–1265, 2007. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Demirtas L, Turkmen K, Kandemir FM, Ozkaraca M, Kucukler S, Gürbüzel M, Comaklı S. The possible role of interleukin-33 as a new player in the pathogenesis of contrast-induced nephropathy in diabetic rats. Ren Fail 38: 952–960, 2016. doi: 10.3109/0886022X.2016.1165034. [DOI] [PubMed] [Google Scholar]
  • 43.Dousdampanis P, Trigka K, Mouzaki A. Tregs and kidney: from diabetic nephropathy to renal transplantation. World J Transplant 6: 556–563, 2016. doi: 10.5500/wjt.v6.i3.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Edwards JP, Hand TW, Morais da Fonseca D, Glass DD, Belkaid Y, Shevach EM. The GARP/Latent TGF-β1 complex on Treg cells modulates the induction of peripherally derived Treg cells during oral tolerance. Eur J Immunol 46: 1480–1489, 2016. doi: 10.1002/eji.201546204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Edwards JP, Thornton AM, Shevach EM. Release of active TGF-β1 from the latent TGF-β1/GARP complex on T regulatory cells is mediated by integrin β8. J Immunol 193: 2843–2849, 2014. doi: 10.4049/jimmunol.1401102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Eller K, Kirsch A, Wolf AM, Sopper S, Tagwerker A, Stanzl U, Wolf D, Patsch W, Rosenkranz AR, Eller P. Potential role of regulatory T cells in reversing obesity-linked insulin resistance and diabetic nephropathy. Diabetes 60: 2954–2962, 2011. doi: 10.2337/db11-0358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Eller K, Weber T, Pruenster M, Wolf AM, Mayer G, Rosenkranz AR, Rot A. CCR7 deficiency exacerbates injury in acute nephritis due to aberrant localization of regulatory T cells. J Am Soc Nephrol 21: 42–52, 2010. doi: 10.1681/ASN.2009020133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Eller K, Wolf D, Huber JM, Metz M, Mayer G, McKenzie AN, Maurer M, Rosenkranz AR, Wolf AM. IL-9 production by regulatory T cells recruits mast cells that are essential for regulatory T cell-induced immune suppression. J Immunol 186: 83–91, 2011. doi: 10.4049/jimmunol.1001183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Enghard P, Rieder C, Kopetschke K, Klocke JR, Undeutsch R, Biesen R, Dragun D, Gollasch M, Schneider U, Aupperle K, Humrich JY, Hiepe F, Backhaus M, Radbruch AH, Burmester GR, Riemekasten G. Urinary CD4 T cells identify SLE patients with proliferative lupus nephritis and can be used to monitor treatment response. Ann Rheum Dis 73: 277–283, 2014. doi: 10.1136/annrheumdis-2012-202784. [DOI] [PubMed] [Google Scholar]
  • 50.Epstein FH. Hypoxia of the renal medulla. Q J Med 57: 807–810, 1985. [PubMed] [Google Scholar]
  • 51.Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res Clin Pract 105: 141–150, 2014. doi: 10.1016/j.diabres.2014.04.006. [DOI] [PubMed] [Google Scholar]
  • 52.Feng Y, Arvey A, Chinen T, van der Veeken J, Gasteiger G, Rudensky AY. Control of the inheritance of regulatory T cell identity by a cis element in the Foxp3 locus. Cell 158: 749–763, 2014. doi: 10.1016/j.cell.2014.07.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fife BT, Bluestone JA. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol Rev 224: 166–182, 2008. doi: 10.1111/j.1600-065X.2008.00662.x. [DOI] [PubMed] [Google Scholar]
  • 54.Fisson S, Darrasse-Jèze G, Litvinova E, Septier F, Klatzmann D, Liblau R, Salomon BL. Continuous activation of autoreactive CD4+ CD25+ regulatory T cells in the steady state. J Exp Med 198: 737–746, 2003. doi: 10.1084/jem.20030686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Fleming JO, Weinstock JV. Clinical trials of helminth therapy in autoimmune diseases: rationale and findings. Parasite Immunol 37: 277–292, 2015. doi: 10.1111/pim.12175. [DOI] [PubMed] [Google Scholar]
  • 56.Floess S, Freyer J, Siewert C, Baron U, Olek S, Polansky J, Schlawe K, Chang HD, Bopp T, Schmitt E, Klein-Hessling S, Serfling E, Hamann A, Huehn J. Epigenetic control of the foxp3 locus in regulatory T cells. PLoS Biol 5: e38, 2007. doi: 10.1371/journal.pbio.0050038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4: 330–336, 2003. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 58.Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 6: 1142–1151, 2005. doi: 10.1038/ni1263. [DOI] [PubMed] [Google Scholar]
  • 59.Fontenot JD, Rudensky AY. A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor Foxp3. Nat Immunol 6: 331–337, 2005. doi: 10.1038/ni1179. [DOI] [PubMed] [Google Scholar]
  • 60.Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Kuchroo VK, Sharpe AH. PD-L1 regulates the development, maintenance, and function of induced regulatory T cells. J Exp Med 206: 3015–3029, 2009. doi: 10.1084/jem.20090847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Friedline RH, Brown DS, Nguyen H, Kornfeld H, Lee J, Zhang Y, Appleby M, Der SD, Kang J, Chambers CA. CD4+ regulatory T cells require CTLA-4 for the maintenance of systemic tolerance. J Exp Med 206: 421–434, 2009. doi: 10.1084/jem.20081811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Furie R, Nicholls K, Cheng TT, Houssiau F, Burgos-Vargas R, Chen SL, Hillson JL, Meadows-Shropshire S, Kinaszczuk M, Merrill JT. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol 66: 379–389, 2014. doi: 10.1002/art.38260. [DOI] [PubMed] [Google Scholar]
  • 63.Furtado GC, Curotto de Lafaille MA, Kutchukhidze N, Lafaille JJ. Interleukin 2 signaling is required for CD4+ regulatory T cell function. J Exp Med 196: 851–857, 2002. doi: 10.1084/jem.20020190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 504: 446–450, 2013. doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
  • 65.Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Satpute SR, Crow MT, King LS, Rabb H. Foxp3+ regulatory T cells participate in repair of ischemic acute kidney injury. Kidney Int 76: 717–729, 2009. doi: 10.1038/ki.2009.259. [DOI] [PubMed] [Google Scholar]
  • 66.Gandolfo MT, Jang HR, Bagnasco SM, Ko GJ, Agreda P, Soloski MJ, Crow MT, Rabb H. Mycophenolate mofetil modifies kidney tubular injury and Foxp3+ regulatory T cell trafficking during recovery from experimental ischemia-reperfusion. Transpl Immunol 23: 45–52, 2010. doi: 10.1016/j.trim.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Gavin MA, Rasmussen JP, Fontenot JD, Vasta V, Manganiello VC, Beavo JA, Rudensky AY. Foxp3-dependent programme of regulatory T-cell differentiation. Nature 445: 771–775, 2007. doi: 10.1038/nature05543. [DOI] [PubMed] [Google Scholar]
  • 68.Ghali JR, Alikhan MA, Holdsworth SR, Kitching AR. Induced regulatory T cells are phenotypically unstable and do not protect mice from rapidly progressive glomerulonephritis. Immunology 150: 100–114, 2017. doi: 10.1111/imm.12671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol 174: 1783–1786, 2005. doi: 10.4049/jimmunol.174.4.1783. [DOI] [PubMed] [Google Scholar]
  • 70.Grinberg-Bleyer Y, Baeyens A, You S, Elhage R, Fourcade G, Gregoire S, Cagnard N, Carpentier W, Tang Q, Bluestone J, Chatenoud L, Klatzmann D, Salomon BL, Piaggio E. IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J Exp Med 207: 1871–1878, 2010. doi: 10.1084/jem.20100209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Groux H, O’Garra A, Bigler M, Rouleau M, Antonenko S, de Vries JE, Roncarolo MG. A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 389: 737–742, 1997. doi: 10.1038/39614. [DOI] [PubMed] [Google Scholar]
  • 72.Gunnlaugsdottir B, Maggadottir SM, Skaftadottir I, Ludviksson BR. The ex vivo induction of human CD103+ CD25hi Foxp3+ CD4+ and CD8+ Tregs is IL-2 and TGF-β1 dependent. Scand J Immunol 77: 125–134, 2013. doi: 10.1111/sji.12009. [DOI] [PubMed] [Google Scholar]
  • 73.Haase VH. Mechanisms of hypoxia responses in renal tissue. J Am Soc Nephrol 24: 537–541, 2013. doi: 10.1681/ASN.2012080855. [DOI] [PubMed] [Google Scholar]
  • 74.Haribhai D, Lin W, Relland LM, Truong N, Williams CB, Chatila TA. Regulatory T cells dynamically control the primary immune response to foreign antigen. J Immunol 178: 2961–2972, 2007. doi: 10.4049/jimmunol.178.5.2961. [DOI] [PubMed] [Google Scholar]
  • 75.Haxhinasto S, Mathis D, Benoist C. The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3+ cells. J Exp Med 205: 565–574, 2008. doi: 10.1084/jem.20071477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, Jin Y, Gan Y, Hu X, Jia R, Xu C, Hou Z, Leong YA, Zhu L, Feng J, An Y, Jia Y, Li C, Liu X, Ye H, Ren L, Li R, Yao H, Li Y, Chen S, Zhang X, Su Y, Guo J, Shen N, Morand EF, Yu D, Li Z. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med 22: 991–993, 2016. doi: 10.1038/nm.4148. [DOI] [PubMed] [Google Scholar]
  • 77.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 299: 1057–1061, 2003. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 78.Horvath C, Andrews L, Baumann A, Black L, Blanset D, Cavagnaro J, Hastings KL, Hutto DL, MacLachlan TK, Milton M, Reynolds T, Roberts S, Rogge M, Sims J, Treacy G, Warner G, Green JD. Storm forecasting: additional lessons from the CD28 superagonist TGN1412 trial. Nat Rev Immunol 12: 740, 2012. doi: 10.1038/nri3302. [DOI] [PubMed] [Google Scholar]
  • 79.Horwitz DA, Zheng SG, Wang J, Gray JD. Critical role of IL-2 and TGF-β in generation, function and stabilization of Foxp3+CD4+ Treg. Eur J Immunol 38: 912–915, 2008. doi: 10.1002/eji.200738109. [DOI] [PubMed] [Google Scholar]
  • 80.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 7: 401–410, 2006. doi: 10.1038/ni1318. [DOI] [PubMed] [Google Scholar]
  • 81.Hu J, Zhang L, Wang N, Ding R, Cui S, Zhu F, Xie Y, Sun X, Wu D, Hong Q, Li Q, Shi S, Liu X, Chen X. Mesenchymal stem cells attenuate ischemic acute kidney injury by inducing regulatory T cells through splenocyte interactions. Kidney Int 84: 521–531, 2013. doi: 10.1038/ki.2013.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Huang B, Zhao J, Lei Z, Shen S, Li D, Shen GX, Zhang GM, Feng ZH. miR-142-3p restricts cAMP production in CD4+CD25 T cells and CD4+CD25+ TREG cells by targeting AC9 mRNA. EMBO Rep 10: 180–185, 2009. doi: 10.1038/embor.2008.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, Powell JD, Pardoll DM, Drake CG, Vignali DA. Role of LAG-3 in regulatory T cells. Immunity 21: 503–513, 2004. doi: 10.1016/j.immuni.2004.08.010. [DOI] [PubMed] [Google Scholar]
  • 84.Huehn J, Beyer M. Epigenetic and transcriptional control of Foxp3+ regulatory T cells. Semin Immunol 27: 10–18, 2015. doi: 10.1016/j.smim.2015.02.002. [DOI] [PubMed] [Google Scholar]
  • 85.Humphreys NE, Xu D, Hepworth MR, Liew FY, Grencis RK. IL-33, a potent inducer of adaptive immunity to intestinal nematodes. J Immunol 180: 2443–2449, 2008. doi: 10.4049/jimmunol.180.4.2443. [DOI] [PubMed] [Google Scholar]
  • 86.Hünig T. Response to ‘Storm forecasting: additional lessons from the CD28 superagonist TGN1412 trial’. Nat Rev Immunol 12: 317–318, 2012. doi: 10.1038/nri3192-c2. [DOI] [PubMed] [Google Scholar]
  • 87.Jang HR, Rabb H. Immune cells in experimental acute kidney injury. Nat Rev Nephrol 11: 88–101, 2015. doi: 10.1038/nrneph.2014.180. [DOI] [PubMed] [Google Scholar]
  • 88.Janson PC, Winerdal ME, Marits P, Thörn M, Ohlsson R, Winqvist O. FOXP3 promoter demethylation reveals the committed Treg population in humans. PLoS One 3: e1612, 2008. doi: 10.1371/journal.pone.0001612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jaworska K, Ratajczak J, Huang L, Whalen K, Yang M, Stevens BK, Kinsey GR. Both PD-1 ligands protect the kidney from ischemia reperfusion injury. J Immunol 194: 325–333, 2015. doi: 10.4049/jimmunol.1400497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Johnston RJ, Choi YS, Diamond JA, Yang JA, Crotty S. STAT5 is a potent negative regulator of TFH cell differentiation. J Exp Med 209: 243–250, 2012. doi: 10.1084/jem.20111174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Jordan MS, Boesteanu A, Reed AJ, Petrone AL, Holenbeck AE, Lerman MA, Naji A, Caton AJ. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat Immunol 2: 301–306, 2001. doi: 10.1038/86302. [DOI] [PubMed] [Google Scholar]
  • 92.Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 30: 531–564, 2012. doi: 10.1146/annurev.immunol.25.022106.141623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Josefowicz SZ, Wilson CB, Rudensky AY. Cutting edge: TCR stimulation is sufficient for induction of Foxp3 expression in the absence of DNA methyltransferase 1. J Immunol 182: 6648–6652, 2009. doi: 10.4049/jimmunol.0803320. [DOI] [PubMed] [Google Scholar]
  • 94.Kaneko Y, Nimmerjahn F, Madaio MP, Ravetch JV. Pathology and protection in nephrotoxic nephritis is determined by selective engagement of specific Fc receptors. J Exp Med 203: 789–797, 2006. doi: 10.1084/jem.20051900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kang HK, Michaels MA, Berner BR, Datta SK. Very low-dose tolerance with nucleosomal peptides controls lupus and induces potent regulatory T cell subsets. J Immunol 174: 3247–3255, 2005. doi: 10.4049/jimmunol.174.6.3247. [DOI] [PubMed] [Google Scholar]
  • 96.Kang SW, Kim SH, Lee N, Lee WW, Hwang KA, Shin MS, Lee SH, Kim WU, Kang I. 1,25-Dihyroxyvitamin D3 promotes FOXP3 expression via binding to vitamin D response elements in its conserved noncoding sequence region. J Immunol 188: 5276–5282, 2012. doi: 10.4049/jimmunol.1101211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kasagi S, Kawano S, Kumagai S. PD-1 and autoimmunity. Crit Rev Immunol 31: 265–295, 2011. doi: 10.1615/CritRevImmunol.v31.i4.10. [DOI] [PubMed] [Google Scholar]
  • 98.Kerdiles YM, Stone EL, Beisner DR, McGargill MA, Ch’en IL, Stockmann C, Katayama CD, Hedrick SM. Foxo transcription factors control regulatory T cell development and function. Immunity 33: 890–904, 2010. doi: 10.1016/j.immuni.2010.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Kim H, Lee G, Park S, Chung HS, Lee H, Kim JY, Nam S, Kim SK, Bae H. Bee venom mitigates cisplatin-induced nephrotoxicity by regulating CD4+CD25+Foxp3+ regulatory T cells in mice. Evid Based Complement Alternat Med 2013: 879845, 2013. 10.1155/2013/879845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kim H, Lee H, Lee G, Jang H, Kim SS, Yoon H, Kang GH, Hwang DS, Kim SK, Chung HS, Bae H. Phospholipase A2 inhibits cisplatin-induced acute kidney injury by modulating regulatory T cells by the CD206 mannose receptor. Kidney Int 88: 550–559, 2015. doi: 10.1038/ki.2015.147. [DOI] [PubMed] [Google Scholar]
  • 101.Kim HP, Leonard WJ. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J Exp Med 204: 1543–1551, 2007. doi: 10.1084/jem.20070109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Kim MG, Jung Cho E, Won Lee J, Sook Ko Y, Young Lee H, Jo SK, Cho WY, Kim HK. The heat-shock protein-70-induced renoprotective effect is partially mediated by CD4+CD25+Foxp3+ regulatory T cells in ischemia/reperfusion-induced acute kidney injury. Kidney Int 85: 62–71, 2014. doi: 10.1038/ki.2013.277. [DOI] [PubMed] [Google Scholar]
  • 103.Kim MG, Koo TY, Yan JJ, Lee E, Han KH, Jeong JC, Ro H, Kim BS, Jo SK, Oh KH, Surh CD, Ahn C, Yang J. IL-2/anti-IL-2 complex attenuates renal ischemia-reperfusion injury through expansion of regulatory T cells. J Am Soc Nephrol 24: 1529–1536, 2013. doi: 10.1681/ASN.2012080784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kim MG, Lee SY, Ko YS, Lee HY, Jo SK, Cho WY, Kim HK. CD4+CD25+ regulatory T cells partially mediate the beneficial effects of FTY720, a sphingosine-1-phosphate analogue, during ischaemia/reperfusion-induced acute kidney injury. Nephrol Dial Transplant 26: 111–124, 2011. doi: 10.1093/ndt/gfq480. [DOI] [PubMed] [Google Scholar]
  • 105.Kinsey GR, Huang L, Jaworska K, Khutsishvili K, Becker DA, Ye H, Lobo PI, Okusa MD. Autocrine adenosine signaling promotes regulatory T cell-mediated renal protection. J Am Soc Nephrol 23: 1528–1537, 2012. doi: 10.1681/ASN.2012010070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kinsey GR, Huang L, Vergis AL, Li L, Okusa MD. Regulatory T cells contribute to the protective effect of ischemic preconditioning in the kidney. Kidney Int 77: 771–780, 2010. doi: 10.1038/ki.2010.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kinsey GR, Sharma R, Huang L, Li L, Vergis AL, Ye H, Ju ST, Okusa MD. Regulatory T cells suppress innate immunity in kidney ischemia-reperfusion injury. J Am Soc Nephrol 20: 1744–1753, 2009. doi: 10.1681/ASN.2008111160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Kinsey GR, Sharma R, Okusa MD. Regulatory T cells in AKI. J Am Soc Nephrol 24: 1720–1726, 2013. doi: 10.1681/ASN.2013050502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Kitoh A, Ono M, Naoe Y, Ohkura N, Yamaguchi T, Yaguchi H, Kitabayashi I, Tsukada T, Nomura T, Miyachi Y, Taniuchi I, Sakaguchi S. Indispensable role of the Runx1-Cbfβ transcription complex for in vivo-suppressive function of FoxP3+ regulatory T cells. Immunity 31: 609–620, 2009. doi: 10.1016/j.immuni.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 110.Kluger MA, Melderis S, Nosko A, Goerke B, Luig M, Meyer MC, Turner JE, Meyer-Schwesinger C, Wegscheid C, Tiegs G, Stahl RA, Panzer U, Steinmetz OM. Treg17 cells are programmed by Stat3 to suppress Th17 responses in systemic lupus. Kidney Int 89: 158–166, 2016. doi: 10.1038/ki.2015.296. [DOI] [PubMed] [Google Scholar]
  • 111.Kluger MA, Meyer MC, Nosko A, Goerke B, Luig M, Wegscheid C, Tiegs G, Stahl RA, Panzer U, Steinmetz OM. RORγt+Foxp3+ cells are an independent bifunctional regulatory T cell lineage and mediate crescentic GN. J Am Soc Nephrol 27: 454–465, 2016. doi: 10.1681/ASN.2014090880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kluger MA, Nosko A, Ramcke T, Goerke B, Meyer MC, Wegscheid C, Luig M, Tiegs G, Stahl RA, Steinmetz OM. RORγt expression in Tregs promotes systemic lupus erythematosus via IL-17 secretion, alteration of Treg phenotype and suppression of Th2 responses. Clin Exp Immunol 188: 63–78, 2017. doi: 10.1111/cei.12905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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 10: 595–602, 2009. doi: 10.1038/ni.1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Koo TY, Lee JG, Yan JJ, Jang JY, Ju KD, Han M, Oh KH, Ahn C, Yang J. The P2X7 receptor antagonist, oxidized adenosine triphosphate, ameliorates renal ischemia-reperfusion injury by expansion of regulatory T cells. Kidney Int 92: 415–431, 2017. doi: 10.1016/j.kint.2017.01.031. [DOI] [PubMed] [Google Scholar]
  • 115.Kornete M, Mason ES, Piccirillo CA. Immune regulation in T1D and T2D: prospective role of Foxp3+ Treg cells in disease pathogenesis and treatment. Front Endocrinol (Lausanne) 4: 76, 2013. doi: 10.3389/fendo.2013.00076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.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 6: 1219–1227, 2005. doi: 10.1038/ni1265. [DOI] [PubMed] [Google Scholar]
  • 117.Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, Schickel JN, Tran DQ, Stoddard J, Zhang Y, Frucht DM, Dumitriu B, Scheinberg P, Folio LR, Frein CA, Price S, Koh C, Heller T, Seroogy CM, Huttenlocher A, Rao VK, Su HC, Kleiner D, Notarangelo LD, Rampertaap Y, Olivier KN, McElwee J, Hughes J, Pittaluga S, Oliveira JB, Meffre E, Fleisher TA, Holland SM, Lenardo MJ, Tangye SG, Uzel G. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345: 1623–1627, 2014. doi: 10.1126/science.1255904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kuhn A, Beissert S, Krammer PH. CD4+CD25+ regulatory T cells in human lupus erythematosus. Arch Dermatol Res 301: 71–81, 2009. doi: 10.1007/s00403-008-0891-9. [DOI] [PubMed] [Google Scholar]
  • 119.Kulpa DA, Lawani M, Cooper A, Peretz Y, Ahlers J, Sékaly RP. PD-1 coinhibitory signals: the link between pathogenesis and protection. Semin Immunol 25: 219–227, 2013. doi: 10.1016/j.smim.2013.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lai LW, Yong KC, Lien YH. Pharmacologic recruitment of regulatory T cells as a therapy for ischemic acute kidney injury. Kidney Int 81: 983–992, 2012. doi: 10.1038/ki.2011.412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lang PA, Merkler D, Funkner P, Shaabani N, Meryk A, Krings C, Barthuber C, Recher M, Brück W, Häussinger D, Ohashi PS, Lang KS. Oxidized ATP inhibits T-cell-mediated autoimmunity. Eur J Immunol 40: 2401–2408, 2010. doi: 10.1002/eji.200939838. [DOI] [PubMed] [Google Scholar]
  • 122.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, Peterson DA, Stappenbeck TS, Hsieh CS. Peripheral education of the immune system by colonic commensal microbiota. Nature 478: 250–254, 2011. doi: 10.1038/nature10434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lathrop SK, Santacruz NA, Pham D, Luo J, Hsieh CS. Antigen-specific peripheral shaping of the natural regulatory T cell population. J Exp Med 205: 3105–3117, 2008. doi: 10.1084/jem.20081359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Meylan F, Siegel R, Hennighausen L, Shevach EM, O’shea JJ. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26: 371–381, 2007. doi: 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 125.Le Mercier I, Lines JL, Noelle RJ. Beyond CTLA-4 and PD-1, the generation Z of negative checkpoint regulators. Front Immunol 6: 418, 2015. doi: 10.3389/fimmu.2015.00418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Lee H, Kim H, Lee G, Chung HS, Bae H. Curcumin attenuates lupus nephritis upon interaction with regulatory T cells in New Zealand Black/White mice. Br J Nutr 110: 69–76, 2013. doi: 10.1017/S0007114512004734. [DOI] [PubMed] [Google Scholar]
  • 127.Lee H, Nho D, Chung HS, Lee H, Shin MK, Kim SH, Bae H. CD4+CD25+ regulatory T cells attenuate cisplatin-induced nephrotoxicity in mice. Kidney Int 78: 1100–1109, 2010. doi: 10.1038/ki.2010.139. [DOI] [PubMed] [Google Scholar]
  • 128.Lee SY, Cho ML, Oh HJ, Ryu JG, Park MJ, Jhun JY, Park MK, Stone JC, Ju JH, Hwang SY, Park SH, Surh CD, Kim HY. Interleukin-2/anti-interleukin-2 monoclonal antibody immune complex suppresses collagen-induced arthritis in mice by fortifying interleukin-2/STAT5 signalling pathways. Immunology 137: 305–316, 2012. doi: 10.1111/imm.12008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lee SY, Lee YS, Choi HM, Ko YS, Lee HY, Jo SK, Cho WY, Kim HK. Distinct pathophysiologic mechanisms of septic acute kidney injury: role of immune suppression and renal tubular cell apoptosis in murine model of septic acute kidney injury. Crit Care Med 40: 2997–3006, 2012. doi: 10.1097/CCM.0b013e31825b912d. [DOI] [PubMed] [Google Scholar]
  • 130.Lee VW, Harris DC. Adriamycin nephropathy: a model of focal segmental glomerulosclerosis. Nephrology (Carlton) 16: 30–38, 2011. doi: 10.1111/j.1440-1797.2010.01383.x. [DOI] [PubMed] [Google Scholar]
  • 131.Levine AG, Arvey A, Jin W, Rudensky AY. Continuous requirement for the TCR in regulatory T cell function. Nat Immunol 15: 1070–1078, 2014. doi: 10.1038/ni.3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Li D, Kong C, Tsun A, Chen C, Song H, Shi G, Pan W, Dai D, Shen N, Li B. MiR-125a-5p decreases the sensitivity of Treg cells toward IL-6-mediated conversion by inhibiting IL-6R and STAT3 expression. Sci Rep 5: 14615, 2015. doi: 10.1038/srep14615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Li MO, Sanjabi S, Flavell RA. Transforming growth factor-β controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 25: 455–471, 2006. doi: 10.1016/j.immuni.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 134.Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-β1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 26: 579–591, 2007. doi: 10.1016/j.immuni.2007.03.014. [DOI] [PubMed] [Google Scholar]
  • 135.Li X, Liang Y, LeBlanc M, Benner C, Zheng Y. Function of a Foxp3 cis-element in protecting regulatory T cell identity. Cell 158: 734–748, 2014. doi: 10.1016/j.cell.2014.07.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Liang H, Xu F, Wen XJ, Liu HZ, Wang HB, Zhong JY, Yang CX, Zhang B. Interleukin-33 signaling contributes to renal fibrosis following ischemia reperfusion. Eur J Pharmacol 812: 18–27, 2017. doi: 10.1016/j.ejphar.2017.06.031. [DOI] [PubMed] [Google Scholar]
  • 137.Liang Y, Li Y, Kuang Q, Ding X, Wei Z, Fang Y. Superagonistic CD28 protects against renal ischemic injury by expansion of regulatory T-cell. Am J Nephrol 45: 389–399, 2017. doi: 10.1159/000470918. [DOI] [PubMed] [Google Scholar]
  • 138.Liao G, Nayak S, Regueiro JR, Berger SB, Detre C, Romero X, de Waal Malefyt R, Chatila TA, Herzog RW, Terhorst C. GITR engagement preferentially enhances proliferation of functionally competent CD4+CD25+FoxP3+ regulatory T cells. Int Immunol 22: 259–270, 2010. doi: 10.1093/intimm/dxq001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Liew FY. IL-33: a Janus cytokine. Ann Rheum Dis 71, Suppl 2: i101–i104, 2012. doi: 10.1136/annrheumdis-2011-200589. [DOI] [PubMed] [Google Scholar]
  • 140.Lin W, Haribhai D, Relland LM, Truong N, Carlson MR, Williams CB, Chatila TA. Regulatory T cell development in the absence of functional Foxp3. Nat Immunol 8: 359–368, 2007. doi: 10.1038/ni1445. [DOI] [PubMed] [Google Scholar]
  • 141.Lio CW, Hsieh CS. A two-step process for thymic regulatory T cell development. Immunity 28: 100–111, 2008. doi: 10.1016/j.immuni.2007.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Lippe R, Ohl K, Varga G, Rauen T, Crispin JC, Juang YT, Kuerten S, Tacke F, Wolf M, Roebrock K, Vogl T, Verjans E, Honke N, Ehrchen J, Foell D, Skryabin B, Wagner N, Tsokos GC, Roth J, Tenbrock K. CREMα overexpression decreases IL-2 production, induces a TH17 phenotype and accelerates autoimmunity. J Mol Cell Biol 4: 121–123, 2012. doi: 10.1093/jmcb/mjs004. [DOI] [PubMed] [Google Scholar]
  • 143.Liston A, Lu LF, O’Carroll D, Tarakhovsky A, Rudensky AY. Dicer-dependent microRNA pathway safeguards regulatory T cell function. J Exp Med 205: 1993–2004, 2008. doi: 10.1084/jem.20081062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Liston A, Masters SL. Homeostasis-altering molecular processes as mechanisms of inflammasome activation. Nat Rev Immunol 17: 208–214, 2017. doi: 10.1038/nri.2016.151. [DOI] [PubMed] [Google Scholar]
  • 145.Liu R, Zhou Q, La Cava A, Campagnolo DI, Van Kaer L, Shi FD. Expansion of regulatory T cells via IL-2/anti-IL-2 mAb complexes suppresses experimental myasthenia. Eur J Immunol 40: 1577–1589, 2010. doi: 10.1002/eji.200939792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Liu Y, Zhang P, Li J, Kulkarni AB, Perruche S, Chen W. A critical function for TGF-β signaling in the development of natural CD4+CD25+Foxp3+ regulatory T cells. Nat Immunol 9: 632–640, 2008. doi: 10.1038/ni.1607. [DOI] [PubMed] [Google Scholar]
  • 147.Lowther DE, Goods BA, Lucca LE, Lerner BA, Raddassi K, van Dijk D, Hernandez AL, Duan X, Gunel M, Coric V, Krishnaswamy S, Love JC, Hafler DA. PD-1 marks dysfunctional regulatory T cells in malignant gliomas. JCI Insight 1: pii: e85935, 2016. doi: 10.1172/jci.insight.85935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Lu J, Cao Q, Zheng D, Sun Y, Wang C, Yu X, Wang Y, Lee VW, Zheng G, Tan TK, Wang X, Alexander SI, Harris DC, Wang Y. Discrete functions of M2a and M2c macrophage subsets determine their relative efficacy in treating chronic kidney disease. Kidney Int 84: 745–755, 2013. doi: 10.1038/ki.2013.135. [DOI] [PubMed] [Google Scholar]
  • 149.Lu LF, Thai TH, Calado DP, Chaudhry A, Kubo M, Tanaka K, Loeb GB, Lee H, Yoshimura A, Rajewsky K, Rudensky AY. Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein. Immunity 30: 80–91, 2009. doi: 10.1016/j.immuni.2008.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Lünemann A, Lünemann JD, Münz C. Regulatory NK-cell functions in inflammation and autoimmunity. Mol Med 15: 352–358, 2009. doi: 10.2119/molmed.2009.00035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lv K, Zhang Y, Zhang M, Zhong M, Suo Q. Galectin-9 promotes TGF-β1-dependent induction of regulatory T cells via the TGF-β/Smad signaling pathway. Mol Med Rep 7: 205–210, 2013. doi: 10.3892/mmr.2012.1125. [DOI] [PubMed] [Google Scholar]
  • 152.Mahajan D, Wang Y, Qin X, Wang Y, Zheng G, Wang YM, Alexander SI, Harris DC. CD4+CD25+ regulatory T cells protect against injury in an innate murine model of chronic kidney disease. J Am Soc Nephrol 17: 2731–2741, 2006. doi: 10.1681/ASN.2005080842. [DOI] [PubMed] [Google Scholar]
  • 153.Malek TR, Yu A, Vincek V, Scibelli P, Kong L. CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rβ-deficient mice. Implications for the nonredundant function of IL-2. Immunity 17: 167–178, 2002. doi: 10.1016/S1074-7613(02)00367-9. [DOI] [PubMed] [Google Scholar]
  • 154.Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-β receptor. Immunity 25: 441–454, 2006. doi: 10.1016/j.immuni.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 155.Matsuoka K, Koreth J, Kim HT, Bascug G, McDonough S, Kawano Y, Murase K, Cutler C, Ho VT, Alyea EP, Armand P, Blazar BR, Antin JH, Soiffer RJ, Ritz J. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med 5: 179ra43, 2013. doi: 10.1126/scitranslmed.3005265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Matta BM, Lott JM, Mathews LR, Liu Q, Rosborough BR, Blazar BR, Turnquist HR. IL-33 is an unconventional Alarmin that stimulates IL-2 secretion by dendritic cells to selectively expand IL-33R/ST2+ regulatory T cells. J Immunol 193: 4010–4020, 2014. doi: 10.4049/jimmunol.1400481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol 12: 991–1045, 1994. doi: 10.1146/annurev.iy.12.040194.005015. [DOI] [PubMed] [Google Scholar]
  • 158.Maynard CL, Harrington LE, Janowski KM, Oliver JR, Zindl CL, Rudensky AY, Weaver CT. Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3 precursor cells in the absence of interleukin 10. Nat Immunol 8: 931–941, 2007. doi: 10.1038/ni1504. [DOI] [PubMed] [Google Scholar]
  • 159.McHugh RS, Whitters MJ, Piccirillo CA, Young DA, Shevach EM, Collins M, Byrne MC. CD4+CD25+ immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity 16: 311–323, 2002. doi: 10.1016/S1074-7613(02)00280-7. [DOI] [PubMed] [Google Scholar]
  • 160.McIntire KR, Sell S, Miller JF. Pathogenesis of the post-neonatal thymectomy wasting syndrome. Nature 204: 151–155, 1964. doi: 10.1038/204151a0. [DOI] [PubMed] [Google Scholar]
  • 161.Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG, Rathmell JC. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 186: 3299–3303, 2011. doi: 10.4049/jimmunol.1003613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mizui M, Koga T, Lieberman LA, Beltran J, Yoshida N, Johnson MC, Tisch R, Tsokos GC. IL-2 protects lupus-prone mice from multiple end-organ damage by limiting CD4CD8 IL-17-producing T cells. J Immunol 193: 2168–2177, 2014. doi: 10.4049/jimmunol.1400977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Mora C, Navarro JF. Inflammation and diabetic nephropathy. Curr Diab Rep 6: 463–468, 2006. doi: 10.1007/s11892-006-0080-1. [DOI] [PubMed] [Google Scholar]
  • 164.Murawski MR, Litherland SA, Clare-Salzler MJ, Davoodi-Semiromi A. Upregulation of Foxp3 expression in mouse and human Treg is IL-2/STAT5 dependent: implications for the NOD STAT5B mutation in diabetes pathogenesis. Ann N Y Acad Sci 1079: 198–204, 2006. doi: 10.1196/annals.1375.031. [DOI] [PubMed] [Google Scholar]
  • 165.Naghavian R, Ghaedi K, Kiani-Esfahani A, Ganjalikhani-Hakemi M, Etemadifar M, Nasr-Esfahani MH. miR-141 and miR-200a, revelation of new possible players in modulation of Th17/Treg differentiation and pathogenesis of multiple sclerosis. PLoS One 10: e0124555, 2015. doi: 10.1371/journal.pone.0124555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Nishizuka Y, Sakakura T. Thymus and reproduction: sex-linked dysgenesia of the gonad after neonatal thymectomy in mice. Science 166: 753–755, 1969. doi: 10.1126/science.166.3906.753. [DOI] [PubMed] [Google Scholar]
  • 167.Nosko A, Kluger MA, Diefenhardt P, Melderis S, Wegscheid C, Tiegs G, Stahl RA, Panzer U, Steinmetz OM. T-Bet enhances regulatory T cell fitness and directs control of Th1 responses in crescentic GN. J Am Soc Nephrol 28: 185–196, 2017. doi: 10.1681/ASN.2015070820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Nurieva RI, Podd A, Chen Y, Alekseev AM, Yu M, Qi X, Huang H, Wen R, Wang J, Li HS, Watowich SS, Qi H, Dong C, Wang D. STAT5 protein negatively regulates T follicular helper (Tfh) cell generation and function. J Biol Chem 287: 11234–11239, 2012. doi: 10.1074/jbc.M111.324046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.O’Garra A, Vieira P. Regulatory T cells and mechanisms of immune system control. Nat Med 10: 801–805, 2004. doi: 10.1038/nm0804-801. [DOI] [PubMed] [Google Scholar]
  • 170.Ohkura N, Kitagawa Y, Sakaguchi S. Development and maintenance of regulatory T cells. Immunity 38: 414–423, 2013. doi: 10.1016/j.immuni.2013.03.002. [DOI] [PubMed] [Google Scholar]
  • 171.Ohta A, Kini R, Ohta A, Subramanian M, Madasu M, Sitkovsky M. The development and immunosuppressive functions of CD4+ CD25+ FoxP3+ regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol 3: 190, 2012. doi: 10.3389/fimmu.2012.00190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Okusa MD. A(2A) adenosine receptor: a novel therapeutic target in renal disease. Am J Physiol Renal Physiol 282: F10–F18, 2002. [DOI] [PubMed] [Google Scholar]
  • 173.Ouyang W, Beckett O, Ma Q, Paik JH, DePinho RA, Li MO. Foxo proteins cooperatively control the differentiation of Foxp3+ regulatory T cells. Nat Immunol 11: 618–627, 2010. doi: 10.1038/ni.1884. [DOI] [PubMed] [Google Scholar]
  • 174.Ouyang W, Liao W, Luo CT, Yin N, Huse M, Kim MV, Peng M, Chan P, Ma Q, Mo Y, Meijer D, Zhao K, Rudensky AY, Atwal G, Zhang MQ, Li MO. Novel Foxo1-dependent transcriptional programs control Treg cell function. Nature 491: 554–559, 2012. doi: 10.1038/nature11581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Owen KL, Shibata T, Izui S, Walker SE. Recombinant interleukin-2 therapy of systemic lupus erythematosus in the New Zealand black/New Zealand white mouse. J Biol Response Mod 8: 366–374, 1989. [PubMed] [Google Scholar]
  • 176.Park CW. Diabetic kidney disease: from epidemiology to clinical perspectives. Diabetes Metab J 38: 252–260, 2014. doi: 10.4093/dmj.2014.38.4.252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Park JH, Jang HR, Kim DH, Kwon GY, Lee JE, Huh W, Choi SJ, Oh W, Oh HY, Kim YG. Early, but not late, treatment with human umbilical cord blood-derived mesenchymal stem cells attenuates cisplatin nephrotoxicity through immunomodulation. Am J Physiol Renal Physiol 313: F984–F996, 2017. doi: 10.1152/ajprenal.00097.2016. [DOI] [PubMed] [Google Scholar]
  • 178.Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD, Arancibia-Cárcamo CV, Sobel RA, Rudensky AY, Kuchroo VK, Freeman GJ, Sharpe AH. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med 212: 1603–1621, 2015. doi: 10.1084/jem.20141030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Paust HJ, Ostmann A, Erhardt A, Turner JE, Velden J, Mittrücker HW, Sparwasser T, Panzer U, Tiegs G. Regulatory T cells control the Th1 immune response in murine crescentic glomerulonephritis. Kidney Int 80: 154–164, 2011. doi: 10.1038/ki.2011.108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Perazella MA. Renal vulnerability to drug toxicity. Clin J Am Soc Nephrol 4: 1275–1283, 2009. doi: 10.2215/CJN.02050309. [DOI] [PubMed] [Google Scholar]
  • 181.Piccirillo CA, Shevach EM. Naturally-occurring CD4+CD25+ immunoregulatory T cells: central players in the arena of peripheral tolerance. Semin Immunol 16: 81–88, 2004. doi: 10.1016/j.smim.2003.12.003. [DOI] [PubMed] [Google Scholar]
  • 182.Polansky JK, Kretschmer K, Freyer J, Floess S, Garbe A, Baron U, Olek S, Hamann A, von Boehmer H, Huehn J. DNA methylation controls Foxp3 gene expression. Eur J Immunol 38: 1654–1663, 2008. doi: 10.1002/eji.200838105. [DOI] [PubMed] [Google Scholar]
  • 183.Pop SM, Wong CP, Culton DA, Clarke SH, Tisch R. Single cell analysis shows decreasing FoxP3 and TGFβ1 coexpressing CD4+CD25+ regulatory T cells during autoimmune diabetes. J Exp Med 201: 1333–1346, 2005. doi: 10.1084/jem.20042398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Porter CM, Horvath-Arcidiacono JA, Singh AK, Horvath KA, Bloom ET, Mohiuddin MM. Characterization and expansion of baboon CD4+CD25+ Treg cells for potential use in a non-human primate xenotransplantation model. Xenotransplantation 14: 298–308, 2007. doi: 10.1111/j.1399-3089.2007.00416.x. [DOI] [PubMed] [Google Scholar]
  • 185.Rajewsky K. Clonal selection and learning in the antibody system. Nature 381: 751–758, 1996. doi: 10.1038/381751a0. [DOI] [PubMed] [Google Scholar]
  • 186.Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, Francisco L, Sharpe AH, Powrie F. Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. J Immunol 177: 4376–4383, 2006. doi: 10.4049/jimmunol.177.7.4376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Read S, Malmström V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25+CD4+ regulatory cells that control intestinal inflammation. J Exp Med 192: 295–302, 2000. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Riedel JH, Becker M, Kopp K, Düster M, Brix SR, Meyer-Schwesinger C, Kluth LA, Gnirck AC, Attar M, Krohn S, Fehse B, Stahl RAK, Panzer U, Turner JE. IL-33-mediated expansion of type 2 innate lymphoid cells protects from progressive glomerulosclerosis. J Am Soc Nephrol 28: 2068–2080, 2017. doi: 10.1681/ASN.2016080877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Ronchetti S, Zollo O, Bruscoli S, Agostini M, Bianchini R, Nocentini G, Ayroldi E, Riccardi C. GITR, a member of the TNF receptor superfamily, is costimulatory to mouse T lymphocyte subpopulations. Eur J Immunol 34: 613–622, 2004. doi: 10.1002/eji.200324804. [DOI] [PubMed] [Google Scholar]
  • 190.Rosenzwajg M, Languille E, Debiec H, Hygino J, Dahan K, Simon T, Klatzmann D, Ronco P. B- and T-cell subpopulations in patients with severe idiopathic membranous nephropathy may predict an early response to rituximab. Kidney Int 92: 227–237, 2017. doi: 10.1016/j.kint.2017.01.012. [DOI] [PubMed] [Google Scholar]
  • 191.Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity 42: 607–612, 2015. doi: 10.1016/j.immuni.2015.04.005. [DOI] [PubMed] [Google Scholar]
  • 192.Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, Treuting P, Siewe L, Roers A, Henderson WR Jr, Muller W, Rudensky AY. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28: 546–558, 2008. doi: 10.1016/j.immuni.2008.02.017. [DOI] [PubMed] [Google Scholar]
  • 193.Rudra D, Egawa T, Chong MM, Treuting P, Littman DR, Rudensky AY. Runx-CBFβ complexes control expression of the transcription factor Foxp3 in regulatory T cells. Nat Immunol 10: 1170–1177, 2009. doi: 10.1038/ni.1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, Sene D, Cacoub P, Klatzmann D. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365: 2067–2077, 2011. doi: 10.1056/NEJMoa1105143. [DOI] [PubMed] [Google Scholar]
  • 195.Sakaguchi S, Ono M, Setoguchi R, Yagi H, Hori S, Fehervari Z, Shimizu J, Takahashi T, Nomura T. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol Rev 212: 8–27, 2006. doi: 10.1111/j.0105-2896.2006.00427.x. [DOI] [PubMed] [Google Scholar]
  • 196.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor α-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155: 1151–1164, 1995. [PubMed] [Google Scholar]
  • 197.Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity 12: 431–440, 2000. doi: 10.1016/S1074-7613(00)80195-8. [DOI] [PubMed] [Google Scholar]
  • 198.Sauer S, Bruno L, Hertweck A, Finlay D, Leleu M, Spivakov M, Knight ZA, Cobb BS, Cantrell D, O’Connor E, Shokat KM, Fisher AG, Merkenschlager M. T cell receptor signaling controls Foxp3 expression via PI3K, Akt, and mTOR. Proc Natl Acad Sci USA 105: 7797–7802, 2008. doi: 10.1073/pnas.0800928105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Scalapino KJ, Daikh DI. CTLA-4: a key regulatory point in the control of autoimmune disease. Immunol Rev 223: 143–155, 2008. doi: 10.1111/j.1600-065X.2008.00639.x. [DOI] [PubMed] [Google Scholar]
  • 200.Scalapino KJ, Tang Q, Bluestone JA, Bonyhadi ML, Daikh DI. Suppression of disease in New Zealand Black/New Zealand White lupus-prone mice by adoptive transfer of ex vivo expanded regulatory T cells. J Immunol 177: 1451–1459, 2006. doi: 10.4049/jimmunol.177.3.1451. [DOI] [PubMed] [Google Scholar]
  • 201.Schiering C, Krausgruber T, Chomka A, Fröhlich A, Adelmann K, Wohlfert EA, Pott J, Griseri T, Bollrath J, Hegazy AN, Harrison OJ, Owens BMJ, Löhning M, Belkaid Y, Fallon PG, Powrie F. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 513: 564–568, 2014. doi: 10.1038/nature13577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Schlenner SM, Weigmann B, Ruan Q, Chen Y, von Boehmer H. Smad3 binding to the foxp3 enhancer is dispensable for the development of regulatory T cells with the exception of the gut. J Exp Med 209: 1529–1535, 2012. doi: 10.1084/jem.20112646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Schmidt AM, Lu W, Sindhava VJ, Huang Y, Burkhardt JK, Yang E, Riese MJ, Maltzman JS, Jordan MS, Kambayashi T. Regulatory T cells require TCR signaling for their suppressive function. J Immunol 194: 4362–4370, 2015. doi: 10.4049/jimmunol.1402384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Sebastian M, Lopez-Ocasio M, Metidji A, Rieder SA, Shevach EM, Thornton AM. Helios controls a limited subset of regulatory T cell functions. J Immunol 196: 144–155, 2016. doi: 10.4049/jimmunol.1501704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, Solomon M, Selby W, Alexander SI, Nanan R, Kelleher A, Fazekas de St Groth B. Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med 203: 1693–1700, 2006. doi: 10.1084/jem.20060468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Sgouroudis E, Albanese A, Piccirillo CA. Impact of protective IL-2 allelic variants on CD4+ Foxp3+ regulatory T cell function in situ and resistance to autoimmune diabetes in NOD mice. J Immunol 181: 6283–6292, 2008. doi: 10.4049/jimmunol.181.9.6283. [DOI] [PubMed] [Google Scholar]
  • 207.Sharma MD, Baban B, Chandler P, Hou DY, Singh N, Yagita H, Azuma M, Blazar BR, Mellor AL, Munn DH. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J Clin Invest 117: 2570–2582, 2007. doi: 10.1172/JCI31911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Sharma R, Bagavant H, Jarjour WN, Sung SS, Ju ST. The role of Fas in the immune system biology of IL-2Rα knockout mice: interplay among regulatory T cells, inflammation, hemopoiesis, and apoptosis. J Immunol 175: 1965–1973, 2005. doi: 10.4049/jimmunol.175.3.1965. [DOI] [PubMed] [Google Scholar]
  • 209.Sharma R, Fu SM, Ju ST. IL-2: a two-faced master regulator of autoimmunity. J Autoimmun 36: 91–97, 2011. doi: 10.1016/j.jaut.2011.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Sharma R, Jarjour WN, Zheng L, Gaskin F, Fu SM, Ju ST. Large functional repertoire of regulatory T-cell suppressible autoimmune T cells in scurfy mice. J Autoimmun 29: 10–19, 2007. doi: 10.1016/j.jaut.2007.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Sharma R, Ju AC, Kung JT, Fu SM, Ju ST. Rapid and selective expansion of nonclonotypic T cells in regulatory T cell-deficient, foreign antigen-specific TCR-transgenic scurfy mice: antigen-dependent expansion and TCR analysis. J Immunol 181: 6934–6941, 2008. doi: 10.4049/jimmunol.181.10.6934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Sharma R, Sharma PR, Kim YC, Leitinger N, Lee JK, Fu SM, Ju ST. IL-2-controlled expression of multiple T cell trafficking genes and Th2 cytokines in the regulatory T cell-deficient scurfy mice: implication to multiorgan inflammation and control of skin and lung inflammation. J Immunol 186: 1268–1278, 2011. doi: 10.4049/jimmunol.1002677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Sharma R, Sung SS, Abaya CE, Ju AC, Fu SM, Ju ST. IL-2 regulates CD103 expression on CD4+ T cells in Scurfy mice that display both CD103-dependent and independent inflammation. J Immunol 183: 1065–1073, 2009. doi: 10.4049/jimmunol.0804354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Sharma R, Sung SS, Gaskin F, Fu SM, Ju ST. A novel function of IL-2: chemokine/chemoattractant/retention receptor genes induction in Th subsets for skin and lung inflammation. J Autoimmun 38: 322–331, 2012. doi: 10.1016/j.jaut.2012.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Sharma R, Sung SS, Ju CY, Deshmukh US, Fu SM, Ju ST. Regulatory T-cell (Treg) hybridoma as a novel tool to study Foxp3 regulation and Treg fate. J Autoimmun 37: 113–121, 2011. doi: 10.1016/j.jaut.2011.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Sharma R, Zheng L, Deshmukh US, Jarjour WN, Sung SS, Fu SM, Ju ST. A regulatory T cell-dependent novel function of CD25 (IL-2Rα) controlling memory CD8+ T cell homeostasis. J Immunol 178: 1251–1255, 2007. doi: 10.4049/jimmunol.178.3.1251. [DOI] [PubMed] [Google Scholar]
  • 217.Shevach EM. Biological functions of regulatory T cells. Adv Immunol 112: 137–176, 2011. doi: 10.1016/B978-0-12-387827-4.00004-8. [DOI] [PubMed] [Google Scholar]
  • 218.Shevach EM. From vanilla to 28 flavors: multiple varieties of T regulatory cells. Immunity 25: 195–201, 2006. doi: 10.1016/j.immuni.2006.08.003. [DOI] [PubMed] [Google Scholar]
  • 219.Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S. Stimulation of CD25+CD4+ regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol 3: 135–142, 2002. doi: 10.1038/ni759. [DOI] [PubMed] [Google Scholar]
  • 220.Shin JI, Park SJ, Saleem MA. The beneficial effect of abatacept in lupus nephritis may include stabilization of β1 integrin activation in podocytes and Treg cell repopulation: comment on the article by Furie et al. Arthritis Rheumatol 66: 2913–2914, 2014. doi: 10.1002/art.38740. [DOI] [PubMed] [Google Scholar]
  • 221.Singh K, Hjort M, Thorvaldson L, Sandler S. Concomitant analysis of Helios and Neuropilin-1 as a marker to detect thymic derived regulatory T cells in naïve mice. Sci Rep 5: 7767, 2015. doi: 10.1038/srep07767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Śledzińska A, Hemmers S, Mair F, Gorka O, Ruland J, Fairbairn L, Nissler A, Müller W, Waisman A, Becher B, Buch T. TGF-β signalling is required for CD4+ T cell homeostasis but dispensable for regulatory T cell function. PLoS Biol 11: e1001674, 2013. doi: 10.1371/journal.pbio.1001674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Smyk-Pearson SK, Bakke AC, Held PK, Wildin RS. Rescue of the autoimmune scurfy mouse by partial bone marrow transplantation or by injection with T-enriched splenocytes. Clin Exp Immunol 133: 193–199, 2003. doi: 10.1046/j.1365-2249.2003.02217.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Stephens GL, McHugh RS, Whitters MJ, Young DA, Luxenberg D, Carreno BM, Collins M, Shevach EM. Engagement of glucocorticoid-induced TNFR family-related receptor on effector T cells by its ligand mediates resistance to suppression by CD4+CD25+ T cells. J Immunol 173: 5008–5020, 2004. doi: 10.4049/jimmunol.173.8.5008. [DOI] [PubMed] [Google Scholar]
  • 225.Strauss L, Bergmann C, Whiteside TL. Human circulating CD4+CD25highFoxp3+ regulatory T cells kill autologous CD8+ but not CD4+ responder cells by Fas-mediated apoptosis. J Immunol 182: 1469–1480, 2009. doi: 10.4049/jimmunol.182.3.1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Stremska ME, Jose S, Sabapathy V, Huang L, Bajwa A, Kinsey GR, Sharma PR, Mohammad S, Rosin DL, Okusa MD, Sharma R. IL233, a novel IL-2 and IL-33 hybrid cytokine, ameliorates renal injury. J Am Soc Nephrol 28: 2681–2693, 2017. doi: 10.1681/ASN.2016121272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med 204: 1775–1785, 2007. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Szurek E, Cebula A, Wojciech L, Pietrzak M, Rempala G, Kisielow P, Ignatowicz L. differences in expression level of Helios and Neuropilin-1 do not distinguish thymus-derived from extrathymically-induced CD4+Foxp3+ regulatory T cells. PLoS One 10: e0141161, 2015. doi: 10.1371/journal.pone.0141161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.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 6: 152–162, 2005. doi: 10.1038/ni1160. [DOI] [PubMed] [Google Scholar]
  • 230.Takabatake Y, Li XK, Mizui M, Miyasato K, Matsui I, Kawada N, Imai E, Hünig T, Takahara S, Wada T, Furuichi K, Rakugi H, Isaka Y. A superagonistic monoclonal antibody for CD28 ameliorates crescentic glomerulonephritis in Wistar-Kyoto rats. Mol Med 17: 686–696, 2011. doi: 10.2119/molmed.2010.00229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N, Mak TW, Sakaguchi S. Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 192: 303–310, 2000. doi: 10.1084/jem.192.2.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Tang Q, Henriksen KJ, Boden EK, Tooley AJ, Ye J, Subudhi SK, Zheng XX, Strom TB, Bluestone JA. Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells. J Immunol 171: 3348–3352, 2003. doi: 10.4049/jimmunol.171.7.3348. [DOI] [PubMed] [Google Scholar]
  • 233.Tarbell KV, Yamazaki S, Steinman RM. The interactions of dendritic cells with antigen-specific, regulatory T cells that suppress autoimmunity. Semin Immunol 18: 93–102, 2006. doi: 10.1016/j.smim.2006.01.009. [DOI] [PubMed] [Google Scholar]
  • 234.Thomson CW, Lee BP, Zhang L. Double-negative regulatory T cells: non-conventional regulators. Immunol Res 35: 163–178, 2006. doi: 10.1385/IR:35:1:163. [DOI] [PubMed] [Google Scholar]
  • 235.Thornton AM, Donovan EE, Piccirillo CA, Shevach EM. Cutting edge: IL-2 is critically required for the in vitro activation of CD4+CD25+ T cell suppressor function. J Immunol 172: 6519–6523, 2004. doi: 10.4049/jimmunol.172.11.6519. [DOI] [PubMed] [Google Scholar]
  • 236.Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, Shevach EM. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol 184: 3433–3441, 2010. doi: 10.4049/jimmunol.0904028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Thornton AM, Shevach EM. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J Exp Med 188: 287–296, 1998. doi: 10.1084/jem.188.2.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.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 9: 194–202, 2008. doi: 10.1038/ni1549. [DOI] [PubMed] [Google Scholar]
  • 239.Tran DQ, Andersson J, Wang R, Ramsey H, Unutmaz D, Shevach EM. GARP (LRRC32) is essential for the surface expression of latent TGF-β on platelets and activated FOXP3+ regulatory T cells. Proc Natl Acad Sci USA 106: 13445–13450, 2009. doi: 10.1073/pnas.0901944106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Tsokos GC. Systemic lupus erythematosus. N Engl J Med 365: 2110–2121, 2011. doi: 10.1056/NEJMra1100359. [DOI] [PubMed] [Google Scholar]
  • 241.Tsokos GC, Thai TH. Interleukin-2 in systemic autoimmunity hits the micro way. Arthritis Rheum 64: 3494–3497, 2012. doi: 10.1002/art.34597. [DOI] [PubMed] [Google Scholar]
  • 242.Turka LA, Walsh PT. IL-2 signaling and CD4+ CD25+ Foxp3+ regulatory T cells. Front Biosci 13: 1440–1446, 2008. doi: 10.2741/2773. [DOI] [PubMed] [Google Scholar]
  • 243.Van Vleet TR, Schnellmann RG. Toxic nephropathy: environmental chemicals. Semin Nephrol 23: 500–508, 2003. doi: 10.1016/S0270-9295(03)00094-9. [DOI] [PubMed] [Google Scholar]
  • 244.Vigna-Perez M, Hernández-Castro B, Paredes-Saharopulos O, Portales-Pérez D, Baranda L, Abud-Mendoza C, González-Amaro R. Clinical and immunological effects of Rituximab in patients with lupus nephritis refractory to conventional therapy: a pilot study. Arthritis Res Ther 8: R83, 2006. doi: 10.1186/ar1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Viret C, Janeway CA Jr. MHC and T cell development. Rev Immunogenet 1: 91–104, 1999. [PubMed] [Google Scholar]
  • 246.von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, Enghard P, Sawitzki B, Hiepe F, Radbruch A, Burmester GR, Riemekasten G, Humrich JY. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis 75: 1407–1415, 2016. doi: 10.1136/annrheumdis-2015-207776. [DOI] [PubMed] [Google Scholar]
  • 247.Vuddamalay Y, van Meerwijk JP. CD28 and CD28lowCD8+ regulatory T cells: of mice and men. Front Immunol 8: 31, 2017. doi: 10.3389/fimmu.2017.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Walker LS, Chodos A, Eggena M, Dooms H, Abbas AK. Antigen-dependent proliferation of CD4+ CD25+ regulatory T cells in vivo. J Exp Med 198: 249–258, 2003. doi: 10.1084/jem.20030315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Wang YM, Alexander SI. CD8 regulatory T cells: what’s old is now new. Immunol Cell Biol 87: 192–193, 2009. doi: 10.1038/icb.2009.8. [DOI] [PubMed] [Google Scholar]
  • 250.Wang YM, Bakhtiar M, Alexander SI. ILC2: there’s a new cell in town. J Am Soc Nephrol 28: 1953–1955, 2017. doi: 10.1681/ASN.2017040398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Wang YM, McRae JL, Robson SC, Cowan PJ, Zhang GY, Hu M, Polhill T, Wang Y, Zheng G, Wang Y, Lee VW, Unwin RJ, Harris DC, Dwyer KM, Alexander SI. Regulatory T cells participate in CD39-mediated protection from renal injury. Eur J Immunol 42: 2441–2451, 2012. doi: 10.1002/eji.201242434. [DOI] [PubMed] [Google Scholar]
  • 252.Wang YM, Zhang GY, Wang Y, Hu M, Wu H, Watson D, Hori S, Alexander IE, Harris DC, Alexander SI. Foxp3-transduced polyclonal regulatory T cells protect against chronic renal injury from adriamycin. J Am Soc Nephrol 17: 697–706, 2006. doi: 10.1681/ASN.2005090978. [DOI] [PubMed] [Google Scholar]
  • 253.Ward MM. Changes in the incidence of end-stage renal disease due to lupus nephritis, 1982–1995. Arch Intern Med 160: 3136–3140, 2000. doi: 10.1001/archinte.160.20.3136. [DOI] [PubMed] [Google Scholar]
  • 254.Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, Levy-Lahad E, Mazzella M, Goulet O, Perroni L, Bricarelli FD, Byrne G, McEuen M, Proll S, Appleby M, Brunkow ME. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet 27: 18–20, 2001. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 255.Williams LM, Rudensky AY. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol 8: 277–284, 2007. doi: 10.1038/ni1437. [DOI] [PubMed] [Google Scholar]
  • 256.Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, Nomura T, Sakaguchi S. CTLA-4 control over Foxp3+ regulatory T cell function. Science 322: 271–275, 2008. doi: 10.1126/science.1160062. [DOI] [PubMed] [Google Scholar]
  • 257.Wohlfert EA, Grainger JR, Bouladoux N, Konkel JE, Oldenhove G, Ribeiro CH, Hall JA, Yagi R, Naik S, Bhairavabhotla R, Paul WE, Bosselut R, Wei G, Zhao K, Oukka M, Zhu J, Belkaid Y. GATA3 controls Foxp3+ regulatory T cell fate during inflammation in mice. J Clin Invest 121: 4503–4515, 2011. doi: 10.1172/JCI57456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Wolf D, Hochegger K, Wolf AM, Rumpold HF, Gastl G, Tilg H, Mayer G, Gunsilius E, Rosenkranz AR. CD4+CD25+ regulatory T cells inhibit experimental anti-glomerular basement membrane glomerulonephritis in mice. J Am Soc Nephrol 16: 1360–1370, 2005. doi: 10.1681/ASN.2004100837. [DOI] [PubMed] [Google Scholar]
  • 259.Wolf M, Schimpl A, Hünig T. Control of T cell hyperactivation in IL-2-deficient mice by CD4+CD25 and CD4+CD25+ T cells: evidence for two distinct regulatory mechanisms. Eur J Immunol 31: 1637–1645, 2001. doi:. [DOI] [PubMed] [Google Scholar]
  • 260.Wuest TY, Willette-Brown J, Durum SK, Hurwitz AA. The influence of IL-2 family cytokines on activation and function of naturally occurring regulatory T cells. J Leukoc Biol 84: 973–980, 2008. doi: 10.1189/jlb.1107778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Xie YF, Shu R, Jiang SY, Song ZC, Guo QM, Dong JC, Lin ZK. miRNA-146 negatively regulates the production of pro-inflammatory cytokines via NF-κB signalling in human gingival fibroblasts. J Inflamm (Lond) 11: 38, 2014. doi: 10.1186/s12950-014-0038-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Xing Q, Su H, Cui J, Wang B. Role of Treg cells and TGF-β1 in patients with systemic lupus erythematosus: a possible relation with lupus nephritis. Immunol Invest 41: 15–27, 2012. doi: 10.3109/08820139.2011.578189. [DOI] [PubMed] [Google Scholar]
  • 263.Xing Q, Wang B, Su H, Cui J, Li J. Elevated Th17 cells are accompanied by FoxP3+ Treg cells decrease in patients with lupus nephritis. Rheumatol Int 32: 949–958, 2012. doi: 10.1007/s00296-010-1771-0. [DOI] [PubMed] [Google Scholar]
  • 264.Xu J, Su HL, Wang JH, Zhang CH. [Role of CD4+CD25+Foxp3+ regulatory T cells in type 2 diabetic nephropathy]. Nan Fang Yi Ke Da Xue Xue Bao 29: 137–139, 2009. [PubMed] [Google Scholar]
  • 265.Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-Bucktrout S, Anthony BA, Sverdrup FM, Head R, Kuster DJ, Ruminski P, Weiss D, Von Schack D, Bluestone JA. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med 209: 1713–1722, 2012. doi: 10.1084/jem.20120822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Yamanouchi J, Rainbow D, Serra P, Howlett S, Hunter K, Garner VE, Gonzalez-Munoz A, Clark J, Veijola R, Cubbon R, Chen SL, Rosa R, Cumiskey AM, Serreze DV, Gregory S, Rogers J, Lyons PA, Healy B, Smink LJ, Todd JA, Peterson LB, Wicker LS, Santamaria P. Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity. Nat Genet 39: 329–337, 2007. doi: 10.1038/ng1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Yan JJ, Lee JG, Jang JY, Koo TY, Ahn C, Yang J. IL-2/anti-IL-2 complexes ameliorate lupus nephritis by expansion of CD4+CD25+Foxp3+ regulatory T cells. Kidney Int 91: 603–615, 2017. doi: 10.1016/j.kint.2016.09.022. [DOI] [PubMed] [Google Scholar]
  • 268.Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, Watford WT, Laurence A, Robinson GW, Shevach EM, Moriggl R, Hennighausen L, Wu C, O’Shea JJ. Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109: 4368–4375, 2007. doi: 10.1182/blood-2006-11-055756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Yu X, Harden K, Gonzalez LC, Francesco M, Chiang E, Irving B, Tom I, Ivelja S, Refino CJ, Clark H, Eaton D, Grogan JL. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 10: 48–57, 2009. doi: 10.1038/ni.1674. [DOI] [PubMed] [Google Scholar]
  • 270.Zeng H, Yang K, Cloer C, Neale G, Vogel P, Chi H. mTORC1 couples immune signals and metabolic programming to establish Treg-cell function. Nature 499: 485–490, 2013. doi: 10.1038/nature12297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Zhang BY, Fang Y, Jiao XY, Wu S, Cai JR, Zou JZ, Ding XQ. Delayed ischaemic preconditioning in the presence of galectin-9 protects against renal ischaemic injury through a regulatory T-cell dependent mechanism. Nephrology (Carlton) 21: 828–834, 2016. doi: 10.1111/nep.12680. [DOI] [PubMed] [Google Scholar]
  • 272.Zhang Y, Maksimovic J, Naselli G, Qian J, Chopin M, Blewitt ME, Oshlack A, Harrison LC. Genome-wide DNA methylation analysis identifies hypomethylated genes regulated by FOXP3 in human regulatory T cells. Blood 122: 2823–2836, 2013. doi: 10.1182/blood-2013-02-481788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Zhao DM, Thornton AM, DiPaolo RJ, Shevach EM. Activated CD4+CD25+ T cells selectively kill B lymphocytes. Blood 107: 3925–3932, 2006. doi: 10.1182/blood-2005-11-4502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Zheng L, Sharma R, Gaskin F, Fu SM, Ju ST. A novel role of IL-2 in organ-specific autoimmune inflammation beyond regulatory T cell checkpoint: both IL-2 knockout and Fas mutation prolong lifespan of Scurfy mice but by different mechanisms. J Immunol 179: 8035–8041, 2007. doi: 10.4049/jimmunol.179.12.8035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Zheng L, Sharma R, Kung JT, Deshmukh US, Jarjour WN, Fu SM, Ju ST. Pervasive and stochastic changes in the TCR repertoire of regulatory T-cell-deficient mice. Int Immunol 20: 517–523, 2008. doi: 10.1093/intimm/dxn017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Zheng Y, Chaudhry A, Kas A, deRoos P, Kim JM, Chu TT, Corcoran L, Treuting P, Klein U, Rudensky AY. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control TH2 responses. Nature 458: 351–356, 2009. doi: 10.1038/nature07674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, Rudensky AY. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 463: 808–812, 2010. doi: 10.1038/nature08750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Zhou J, Bashey A, Zhong R, Corringham S, Messer K, Pu M, Ma W, Chut T, Soiffer R, Mitrovich RC, Lowy I, Ball ED. CTLA-4 blockade following relapse of malignancy after allogeneic stem cell transplantation is associated with T cell activation but not with increased levels of T regulatory cells. Biol Blood Marrow Transplant 17: 682–692, 2011. doi: 10.1016/j.bbmt.2010.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Zhou L, Lopes JE, Chong MM, Ivanov II, Min R, Victora GD, Shen Y, Du J, Rubtsov YP, Rudensky AY, Ziegler SF, Littman DR. TGF-β-induced Foxp3 inhibits TH17 cell differentiation by antagonizing RORgammat function. Nature 453: 236–240, 2008. doi: 10.1038/nature06878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus MT, Bluestone JA. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp Med 205: 1983–1991, 2008. doi: 10.1084/jem.20080707. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES