Abstract
Interleukin-2 (IL-2) is a critical cytokine for T cell peripheral tolerance and immunity. Here we review how IL-2 interaction with the high affinity IL-2 receptor (IL-2R) supports the development and homeostasis of regulatory T cells and contributes to the differentiation of helper, cytotoxic and memory T cells. A critical element for each T cell population is the expression of CD25 (Il2rα), which heightens the receptor affinity for IL-2. Signaling through the high affinity IL-2R also reinvigorates CD8+ exhausted T (Tex) cells in response to checkpoint blockade. We consider the molecular underpinnings reflecting how IL-2R signaling impacts these various T cell subsets and the implications for enhancing IL-2-dependent immunotherapy of autoimmunity, other inflammatory disorders, and cancer.
In Brief
Our knowledge concerning the contributions of IL-2 in T cell biology, particularly its role in the development and functional activity of regulatory, effector, and memory T cells and re-invigorating exhausted T cells continue to advance. These cells depend on distinctive IL-2R signaling thresholds, providing a pharmacological window for the immunotherapy of autoimmunity and cancer. Here we report on the current knowledge of the functions of IL-2 and its receptor in T cells and how this understanding is transforming therapeutic strategies.
Introduction
Since its discovery as a growth factor for antigen-activated T cells, IL-2 and its receptor (IL-2R) have been under intensive investigation for over 45 years. This cytokine–receptor pair critically regulates tolerance and immunity1,2, largely by affecting regulatory (Treg), effector (Teff) and memory T ™ cells. Activated T cells, particularly CD4+ cells, are the main source of IL-2. Under most physiological conditions, IL-2 exerts its biological effect through the high affinity IL-2R (Kd 10−11M), which consists of three subunits, IL-2Rα (CD25), IL-2Rβ (CD122) and the common gamma chain (γc; CD132)1 (Fig. 1A). IL-2 binding to CD25 (Kd 10−8 M), leads to a small conformational change in IL-2, and the CD25/IL-2 complex is presented in cis for binding to CD122 and CD1323. The high affinity IL-2R is constitutively expressed on Treg cells, is induced by antigen on Teff and TM cells and is also found on ILC2s4 and a small subset of regulatory NK cells5. Alternatively, when IL-2 concentrations are high enough, IL-2 binds to cells, e.g., most NK cells and memory-phenotypic CD8+ T cells, that express the intermediate affinity (Kd 10−9 M) IL-2R, consisting of only CD122 and CD132 (Fig. 1A). However, physiologic concentrations of IL-2 at homeostasis are generally not sufficient to provide adequate signaling through CD122/CD132 alone.
Figure 1. The IL-2 receptor and IL-2 signaling pathways.
A) The subunits of the intermediate and high affinity IL-2R and the main cell types that express them. B) The basic signaling pathways downstream of the IL-2R and their affect on Teff versus Treg cells. STAT5 is predominant in Tregs due to high PTEN, which abrogates PI3K-Akt-mTOR. Created with BioRender.com.
Proximal IL-2R signaling depends upon the cytoplasmic regions of CD122 and CD132, which engage three main pathways in Teff cells: STAT5, ERK-MAP kinase, and PI3K-Akt-mTOR6 (Fig. 1B). In Treg cells, STAT5 is the dominant pathway due to high levels of PTEN, which attenuates the PI3K-Akt-mTOR pathway7. The activation of these pathways requires tyrosine phosphorylation of CD122 and CD132 by JAK1 and JAK3, which recruit the Shc adaptor and STAT5A and STAT5B to the cytoplasmic tail of CD122. Subsequent phosphorylation of STAT5 leads to dimerization and tetramerization of STAT5 that allows localization to the nucleus, where they bind DNA to regulate gene transcription8,9. Although STAT5A and STAT5B have extensive structural homology and largely overlap in their DNA binding targets, they have some critical non-overlapping functional roles. STAT5B has a dominant role in Treg and Teff cell responses and the asymmetric expression of STAT5A and STAT5B seems to underpin phenotypic differences between each paralog10. Yet, IL-2R signaling is more complex than proximal JAK1/3-STAT5 activation followed by downstream gene activation. Evaluation of the phospho-proteome of IL-2-stimulated activated CD8+ T cells revealed that over 600 intracellular proteins are modified by tyrosine, serine, or threonine phosphorylation11. Of these modifications, 90% are due to JAK-dependent IL-2R signaling, with the remaining 10% dependent on SRC kinases.
Considerable interest remains in IL-2 as it has high therapeutic potential. Low-dose IL-2 offers a means to re-regulate the immune system in the context of autoimmunity, graft vs. host disease (GVHD), and other inflammatory disorders. Higher amounts of IL-2 still hold promise to boost cancer immunotherapy. Indeed, IL-2 remains an important target of bioengineering to enhance its selectivity toward the high affinity IL-2R on Treg cells vs. the intermediate affinity IL-2R on NK and CD8+ memory-phenotypic T cells. Reviews have covered in detail engineered IL-2s and recombinant IL-2 (rIL-2) in clinical trials of autoimmunity and cancer12,13. This review focuses on the mechanisms by which IL-2 impacts the development, homeostasis, and function of Treg, Teff and TM cells, including how IL-2R signaling strength controls the development of these cells. This review also discusses how our current understanding of IL-2 and its receptor impact efforts to harness IL-2 as a therapeutic agent.
IL-2 in Treg development
Mice that lack IL-2, CD25, or CD122 develop a lymphoproliferative disorder that results in lethal autoimmunity14–16, illustrating that IL-2R signaling is critical for self-tolerance17. Initially, CD25 was recognized as a marker of activated Teff cells and later was found to also mark Treg cells18. Subsequent findings showed that the autoimmunity seen in IL-2/IL-2R-deficient mice was caused by reduced Treg cells and that thymic-directed reconstitution of IL-2R signaling corrected this Treg cell defect19. Importantly, the transfer of wild-type (WT) Treg cells into CD122-deficient mice fully corrected their lethal autoimmunity, which established that IL-2R signaling is essential for Treg cells20. The interpretation of these initial findings suggested that IL-2 is essential for thymic Treg development20. Others favored that IL-2 was essential for Treg cell survival in the periphery21–23. We now know that IL-2 plays an integral role in both Treg cell development and survival, as discussed below.
Treg cells develop in the thymus from CD4+CD8− “single positive” (CD4SP) thymocytes due to TCR, CD28 co-stimulatory and IL-2R signaling24,25 (Fig. 2). Positively selected thymocytes migrate into the medulla where high affinity recognition of self-antigens in the context of co-stimulatory molecules causes negative selection26. Cells exhibiting a moderate affinity will survive negative selection and induce CD25 expression, representing potential precursors for Foxp3+ Treg cells27,28. IL-2 from self-reactive T cells acts through the high affinity IL-2R on developing Treg cells to prevent negative selection29,30 and then drives functional maturation of Foxp3+ Treg cells31,32 largely through STAT5 activation33–35. Correspondingly, thymic development is aborted in mice with Treg-specific conditional deficiency of CD2531,32. These developing Treg cells lack functionality, leading to rapid lethal autoimmunity akin to that seen in Scurfy and Foxp3-deficient mice. These studies indicate a strict requirement for IL-2R signaling for functional Foxp3+ Treg cells during thymic development.
Figure 2. IL-2 provides two types of essential signals for Treg development in the thymus.
The parallel Treg development model (A) consists of two CD25−Foxp3lo and CD25+Foxp3− Treg progenitors (TregP) that each derive mature Treg cells. The two TregP arise from a common pool of CD25−Foxp3− CD4SP progenitor cells that also gives rise to self-reactive CD4+IL-2+ T cells. The serial Treg development model (B) proposes that TregP and self-reactive CD4+IL-2+ T cells arise from a common CD25+Foxp3− precursor population and that fate is determined by duration of agonist signals. Mature Treg cells are derived from a single CD25loFoxp3int precursor population. Notably, Foxp3int expression is achieved independent of IL-2 in the serial model. In both models, IL-2 secreted by self-reactive CD4+ T cells provides two signals to TregP for Treg differentiation; Step #1 upregulates CD25 and Foxp3 and Step #2 drives functional maturation of Treg cells. Created with BioRender.com.
Comparison of Treg development in mice with a germline defect in CD25 (CD25gKO), versus a conditional CD25 deficiency limited to Treg lineage cells, i.e., Foxp3Cre CD25fl/fl mice (CD25cKO), is informative concerning the role of IL-2 during thymic development31. First, even though CD25 was effectively deleted in both models, the overall defect in Treg development is more pronounced in CD25gKO mice, although Foxp3lo cells are readily detected and some of these cells show STAT5 activation. Thus, the development of Foxp3lo Treg cells is likely due to the TCR and IL-7R or IL-15R signaling rather than IL-2R signaling (Fig. 2). However, IL-7 and IL-15 are not sufficient to rescue Treg development in mice lacking IL-2, CD25, or CD12232,36–38. Second, developing Treg cells from CD25cKO mice have increased Foxp3 compared to CD25gKO mice, but lower than in WT mice. These findings reflect that IL-2 acts at two steps of thymic development (Fig. 2). At step 1, IL-2 helps to promote the survival and proliferation of developing Treg cells. Continual IL-2R signaling at step 2 leads to functional programming of Treg cells.
The precise stage(s) during Treg development where IL-2 is required and the molecular consequences of its signaling are unclear. Treg precursors (TregP) are modeled to consist of either CD25+ Foxp3− or CD25− Foxp3lo cells, both having passed negative selection39. These two cell populations represent two parallel pathways of Treg development40,41 (Fig. 2k). The Foxp3− CD25+ subset is more dependent on TCR and co-stimulatory signaling, which induce expression of CD25. Conversely, Foxp3lo CD25− TregP cells are purported to rely on multiple signals for their development, such as IL-1542, NF-κB and/or IL-440. Subsequent signaling through the high affinity IL-2R then generates fully functional Foxp3hi Treg cells from both progenitors. The finding that the TCR repertoire of CD25− Foxp3lo TregP cells has little overlap with CD25+ Foxp3− TregP40 cells provides support for this parallel pathway model.
Another study challenges this parallel model and instead suggests that a common CD25+ Foxp3− precursor cell develops into CD25lo Foxp3int TregP cells, then into functional CD25+ Foxp3hi Treg cells in a serial, stepwise manner43 (Fig. 2B). The generation of CD25lo Foxp3int TregP cells is dependent on interruption of TCR and co-stimulatory agonist signals during negative selection, then IL-2R signaling in CD25lo Foxp3int TregP cells subsequently generates CD25+ Foxp3hi Treg cells. This is referred to as the ‘primary development pathway’ and is responsible for the majority of Treg development. Notably, initial Foxp3 induction is not dependent on IL-2 in this model, providing an explanation as to why IL-2 or IL-2R germline knockouts still generate sub-functional Foxp3-expressing Treg cells21,22,24,31. The authors also argue for an ‘alternative Treg development pathway’ that allows for CD25+ Foxp3− precursors to induce Foxp3 expression, bypass the CD25lo Foxp3int stage, and become mature Treg cells under supraphysiologic concentrations of IL-2. This observation highlights the importance of IL-2 concentrations in driving Treg development from distinct precursors and may reconcile differences between this and the parallel pathway model.
The expression of Foxp3 is critical for enforcing the Treg suppressive program and requires STAT533. A conserved non-coding sequence (CNS0) downstream of the Foxp3 promoter includes a STAT5 response element to allow for Foxp3 induction by STAT5 in developing thymocytes44,45. CNS0 acts in concert with intronic CNS3 to maintain Foxp3 expression45, whereas CNS2 maintains the heritability of Foxp3 and requires demethylation for Foxp3 stability46–48.
The IL-2-dependent transcriptome of thymic Treg cells includes ~2000 genes, indicating that IL-2R signaling broadly affects developing Treg cells31. Foxp3 and other well-known IL-2-dependent genes49, e.g., Socs1, Socs2, Cish, and Myc, require IL-2R signaling in thymic Treg cells. In addition, most transcripts related to Treg function are reduced after conditional knockout of CD25, consistent with a role for IL-2 in functional maturation31. Since transcripts related to TCR are also impacted after CD25 ablation, IL-2 may integrate signaling induced by TCR and co-stimulation during Treg development to generate functional Treg cells31.
Along with mainstream thymic development, peripheral naïve CD4+ T cells can be activated to become Foxp3+ Treg cells. Treg cells induced in vitro are referred to as iTreg cells. When this occurs in peripheral tissues, they are designated as pTreg cells50. IL-2 and TGF-β support the development of iTreg cells, which are immunosuppressive in vitro and in vivo51–53. However, iTregs display methylation of CNS2, which jeopardizes the stability of Foxp346,47,54. Notably, expression of Foxp3 and suppressive function after adoptive transfer of iTreg cells in vivo depends on endogenous IL-2 secreted by CD4+ Teff cells54.
Many mediators, including IL-2, TGF-β, retinoic acid, short chain fatty acids produced by commensal organisms, and food antigens support the development of pTreg cells55. TCR repertoire analysis indicates that the large majority of Treg cells are of thymic origin and show little overlap with naive CD4+ T cells56. In contrast, the TCR repertoire of pTreg cells by necessity overlaps with conventional CD4+ T cells and serves a nonredundant role in expanding the specificities of Treg cells57. pTreg cells are thought to support tolerance toward food antigens, commensal organisms and a developing fetus58,59. ILC3-derived IL-2 supports Foxp3 induction and pTreg formation in the intestinal tract, which is dependent on IL-1β from intestinal macrophages sensing commensal organisms60. Thus, pTreg cells adapt to environmental cues to maintain self-tolerance.
IL-2 in the peripheral homeostasis of Treg cells
The essential role for IL-2 during thymic Treg development complicates the extent to which peripheral mature Treg cells depend on IL-2. This issue has been resolved. Mice with conditional ablation of CD25 induced selectively in Treg cells31,61 gradually lose Treg cells over 3-4 months. Thus, without high affinity IL-2R signaling, all Treg cells fail to persist. The lack of persistence is not due to impaired proliferation but enhanced apoptosis, mitochondrial dysfunction, and impaired glycolytic metabolism. Gene expression profiling also points to a role for cholesterol biosynthesis that may support oxidative phosphorylation for IL-2-driven homeostasis of Treg cells31,62. As proliferation of peripheral Treg cells in the absence of IL-2R signaling is not obviously impaired, other inputs, such as TCR and co-stimulatory signaling or cytokines like IL-7 may help maintain such Treg cells for some time37,61.
In addition to controlling survival, IL-2 programs Treg cell functions63. Expression of constitutively active STAT5 in Treg cells enhances their suppressive capacity32. Moreover, suppression of experimental autoimmune encephalomyelitis by CD25-deficient Treg cells is less effective when compared to WT Treg cells61. Conversely, administration of an IL-2 agonist that delivers persistent IL-2R signaling alters Treg cell gene expression that enhances suppressive function, proliferation, oxidative phosphorylation, chromatin accessibility and recruitment of DNA-binding proteins62. This type of IL-2R signaling also supports expression of chemokine receptors that facilities trafficking of Treg cells into inflamed tissues64.
The Treg lineage is highly stable and self-renewing at homeostasis, as evidence by fate-mapped Treg cells that persist for over 8 months65. Targeted Foxp3 deletion in mature Treg cells causes them to become IFNγ- or IL-2-producing Teff cells, illustrating the central role of Foxp3 in maintaining Treg lineage66. Inflammation destabilizes Treg cells, whereby they lose suppressive function and/or Foxp3 expression and become Teff cells, or ‘ex-Treg cells’67. During inflammation, the relative amounts of inflammatory cytokines and IL-2 may determine Treg fate. When Treg cells express a hypomorphic variant of CD25, they are less effective in suppressing colitis, become unstable and lose lineage commitment68. In contrast, under conditions without obvious inflammation, peripheral Treg cells that lack IL-2R signaling for many weeks readily express near WT amounts (80-90%) of Foxp3 and exhibit minimal signs of conversion to ex-Treg cells in secondary lymphoid tissue and the gut mucosa31,61. The intestine is an important site of Treg instability, particularly with conversion into TH17 cells69. Thus, the loss of IL-2R signaling does not inherently promote Treg instability, but rather may depend on an accompanying inflammatory response.
Mature Treg cells in the periphery display extensive heterogeneity in their activation, localization, survival, proliferation, and suppressive function. These are shaped by numerous molecular factors, one of which is IL-270–73. CD62Lhi CCR7+ resting or central Treg (cTreg) cells reside in secondary lymphoid tissues, whereas CD62Llo ICOShi activated or effector Treg (eTreg) cells home to tissues71,72,74,75. When compared to cTreg cells, eTreg cells exhibit higher proliferation and suppressive function70. Although both Treg subsets depend on IL-2 for their maintenance, cTreg cells decline more rapidly than eTreg cells in the absence of peripheral IL-2R signaling31,32,61,74. This longer persistence of eTreg cells likely reflects signals from the TCR and CD28. Klrg1+ eTreg cells are terminally differentiated eTreg cells that develop after 8-9 cells divisions and are highly dependent on IL-276, perhaps due to the direct action of IL-2, synergy of IL-2 and TCR signaling, or IL-2-dependent programming in cTreg cells.
T follicular regulatory (TFR) cells have a crucial role in germinal center homeostasis by mitigating the generation of self-reactive B cells. IL-2/STAT5 engagement of Blimp-1 promotes IL-10-producing eTreg cells, and suppresses Bcl-6 to prevent TFR development77. Thus, low or absent IL-2 signaling is required for Bcl-6 induction and TFR differentiation77. Bach2 also acts to restrain STAT5 activation and promote TFR differentiation78. Whether IL-2 signaling limits development of RORγt+ Foxp3+ TR17 cells79, the counterpart to TH17 cells that develop in environments with low IL-280, remains to be determined.
Cellular sources of IL-2
Treg cells require paracrine IL-2 because Foxp3 represses IL-2 transcription81. Although antigen-activated CD4+ T cells are considered the main producers of IL-2, other cell types, including dendritic cells (DCs), B cells, CD8+ T cells and ILCs produce IL-2 at lower amounts82. Thymic Treg cells adapt to limited IL-2 by a somewhat higher sensitivity to IL-2 than peripheral Treg cells83. Treg cells are found in a niche within the thymic medulla where they are thought to receive IL-2 signals from DCs84. However, conditional deletion of IL-2 in T cells, but not DCs, impairs Treg development41,85, consistent with most IL-2 secretion coming from newly generated CD4+ self-reactive thymocytes85. IL-2 production by self-reactive T cells is induced by strong TCR signaling in a niche whereby developing Treg cells constrain the development of additional IL-2-secreting cells, which in turn also limits Treg development41.
Thymic DCs express CD25, but not CD122, and therefore do not respond to IL-2. Rather, CD25-bearing DCs may bind IL-2 produced by self-reactive T cells. Although IL-2 binding solely to CD25 has fast on and off kinetics and is unstable, this type of interaction of IL-2 with DCs may help to retain IL-2 in the DC-Treg niche and potentially trans-present IL-2 to Treg cells85,86.
Cell-targeted deletion of IL-2 also demonstrates that the main source of IL-2 contributing to Treg homeostasis in the periphery is largely T cells85 (Fig. 3), with a possible limited role for DCs in mucosal-associated draining lymph nodes, such as the mesenteric lymph node85. This T cell-derived IL-2 for Treg cells does not rely on activation by foreign antigen, but rather by self-antigens87. Imaging studies of lymph nodes show that IL-2-responsive Treg cells, as measured by activated STAT5, are found in discrete clusters with rare IL-2-secreting self-reactive T cells and DCs87. Consequently, Treg cells in the cluster express high levels of suppressive molecules and preferentially consume IL-2 to actively suppress the IL-2-secreting autoreactive T cells. Thus, IL-2 from autoreactive T cells contributes to Treg homeostasis, but this co-localization of Treg cells with autoreactive T cells provides a feedback circuit whereby Treg cells suppress and reduce the frequency of those self-reactive T cells (Fig. 3). This feedback circuit represents one important way in which T cell immune homeostasis and tolerance is maintained.
Figure 3. Peripheral homeostasis is maintained through Treg sequestration of IL-2.
Autoreactive PD-1+ CD4+ T cells secrete IL-2 upon binding self-antigen presented by dendritic cells. Treg cells bind and sequester this IL-2 to support their homeostasis and suppressive activity and limit IL-2R signaling in autoreactive CD4+ T cells. When IL-2 concentrations become high enough to sustain IL-2R signaling in autoreactive T cells, tolerance is broken. Created with BioRender.com.
IL-2 in the differentiation of CD4+ T helper cell subsets
Naïve CD4+ T cells are clonally-restricted precursors that differentiate into a wide array of specialized effectors upon encounter with their cognate antigen, co-stimulation and cytokine cues. When naïve T cells encounter their antigen in the context of co-stimulation, their initial activation is characterized by IL-2 secretion and expression of CD25 and CD122, making them responsive to autocrine and paracrine IL-2. Binding of IL-2 in the context of TCR signaling promotes T cell proliferation, but also contributes to programming the differentiation of helper and cytotoxic subsets88. These subsets can be broadly grouped into T follicular helper cells (TFH), T helper subsets (TH1, TH2 and TH17), and cytotoxic T lymphocytes (CTLs), although this is not comprehensive, and considerable heterogeneity and plasticity exists with each of these groups89.
TH1 cells are important for immunity against viral and intracellular bacterial pathogens. IL-2 drives TH1 survival and differentiation by increasing expression of IL12RB2 through activated STAT5, allowing IL-12 induction of T-bet via STAT490,91. IL-2 also remodels the metabolism of activated cells by engaging HIF-1α and c-Myc to drive glycolytic activity, which subsequently augments IFNγ secretion, the signature TH1 cytokine92–94. This is likely mediated through IL-2-dependent activation of mTORC, which contributes to increased T-bet expression through protein kinase C95,96.
TH2 polarization is required for protective immunity against parasites while excessive TH2 activity may predispose one to asthma and allergy. The TH2 response is engaged by IL-4, which activates STAT6 to induce GATA3, the TH2 lineage-determining transcription factor97,98. IL-2 contributes to TH2 cell differentiation by STAT5-mediated upregulation of IL4RA and by increasing the accessibility of the IL4 locus99,100.
If IL-2R signaling in recently activated CD4+ T cells remains high, the prolonged activation of STAT5 and Blimp-1 suppresses the expression of Bcl-6, which opposes differentiation into TFH cells101–105. In contrast, recently activated T cells that receive strong TCR and IL-6 signals decrease CD122 expression and become hyporesponsive to IL-2. This setting, coupled with sequestration of IL-2106, establishes an environment with low amounts of IL-2 signaling, which favors TFH differentiation107. Once Bcl-6 is expressed, it directly represses the HIF-1α - and c-Myc-driven glycolytic activity involved in TH1 function93.
TFH cells are critical in the development of germinal centers to shape antibody responses to infectious agents and vaccination108. Correspondingly, modulation of IL-2 affects this response. For example, the levels of IL-2 are relatively high in neonates from autoreactive CD4+ T cells prior to Treg cell egress from the thymus. When IL-2 is transiently blocked with anti-IL-2 during this time, the TFH response is augmented, leading to improved immunity to respiratory syncytial virus (RSV) and faster clearance of subsequent RSV rechallenge109. Furthermore, high TFH/Treg ratios correlate with autoimmunity. Exogenous low-dose IL-2 lowers this ratio, likely by limiting TFH differentiation110 but does not elevate other T helper responses.
TH17 cells normally function to protect the host from extracellular bacteria and fungi but upon hyperactivation, are implicated in multiple autoinflammatory processes. TH17 are characterized by their secretion of IL-17 and expression of the transcription factor RORγt111. Like TFH, the presence of IL-2 also restrains the differentiation of TH17 cells. Some mutations in IL2RA result in hyperresponsiveness to IL-2 and are associated with inflammatory bowel disease due to increased CD4+ TH1 polarization, higher concentrations of IFNγ and a loss of TH17 cells in the intestine112,113. Mechanistically, IL-2R signaling leads to accumulation of STAT5 binding at the IL17 locus, which outcompetes STAT3, inhibiting transcription of IL17114. PTEN inhibits IL-2-mediated suppression of IL17 by enforcing the phosphorylation of STAT3 over STAT5115. Additionally, Batf antagonizes STAT5 recruitment of Ets1-Runx1 to TH1 and Treg-specific gene loci, thereby favoring Th17 differentiation116. Similar to TFH, low-dose IL-2 decreases the ratio of TH17/Treg in patients with autoimmune diseases, which correlates with improved therapeutic responses110. Thus, along with its essential role of IL-2 for Tregs, IL-2 also critically shapes proliferation and differentiation of CD4+ Teff cells with specialized functional activity.
IL-2 in differentiation of CD8+ Teff, TM and exhausted (Tex) cells
In the context of primary infection or antigen exposure, naïve CD8+ T cells undergo expansion, contraction, and memory phases. The balance between short-lived Teff and memory precursors is regulated by duration of antigen exposure and inflammatory cytokines as well as the concentration and timing of IL-2 (Fig. 4B). CD8+ Teff cells with high amounts of CD25 during the primary response exhibit robust proliferation and develop high effector function yet are more prone to apoptosis117. In contrast, activated CD8+ T cells that express CD25 at relatively low amounts during the primary response generate moderate IL-2R signaling, resulting in CD62L+ CD127+ TM cell development117.
Figure 4. CD25 levels tune immune cell responses to endogenous and exogenous IL-2.
A) The ability of IL-2 to promote or inhibit the functional activity of the indicated cells depends on the concentration of IL-2 and the levels of CD25. B) High IL-2 causes differentiation of short-lived effector CD8+ T cells. Moderate IL-2 supports differentiation into memory precursors. Programming memory recall responses requires autocrine IL-2 signaling. Exogenous IL-2 directed to the high affinity IL-2R induces highly functional effector-like cells from the Tex lineage; exogenous IL-2 directed to the intermediate affinity IL-2R leads to proliferation of stem-like Tex cells, which can give rise to terminally differentiated Tex cells.Created with BioRender.com.
IL-2-dependent regulation of Eomes and Blimp-1 contribute to the TM vs. Teff fate decision during priming118–121. Moderate IL-2R signaling acts through Akt and STAT5 to support expression of Eomes, and coupled with Bcl-6 and Id3, promotes TM cells120,122–124. High IL-2R signaling induces expression of Blimp-1, and coupled with T-bet and Id2, supports development of highly functional short-lived CD8+ Teff cells117,124. Since Bcl-6 and Blimp-1 reciprocally repress each other, the TM fate is enforced by Bcl-6 repressing Blimp-1. At higher IL-2R signaling, greater levels of Blimp-1 are induced, leading to the repression of Bcl-6, to drive development of CTLs. Another consequence of IL-2-dependent STAT5 activation is increased expression of granzyme B and perforin-1, which are mediators of CTL activity120,125,126. Activated CD4+ T cells are not only cytokine-producing helper cells but under some circumstances have the potential to develop into CTLs127–129, seemly independent of the CD4+ T helper program. The same pathways active in activated CD8+ T cells, as discussed above, also function in CD4+ T cells to generate CTL activity127,130.
CD4+ Teff cells are canonically the main source of IL-2 for CD8+ Teff development. However, IL-2-dependent recall expansion by memory cells can form in the absence of CD4+ T cell “help”131,132. A subset of antigen-activated CD8+ T cells produce autocrine IL-2 early in the response after initial priming133–135 (Fig 4B). This autocrine IL-2-dependent STAT5 signaling supports differentiation into stem-like memory cells while protecting these cells from terminal Teff differentiation and eventual cell death135,136.
The CD8+ Tex lineage consist of dysfunctional T cells, where chronic antigen exposure results in high expression of co-inhibitory receptors (IRs), low effector function, and reduced proliferative capacity137,138. CD8+ Tex cells lose the ability to produce IL-2 followed by TNFα and IFNγ, whereas expression of granzyme B is more variable137–140. The CD8+ Tex lineage consists of two major developmental stages (Fig. 4B). TCF1hi progenitor-or stem-like Tex cells exhibit low effector activity and are the major population that expands to anti-PD-1/PD-L1 therapy141–145. These cells develop into TCF1lo terminally differentiated Tex cells, which are short-lived and express higher levels of PD-1, other IRs, and granzyme141–145.
The strength and duration of IL-2R signaling influence CD8+ Tex cell development. In contrast to CD25, relatively high levels of CD122 persist on CD8+ Tex cells117,146. Infrequent exposure to IL-2 and transient STAT5 activation promotes development into the Tex lineage through Blimp-1 and subsequent induction of IRs146. In mouse tumor models, intermittent IL-2 and transient STAT5 activation causes an accumulation of 5-hydroxytryptophan, leading to nuclear translocation of the aryl hydrocarbon receptor (AhR)147. Transcriptional regulation by AhR increases IR expression while decreasing cytokine production, both hallmarks of CD8+ Tex cells147. Membrane-spanning sushi domain-containing protein 2 (SUSD2) also acts to promote CD8+ Tex development through its action as a negative regulator of STAT5 signaling148. Correspondingly, after deletion of SUSD2, tumor antigen-specific CD8+ T cells and CAR T cells elicit more productive antitumor responses than their WT counterparts148.
The action of some engineered IL-2s also supports a role for IL-2 in Tex development. When compared to WT IL-2, the IL-2 partial agonist H9T induces lower STAT5 activation but does not affect IL-2R-dependent PI3K-Akt or ERK-MAPK signaling149. This attenuated IL-2 activity favors development of TCF1+ CD8+ stem-like cells that express lower levels of exhaustion markers and supports more effective antitumor responses (Fig. 4B). These types of effects were recapitulated in additional studies with other engineered IL-2s with selectivity toward CD122/CD132150,151. These IL-2 agonists promote low moderate IL-2R signaling and antitumor activity by Tex cells, but they do not lead to transcriptional reprogramming and full recovery of Teff activity.
Persistent IL-2R signaling in combination with anti-PD-1 more effectively reverses the Tex state by inducing highly functional CD8+ Teff cells with robust antitumor and antiviral capacity152–156 (Fig. 4B). These enhanced responses are due to epigenetic reprogramming of TCF1+ Tex stem cells leading to a unique transcriptional signature similar to canonical Teff cells. To achieve this effect, IL-2 is delivered to the TCF1+ Tex cells through their expression of CD25 or by linking a CD122/CD132-selective IL-2 mutein to anti-PD-1152–154,156. Although anti-PD-1 alone minimally affects the gene program of Tex cells, these IL-2-dependent effects on TCF1+ Tex cells synergize with anti-PD-1, leading to expansion of reinvigorated Tex cells. This effect, even in the PD-1-directed IL-2 variant, is a combination of both anti-PD-1- and IL-2-specific effects, but most cellular changes seem to be driven by IL-2. Similar to providing persistent IL-2R signaling, transduction of LCMV-specific CD8+ T cells with constitutively active STAT5A antagonizes the Tox-dependent Tex lineage, rescues polyfunctionality, and induces durable effector-NK-like cells from CD8+ Tex cells157. Thus, IL-2R signaling through STAT5 appears to reprogram TCF1+ Tex cells to reinvigorate their response to anti-PD-1 to substantially enhance their functional activity.
The molecular basis for distinct IL-2R signaling thresholds in the development of Treg, Teff and TM cells.
Cell surface amounts of CD25 are one important factor that impacts the sensitivities of T cells to IL-2158,159. The stimulation of Treg cells and naïve CD4+ and CD8+ T cells by TCR and IL-2R signaling results in very high levels of IL-2R subunits in vitro. However, the expression of these subunits is much more nuanced in vivo (Fig. 4A). At homeostasis, mouse and human Treg cells express the highest amount of CD25, likely through continual sensing of self-antigen and IL-2, and consequently, these cells express the highest amount of the high affinity IL-2R. In normal mice, very few other T cells express the CD25 whereas in healthy humans, some naïve and memory-phenotypic CD4+ and CD8+ T cells express low amounts of the high affinity IL-2R. After naïve CD4+ and CD8+ T cells encounter antigen in vivo, CD25 and CD122 are induced, resulting in the expression of the high affinity IL-2R160, but these levels are typically lower than detected after stimulation in vitro. Thus, under most circumstances, even during immune activation, Treg cells express the highest amounts of the high affinity IL-2R, although some Teff cells may transiently express similar levels.
High levels of the high affinity IL-2R allow Treg cells to preferentially respond to IL-2. When human PBMCs were examined, proximal IL-2R signaling, as measured by activation of STAT5, was induced with 10-20-fold lower levels of IL-2 in Treg cells than CD45RO+ memory-phenotypic CD4+ T cells83,161. Importantly, downstream gene activation, which requires integration of IL-2R signaling for a period of time, required 100-fold less IL-2 in Treg cells83,171. This high level of sensitivity to IL-2 forms the basis of using low-dose IL-2 to boost Treg cells in autoimmune diseases.
High CD25 expression does not fully account for the low IL-2R signaling threshold in Treg cells162. Serine/threonine protein phosphatase 2A (PP2A) activity is enhanced in Treg cells, which potentiates proximal IL-2R signaling159,163. PP2A directly enhances IL-2R signaling by preventing the loss of CD122 through limiting Adam-10-dependent cleavage of cell surface CD122. PP2A also enhances gene expression in Treg cells related to survival, activation, immunosuppressive function, and IL-2R signaling. Similarly, Mst1 and Mst2 are serine-threonine kinases that are essential to maintain Treg sensitivity to IL-2 by promoting STAT5 activation164.
Mutations of three key tyrosine residues to phenylalanine on the intracytoplasmic tail of CD122, which are critical to the interaction of the Shc adapter and STAT5 to CD122, lead to markedly diminished, but not abrogated, proximal IL-2R signaling in T lymphocytes83. Notably, thymic Treg cell development and peripheral homeostasis is outwardly normal whereas Teff cells are markedly impaired. These mutations affect the expression of 300-400 genes in Treg and Teff cells. However, key IL-2-dependent genes of Treg cells, e.g., Foxp3, Il2ra, and Socs2, are normally expressed. Thus, low proximal IL-2R signaling is sufficient to support some essential elements required for Treg but not Teff cells. Similar results are reported for mice harboring a point hypomorphic mutation in CD25 or treated with anti-IL-2, leading to reduced CD25 expression and proximal IL-2R signaling68,162. Mice with hypomorphic CD25 show relatively normal Treg development under homeostatic conditions, but their Treg cells do not effectively suppress colitis in the T cell transfer model. This reduced control of autoimmunity may be due to inflammatory conditions and/or autoreactive T cells with normal IL-2R signaling.
Another feature of Treg cells is their low expression of the IL-7R165, in part due to Foxp3 repression of IL7RA (CD127)166, which limits their responsiveness to IL-7. The IL-2R and IL-7R share CD132 as a receptor subunit. Surface plasmon resonance showed that CD127 binds to CD132 in the absence if IL-7167. Thus, IL-7 competition for and/or CD127 sequestering of CD132 may act to impede IL-2R signaling for naïve and memory T cells or memory Treg cells that express a high amount of CD127, while this mechanism is less relevant for CD127lo Treg cells166–168. Such competition may also fine-tune the response of Teff and TM cells, where high vs. moderate strength of IL-2R signaling support CD8+ Teff vs TM cell fate decision, respectively.
Single nucleotide polymorphisms (SNPs) in IL2, IL2RA, and IL2RB are linked to susceptibility to several autoimmune diseases. Most SNPs are found in enhancers, consistent with their effects on altering gene transcription169. The precise role of SNPs in autoimmunity is difficult to discern as their effects are subtle and likely promote autoimmunity in conjunction with other SNPs and environmental triggers. With respect to IL2RA, one SNP (rs12722495) associated with type 1 diabetes (T1D) lowers CD25 expression in CD4+ Teff and Treg cells, resulting in lower IL-2-dependent STAT5 activation and Foxp3 expression by Treg cells170. Low Treg suppressive activity due to this SNP likely contributes to T1D. Another SNP (rs61839660) associated with Crohn’s disease delays the expression of CD25, which in turn increases inflammatory TH17 cells113,171,172.
TCR and IL-2R signaling are major inducers of IL2RA transcription. Although TCR signaling is sufficient to induce CD25 expression, IL-2 further enhances CD25 amounts. TCR regulates IL2RA in part through activation of NFAT, NF-κB, and AP-1 whereas IL-2 regulates IL2RA through STAT5173. The IL2RA locus is one of the most STAT5-responsive genes, containing 17 binding regions in humans49. In Treg cells, IL2RA transcription is further enhanced and maintained by Foxp3 in coordination with Runx1 and NFAT174–176. Binding to the locus requires the coordination of STAT5 super-enhancer sequences found upstream and downstream of the promoter region. The extent that these enhancers bind STAT5 dictates the amount of IL2RA transcription and cell surface expression to fine-tune responses to environmental levels of IL-2 (Fig. 4A).
Immunotherapy of autoimmunity and cancer with IL-2
There remains much interest and activity to harness IL-2 to manipulate T cells in patients with autoimmunity and cancer. Two main principles driving this effort are: (1) selective targeting of the high affinity IL-2R on Treg cells with low-doses of IL-2; (2) higher doses targeting the intermediate affinity IL-2R on memory-phenotypic CD8+ T and NK cells can boost anti-cancer responses while minimizing the off-target expansion of tumor-infiltrating Treg cells associated with poor prognosis177. In both situations, IL-2 or IL-2R has been engineered in various ways to enhance selectivity toward the high vs. intermediate affinity IL-2R. The five main classes of engineered IL-2s include: muteins, IL-2/anti-IL-2 complexes, PEGylated IL-2, IL-2/CD25 fusion proteins, and orthogonal IL-2. For details, refer to reviews on these biologics and clinical testing12,13. Below discusses some of the issues uncovered in applying IL-2 to immunotherapy of autoimmunity and cancer.
Low dose (LD) rIL-2 was first used in clinical trials for chronic GVHD and hepatitis C virus-induced cryoglobulinemic vasculitis178,179. Such trials have also been completed for patients with T1D, alopecia areata, rheumatoid arthritis, systemic lupus erythematosus (SLE), psoriasis, granulomatosis with polyangiitis, Takayasu’s disease, and inflammatory bowel disease, and many others are ongoing13,180,181. From these trials, LD rIL-2 is 1-1.5 million IU of rIL-2 administered at frequencies ranging between daily to every other week. However, somewhat higher doses are administered in chronic GVHD182. These trials indicate that LD rIL-2 therapy is safe and increases Treg cells in all patients but at variable levels through an undetermined mechanism. Many patients show clinical improvement, but not complete resolution of disease. Importantly, self-reactive T cells are not reactivated, reflecting that human Treg cells are at least 100-fold more responsive than CD4+ CD45RO+ Teff cells to low amounts of rIL-2161. Treg cells from patients undergoing LD rIL-2 therapy exhibit a more activated phenotype and are more suppressive in vitro. Some evidence also suggests increased thymic Treg cell output and lower abundance of TFH and TH17 cells110,183, but the enhancement of the Treg compartment is thought to be the primary therapeutic mechanism.
Several observations indicate that LD rIL-2 may have some limitations. First, although clinical symptoms improved, LD rIL-2 has not shown off-drug efficacy182. Thus, LD rIL-2 is unlikely to induce long-lasting immune tolerance. Second, clear indication of clinical efficacy has not been robustly established in several phase 2 trials in SLE and alopecia areata180,184,185. Third, besides Treg cells, ILC2s186 and the CD56hi subpopulation of NK cells187,188 also respond to LD rIL-2. Both populations express the high affinity IL-2R and their expansion is an off-target effect (Fig. 4A). Virtually all trials to date have relied on using rIL-2, which has poor pharmacokinetics and pharmacodynamics that require frequent administration to maintain Treg increases.
The development of novel IL-2 agonists may enhance the efficacy of LD IL-2 and several are in clinical testing12,13. The IL-2 muteins (efavaleukin, RG-7835, CC-92252), which are fused to Fc, or pegylated IL-2 (NKTR-358), show more selective binding to the high affinity IL-2R by lowering the interaction with CD122 and may reduce off-target effects189,190. However, RG-7835 and CC-92252 did not meet efficacy criteria perhaps due to altered binding to the IL-2R. The IL-2/CD25 fusion protein targets the high affinity IL-2R through slow and steady dissociation of IL-2/CD25 monomers from more numerous transdimers and achieves selectivity toward the high affinity IL-2R by limiting the availability of active monomers191. IL-2/CD25 (BMS-986326) is currently being tested in normal subjects (NCT04736134) and patients with SLE (NCT06013995).
Off-target responses by ILC2s and CD56hi NK cells may be difficult to avoid, even with engineered IL-2s. ILC2-derived IL-5 promotes inflammation and is likely responsible for the eosinophilia that accompanies LD rIL-2 therapy186. However, ILC2s produce amphiregulin, which may facilitate tissue repair192. In addition, IL-2 in conjunction with retinoic acid and IL-33 induces a regulatory subset of ILC2s that secretes IL-10 that may decrease inflammation and represents another potential beneficial off-target effect of LD rIL-2193. CD56hi NK cells produce IFNγ, which is not desirable during LD rIL-2187, yet CD56hi NK cells exhibit anti-proliferative activity on Teff cells, which could also be beneficial194. The extent that stimulation of these cells is a detriment or a benefit during LD IL-2 needs to be carefully considered.
rIL-2 was the first approved immunotherapy for use in renal cell carcinoma and melanoma195,196. A few (~5-10%) patients were cured of these cancers, but most did not respond to therapy. Checkpoint blockade is the immunotherapy of choice for these and other malignancies due to improved response rates and relatively fewer side effects. Yet, many patients do not respond to checkpoint blockade, and for those that do, remission does not result in a cure. Thus, there remains considerable interest to use IL-2 in cancer immunotherapy due to its ability to promote development and expansion of highly functional Teff cells. Most efforts are focused on using engineered IL-2s to improve bioavailability, limit toxicities, and direct their activity toward lymphoid cells that express the intermediate affinity IL-2R, largely memory phenotypic CD8+ T and NK cells197,198. These types of engineered IL-2 agonists limit expansion of Treg cells, which have the potential to suppress antitumor responses. Another approach is to target IL-2 or engineered IL-2s to the tumor microenvironment, often by linking it to tumor-targeting antibodies or by inducing the release of bioactive IL-2 due to properties inherent to the tumor, such as tumor-specific proteases or high pH13.
Numerous trials are underway testing these novel IL-2-based approaches13,199. Several trials offer cautionary notes concerning IL-2s with selectivity for the intermediate affinity IL-2R. In a phase 3 trial, patients with metastatic melanoma were treated with anti-PD-1 (Nivolumab) or the combination of Nivolumab and a pegylated IL-2 (Bempegaldesleukin) with selectivity toward the intermediate affinity IL-2R. The combination therapy offered no benefit beyond what occurred with anti-PD-1 monotherapy (NCT03635983)200. A similar phase 3 trial (NCT03729245) in patients with metastatic renal cell carcinoma also did not reveal added benefit from the combination group. Some of these disappointing results might reflect the pharmacokinetics and pharmacodynamics specific to the engineered IL-2s used.
The IL-2-dependent reversal of Tex cells in conjunction with anti-PD-1 in preclinical models (as discussed above) revealed highly effective control of antitumor responses154,156. In a side-by-side comparison between IL-2/CD25 (selective toward the high affinity IL-2R), and IL-2/anti-IL-2 complexes (selective toward the intermediate affinity IL-2R), the former developed substantially more effective antitumor responses as a monotherapy or in combination with anti-PD-1154. Although Treg cells may expand in the periphery, notably, the ratio of CD8+ T cells to Treg cells in the tumor microenvironment was high152–154,156. Indeed, this might reflect a goldilocks situation, where an IL-2 analog directed to the high affinity IL-2R promotes robust antitumor responses with checkpoint blockade while an accompanying increase in Treg cells in the periphery suppresses off-target autoimmune-like effects that often accompany checkpoint blockade201. The critical issue is whether these promising preclinical studies translate when tested in patients with cancer.
Concluding remarks
We have a very detailed understanding concerning the role of varied IL-2R signaling for the development and homeostasis of Treg cells and for the development of various helper and cytotoxic T cell subsets. The IL-2R also provides important signals related to development of CD8+ TM and Tex cells fates. In all these cases, these cells are influenced by the levels of cell surface CD25 and thus, by the ability of T cells to capture typically limiting amounts of IL-2 through the high affinity IL-2R. Despite our current knowledge, we still have a cursory understanding of the molecular basis by which IL-2R signaling controls the above cell populations. This is especially true for thymic development of Treg cells, TR17 cells, and reprogramming of Tex cells. Little attention has been placed on how IL-2 impacts CD4+ TM202–204. Moreover, IL-2R signaling is more complex than the current paradigm (Fig. 1B), as emphasized by the plethora of IL-2-induced phosphorylated proteins in CD8+ T cells11. The ability to perform high resolution multiplex imaging, as done for Treg cells in lymph nodes87, offers the opportunity to examine the physiological and therapeutic contribution of IL-2R signaling on the immune landscape in lymphoid and non-lymphoid tissues.
The current state of the field makes it plain that IL-2 as a therapeutic should target the high affinity IL-2R. This is relatively straightforward for Treg cells, where a significant pharmacological window for selectivity over Teff cells exists. However, LD rIL-2 therapy likely requires more persistent signaling that may be delivered by newer IL-2 analogs. The effectiveness of this therapy may also be enhanced when used in combination with other immune modulators that lower inflammation and/or impair activation of autoreactive T cells. Although LD rIL-2 avoids Teff cells, activation of ILC2s and CD56hi NK cells often occurs. Whether these cells appreciably hinder or contribute to any therapeutic benefit needs careful consideration.
Targeting the high affinity IL-2R may also be critical when using IL-2 in cancer immunotherapy. IL-2 agonists, designed to selectively target the high amounts of the intermediate affinity IL-2R on memory-phenotypic T cells, are essentially a surrogate for IL-15. This type of IL-2 is less effective in generating antitumor responses than IL-2 agonists that target the high affinity IL-2R on tumor-specific CD8+ Teff cells153,154,156,205,206.Targeting the high affinity IL-2R is also effective in reinvigorating CD8+ Tex cells in the context of checkpoint blockade to drive antiviral and antitumor responses in preclinical studies153,154,156. In these type of studies, substantial increases in antigen-reactive T cells with high functional activity occurred in the tumor microenvironment such that the Teff cells outnumbered Treg cells, even though Treg cells are expected to respond to the higher doses of IL-2. Why Treg cells do not interfere with this productive immune response requires further investigation. Moreover, these promising preclinical findings warrant testing in cancer patients. Our aim in this review was to highlight our understanding of how IL-2 critically impacts Treg, Teff, and TM cells and provide a forward-looking point regarding IL-2 as a therapeutic.
Highlights:
IL-2 shapes the development and function of regulatory, effector and memory T cells.
The amount of CD25 determines which cells access and respond to IL-2.
IL-2 reprograms exhausted CD8+ T cells to rescue effector function.
Targeting the high affinity IL-2R is the key to therapy for autoimmunity and cancer.
Acknowledgements
Our work is funded by the NIH (R01AI148675, R01131648, R21AI159489), Florida Department of Health (21B03), and Bristol Myers Squibb to TRM and the NIH (F30AI157211) to ANS.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Competing Interests
The University of Miami and Thomas Malek have a patent pending on IL-2/CD25 fusion proteins and this technology has been licensed to Bristol Myers Squibb by the University of Miami for commercial development. Both receive royalties for its commercialization.
References
- 1.Liao W, Lin J-X, and Leonard WJ (2013). Interleukin-2 at the Crossroads of Effector Responses, Tolerance, and Immunotherapy. Immunity 38, 13–25. 10.1016/j.immuni.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Malek TR, and Castro I (2010). Interleukin-2 Receptor Signaling: At the Interface between Tolerance and Immunity. Immunity 33, 153–165. 10.1016/j.immuni.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wang X, Rickert M, and Garcia KC (2005). Structure of the Quaternary Complex of Interleukin-2 with Its α, ß, and γ c Receptors. Science 310, 1159–1163. 10.1126/science.1117893. [DOI] [PubMed] [Google Scholar]
- 4.Mjösberg JM, Trifari S, Crellin NK, Peters CP, Van Drunen CM, Piet B, Fokkens WJ, Cupedo T, and Spits H (2011). Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat Immunol 12, 1055–1062. 10.1038/ni.2104. [DOI] [PubMed] [Google Scholar]
- 5.Caligiuri MA, Zmuidzinas A, Manley TJ, Levine H, Smith KA, and Ritz J (1990). Functional consequences of interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer cell subset with high affinity receptors. J Exp Med 171, 1509–1526. 10.1084/jem.171.5.1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ross SH, and Cantrell DA (2018). Signaling and Function of Interleukin-2 in T Lymphocytes. Annu Rev Immunol 36, 411–433. 10.1146/annurev-immunol-042617-053352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Walsh PT (2006). PTEN inhibits IL-2 receptor-mediated expansion of CD4+CD25+ Tregs. J Clin Invest 116(9), 2521–2531. 10.1172/JCI28057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mandal M, Powers SE, Maienschein-Cline M, Bartom ET, Hamel KM, Kee BL, Dinner AR, and Clark MR (2011). Epigenetic repression of the Igk locus by STAT5-mediated recruitment of the histone methyltransferase Ezh2. Nat Immunol 12, 1212–1220. 10.1038/ni.2136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lin J-X, Li P, Liu D, Jin HT, He J, Rasheed MAU, Rochman Y, Wang L, Cui K, Liu C, et al. (2012). Critical Role of STAT5 Transcription Factor Tetramerization for Cytokine Responses and Normal Immune Function. Immunity 36, 586–599. 10.1016/j.immuni.2012.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Villarino A, Laurence A, Robinson GW, Bonelli M, Dema B, Afzali B, Shih H-Y, Sun H-W, Brooks SR, Hennighausen L, et al. (2016). Signal transducer and activator of transcription 5 (STAT5) paralog dose governs T cell effector and regulatory functions. eLife 5, e08384. 10.7554/eLife.08384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ross SH, Rollings C, Anderson KE, Hawkins PT, Stephens LR, and Cantrell DA (2016). Phosphoproteomic Analyses of Interleukin 2 Signaling Reveal Integrated JAK Kinase-Dependent and -Independent Networks in CD8 + T Cells. Immunity 45, 685–700. 10.1016/j.immuni.2016.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Hernandez R, Põder J, LaPorte KM, and Malek TR (2022). Engineering IL-2 for immunotherapy of autoimmunity and cancer. Nat Rev Immunol 22, 614–628. 10.1038/S41577-022-00680-w. [DOI] [PubMed] [Google Scholar]
- 13.Raeber ME, Sahin D, Karakus U, and Boyman O (2023). A systematic review of interleukin-2-based immunotherapies in clinical trials for cancer and autoimmune diseases. eBioMedicine 90, 104539. 10.1016/j.ebiom.2023.104539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Willerford DM, Chen J, Ferry JA, Davidson L, Ma A, and Alt FW (1995). Interleukin-2 receptor α chain regulates the size and content of the peripheral lymphoid compartment. Immunity 3, 521–530. 10.1016/1074-7613(95)90180-9. [DOI] [PubMed] [Google Scholar]
- 15.Sadlack B, Merz H, Schorle H, Schimpl A, Feller AC, and Horak I (1993). Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 75, 253–261. 10.1016/0092-8674(93)80067-0. [DOI] [PubMed] [Google Scholar]
- 16.Suzuki H, Kündig TM, Furlonger C, Wakeham A, Timms E, Matsuyama T, Schmits R, Simard JJ, Ohashi PS, and Griesser H (1995). Deregulated T cell activation and autoimmunity in mice lacking interleukin-2 receptor beta. Science 268, 1472–1476. 10.1126/science7770771. [DOI] [PubMed] [Google Scholar]
- 17.Sadlack B, Löhler J, Schorle H, Klebb G, Haber H, Sickel E, Noelle RJ, and Horak I (1995). Generalized autoimmune disease in interleukin-2-deficient mice is triggered by an uncontrolled activation and proliferation of CD4 + T cells. Eur J Immunol 25, 3053–3059. 10.1002/eji1830251111. [DOI] [PubMed] [Google Scholar]
- 18.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, and Toda M (1995). Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155, 1151–1164. [PubMed] [Google Scholar]
- 19.Malek TR, Porter BO, Codias EK, Scibelli P, and Yu A (2000). Normal Lymphoid Homeostasis and Lack of Lethal Autoimmunity in Mice Containing Mature T Cells with Severely Impaired IL-2 Receptors. J Immunol 164, 2905–2914. 10.4049/jimmunol.164.6.2905. [DOI] [PubMed] [Google Scholar]
- 20.Malek TR, Yu A, Vincek V, Scibelli P, and Kong L (2002). CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2. Immunity 17, 167–178. 10.1016/s1074-7613(02)00367-9. [DOI] [PubMed] [Google Scholar]
- 21.Fontenot JD, Rasmussen JP, Gavin MA, and Rudensky AY (2005). A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 6, 1142–1151. 10.1038/ni1263. [DOI] [PubMed] [Google Scholar]
- 22.D’Cruz LM, and Klein L (2005). Development and function of agonist-induced CD25+Foxp3+ regulatory T cells in the absence of interleukin 2 signaling. Nat Immunol 6, 1152–1159. 10.1038/ni1264. [DOI] [PubMed] [Google Scholar]
- 23.Setoguchi R, Hori S, Takahashi T, and Sakaguchi S (2005). Homeostatic maintenance of natural Foxp3 + CD25+ CD4+ regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med 201, 723–735. 10.1084/jem.20041982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Burchill MA, Yang J, Vogtenhuber C, Blazar BR, and Farrar MA (2007). IL-2 Receptor β-Dependent STAT5 Activation Is Required for the Development of Foxp3+ Regulatory T Cells. J Immunol 178, 280–290. 10.4049/jimmunol.178.1.280. [DOI] [PubMed] [Google Scholar]
- 25.Hsieh C-S, Lee H-M, and Lio C-WJ (2012). Selection of regulatory T cells in the thymus. Nat Rev Immunol 12, 157–167. 10.1038/nri3155. [DOI] [PubMed] [Google Scholar]
- 26.Roberts JL, Sharrow SO, and Singer A (1990). Clonal deletion and clonal anergy in the thymus induced by cellular elements with different radiation sensitivities. J Exp Med 171, 935–940. 10.1084/jem171.3.935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ramsdell F, Lantz T, and Fowlkes BJ (1989). A nondeletional mechanism of thymic self tolerance. Science 246, 1038–1041. 10.1126/science.2511629. [DOI] [PubMed] [Google Scholar]
- 28.Stritesky GL, Jameson SC, and Hogquist KA (2012). Selection of self-reactive T cells in the thymus. Annu Rev Immunol 30, 95–114. 10.1146/annurev-immunol-020711-075035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hu DY, Wirasinha RC, Goodnow CC, and Daley SR (2017). IL-2 prevents deletion of developing T-regulatory cells in the thymus. Cell Death Differ 24, 1007–1016. 10.1038/cdd.2017.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tai X, Erman B, Alag A, Mu J, Kimura M, Katz G, Guinter T, McCaughtry T, Etzensperger R, Feigenbaum L, et al. (2013). Foxp3 transcription factor is proapoptotic and lethal to developing regulatory T cells unless counterbalanced by cytokine survival signals. Immunity 38, 1116–1128. 10.1016/j.immuni.2013.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Toomer KH, Lui JB, Altman NH, Ban Y, Chen X, and Malek TR (2019). Essential and non-overlapping IL-2Rα-dependent processes for thymic development and peripheral homeostasis of regulatory T cells. NatCommun 10, 1037. 10.1038/s41467-019-08960-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, Gasteiger G, Feng Y, Fontenot JD, and Rudensky AY (2016). An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17, 1322–1333. 10.1038/ni.3540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yao Z, Kanno Y, Kerenyi M, Stephens G, Durant L, Watford WT, Laurence A, Robinson GW, Shevach EM, Moriggl R, et al. (2007). Nonredundant roles for Stat5a/b in directly regulating Foxp3. Blood 109, 4368–4375. 10.1182/blood-2006-11-055756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Burchill MA, Goetz CA, Prlic M, O’Neil JJ, Harmon IR, Bensinger SJ, Turka LA, Brennan P, Jameson SC, and Farrar MA (2003). Distinct effects of STAT5 activation on CD4+ and CD8+ T cell homeostasis: development of CD4+CD25+ regulatory T cells versus CD8+ memory T cells. J Immunol 171, 5853–5864. 10.4049/jimmunol.171.11.5853. [DOI] [PubMed] [Google Scholar]
- 35.Yao Z, Cui Y, Watford WT, Bream JH, Yamaoka K, Hissong BD, Li D, Durum SK, Jiang Q, Bhandoola A, et al. (2006). Stat5a/b are essential for normal lymphoid development and differentiation. Proc Natl Acad Sci U.S.A 103, 1000–1005. 10.1073/pnas.0507350103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Apert C, Galindo-Albarrán AO, Castan S, Detraves C, Michaud H, McJannett N, Haegeman B, Fillatreau S, Malissen B, Holländer G, et al. (2022). IL-2 and IL-15 drive intrathymic development of distinct periphery-seeding CD4+Foxp3+ regulatory T lymphocytes. Front Immunol 13, 965303. 10.3389/fimmu.2022.965303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bayer AL, Lee JY, De La Barrera A, Surh CD, and Malek TR (2008). A function for IL-7R for CD4+CD25+Foxp3+ T regulatory cells. J Immunol 181, 225–234. 10.4049/jimmunol181.1.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Vang KB, Yang J, Mahmud SA, Burchill MA, Vegoe AL, and Farrar MA (2008). IL-2, -7, and -15, but not thymic stromal lymphopoeitin, redundantly govern CD4+Foxp3+ regulatory T cell development. J Immunol 181, 3285–3290. 10.4049/jimmunol.181.5.3285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tai X, and Singer A (2014). Basis of Treg development in the thymus. Cell Cycle 13, 501–502. 10.4161/CC.27787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Owen DL, Mahmud SA, Sjaastad LE, Williams JB, Spanier JA, Simeonov DR, Ruscher R, Huang W, Proekt I, Miller CN, et al. (2019). Thymic regulatory T cells arise via two distinct developmental programs. Nat Immunol 20, 195–205. 10.1038/S41590-018-0289-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hemmers S, Schizas M, Azizi E, Dikiy S, Zhong Y, Feng Y, Altan-Bonnet G, and Rudensky AY (2019). IL-2 production by self-reactive CD4 thymocytes scales regulatory T cell generation in the thymus. J Exp Med 216, 2466–2478. 10.1084/jem.20190993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Marshall D, Sinclair C, Tung S, and Seddon B (2014). Differential requirement for IL-2 and IL-15 during bifurcated development of thymic regulatory T cells. J Immunol 193, 5525–5533. 10.4049/jimmunol.1402144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tai X, Indart A, Rojano M, Guo J, Apenes N, Kadakia T, Craveiro M, Alag A, Etzensperger R, Badr ME, et al. (2023). How autoreactive thymocytes differentiate into regulatory versus effector CD4+ T cells after avoiding clonal deletion. Nat Immunol 24, 637–651. 10.1038/s41590-023-01469-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Dikiy S, Li J, Bai L, Jiang M, Janke L, Zong X, Hao X, Hoyos B, Wang Z-M, Xu B, et al. (2021). A distal Foxp3 enhancer enables interleukin-2 dependent thymic Treg cell lineage commitment for robust immune tolerance. Immunity 54, 931–946.e11. 10.1016/j.immuni.2021.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kawakami R, Kitagawa Y, Chen KY, Arai M, Ohara D, Nakamura Y, Yasuda K, Osaki M, Mikami N, Lareau CA, et al. (2021). Distinct Foxp3 enhancer elements coordinate development, maintenance, and function of regulatory T cells. Immunity 54, 947–961.e8. 10.1016/j.immuni.2021.04.005. [DOI] [PubMed] [Google Scholar]
- 46.Feng Y, Arvey A, Chinen T, van der Veeken J, Gasteiger G, and Rudensky AY (2014). Control of the inheritance of regulatory T cell identity by a cis element in the Foxp3 locus. Cell 158, 749–763. 10.1016/j.cell.2014.07.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Nagar M, Vernitsky H, Cohen Y, Dominissini D, Berkun Y, Rechavi G, Amariglio N, and Goldstein I (2008). Epigenetic inheritance of DNA methylation limits activation-induced expression of FoxP3 in conventional human CD25-CD4+ T cells. Int Immunol 20, 1041–1055. 10.1093/intimm/dxn062. [DOI] [PubMed] [Google Scholar]
- 48.Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, and Rudensky AY (2010). Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 463, 808–812. 10.1038/nature08750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li P, Mitra S, Spolski R, Oh J, Liao W, Tang Z, Mo F, Li X, West EE, Gromer D, et al. (2017). STAT5-mediated chromatin interactions in superenhancers activate IL-2 highly inducible genes: Functional dissection of the Il2ra gene locus. Proc Natl Acad Sci U.S.A 114, 12111–12119. 10.1073/pnas.1714019114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Shevach EM, and Thornton AM (2014). tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev 259, 88–102. 10.1111/imr.12160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen W, Jin W, Hardegen N, Lei K, Li L, Marinos N, McGrady G, and Wahl SM (2003). 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. 10.1084/jem.20030152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.DiPaolo RJ, Brinster C, Davidson TS, Andersson J, Glass D, and Shevach EM (2007). Autoantigen-specific TGFβ-induced Foxp3+ regulatory T cells prevent autoimmunity by inhibiting dendritic cells from activating autoreactive T cells. J Immunol 179, 4685–4693. 10.4049/jimmunol179.7.4685. [DOI] [PubMed] [Google Scholar]
- 53.Huter EN, Punkosdy GA, Glass DD, Cheng LI, Ward JM, and Shevach EM (2008). TGF-β-induced Foxp3 + regulatory T cells rescue scurfy mice. Eur J Immunol 38, 1814–1821. 10.1002/eji.200838346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chen O, Kim YC, Laurence A, Punkosdy GA, and Shevach EM (2011). IL-2 controls the stability of Foxp3 expression in TGF-β–induced Foxp3+ T cells in vivo. J Immunol 186, 6329–6337. 10.4049/jimmunol.1100061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Bertolini TB, Biswas M, Terhorst C, Daniell H, Herzog RW, and Piñeros AR (2021). Role of orally induced regulatory T cells in immunotherapy and tolerance. Cell Immunol 359, 104251. 10.1016/j.cellimm.2020.104251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Pacholczyk R, Ignatowicz H, Kraj P, and Ignatowicz L (2006). Origin and T cell receptor diversity of Foxp3+CD4+CD25+ T cells. Immunity 25, 249–259. 10.1016/j.immuni.2006.05.016. [DOI] [PubMed] [Google Scholar]
- 57.Haribhai D, Williams JB, Jia S, Nickerson D, Schmitt EG, Edwards B, Ziegelbauer J, Yassai M, Li S-H, Relland LM, et al. (2011). A requisite role for induced regulatory T cells in tolerance based on expanding antigen receptor diversity. Immunity 35, 109–122. 10.1016/j.immuni.2011.03.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio C-W, Santacruz N, Peterson DA, Stappenbeck TS, and Hsieh C-S (2011). Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254. 10.1038/nature10434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Samstein RM, Josefowicz SZ, Arvey A, Treuting PM, and Rudensky AY (2012). Extrathymic generation of regulatory T cells in placental mammals mitigates maternal-fetal conflict. Cell 150, 29–38. 10.1016/j.cell.2012.05.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Zhou L, Chu C, Teng F, Bessman NJ, Goc J, Santosa EK, Putzel GG, Kabata H, Kelsen JR, Baldassano RN, et al. (2019). Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature 568, 405–409. 10.1038/s41586-019-1082-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Fan MY, Low JS, Tanimine N, Finn KK, Priyadharshini B, Germana SK, Kaech SM, and Turka LA (2018). Differential roles of IL-2 signaling in developing versus mature Tregs. Cell Rep 25, 1204–1213.e4. 10.1016/j.celrep.2018.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Moro A, Gao Z, Wang L, Yu A, Hsiung S, Ban Y, Yan A, Sologon CM, Chen XS, and Malek TR (2022). Dynamic transcriptional activity and chromatin remodeling of regulatory T cells after varied duration of interleukin-2 receptor signaling. Nat Immunol 23, 802–813. 10.1038/S41590-022-01179-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Dikiy S, and Rudensky AY (2023). Principles of regulatory T cell function. Immunity 56, 240–255. 10.1016/j.immuni.2023.01.004. [DOI] [PubMed] [Google Scholar]
- 64.Ward NC, Lui JB, Hernandez R, Yu L, Struthers M, Xie J, Santos Savio A, Dwyer CJ, Hsiung S, Yu A, et al. (2020). Persistent IL-2 receptor signaling by IL-2/CD25 fusion protein controls diabetes in NOD mice by multiple mechanisms. Diabetes 69, 2400–2413. 10.2337/db20-0186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, Treuting P, Siewe L, Roers A, Henderson WR, et al. (2008). Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558. 10.1016/j.immuni.2008.02.017. [DOI] [PubMed] [Google Scholar]
- 66.Williams LM, and Rudensky AY (2007). Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat Immunol 8, 277–284. 10.1038/ni1437. [DOI] [PubMed] [Google Scholar]
- 67.Zhou X, Bailey-Bucktrout SL, Jeker LT, Penaranda C, Martínez-Llordella M, Ashby M, Nakayama M, Rosenthal W, and Bluestone JA (2009). Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol 10, 1000–1007. 10.1038/ni.1774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Permanyer M, Bošnjak B, Glage S, Friedrichsen M, Floess S, Huehn J, Patzer GE, Odak I, Eckert N, Zargari R, et al. (2021). Efficient IL-2R signaling differentially affects the stability, function, and composition of the regulatory T-cell pool. Cell Mol Immunol 18, 398–414. 10.1038/s41423-020-00599-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Xu L, Kitani A, Fuss I, and Strober W (2007). Cutting Edge: Regulatory T Cells Induce CD4+CD25−Foxp3− T Cells or Are Self-Induced to Become Th17 Cells in the Absence of Exogenous TGF-β. J Immunol 178, 6725–6729. 10.4049/jimmunol.178.11.6725. [DOI] [PubMed] [Google Scholar]
- 70.Toomer KH, Yuan X, Yang J, Dee MJ, Yu A, and Malek TR (2016). Developmental progression and interrelationship of central and effector regulatory T cell subsets. J Immunol 196, 3665–3676. 10.4049/jimmunol.1500595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.DiSpirito JR, Zemmour D, Ramanan D, Cho J, Zilionis R, Klein AM, Benoist C, and Mathis D (2018). Molecular diversification of regulatory T cells in nonlymphoid tissues. Sci Immunol 3, eaat5861. 10.1126/sciimmunol.aat5861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zemmour D, Zilionis R, Kiner E, Klein AM, Mathis D, and Benoist C (2018). Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat Immunol 19, 291–301. 10.1038/s41590-018-0051-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Levine AG, Arvey A, Jin W, and Rudensky AY (2014). Continuous requirement for the TCR in regulatory T cell function. Nat Immunol 15, 1070–1078. 10.1038/ni.3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Smigiel KS, Richards E, Srivastava S, Thomas KR, Dudda JC, Klonowski KD, and Campbell DJ (2014). CCR7 provides localized access to IL-2 and defines homeostatically distinct regulatory T cell subsets. J Exp Med 211, 121–136. 10.1084/jem.20131142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kornete M, Sgouroudis E, and Piccirillo CA (2012). ICOS-dependent homeostasis and function of Foxp3+ regulatory T cells in islets of nonobese diabetic mice. J Immunol 188, 1064–1074. 10.4049/jimmunol1101303. [DOI] [PubMed] [Google Scholar]
- 76.Cheng G, Yuan X, Tsai MS, Podack ER, Yu A, and Malek TR (2012). IL-2 receptor signaling is essential for the development of Klrg1+ terminally differentiated T regulatory cells. J Immunol 189, 1780–1791. 10.4049/jimmunol1103768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Botta D, Fuller MJ, Marquez-Lago TT, Bachus H, Bradley JE, Weinmann AS, Zajac AJ, Randall TD, Lund FE, León B, et al. (2017). Dynamic regulation of T follicular regulatory cell responses by interleukin 2 during influenza infection. Nat Immunol 18, 1249–1260. 10.1038/ni.3837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zhang H, Dai D, Hu Q, Yang F, Xue Y, Li F, Shen N, Zhang M, and Huang C (2021). Bach2 attenuates IL-2R signaling to control Treg homeostasis and Tfr development. Cell Rep 35, 109096. 10.1016/j.celrep.2021.109096. [DOI] [PubMed] [Google Scholar]
- 79.Sefik E, Geva-Zatorsky N, Oh S, Konnikova L, Zemmour D, McGuire AM, Burzyn D, Ortiz-Lopez A, Lobera M, Yang J, et al. (2015). Individual intestinal symbionts induce a distinct population of RORγ + regulatory T cells. Science 349, 993–997. 10.1126/scienceaaa9420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, Blank RB, Meylan F, Siegel R, Hennighausen L, et al. (2007). Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity 26, 371–381. 10.1016/j.immuni.2007.02.009. [DOI] [PubMed] [Google Scholar]
- 81.Popmihajlov Z, and Smith KA (2008). Negative feedback regulation of T cells via interleukin-2 and Foxp3 reciprocity. PLoS ONE 3, e1581. 10.1371/journal.pone.0001581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Whyte CE, Singh K, Burton OT, Aloulou M, Kouser L, Veiga RV, Dashwood A, Okkenhaug H, Benadda S, Moudra A, et al. (2022). Context-dependent effects of IL-2 rewire immunity into distinct cellular circuits. J Exp Med 219, e20212391. 10.1084/jem.20212391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yu A, Zhu L, Altman NH, and Malek TR (2009). A low interleukin-2 receptor signaling threshold supports the development and homeostasis of T regulatory cells. Immunity 30, 204–217. 10.1016/j.immuni.2008.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Weist BM, Kurd N, Boussier J, Chan SW, and Robey EA (2015). Thymic regulatory T cell niche size is dictated by limiting IL-2 from antigen-bearing dendritic cells and feedback competition. Nature Immunol 16, 635–641. doi: 10.1038/ni.3171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Owen DL, Mahmud SA, Vang KB, Kelly RM, Blazar BR, Smith KA, and Farrar MA (2018). Identification of cellular sources of IL-2 needed for regulatory T cell development and homeostasis. J Immunol 200, 3926–3933. 10.4049/jimmunol.1800097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wuest SC, Edwan JH, Martin JF, Han S, Perry JSA, Cartagena CM, Matsuura E, Maric D, Waldmann TA, and Bielekova B (2011). A role for interleukin-2 transpresentation in dendritic cell–mediated T cell activation in humans, as revealed by daclizumab therapy. Nat Med 17, 604–609. 10.1038/nm.2365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wong HS, Park K, Gola A, Baptista AP, Miller CH, Deep D, Lou M, Boyd LF, Rudensky AY, Savage PA, et al. (2021). A local regulatory T cell feedback circuit maintains immune homeostasis by pruning self-activated T cells. Cell 184, 3981–3997.e22. 10.1016/j.cell.2021.05.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jones DM, Read KA, and Oestreich KJ (2020). Dynamic roles for IL-2-STAT5 signaling in effector and regulatory CD4+ T cell populations. J Immunol 205, 1721–1730. 10.4049/jimmunol.2000612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Saravia J, Chapman NM, and Chi H (2019). Helper T cell differentiation. Cell Mol Immunol 16, 634–643. 10.1038/s41423-019-0220-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Szabo SJ, Kim ST, Costa GL, Zhang X, Fathman CG, and Glimcher LH (2000). A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100, 655–669. 10.1016/S0092-8674(00)80702-3. [DOI] [PubMed] [Google Scholar]
- 91.Liao W, Lin J-X, Wang L, Li P, and Leonard WJ (2011). Modulation of cytokine receptors by IL-2 broadly regulates differentiation into helper T cell lineages. Nat Immunol 12, 551–559. 10.1038/ni.2030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Chang C-H, Curtis JD, Maggi LB, Faubert B, Villarino AV, O’Sullivan D, Huang SC-C, van der Windt GJW, Blagih J, Qiu J, et al. (2013). Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251. 10.1016/j.cell.2013.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Oestreich KJ, Read KA, Gilbertson SE, Hough KP, McDonald PW, Krishnamoorthy V, and Weinmann AS (2014). Bcl-6 directly represses the gene program of the glycolysis pathway. Nat Immunol 15, 957–964. 10.1038/ni.2985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Villarino AV, Laurence AD, Davis FP, Nivelo L, Brooks SR, Sun H-W, Jiang K, Afzali B, Frasca D, Hennighausen L, et al. (2022). A central role for STAT5 in the transcriptional programing of T helper cell metabolism. Sci Immunol 7, eabl9467. 10.1126/sciimmunol.abl9467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Lee K, Gudapati P, Dragovic S, Spencer C, Joyce S, Killeen N, Magnuson MA, and Boothby M (2010). Mammalian Target of Rapamycin Protein Complex 2 regulates differentiation of Th1 and Th2 cell subsets via distinct signaling pathways. Immunity 32, 743–753. 10.1016/j.immuni.2010.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Ray JP, Staron MM, Shyer JA, Ho P-C, Marshall HD, Gray SM, Laidlaw BJ, Araki K, Ahmed R, Kaech SM, et al. (2015). The interleukin-2-mTORC1 kinase axis defines the signaling, differentiation, and metabolism of T helper 1 and follicular B helper T cells. Immunity 43, 690–702. 10.1016/j.immuni.2015.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ouyang W, Löhning M, Gao Z, Assenmacher M, Ranganath S, Radbruch A, and Murphy KM (2000). Stat6-independent GATA-3 autoactivation directs IL-4-independent Th2 development and commitment. Immunity 12, 21–31. 10.1016/S1074-7613(00)80156-9. [DOI] [PubMed] [Google Scholar]
- 98.Zheng W, and Flavell RA (1997). The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell 89, 587–596. 10.1016/S0092-8674(00)80240-8. [DOI] [PubMed] [Google Scholar]
- 99.Liao W, Schones DE, Oh J, Cui Y, Cui K, Roh T-Y, Zhao K, and Leonard WJ (2008). Priming for T helper type 2 differentiation by interleukin 2–mediated induction of interleukin 4 receptor α-chain expression. Nat Immunol 9, 1288–1296. 10.1038/ni.1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Zhu J, Cote-Sierra J, Guo L, and Paul WE (2003). Stat5 activation plays a critical role in Th2 differentiation. Immunity 19, 739–748. 10.1016/S1074-7613(03)00292-9. [DOI] [PubMed] [Google Scholar]
- 101.Ballesteros-Tato A, León B, Graf BA, Moquin A, Adams PS, Lund FE, and Randall TD (2012). Interleukin-2 inhibits germinal center formation by limiting T follicular helper cell differentiation. Immunity 36, 847–856. 10.1016/j.immuni.2012.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.DiToro D, Winstead CJ, Pham D, Wtte S, Andargachew R, Singer JR, Wlson CG, Zindl CL, Luther RJ, Silberger DJ, et al. (2018). Differential IL-2 expression defines developmental fates of follicular versus nonfollicular helper T cells. Science 361, eaao2933. 10.1126/scienceaao2933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, Dent AL, Craft J, and Crotty S (2009). Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science 325, 1006–1010. 10.1126/science1175870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Johnston RJ, Choi YS, Diamond JA, Yang JA, and Crotty S (2012). STAT5 is a potent negative regulator of TFH cell differentiation. J Exp Med 209, 243–250. 10.1084/jem.20111174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Nurieva RI, Podd A, Chen Y, Alekseev AM, Yu M, Qi X, Huang H, Wen R, Wang J, Li HS, et al. (2012). STAT5 protein negatively regulates T follicular helper (Tfh) cell generation and function. J Biol Chem 287, 11234–11239. 10.1074/jbc.M111.324046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.León B, Bradley JE, Lund FE, Randall TD, and Ballesteros-Tato A (2014). FoxP3+ regulatory T cells promote influenza-specific Tfh responses by controlling IL-2 availability. Nat Commun 5, 3495. 10.1038/ncomms4495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Papillion A, Powell MD, Chisolm DA, Bachus H, Fuller MJ, Weinmann AS, Villarino A, O’Shea JJ, León B, Oestreich KJ, et al. (2019). Inhibition of IL-2 responsiveness by IL-6 is required for the generation of GC-T FH cells. Sci Immunol 4, eaaw7636. 10.1126/sciimmunol.aaw7636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Crotty S. (2019). T follicular helper cell biology: a decade of discovery and diseases. Immunity 50, 1132–1148. 10.1016/j.immuni.2019.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Pyle CJ, Labeur-lurman L, Groves HT, Puttur F, Lloyd CM, Tregoning JS, and Harker JA (2021). Enhanced IL-2 in early life limits the development of TFH and protective antiviral immunity. J Exp Med 218, e20201555. 10.1084/jem.20201555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, Jin Y, Gan Y, Hu X, Jia R, et al. (2016). Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med 22, 991–993. 10.1038/nm.4148. [DOI] [PubMed] [Google Scholar]
- 111.Maddur MS, Miossec P, Kaveri SV, and Bayry J (2012). Th17 Cells. Am J Pathol 181, 8–18. 10.1016/j.ajpath.2012.03.044. [DOI] [PubMed] [Google Scholar]
- 112.Joosse ME, Charbit-Henrion F, Boisgard R, Raatgeep R(H)C, Lindenbergh-Kortleve DJ, Costes LMM, Nugteren S, Guegan N, Parlato M, Veenbergen S, et al. (2021). Duplication of the IL2RA locus causes excessive IL-2 signaling and may predispose to very early onset colitis. Mucosal Immunol 14, 1172–1182. 10.1038/s41385-021-00423-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Goldberg R, Clough JN, Roberts LB, Sanchez J, Kordasti S, Petrov N, Hertweck A, Lorenc A, Jackson I, Tasker S, et al. (2021). A Crohn’s disease-associated IL2RA enhancer variant determines the balance of T cell immunity by regulating responsiveness to IL-2 signalling. J Crohn’s Colitis 15, 2054–2065. 10.1093/ecco-jcc/jjab103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yang X-P, Ghoreschi K, Steward-Tharp SM, Rodriguez-Canales J, Zhu J, Grainger JR, Hirahara K, Sun H-W, Wei L, Vahedi G, et al. (2011). Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol 12, 247–254. 10.1038/ni.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kim HS, Jang SW, Lee W, Kim K, Sohn H, Hwang SS, and Lee GR (2017). PTEN drives Th17 cell differentiation by preventing IL-2 production. J Exp Med 214, 3381–3398. 10.1084/jem.20170523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Pham D, Silberger DJ, Nguyen KN, Gao M, Weaver CT, and Hatton RD (2023). Batf stabilizes Th17 cell development via impaired Stat5 recruitment of Ets1-Runx1 complexes. EMBO J 42, e109803. 10.15252/embj.2021109803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kalia V, Sarkar S, Subramaniam S, Haining WN, Smith KA, and Ahmed R (2010). Prolonged interleukin-2Rα expression on virus-specific CD8+ T cells favors terminal-effector differentiation in vivo. Immunity 32, 91–103. 10.1016/j.immuni.2009.11.010. [DOI] [PubMed] [Google Scholar]
- 118.Gong D, and Malek TR (2007). Cytokine-dependent Blimp-1 expression in activated T cells inhibits IL-2 production. J Immunol 178, 242–252. 10.4049/jimmunol178.1.242. [DOI] [PubMed] [Google Scholar]
- 119.Malek TR (2008). The biology of interleukin-2. Annu Rev Immunol 26, 453–479. 10.1146/annurev.immunol.26.021607.090357. [DOI] [PubMed] [Google Scholar]
- 120.Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ, and Rao A (2010). Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 32, 79–90. 10.1016/j.immuni.2009.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Calame K. (2008). Activation-dependent induction of Blimp-1. Curr Opin Immunol 20, 259–264. 10.1016/j.coi.2008.04.010. [DOI] [PubMed] [Google Scholar]
- 122.Pearce EL, Mullen AC, Martins GA, Krawczyk CM, Hutchins AS, Zediak VP, Banica M, DiCioccio CB, Gross DA, Mao C, et al. (2003). Control of effector CD8 + T cell function by the transcription factor Eomesodermin. Science 302, 1041–1043. 10.1126/science1090148. [DOI] [PubMed] [Google Scholar]
- 123.Hand TW, Cui W, Jung YW, Sefik E, Joshi NS, Chandele A, Liu Y, and Kaech SM (2010). Differential effects of STAT5 and PI3K/AKT signaling on effector and memory CD8 T-cell survival. Proc Natl Acad Sci 107, 16601–16606. 10.1073/pnas1003457107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang CY, Best JA, Knell J, Yang E, Sheridan AD, Jesionek AK, Li HS, Rivera RR, Lind KC, D’Cruz LM, et al. (2011). The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat Immunol 12, 1221–1229. 10.1038/ni.2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Imada K, Bloom ET, Nakajima H, Horvath-Arcidiacono JA, Udy GB, Davey HW, and Leonard WJ (1998). Stat5b is essential for natural killer cell–mediated proliferation and cytolytic activity. J Exp Med 188, 2067–2074. 10.1084/jem188.11.2067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Malek TR, Yu A, Scibelli P, Lichtenheld MG, and Codias EK (2001). Broad programming by IL-2 receptor signaling for extended growth to multiple cytokines and functional maturation of antigen-activated T cells. J Immunol 166, 1675–1683. 10.4049/jimmunol.166.3.1675. [DOI] [PubMed] [Google Scholar]
- 127.Brown DM, Kamperschroer C, Dilzer AM, Roberts DM, and Swain SL (2009). IL-2 and antigen dose differentially regulate perforin- and FasL-mediated cytolytic activity in antigen specific CD4+ T cells. Cell Immunol 257, 69–79. 10.1016/j.cellimm.2009.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Takeuchi A, and Saito T (2017). CD4 CTL, a cytotoxic subset of CD4+ T cells, their differentiation and function. Front Immunol 8. 10.3389/fimmu.2017.00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Tian Y, Sette A, and Weiskopf D (2016). Cytotoxic CD4 T cells: differentiation, function, and application to Dengue virus infection. Front Immunol 7. 10.3389/fimmu.2016.00531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Glimcher LH, Townsend MJ, Sullivan BM, and Lord GM (2004). Recent developments in the transcriptional regulation of cytolytic effector cells. Nat Rev Immunol 4, 900–911. 10.1038/nri1490. [DOI] [PubMed] [Google Scholar]
- 131.Bachmann MF, Wolint P, Walton S, Schwarz K, and Oxenius A (2007). Differential role of IL-2R signaling for CD8 + T cell responses in acute and chronic viral infections. Eur J Immunol 37, 1502–1512. 10.1002/eji.200637023. [DOI] [PubMed] [Google Scholar]
- 132.Obar JJ, Molloy MJ, Jellison ER, Stoklasek TA, Zhang W, Usherwood EJ, and Lefrançois L (2010). CD4 + T cell regulation of CD25 expression controls development of short-lived effector CD8 + T cells in primary and secondary responses. Proc Natl Acad Sci 107, 193–198. 10.1073/pnas.0909945107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Feau S, Arens R, Togher S, and Schoenberger SP (2011). Autocrine IL-2 is required for secondary population expansion of CD8+ memory T cells. Nat Immunol 12, 908–913. 10.1038/ni.2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kahan SM, Bakshi RK, Ingram JT, Hendrickson RC, Lefkowitz EJ, Crossman DK, Harrington LE, Weaver CT, and Zajac AJ (2022). Intrinsic IL-2 production by effector CD8 T cells affects IL-2 signaling and promotes fate decisions, stemness, and protection. Sci Immunol 7, eabl6322. 10.1126/sciimmunol.abl6322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Toumi R, Yuzefpolskiy Y, Vegaraju A, Xiao H, Smith KA, Sarkar S, and Kalia V (2022). Autocrine and paracrine IL-2 signals collaborate to regulate distinct phases of CD8 T cell memory. Cell Rep 39, 110632. 10.1016/j.celrep.2022.110632. [DOI] [PubMed] [Google Scholar]
- 136.Chin SS, Guillen E, Chorro L, Achar S, Ng K, Oberle S, Alfei F, Zehn D, Altan-Bonnet G, Delahaye F, et al. (2022). T cell receptor and IL-2 signaling strength control memory CD8+ T cell functional fitness via chromatin remodeling. Nat Commun 13, 2240. 10.1038/S41467-022-29718-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, Lynn RC, Philip M, Rao A, Restifo NP, et al. (2019). Defining ‘T cell exhaustion.’ Nat Rev Immunol 19, 665–674. 10.1038/s41577-019-0221-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.McLane LM, Abdel-Hakeem MS, and Wherry EJ (2019). CD8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol 37, 457–495. 10.1146/annurev-immunol-041015-055318. [DOI] [PubMed] [Google Scholar]
- 139.Paley MA, Kroy DC, Odorizzi PM, Johnnidis JB, Dolfi DV, Barnett BE, Bikoff EK, Robertson EJ, Lauer GM, Reiner SL, et al. (2012). Progenitor and terminal subsets of CD8 + T cells cooperate to contain chronic viral infection. Science 338, 1220–1225. 10.1126/science1229620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, Polley A, Betts MR, Freeman GJ, Vignali DAA, and Wherry EJ (2009). Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10, 29–37. 10.1038/ni.1679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, Shan Q, Hale JS, Lee J, Nasti TH, et al. (2016). Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421. 10.1038/nature19330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Utzschneider DT, Charmoy M, Chennupati V, Pousse L, Ferreira DP, Calderon-Copete S, Danilo M, Alfei F, Hofmann M, Wieland D, et al. (2016). T Cell Factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427. 10.1016/j.immuni.2016.07.021. [DOI] [PubMed] [Google Scholar]
- 143.Wu T, Ji Y, Moseman EA, Xu HC, Manglani M, Kirby M, Anderson SM, Handon R, Kenyon E, Elkahloun A, et al. (2016). The TCF1-Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci Immunol 1, eaai8593. 10.1126/sciimmunol.aai8593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, Carmona SJ, Scarpellino L, Gfeller D, Pradervand S, et al. (2019). Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211.e10. 10.1016/j.immuni.2018.12.021. [DOI] [PubMed] [Google Scholar]
- 145.Beltra J-C, Manne S, Abdel-Hakeem MS, Kurachi M, Giles JR, Chen Z, Casella V, Ngiow SF, Khan O, Huang YJ, et al. (2020). Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841.e8. 10.1016/j.immuni.2020.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Beltra J-C, Bourbonnais S, Bédard N, Charpentier T, Boulangé M, Michaud E, Boufaied I, Bruneau J, Shoukry NH, Lamarre A, et al. (2016). IL2Rβ-dependent signals drive terminal exhaustion and suppress memory development during chronic viral infection. Proc Natl Acad Sci 113. 10.1073/pnas1604256113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Liu Y, Zhou N, Zhou L, Wang J, Zhou Y, Zhang T, Fang Y, Deng J, Gao Y, Liang X, et al. (2021). IL-2 regulates tumor-reactive CD8+ T cell exhaustion by activating the aryl hydrocarbon receptor. Nat Immunol 22, 358–369. 10.1038/s41590-020-00850-9. [DOI] [PubMed] [Google Scholar]
- 148.Zhao B, Gong W, Ma A, Chen J, Velegraki M, Dong H, Liu Z, Wang L, Okimoto T, Jones DM, et al. (2022). SUSD2 suppresses CD8+ T cell antitumor immunity by targeting IL-2 receptor signaling. Nat Immunol 23, 1588–1599. 10.1038/s41590-022-01326-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Mo F, Yu Z, Li P, Oh J, Spolski R, Zhao L, Glassman CR, Yamamoto TN, Chen Y, Golebiowski FM, et al. (2021). An engineered IL-2 partial agonist promotes CD8+ T cell stemness. Nature 597, 544–548. 10.1038/s41586-021-03861-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Drerup JM, Deng Y, Pandeswara SL, Padrón ÁS, Reyes RM, Zhang X, Mendez J, Liu A, Clark CA, Chen W, et al. (2020). CD122-selective IL2 complexes reduce immunosuppression, promote Treg fragility, and sensitize tumor response to PD-L1 blockade. Cancer Res 80, 5063–5075. 10.1158/0008-5472.CAN-20-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ren Z, Zhang A, Sun Z, Liang Y, Ye J, Qiao J, Li B, and Fu Y-X (2022). Selective delivery of low-affinity IL-2 to PD-1+ T cells rejuvenates antitumor immunity with reduced toxicity. J Clin Invest 132, e153604. 10.1172/JC1153604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Codarri Deak L, Nicolini V, Hashimoto M, Karagianni M, Schwalie PC, Lauener L, Varypataki EM, Richard M, Bommer E, Sam J, et al. (2022). PD-1-cis IL-2R agonism yields better effectors from stem-like CD8+ T cells. Nature 610, 161–172. 10.1038/s41586-022-05192-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hashimoto M, Araki K, Cardenas MA, Li P, Jadhav RR, Kissick HT, Hudson WH, McGuire DJ, Obeng RC, Wieland A, et al. (2022). PD-1 combination therapy with IL-2 modifies CD8+ T cell exhaustion program. Nature 610, 173–181. 10.1038/S41586-022-05257-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.LaPorte KM, Hernandez R, Santos Savio A, and Malek TR (2023). Robust IL-2-dependent antitumor immunotherapy requires targeting the high-affinity IL-2R on tumor-specific CD8 + T cells. J Immunother Cancer 11, e006611. 10.1136/jitc-2022-006611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Tichet M, Wullschleger S, Chryplewicz A, Fournier N, Marcone R, Kauzlaric A, Homicsko K, Deak LC, Umaña P, Klein C, et al. (2023). Bispecific PD1-IL2v and anti-PD-L1 break tumor immunity resistance by enhancing stem-like tumor-reactive CD8+ T cells and reprogramming macrophages. Immunity 56, 162–179.e6. 10.1016/j.immuni.2022.12.006. [DOI] [PubMed] [Google Scholar]
- 156.Wu W, Chia T, Lu J, Li X, Guan J, Li Y, Fu F, Zhou S, Feng Y, Deng J, et al. (2023). IL-2Rα-biased agonist enhances antitumor immunity by invigorating tumor-infiltrating CD25+CD8+ T cells. Nat Cancer 4, 1309–1325. 10.1038/s43018-023-00612-0. [DOI] [PubMed] [Google Scholar]
- 157.Beltra J-C, Abdel-Hakeem MS, Manne S, Zhang Z, Huang H, Kurachi M, Su L, Picton L, Ngiow SF, Muroyama Y, et al. (2023). Stat5 opposes the transcription factor Tox and rewires exhausted CD8+ T cells toward durable effector-like states during chronic antigen exposure. Immunity 56, 2699–2718.e11. 10.1016/j.immuni.2023.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Feinerman O, Jentsch G, Tkach KE, Coward JW, Hathorn MM, Sneddon MW, Emonet T, Smith KA, and Altan-Bonnet G (2010). Single-cell quantification of IL-2 response by effector and regulatory T cells reveals critical plasticity in immune response. Mol Syst Biol 6, 437. 10.1038/msb.2010.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ding Y, Yu A, Tsokos GC, and Malek TR (2019). CD25 and protein phosphatase 2A cooperate to enhance IL-2R signaling in human regulatory T cells. J Immunol 203, 93–104. 10.4049/jimmunol1801570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kim H-P, Kelly J, and Leonard WJ (2001). The basis for IL-2-induced IL-2 receptor α chain gene regulation. Immunity 15, 159–172. 10.1016/S1074-7613(01)00167-4. [DOI] [PubMed] [Google Scholar]
- 161.Yu A, Snowhite I, Vendrame F, Rosenzwajg M, Klatzmann D, Pugliese A, and Malek TR (2015). Selective IL-2 responsiveness of regulatory T cells through multiple intrinsic mechanisms supports the use of low-dose IL-2 therapy in type 1 diabetes. Diabetes 64, 2172–2183. 10.2337/db14-1322. [DOI] [PubMed] [Google Scholar]
- 162.Hayes ET, Hagan CE, Khoryati L, Gavin MA, and Campbell DJ (2020). Regulatory T cells maintain selective access to IL-2 and immune homeostasis despite substantially reduced CD25 function. J Immunol 205, 2667–2678. 10.4049/jimmunol1901520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Sharabi A, Li H, Kasper IR, Pan W, Meidan E, Tsokos MG, Moulton VR, and Tsokos GC (2019). PP2A enables IL-2 signaling by preserving IL-2Rβ chain expression during Treg development. JCI Insight 4, e126294. 10.1172/jci.insight126294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Shi H, Liu C, Tan H, Li Y, Nguyen T-LM, Dhungana Y, Guy C, Vogel P, Neale G, Rankin S, et al. (2018). Hippo kinases Mst1 and Mst2 sense and amplify IL-2R-STAT5 signaling in regulatory T cells to establish stable regulatory activity. Immunity 49, 899–914.e6. 10.1016/j.immuni.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, Solomon M, Selby W, Alexander SI, Nanan R, et al. (2006). Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J Exp Med 203, 1693–1700. 10.1084/jem.20060468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Liu W, Putnam AL, Xu-yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, De St. Groth BF, et al. (2006). CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203, 1701–1711. 10.1084/jem.20060772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Waickman AT, Keller HR, Kim T-H, Luckey MA, Tai X, Hong C, Molina-Paris C, Walsh STR, and Park J-H (2020). The cytokine receptor IL-7Rα impairs IL-2 receptor signaling and constrains the in vitro differentiation of Foxp3+ Treg cells. iScience 23, 101421. 10.1016/j.isci.2020.11421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Gratz IK, Truong H-A, Yang SH-Y, Maurano MM, Lee K, Abbas AK, and Rosenblum MD (2013). Cutting edge: memory regulatory T cells require IL-7 and not IL-2 for their maintenance in peripheral tissues. J Immunol 190, 4483–4487. 10.4049/jimmunol1300212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Farh KK-H, Marson A, Zhu J, Kleinewietfeld M, Housley WJ, Beik S, Shoresh N, Whitton H, Ryan RJH, Shishkin AA, et al. (2015). Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343. 10.1038/nature13835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Garg G, Tyler JR, Yang JHM, Cutler AJ, Downes K, Pekalski M, Bell GL, Nutland S, Peakman M, Todd JA, et al. (2012). Type 1 diabetes-associated IL2RA variation lowers IL-2 signaling and contributes to diminished CD4+CD25+ regulatory T cell function. J Immunol 188, 4644–4653. 10.4049/jimmunol1100272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Gootjes C, Zwaginga JJ, Roep BO, and Nikolic T (2022). Functional impact of risk gene variants on the autoimmune responses in type 1 diabetes. Front Immunol 13, 886736. 10.3389/fimmu.2022.886736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Simeonov DR, Gowen BG, Boontanrart M, Roth TL, Gagnon JD, Mumbach MR, Satpathy AT, Lee Y, Bray NL, Chan AY, et al. (2017). Discovery of stimulation-responsive immune enhancers with CRISPR activation. Nature 549, 111–115. 10.1038/nature23875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Spolski R, Li P, and Leonard WJ (2018). Biology and regulation of IL-2: from molecular mechanisms to human therapy. Nat Rev Immunol 18, 648–659. 10.1038/s41577-018-0046-y. [DOI] [PubMed] [Google Scholar]
- 174.Zheng Y, Josefowicz SZ, Kas A, Chu T-T, Gavin MA, and Rudensky AY (2007). Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature 445, 936–940. 10.1038/nature05563. [DOI] [PubMed] [Google Scholar]
- 175.Wu Y, Borde M, Heissmeyer V, Feuerer M, Lapan AD, Stroud JC, Bates DL, Guo L, Han A, Ziegler SF, et al. (2006). Foxp3 controls regulatory T cell function through cooperation with NFAT. Cell 126, 375–387. 10.1016/j.cell.2006.05.042. [DOI] [PubMed] [Google Scholar]
- 176.Ono M, Yaguchi H, Ohkura N, Kitabayashi I, Nagamura Y, Nomura T, Miyachi Y, Tsukada T, and Sakaguchi S (2007). Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature 446, 685–689. 10.1038/nature05673. [DOI] [PubMed] [Google Scholar]
- 177.Ahmadzadeh M, and Rosenberg SA (2006). IL-2 administration increases CD4+CD25hi Foxp3+ regulatory T cells in cancer patients. Blood 107, 2409–2414. 10.1182/blood-2005-06-2399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Koreth J, Matsuoka K, Kim HT, McDonough SM, Bindra B, Alyea EP, Armand P, Cutler C, Ho VT, Treister NS, et al. (2011). Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med 365, 2055–2066. 10.1056/NEJMoa1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Saadoun D, Rosenzwajg M, Joly F, Six A, Carrat F, Thibault V, Sene D, Cacoub P, and Klatzmann D (2011). Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365, 2067–2077. 10.1056/NEJMoal105143. [DOI] [PubMed] [Google Scholar]
- 180.Humrich JY, Cacoub P, Rosenzwajg M, Pitoiset F, Pham HP, Guidoux J, Leroux D, Vazquez T, Riemekasten G, Smolen JS, et al. (2022). Low-dose interleukin-2 therapy in active systemic lupus erythematosus (LUPIL-2): a multicentre, double-blind, randomised and placebo-controlled phase II trial. Ann Rheum Dis 81, 1685–1694. 10.1136/ard-2022-222501. [DOI] [PubMed] [Google Scholar]
- 181.Rosenzwajg M, Lorenzon R, Cacoub P, Pham HP, Pitoiset F, El Soufi K, Rlbet C, Bernard C, Aractingi S, Banneville B, et al. (2019). Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial. Ann Rheum Dis 78, 209–217. 10.1136/annrheumdis-2018-214229. [DOI] [PubMed] [Google Scholar]
- 182.Matsuoka K, Koreth J, Kim HT, Bascug G, McDonough S, Kawano Y, Murase K, Cutler C, Ho VT, Alyea EP, et al. (2013). Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med 5. 10.1126/scitranslmed.3005265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Miao M, Xiao X, Tian J, Zhufeng Y, Feng R, Zhang R, Chen J, Zhang X, Huang B, Jin Y, et al. (2021). Therapeutic potential of targeting Tfr/Tfh cell balance by low-dose-IL-2 in active SLE: a post hoc analysis from a double-blind RCT study. Arthritis Res Ther 23, 167. 10.1186/si3075-021-02535-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.He J, Zhang R, Shao M, Zhao X, Miao M, Chen J, Liu J, Zhang X, Zhang X, Jin Y, et al. (2020). Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: a randomised, double-blind, placebo-controlled trial. Ann Rheum Dis 79, 141–149. 10.1136/annrheumdis-2019-215396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Le Duff F, Bouaziz J-D, Fontas E, Ticchioni M, Viguier M, Dereure O, Reygagne P, Montaudié H, Lacour J-P, Monestier S, et al. (2021). Low-dose IL-2 for treating moderate to severe alopecia areata: a 52-week multicenter prospective placebo-controlled study assessing its impact on T regulatory cell and NK cell populations. J Invest Dermatol 141, 933–936.e6. 10.1016/j.jid.2020.08.015. [DOI] [PubMed] [Google Scholar]
- 186.Van Gool F, Molofsky AB, Morar MM, Rosenzwajg M, Liang H-E, Klatzmann D, Locksley RM, and Bluestone JA (2014). Interleukin-5-producing group 2 innate lymphoid cells control eosinophilia induced by interleukin-2 therapy. Blood 124, 3572–3576. 10.1182/blood-2014-07-587493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ito S, Bollard CM, Carlsten M, Melenhorst JJ, Biancotto A, Wang E, Chen J, Kotliarov Y, Cheung F, Xie Z, et al. (2014). Ultra-low dose interleukin-2 promotes immune-modulating function of regulatory T cells and natural killer cells in healthy volunteers. Mol Ther 22, 1388–1395. 10.1038/mt.2014.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Kubo T, Romee R, Koreth J, Belizaire R, Kamihara Y, Liu H, Asano T, Whangbo J, Hirakawa M, Nikiforow S, et al. (2018). Low-dose interleukin-2 therapy enhances cytotoxicity of CD56bright NK cells in patients with chronic Gvhd. Blood 132, 606–606. 10.1182/blood-2018-99-109918. [DOI] [Google Scholar]
- 189.Tchao N, Sarkar N, Hu X, Zhang R, Milmont C, Shi Jin Y, Chow V, Kroenke M, Gorski K, Furie R, et al. (2022). AB0432 Efavaleukin alfa, a novel IL-2 mutein, selectively expands regulatory T cells in patients with SLE: final results of a phase 1B multiple ascending dose study. Ann Rheum Dis 81, 1343.3–1344. 10.1136/annrheumdis-2022-eular.2244. [DOI] [Google Scholar]
- 190.Dixit N, Fanton C, Langowski JL, Kirksey Y, Kirk P, Chang T, Cetz J, Dixit V, Kim G, Kuo P, et al. (2021). NKTR-358: A novel regulatory T-cell stimulator that selectively stimulates expansion and suppressive function of regulatory T cells for the treatment of autoimmune and inflammatory diseases. J Transl Autoimmun 4, 100103. 10.1016/j.jtauto.2021.100103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Ward NC, Yu A, Moro A, Ban Y, Chen X, Hsiung S, Keegan J, Arbanas JM, Loubeau M, Thankappan A, et al. (2018). IL-2/CD25: a long-acting fusion protein that promotes immune tolerance by selectively targeting the IL-2 receptor on regulatory T cells. J Immunol 201, 2579–2592. 10.4049/jimmunol1800907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Monticelli LA, Sonnenberg GF, Abt MC, Alenghat T, Ziegler CGK, Doering TA, Angelosanto JM, Laidlaw BJ, Sathaliyawala T, Kubota M, et al. (2012). Innate lymphoid cells promote lung tissue homeostasis following acute influenza virus infection. Nat Immunol 12, 1045–1053. 10.1038/ni.2131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Emami Fard N, Xiao M, and Sehmi R (2023). Regulatory ILC2—role of IL-10 producing ILC2 in asthma. Cells 12, 2556. 10.3390/cellsl2212556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.McQuaid SL, Loughran ST, Power PA, Maguire P, Szczygiel A, and Johnson PA (2020). Low-dose IL-2 induces CD56bright NK regulation of T cells via NKp44 and NKp46. Clin and Exp Immunol 200, 228–241. 10.1111/cei13422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, and White DE (1994). Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 271, 907–913. [PubMed] [Google Scholar]
- 196.Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, and Louie AC (1995). Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 13, 688–696. 10.1200/JCO.1995.13.3.688. [DOI] [PubMed] [Google Scholar]
- 197.Levin AM, Bates DL, Ring AM, Krieg C, Lin JT, Su L, Moraga I, Raeber ME, Bowman GR, Novick P, et al. (2012). Exploiting a natural conformational switch to engineer an interleukin-2 ‘superkine.’ Nature 484, 529–533. 10.1038/nature10975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Sahin D, Arenas-Ramirez N, Rath M, Karakus U, Hümbelin M, Van Gogh M, Borsig L, and Boyman O (2020). An IL-2-grafted antibody immunotherapy with potent efficacy against metastatic cancer. Nat Commun 11, 6440. 10.1038/s41467-020-20220-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Overwijk WW, Tagliaferri MA, and Zalevsky J (2021). Engineering IL-2 to give new life to T cell immunotherapy. Annu Rev Med 72, 281–311. 10.1146/annurev-med-073118-011031. [DOI] [PubMed] [Google Scholar]
- 200.Diab A, Gogas H, Sandhu S, Long GV, Ascierto PA, Larkin J, Sznol M, Franke F, Ciuleanu TE, Pereira C, et al. (2023). Bempegaldesleukin plus Nivolumab in untreated advanced melanoma: the open-label, phase III PIVOT IO 001 trial results. J Clin Oncol 41, 4756–4767. 10.1200/JC0.23.00172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Alissafi T, Hatzioannou A, Legaki AI, Varveri A, and Verginis P (2019). Balancing cancer immunotherapy and immune-related adverse events: The emerging role of regulatory T cells. J Autoimmun 104, 102310. 10.1016/j.jaut.2019.102310. [DOI] [PubMed] [Google Scholar]
- 202.Alam F, Singh A, Flores-Malavet V, Sell S, Cooper AM, Swain SL, McKinstry KK, and Strutt TM (2020). CD25-targeted IL-2 signals promote improved outcomes of influenza infection and boost memory CD4 T cell formation. J Immunol 204, 3307–3314. 10.4049/jimmunol.2000205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Dooms H, Wolslegel K, Lin P, and Abbas AK (2007). Interleukin-2 enhances CD4+ T cell memory by promoting the generation of IL-7Rα–expressing cells. J Exp Med 204, 547–557. 10.1084/jem.20062381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.McKinstry KK, Strutt TM, Bautista B, Zhang W, Kuang Y, Cooper AM, and Swain SL (2014). Effector CD4 T-cell transition to memory requires late cognate interactions that induce autocrine IL-2. Nat Commun 5, 5377. 10.1038/ncomms6377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Hernandez R, LaPorte KM, Hsiung S, Santos Savio A, and Malek TR (2021). High-dose IL-2/CD25 fusion protein amplifies vaccine-induced CD4 + and CD8 + neoantigen-specific T cells to promote antitumor immunity. J Immunother Cancer 9, e002865. 10.1136/jitc-2021-002865. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Hernandez R, Toomer KH, Põder J, Santos Savio A, Hsiung S, and Malek TR (2021). Sustained IL-2R signaling of limited duration by high-dose mIL-2/mCD25 fusion protein amplifies tumor-reactive CD8+ T cells to enhance antitumor immunity. Cancer Immunol Immunother 70, 909–921. 10.1007/s00262-020-02722-5. [DOI] [PMC free article] [PubMed] [Google Scholar]