Summary
CD4+ regulatory T cells (Treg) expressing the forkhead box protein 3 (FOXP3) transcription factor (Tregs) are instrumental for the prevention of autoimmune diseases. There is increasing evidence that the human T regulatory population is highly heterogeneous in phenotype and function. Numerous studies conducted in human autoimmune diseases have shown that Treg cells are impaired either in their suppressive function, in number, or both. However, the contribution of the FOXP3+ Treg subpopulations to the development of autoimmunity has not been delineated in detail. Rare genetic disorders that involve deficits in Treg function can be studied to develop a global idea of the impact of partial or complete deficiency in a specific molecular mechanism involved in Treg function. In patients with reduced Treg numbers (but no functional deficiency), the expansion of autologous Treg cells could be a suitable therapeutic approach: either infusion of in‐vitro autologous expanded cells, infusion of interleukin (IL)‐2/anti‐IL‐2 complex, or both. Treg biology‐based therapies may not be suitable in patients with deficits of Treg function, unless their deficit can be corrected in vivo/in vitro. Finally, it is critical to consider the appropriate stage of autoimmune diseases at which administration of Treg cellular therapy can be most effective. We discuss conflicting data regarding whether Treg cells are more effectual at preventing the initiation of autoimmunity, ameliorating disease progression or curing autoimmunity itself.
Keywords: autoimmune, cell therapy, Treg, inflammation, T regulatory cells
Introduction
CD4+ T regulatory cells (Treg) expressing the forkhead box protein 3 (FOXP3) transcription factor (Treg cells) are capable of suppressing immune responses, especially their initiation, by preventing the activation and proliferation of T and B cells 1. The importance of Treg cells has been demonstrated in animal models whereby their depletion is associated with loss of self‐tolerance and development of severe autoimmunity [e.g. immunodysregulation, polyendocrinopathy enteropathy X‐linked (IPEX) syndrome] 2, 3, 4, 5. Further data in murine models have also demonstrated their ability to prevent progression of and even cure established autoimmune/inflammatory disease 6, 7. Overall, these cells are also considered to play a role in allergy prevention 8, gestational tolerance 9, the promotion of graft tolerance post‐transplantation 10 and the prevention of tumour immune responses 11.
Human Treg cells were initially characterized as CD4+ T cells co‐expressing the interleukin (IL)‐2 receptor alpha chain 12, 13, 14, 15, 16, 17, 18. This was based on murine data demonstrating that depletion of CD4+CD25+ T cells led to the development of severe autoimmunity 2. Since then, Treg cells have been more precisely described as CD4+ T cells expressing the FOXP3 transcription factor in mice 3, 4, 5 and in humans 19. However, while the CD25+FOXP3+ phenotype defines Treg cells in mice, both CD25 and FOXP3 can also be induced upon activation in naive CD4+ T cells (of mice and humans) 20. Intriguingly, CD25 and FOXP3 can also be induced in conventional T cells, although this does not equate to them adopting ‘regulatory’ function 21, 22. All this indicates that the mere combination of CD25 and FOXP3 expression is insufficient to define human Treg cells phenotypically in health and diseases 23.
In recent years, we have progressed to further categorize human CD4+FOXP3+ T cells into three distinct subpopulations based upon their phenotypical and functional differences. These subpopulations are (a) Fraction I: CD45RA+FOXP3lo naive Treg cells, considered equivalent to natural Treg cells arising from the thymus (tTreg cells) and demonstrating immunosuppressive properties in vitro; (b) Fraction II: CD45RA–FOXP3hi activated effector Treg cells, also immunosuppressive in vitro; and (c) Fraction III: CD45RA–FOXP3lo cytokine‐secreting but not immunosuppressive cells 23. We were also able to classify these populations similarly using CD25 (instead of FOXP3) and CD45RO (instead of CD45RA). Since then, further elegant work has demonstrated that the Fraction III population could be further subdivided on the basis of CD127 to identify two subpopulations. The proteomic analyses performed identified these subpopulations to closely resemble memory conventional T cells or effector Treg cells, respectively 24. Importantly, these aforementioned subpopulations can be distinctly identified in healthy and diseased states. For example, CD45RA+FOXP3lo Treg cells are the main Treg population identified in cord blood 23, whereas effector Treg cells are highly prevalent in tumours or in peripheral blood of patients with sarcoidosis 25, 26 or mycosis fungoides 27. Interestingly, a small proportion of CD45RA–FOXP3lo cells have been identified in the peripheral blood of patients with active systemic lupus erythematosus 27 or in some tumours 28. The impact of these cell subpopulations on their respective pathologies is not yet known.
Numerous studies conducted in human autoimmune diseases have shown that Treg cells were impaired either in their suppressive function, in number, or both. A further mechanism involves resistance of conventional T cells to Treg‐mediated suppression via the presence of certain cytokines [tumour necrosis factor (TNF) and IL‐6] in the microenvironment and over‐activated phosphatidylinositol 3‐kinase/protein kinase B (PI3K/Akt) signalling 29. Of note, these findings have been demonstrated in a range of autoimmune diseases 29, 30, 31. The conclusions of those studies have been drawn following different phenotypical definitions of human Treg cells, mainly based on the assumption the either that FOXP3‐expressing and/or CD25high CD4+T cells constitute a single homogeneous population of Tregs 19. Hence, the contribution of the heterogeneous FOXP3+ Treg subpopulations to the development of autoimmunity has not been delineated in detail. In this review, we discuss several unresolved questions 32, 33 and emerging issues regarding the role of Treg cells in human autoimmune and inflammatory diseases.
Heterogeneity of human FOXP3‐expressing CD4+ T cell subsets
There is increasing evidence that the human T regulatory population is highly heterogeneous in phenotype and function 34. While FOXP3‐expressing cells can be roughly separated into three subsets (naive Treg cells, effector Treg cells and FOXP3lo‐activated T cells) 23, there are novel data indicating that these subpopulations can be subdivided phenotypically even further (Fig. 1).
Figure 1.

Human regulatory T cell (Treg) subsets. Thymus produces CD45RA+forkhead box protein 3 (FOXP3)lo naive Treg (nTreg) cells as well as naive CD45RA+ non‐Treg cells. nTreg cells can differentiate into CD45RA–FOXP3hi effector T+ (eTreg) cells, which are potently suppressive, but they can also maintain low levels of FOXP3 and become non‐Treg cells. Effector Treg cells can also convert into non‐Treg cells, while non‐Treg cells can up‐regulate FOXP3 transiently to become induced effector Treg cells. As shown, all FOXP3‐expressing subsets can be divided into subsets by differential expression of surface markers or transcription factors.
CD45RA+ naive Treg cells can be separated into the CD31+ recent thymic emigrant (RTE) and the CD31– naive Treg cell population 35 (Fig. 1). Recent data obtained via cytometry by time of flight (CyTOF) has demonstrated that naive Treg cells can be subdivided based on their expression of CD49b, CD62L and certain chemokine receptors. This has led to their subcategorization as CD49b+, CD49b+CXCR3+CCR4+CCR6+ and CXCR3+RORC2+CD62L+ subsets 36. It is currently unknown how CD31+ RTE Treg cells differentiate into each of these subsets; the prevalence of each subset in human health and disease and how each of those subsets differentiates into other FOXP33+ T cells have also to be determined.
Interestingly, CCR4 can also delineate six different subsets among the Fraction III FOXP3lo non‐Treg cell population when combined with CD127 and CD49d. (Fig. 1). Among these subsets, the CD127+CD49d+CCR4– population contains most of the cytokine‐producing cells (IL‐2, IL‐17 and IFN‐γ), while the CD127–CD49d–CCR4+ contains the lowest number of cytokine‐producing cells. This latter subpopulation is, therefore, functionally and phenotypically most similar to the effector Treg subpopulation, as most effector Treg cells are CD127–CD49d–CCR4+ 24. The origin of Fraction III FOXP3lo cells has not yet been completely elucidated: these cells can be derived from naive Treg cells that fail to up‐regulate high levels of FOXP3 upon activation, for instance, because of weak signal transducer and activator of transcription (STAT)‐5 signalling; they may also be derived from some FOXP3hi effector Treg cells through reduced expression of FOXP3, and they can also be derived from conventional CD4+ T cells that transiently up‐regulate FOXP3 upon activation. Of note, all of these phenotypical changes have been observed in vitro, generally in the presence of low‐dose IL‐2 37.
Effector Treg cells constitute a functionally homogeneous suppressive subset that is highly proliferative in vivo but poorly proliferative in vitro, and prone to apoptosis in the absence of IL‐2 1, 19. However, from a phenotypical perspective, these cells can be subcategorized based on their co‐expression of effector T cell transcription factors such as T‐bet [T helper type 1 (Th1), GATA binding protein 3 (GATA‐3) (Th2) and retinoid‐related orphan receptor γt (RORC)] (Th17) 32. This is supported by murine data indicating that Th1‐, Th2‐ and Th17‐like Treg cells suppress their related effector Th cell counterparts, respectively 38, 39, 40. These Treg subsets also express their effector Th cell counterparts’ chemokine receptors. Hence, Th1‐like Treg cells are CXCR3+, Th2‐like Treg cells are CCR6–CCR4+ and Th17‐like Treg cells are CCR6+CCR4+ 41. A further Treg subset comprise T follicular regulatory (Tfr) cells, which are found in germinal centres and can directly influence B cells. 42 These cells interfere with the interaction between T follicular helper (Tfh) cells and B cells to alter subsequent B cell differentiation towards antibody‐producing plasma cells or a memory phenotype. This interaction requires further study, particularly to improve our understanding of antibody‐mediated autoimmune disease [e.g. thyroiditis, type 1 diabetes (T1D)].
Additionally, recent studies have demonstrated phenotypical heterogeneity between Treg cells in the peripheral circulation and those present in the healthy/diseased tissue 43. For instance, IL‐1 receptor type II (IL‐1R2), a decoy receptor for IL‐1, is highly expressed on breast and colonic Treg cells but not on their peripheral circulation counterparts 44, 45. Similarly, CD15s (sialyl Lewis X) is present on peripherally circulating effector Treg cells but is absent on their pulmonary Treg counterparts 46.
Overall, it is clear that historical studies in human autoimmune and/or inflammatory diseases have been conducted on the basis that circulating FOXP3+ T cells were a homogeneous population 30. On the basis of the data discussed above and access to novel technologies, we anticipate the future determination of the distinct contributions of FOXP3+ Treg subsets to human health and disease (Fig. 1).
The significance of FOXP3+ Treg cells abnormalities in autoimmune diseases: from pernicious anaemia to IPEX
In their seminal publication demonstrating that murine Treg cells displayed the CD4+CD25+ phenotype, Sakaguchi et al. showed that the canonical autoimmune abnormality observed in sick mice depleted of Treg cells was the occurrence of autoimmune gastritis with circulating anti‐parietal autoantibodies 2, a condition that is reminiscent of pernicious anaemia in humans 47. However, the role of Treg cells in pernicious anaemia in humans has not yet been delineated. This knowledge gap can also be extrapolated to other autoimmune diseases whereby the role of Treg cells in their development has been studied using animal or human culture systems that are not necessarily reflective of true human pathology 33.
On one hand, upon review of published literature into human autoimmunity, one may be tempted to conclude that all autoimmune diseases could be characterized by either a deficit in Treg number and/or function or resistance of conventional T cells to Treg‐mediated suppression 29, 30, 31, 48, 49. On the other hand, the only known condition with clear evidence for total depletion of Treg cells is IPEX 3, 4, 5. This condition is provoked by different genetic defects in the FOXP3 gene and is characterized by the occurrence of enteropathy, eczema, T1D, thyroiditis, cytopenia, hepatitis, nephritis and gastritis 50, 51, 52. Indeed, the scurfy mouse model is widely utilized for the study of Treg cells, as equivalent defects in the FOXP3 gene lead to a pathologically similar autoimmune disease. Scurfy mice die within a few weeks after birth, while untreated newborns with IPEX die rapidly, both of severe inflammation, allergy and autoimmunity 3, 4, 5. Hence, it is clear that a complete defect in Treg cells leads to the development of this lethal systemic autoimmune and inflammatory disease.
Due to the rapid progression of IPEX in murine and human newborns (fortunately, a rare condition), the detailed study of Treg cell deficiency in adults with autoimmune disease has remained a challenge. Rudensky et al. and Sparwasser et al. attempted to address this by developing mice with Treg cells bearing the diphtheria toxin receptor 53. Thus, the injection of diphtheria toxin would provoke the complete depletion of Treg cells within days, and thereby lead to a rapidly lethal systemic autoimmune disease that is similar to that observed in IPEX 53, 54.
Overall, it is clear from the experimental observations discussed above that a complete deficiency of Treg cells at any stage of life rapidly leads to a fatal systemic disease. However, it is also clear from the clinical literature that human autoimmune diseases tend not to evolve towards an IPEX‐like condition. Indeed, we would reconcile these findings by hypothesizing that Treg number deficiency and dysfunction in human autoimmune diseases is incomplete at any stage of life and is probably mild or minimal. For example, pernicious anaemia may be associated with some organ‐specific endocrine autoimmune diseases such as Graves’ disease, but rarely to other systemic autoimmune diseases 55. Importantly, neither of these conditions evolve towards an IPEX‐like condition. Although not yet proven, if there is indeed a Treg deficit in pernicious anaemia it is likely to be mild or minimal.
FOXP3+ Treg cell function deficiencies in human autoimmune diseases: lessons from genetic diseases
The lack of validated experimental assays reflective of in‐vivo human Treg biology remains a major limitation in the field. To what extent in‐vitro Treg suppressive activity correlates with in‐vivo Treg function has not yet been established in humans 48. This limitation is important to overcome, as Treg cells have numerous mechanisms of action which require different experimental design and reagents to reliably elicit 56. It is indeed plausible that observed in‐vitro Treg functional deficiency in human autoimmune diseases may be explained by the partial deficiency of one or several mechanisms of suppression. One must also not discount the potential for effector T cells to be resistant to Treg‐mediated suppression mechanisms 29. While the specific roles of these mechanisms can be studied in mice (via different conditional knock‐out models), their corresponding contributions in humans have been mainly elicited using in‐vitro models instead 57. For this reason, patients with rare genetic disorders that involve deficits in Treg function can be studied to develop a global idea of the impact of partial or complete deficiency in a specific molecular mechanism involved in Treg function.
One of the most well‐established mechanisms of action of Treg cells is through their cytotoxic T lymphocyte‐associated protein 4 (CTLA‐4) receptor. Indeed, constitutive expression of CTLA‐4 by Treg cells is instrumental for their in‐vivo suppressive capacity 58. Interestingly, CTLA‐4 haploinsufficiency has been described (albeit rarely) in certain families 59, 60. It is therefore noteworthy that patients with heterozygous non‐sense mutations of CTLA‐4 genes develop a systemic autoimmune disease manifesting as diarrhoea, granulomatous interstitial lung disease, autoimmune cytopaenia, thyroiditis, arthritis and skin disease—all of which are reminiscent of IPEX (but with less severity). Of note, none of these patients studied developed autoimmunity in early infancy, but a significant proportion had their first autoimmune abnormality diagnosed in adulthood. From a cellular perspective, although this mutation could have impacted on the CTLA4‐induction properties and function of all activated T cells, the impact on Treg cells specifically is important. This is because normal Treg cells express disproportionally higher surface and intracellular CTLA4 61. Interestingly, in patients with CTLA‐4 haploinsuffiency, they had higher numbers of Treg cells but their individual expression of CTLA‐4 was reduced, especially after activation 59, 60. Hence, CTLA‐4 haploinsufficiency could be considered as a partial CTLA‐4‐related Treg functional deficiency. Additionally, the unintended manifestations of blocking CTLA‐4 have recently been demonstrated in humans with cancer who are receiving anti‐CTLA‐4 checkpoint blockade therapy 62. These therapies work by boosting effector T cell activity and inhibiting Treg cells; however, pharmacovigilance data suggest that some patients develop enteropathy and colitis similar to that of inflammatory bowel disease. It is important to understand why these patients specifically have effector T cells targeting and infiltrating the gut, as the anti‐CTLA‐4 antibody itself is systemically administered.
There is also another rare genetic disorder, which leads instead to a complete CTLA‐4‐related functional deficiency. This involves a deficiency in lipopolysaccharide‐responsive and beige‐like anchor protein (LRBA), which is an intracellular protein involved in the membrane expression of CTLA‐4 63. This condition is clinically characterized by a systemic autoimmune syndrome that resembles CTLA‐4 haploinsufficiency as well as IPEX (as some patients develop T1D). However, LRBA deficiency onsets in early infancy and, indeed, in a few patients shortly after birth 64.
Similarly, mild reduction in CTLA‐4 expression has also been observed in Treg cells isolated from patients with rheumatoid arthritis (RA). While the CTLA‐4 gene is highly demethylated in normal Treg cells (thus indicating stable CTLA‐4 expression) 65, the CTLA‐4 promoter was instead methylated in Treg cells from RA patients 66.
All the above findings are also important from an age‐related perspective; in comparison to LRBA deficiency, which onsets in early infancy, the onset of RA is usually in adults (older than 45–50 years) 67. Hence, by comparing patients with RA, CTLA‐4 haploinsufficiency, LRBA insufficiency and IPEX we can study the relationship between the intensity of CTLA‐4‐related functional deficiency on Treg cells, the effect on Treg biology and the extent of any clinical presentation (including its severity and time of onset) (Fig. 2).
Figure 2.

Relationship between the intensity of regulatory T cell (Treg) functional deficiency and the severity and time to onset of the disease. By comparing patients with rheumatoid arthritis (RA), cytotoxic T lymphocyte antigen 4 (CTLA‐4) haploinsufficiency, lipopolysaccharide‐responsive and beige‐like anchor protein (LRBA) insufficiency and immunodysregulation polyendocrinopathy enteropathy X‐linked (IPEX), we can study the relationship between the intensity of CTLA‐4 related functional deficiency on Treg cells, the effect on Treg biology and the extent of any clinical presentation (including its severity and time of onset).
Finally, although Treg functional deficiencies have been described in numerous other autoimmune diseases, the detailed molecular mechanism(s) responsible for these deficiencies is/are currently unknown 68. For example, impaired Treg suppression has been described in multiple sclerosis (MS) 69. It is indeed noteworthy that a key mechanism of suppression by Treg cells is related to their high expression of CD25, as Treg cells act as a sink for IL‐2 2, 70, 71. Interestingly, a polymorphism in CD25 has been associated with a high risk for developing MS through a genome‐wide association study (GWAS) 72. Therefore, it is plausible that a CD25‐related mechanism of suppression may be altered in the Treg cells of MS patients. These Treg cells may also be unstable, as loss of CD25 on Treg cells is known to alter FOXP3 expression, Treg cell function and precipitate Th17 effector cell differentiation 73. In parallel with RA patients who have a mild deficiency in CTLA‐4, the extent of the defect in Treg suppression is probably mild or moderate. This is further supported by clinical observations that both RA and MS never evolve toward an IPEX‐like syndrome and rarely occur in early infancy.
Targeting FOXP3+ Treg cells for the control of autoimmune responses
As deficiencies in Treg number/function or resistance of T conventional cells from Treg‐mediated suppression are observed in most human autoimmune diseases, it seems logical to propose stable and functionally superior Treg‐based immunotherapies as a new therapeutic strategy in order to reinstate immune homeostasis. The two Treg‐based therapeutic strategies currently being clinically evaluated in autoimmunity are in‐vivo Treg expansion and infusion of in‐vitro expanded Treg cells 19, 74, 75.
From the perspective of in‐vivo expansion, low‐dose IL‐2 has been evaluated in Phases I and II trials as a therapeutic targeting human Treg cells in order to expand them in vivo within the context of autoimmunity (e.g. T1D, alopecia areata and systemic lupus) or inflammatory conditions (hepatitis C‐related cryoglobulinaemic vasculitis) 76. However, although injection of IL‐2 expands the circulating Treg cell population, it also expands effector cells such as natural killer (NK) cells or eosinophils—thus indicating the lack of Treg specificity 77, 78. This has been overcome through the development of an IL‐2/anti‐IL‐2 complex that can specifically promote the binding of IL‐2 to the high‐affinity receptor of IL‐2 that is expressed by activated Treg cells and promote Treg cell expansion in vivo without modifying other effector cells 79, 80, 81.
A second approach is the autologous expansion of Treg cells in vitro in order to reinfuse a large number of Treg cells into patients. Due to the recognized potential for reduced FOXP3 expression or reduced immunosuppressive capacity upon in‐vitro expansion, it is important to culture the right cell subpopulation in conditions that favour maintenance of Treg phenotype and function 82. This will help to optimize any expansion protocols and more reliably predict the phenotype of the end product 83. The addition of rapamycin to the culture conditions is important to eliminate contaminating conventional effector cells. Finally, we also consider it important to utilize molecules capable of modifying Treg epigenetics (e.g. DNA methyltransferase inhibitors or vitamin C) 37, 84, 85. These molecules work to maintain FOXP3 expression as well as various other Treg‐specific demethylation patterns, which consequently lead to more stable and functional Treg cells 37, 65, 85.
Strategies aiming at increasing the number of autologous Treg cells are suitable for diseases with reduced but fully functional Treg cells. However, it is currently unknown in diseases with deficiencies in Treg function whether the deficiency is present within the entire Treg population (i.e. all Treg cells are impaired) or specific to a distinct subset (indicating that some Treg cells would be functionally impaired while others would be fully functional). In the first case, all expanded Treg cells would be functionally impaired with no beneficial therapeutic effect. In the second case the expansion, either in vitro or in vivo, of the global pool of Treg cells would lead to the expansion of deficient Treg cells and of fully functional Treg cells. It could, therefore, be speculated that the number of expanded fully functional Treg cells would be sufficient to overcome and compensate the functional deficiency of the expanded deficient subset. However, it is important not to overlook the possibility that expanded dysfunctional Treg cells could convert into pathogenic cells when in a proinflammatory environment and also exacerbate disease 86. There are data demonstrating that FOXP3+ human Treg cells can start secreting IL‐17 when exposed to cytokines such as IL‐1β, ‐2, ‐6, ‐15, ‐21 and ‐23 87, 88. These IL‐17‐secreting Treg cells can subsequently lose their anti‐inflammatory function despite continuous FOXP3 expression 88.
A third approach is to utilize peripherally induced Treg cells (pTreg cells or iTreg cells if in vitro), which differentiate from naive CD4+ T cells in the presence of transforming growth factor (TGF)‐β and IL‐2 89. One advantage of these cells is the potential to generate antigen‐specific subsets corresponding to the antigens key to the immunopathogenesis of different autoimmune diseases. However, the partially demethylated nature of FOXP3 gives rise to the instability of FOXP3 expression and subsequent loss of suppressive function 89. The in‐vivo stability of human iTreg cells within a proinflammatory microenvironment needs to be optimized if they are to be considered as a safe and non‐pathogenic clinical product.
In diseases with functional Treg deficiencies, infusion of autologous Treg cells would be feasible if the impaired molecular mechanism of suppression is identified and corrected by the use of small molecules in vitro or via genetic modifications. An alternative strategy could be the infusion of allogeneic expanded Treg cells sourced from cord blood or other healthy donors 90. Another strategy is to give those patients therapeutics that can compensate for the impaired Treg mechanism; e.g. in patients with CTLA‐4 deficiency, the use of CTLA‐4‐Ig has proved effective in preventing autoimmune events 63.
As discussed above, Treg‐based strategies are already being clinically evaluated in some human autoimmune diseases with reported deficiencies in Treg cell numbers 78, 91 (and even in Treg functions) 92, with the assumption that increasing their number would ameliorate or even cure the diseases. The first in‐human trial evaluating Treg cells in autoimmunity was conducted in the setting of T1D 92. Fourteen patients received expanded autologous polyclonal Treg cells (CD25+CD127lo) in a dose‐escalation study (from 0·5 to 26 × 108 cells). The reliability of the expansion process was demonstrated by the purity of the final product, 76–96·9% FOXP3+. Although two patients had serious adverse events of severe hypoglycaemia and ketoacidosis, no directly Treg‐related adverse events were reported. This study was not powered for disease‐specific outcomes, as it was a Phase 1 study—hence, results of future Phase 2 studies are awaited. Further novel work using Treg cells is also ongoing in the contexts of graft‐versus‐host disease and solid organ transplantation 93, 94, 95, 96.
Treg cells are capable of inhibiting the initiation of immune responses, although the evidence regarding their ability to control active autoimmune/inflammatory disease is more controversial 2, 6, 7, 74. There are in‐vitro data demonstrating that Treg cells cannot inhibit the proliferation of preactivated effector T cells 97, and when transferred into mice after pathogenic cells the Treg cells are incapable of preventing the onset of autoimmunity 2 (Fig. 3). Indeed, this resistance of effector T cells to Treg‐mediated suppression is another key mechanism in autoimmunity. The effector T cells are supported by a signalling pleiotrophic microenvironment consisting of TNF, IL‐6, IL‐1β as well as over‐activated intracellular signalling via the PI3K/Akt pathways 29. Additionally, in another murine model of severe colitis, the progression of this disease was ameliorated and reversed when the mice underwent adoptive transfer of Treg cells 7. This effect was inhibited when mice were administered antibodies to IL‐10, CTLA‐4 or TGF‐β. Together, these data suggest that it is not only Treg cells but also the presence of particular cytokines in the microenvironment that can modulate disease progression.
Figure 3.

Timing of regulatory T cell (Treg)‐mediated suppression of immune responses. Treg cells inhibit the initiation of immune responses by preventing the activation of effector cells. One important mechanism of suppression is their ability to adsorb interleukin (IL)‐2 produced by effector cells (top). When effector cells are preactivated, Treg cells are not capable of suppressing the ongoing immune responses. In this case, IL‐2 is massively produced and amplifies the activated effector cell pool and also the Treg cells. They can still suppress some non‐activated cells present in the vicinity by bystander suppression (bottom). Mild beneficial therapeutic effects observed in active diseases treated with Treg biology‐based therapies can be explained by the absence of suppression of activated immune cells and the prevention of the activation of dormant pathogenic cells.
Hence, there was little surprise that only modest benefits are observed in trials evaluating Treg‐based therapies for ongoing autoimmune or inflammatory diseases 78, 98. However, while they are inefficient at controlling activated pathogenic cells, the expanded Treg cells could theoretically prevent the activation of resting pathogenic cells 74, 75. Therefore, some modest beneficial effect can be expected, as Treg cells would inhibit the activation of dormant pathogenic cells. This would indeed be a viable therapeutic approach, as clinical observations have identified the role of steroids and/or immunosuppressants in controlling active autoimmune diseases as they target activated effector cells first. Secondly, when the disease is considered in remission, another line of immunosuppressants and/or of immunomodulatory drugs are given to patients in order to prevent relapses or disease flare‐up 75. As Treg cells are professionally involved in the prevention of the initiation of pathogenic autoimmune responses, we believe that Treg cell‐based treatments should only be considered as a maintenance therapy for the prevention of flares or relapses after the elimination of pathogenic cells. Such strategies would, therefore, be suitable for remitting and relapsing diseases such as MS, RA or anti‐neutrophil cytoplasmic antibodies (ANCA)‐associated vasculitis only during the remission period to prevent relapses.
These trials have also not addressed concerning whether or not it is necessary for infused Treg cells in humans to migrate to the diseased tissue in order to prevent further flares/relapses. This is important in the context of autoimmunity, as although patients have a focal site of inflammation (e.g. joints in RA, gut in inflammatory bowel disease), they also have pathology elsewhere (e.g. extra‐articular/intestinal manifestations) 99, 100. Thus, in order to optimize the Treg therapeutic effect and dose, it may be necessary to culture Treg cells that pre‐emptively express tissue‐specific homing markers (if not, induce this phenotype genetically or pharmacologically). This is also important to minimize any off‐target effects such as the risk of malignancy from interference with anti‐tumour immunity.
Interestingly, the Treg‐based therapeutic strategies available in humans aim at expanding polyclonal Treg cells, without considering their antigen specificity 74, 75. Studies in mice indicate that antigen‐specific Treg cells are more efficient that polyclonal Treg cells in preventing autoimmune diseases. However, if the activation of Treg cells is dependent on the target antigen of their T cell receptors (TCRs), the suppressive function itself is not only antigen‐specific. Once activated, Treg cells can suppress effector T cells with the same or any other antigen specificity (via bystander suppression) 48. However, as Treg cells expanded in vitro for cell therapy are highly activated, it may not be necessary to take into account antigen‐specificity of Treg cells to obtain beneficial results in the settings of human autoimmune diseases.
Conclusions
From the data discussed so far, it is clear that abnormalities in the quantity or function of Treg cells are observed in most, if not all, human autoimmune and/or inflammatory diseases. Although numerous Treg subsets have been defined, their individual contributions to human autoimmune and/or inflammatory diseases are largely unknown. The clinical observations of genetic diseases involving deficiencies in Treg function indicate that the severity and the age of disease onset correlate to the depth of Treg functional impairment.
Treg biology‐based therapies may not be suitable in patients with deficits of Treg function, unless their deficit can be corrected in vivo/in vitro. It is also critical to consider the appropriate stage of autoimmune diseases whereby administration of Treg cellular therapy can be effective. As highlighted, there are conflicting data regarding whether Treg cells are more effectual at preventing the initiation of autoimmunity, ameliorating disease progression or curing autoimmunity itself. This is because, although Treg cells can prevent the initiation of autoimmune responses, they cannot terminate ongoing responses.
We therefore propose here a global sequential therapeutic strategy for autoimmune diseases that includes (1) induction treatments for diseases flares or chronic disease with chronic activity and (2) maintenance treatments that are suitable for diseases during remission phases by utilizing Treg cell biology‐derived therapies to prevent relapses or subsequent flares (Fig. 4).
Figure 4.

Therapeutic strategies for autoimmune diseases with regulatory T cell (Treg) biology‐based treatments. The current gold standard treatment regimen for autoimmune diseases includes an induction phase, which aims to eliminate pathogenic cells. Usually, high‐dose steroids are given intravenously during the first days, followed by oral steroids with a tapering scheme. Immunosuppressants such as cyclophosphamide, mycophenolate or rituximab are also necessary to induce remission. When remission is obtained, it is necessary to maintain mild immunosuppressants such as azathioprine or mycophenolate to prevent the occurrence of subsequent flares or relapses of the disease. In diseases with number deficiency of functional Treg cells, autologous Treg cells infusion or interleukin (IL)‐2/anti‐IL‐2 complexes can be used as a maintenance treatment when the disease is controlled by standard induction treatments. In diseases with dysfunctional Treg cells, if the dysfunctional mechanism of suppression is identified molecules mimicking the Treg function, genetically corrected autologous Treg cells or allogeneic or cord blood Treg cells can be infused as maintenance treatment.
In patients with reduced Treg numbers (but no functional deficiency), the expansion of autologous Treg cells could be a suitable therapeutic approach (either infusion of in‐vitro‐expanded autologous cells, infusion of IL‐2/anti‐IL‐2 complex, or both). In contrast, patients with diseases involving deficiencies of Treg function would benefit from a detailed understanding of the impaired mechanisms of action of their Treg cells. We anticipate the development of suitable therapeutics to correct/reduce the severity of their Treg deficiencies and thereby reduce their disease burden. Another feasible therapeutic option would be to administer functional allogeneic expanded Treg cells.
Disclosures
None declared.
Acknowledgements
M. A. is funded by the EASL Juan Rodes PhD Fellowship. The work is supported by the PHRC programme (AOR17082) and by AFPCA (Association Française de la PolyChondrite Atrophiante).
OTHER ARTICLES PUBLISHED IN THIS REVIEW SERIES
Regulatory T cells: exploring mechanisms for future therapies. Clinical and Experimental Immunology 2019, 197: 11–13.
From stability to dynamics: understanding molecular mechanisms of regulatory T cells through Foxp3 transcriptional dynamics. Clinical and Experimental Immunology 2019, 197: 14–23.
Mechanisms of human FoxP3+ Treg cell development and function in health and disease. Clinical and Experimental Immunology 2019, 197: 36–51.
Methods to manufacture regulatory T cells for cell therapy. Clinical and Experimental Immunology 2019, 197: 52–63.
References
- 1. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008; 133:775–87. [DOI] [PubMed] [Google Scholar]
- 2. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self‐tolerance maintained by activated T cells expressing IL‐2 receptor alpha‐chains (CD25). Breakdown of a single mechanism of self‐tolerance causes various autoimmune diseases. J Immunol 1995; 155:1151–64. [PubMed] [Google Scholar]
- 3. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003; 299:1057–61. [DOI] [PubMed] [Google Scholar]
- 4. Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nat Immunol 2003; 4:337–42. [DOI] [PubMed] [Google Scholar]
- 5. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003; 4:330–6. [DOI] [PubMed] [Google Scholar]
- 6. Nguyen TL, Makhlouf NT, Anthony BA, Teague RM, DiPaolo RJ. In vitro induced regulatory T cells are unique from endogenous regulatory T cells and effective at suppressing late stages of ongoing autoimmunity. PLOS ONE 2014; 9:e104698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Liu H, Hu B, Xu D, Liew FY. CD4+CD25+ regulatory T cells cure murine colitis: the role of IL‐10, TGF‐beta, and CTLA4. J Immunol 2003; 171:5012–7. [DOI] [PubMed] [Google Scholar]
- 8. Palomares O, Yaman G, Azkur AK, Akkoc T, Akdis M, Akdis CA. Role of Treg in immune regulation of allergic diseases. Eur J Immunol 2010; 40:1232–40. [DOI] [PubMed] [Google Scholar]
- 9. La Rocca C, Carbone F, Longobardi S, Matarese G. The immunology of pregnancy: regulatory T cells control maternal immune tolerance toward the fetus. Immunol Lett 2014; 162:41–8. [DOI] [PubMed] [Google Scholar]
- 10. Afzali B, Lombardi G, Lechler RI, Lord GM. The role of T helper 17 (Th17) and regulatory T cells (Treg) in human organ transplantation and autoimmune disease. Clin Exp Immunol 2007; 148:32–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Nishikawa H, Sakaguchi S. Regulatory T cells in tumor immunity. Int J Cancer 2010; 127:759–67. [DOI] [PubMed] [Google Scholar]
- 12. Baecher‐Allan C, Brown JA, Freeman GJ, Hafler DA. CD4+CD25high regulatory cells in human peripheral blood. J Immunol 2001; 167:1245–53. [DOI] [PubMed] [Google Scholar]
- 13. Stephens LA, Mottet C, Mason D, Powrie F. Human CD4(+)CD25(+) thymocytes and peripheral T cells have immune suppressive activity in vitro. Eur J Immunol 2001; 31:1247–54. [DOI] [PubMed] [Google Scholar]
- 14. Taams LS, Smith J, Rustin MH, Salmon M, Poulter LW, Akbar AN. Human anergic/suppressive CD4(+)CD25(+) T cells: a highly differentiated and apoptosis‐prone population. Eur J Immunol 2001; 31:1122–31. [DOI] [PubMed] [Google Scholar]
- 15. Dieckmann D, Plottner H, Berchtold S, Berger T, Schuler G. Ex vivo isolation and characterization of CD4(+)CD25(+) T cells with regulatory properties from human blood. J Exp Med 2001; 193:1303–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Levings MK, Sangregorio R, Roncarolo MG. Human CD25(+)CD4(+) t regulatory cells suppress naive and memory T cell proliferation and can be expanded in vitro without loss of function. J Exp Med 2001; 193:1295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Jonuleit H, Schmitt E, Stassen M, Tuettenberg A, Knop J, Enk AH. Identification and functional characterization of human CD4(+)CD25(+) T cells with regulatory properties isolated from peripheral blood. J Exp Med 2001; 193:1285–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Ng WF, Duggan PJ, Ponchel F et al Human CD4(+)CD25(+) cells: a naturally occurring population of regulatory T cells. Blood 2001; 98:2736–44. [DOI] [PubMed] [Google Scholar]
- 19. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 2010; 10:490–500. [DOI] [PubMed] [Google Scholar]
- 20. Walker MR, Kasprowicz DJ, Gersuk VH, et al Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25– T cells. J Clin Invest 2003; 112:1437–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Morgan ME, van Bilsen JH, Bakker AM et al Expression of FOXP3 mRNA is not confined to CD4+CD25+ T regulatory cells in humans. Hum Immunol 2005; 66:13–20. [DOI] [PubMed] [Google Scholar]
- 22. RodrÃguez‐Perea AL, Arcia ED, Rueda CM, Velilla PA. Phenotypical characterization of regulatory T cells in humans and rodents. Clin Exp Immunol 2016; 185:281–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Miyara M, Yoshioka Y, Kitoh A et al Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009; 30:899–911. [DOI] [PubMed] [Google Scholar]
- 24. Cuadrado E, van den Biggelaar M, de Kivit S et al Proteomic analyses of human regulatory T cells reveal adaptations in signaling pathways that protect cellular identity. Immunity 2018; 48:1046–59 e6. [DOI] [PubMed] [Google Scholar]
- 25. Miyara M, Amoura Z, Parizot C et al The immune paradox of sarcoidosis and regulatory T cells. J Exp Med 2006; 203:359–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Najafi M, Farhood B, Mortezaee K. Contribution of regulatory T cells to cancer: a review. J Cell Physiol 2019; 234: 7983–93. 10.1002/jcp.27553. [DOI] [PubMed] [Google Scholar]
- 27. Miyara M, Chader D, Sage E et al Sialyl Lewis x (CD15s) identifies highly differentiated and most suppressive FOXP3high regulatory T cells in humans. Proc Natl Acad Sci USA 2015; 112:7225–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Saito T, Nishikawa H, Wada H et al Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016; 22:679–84. [DOI] [PubMed] [Google Scholar]
- 29. Mercadante ER, Lorenz UM. Breaking free of control: how conventional T cells overcome regulatory T cell suppression. Front Immunol 2016; 7:193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Miyara M, Gorochov G, Ehrenstein M, Musset L, Sakaguchi S, Amoura Z. Human FoxP3+ regulatory T cells in systemic autoimmune diseases. Autoimmun Rev 2011; 10:744–55. [DOI] [PubMed] [Google Scholar]
- 31. Long SA, Buckner JH. CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. J Immunol 2011; 187:2061–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Dominguez‐Villar M, Hafler DA. Regulatory T cells in autoimmune disease. Nat Immunol 2018; 19:665–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Miyara M, Sakaguchi S. Human FoxP3(+)CD4(+) regulatory T cells: their knowns and unknowns. Immunol Cell Biol 2011; 89:346–51. [DOI] [PubMed] [Google Scholar]
- 34. Mohr A, Malhotra R, Mayer G, Gorochov G, Miyara M. Human FOXP3(+) T regulatory cell heterogeneity. Clin Transl Immunology 2018; 7:e1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Fritzsching B, Oberle N, Pauly E et al Naive regulatory T cells: a novel subpopulation defined by resistance toward CD95L‐mediated cell death. Blood 2006; 108:3371–8. [DOI] [PubMed] [Google Scholar]
- 36. Kunicki MA, Amaya Hernandez LC, Davis KL, Bacchetta R, Roncarolo MG. Identity and diversity of human peripheral Th and T regulatory cells defined by single‐cell mass cytometry. J Immunol 2018; 200:336–46. [DOI] [PubMed] [Google Scholar]
- 37. Miyara M, Chader D, Burlion A et al Combination of IL‐2, rapamycin, DNA methyltransferase and histone deacetylase inhibitors for the expansion of human regulatory T cells. Oncotarget 2017; 8:104733–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Levine AG, Mendoza A, Hemmers S et al Stability and function of regulatory T cells expressing the transcription factor T‐bet. Nature 2017; 546:421–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Zheng Y, Chaudhry A, Kas A et al Regulatory T‐cell suppressor program co‐opts transcription factor IRF4 to control T(H)2 responses. Nature 2009; 458:351–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Chaudhry A, Rudra D, Treuting P et al CD4+ regulatory T cells control TH17 responses in a Stat3‐dependent manner. Science 2009; 326:986–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Duhen T, Duhen R, Lanzavecchia A, Sallusto F, Campbell DJ. Functionally distinct subsets of human FOXP3+ Treg cells that phenotypically mirror effector Th cells. Blood 2012; 119:4430–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Sage PT, Sharpe AH. T follicular regulatory cells. Immunol Rev 2016; 271:246–59. [DOI] [PubMed] [Google Scholar]
- 43. Panduro M, Benoist C, Mathis D. Tissue Tregs. Annu Rev Immunol 2016; 34:609–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Plitas G, Konopacki C, Wu K et al Regulatory T cells exhibit distinct features in human breast cancer. Immunity 2016; 45:1122–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. De Simone M, Arrigoni A, Rossetti G et al Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor‐infiltrating T regulatory cells. Immunity 2016; 45:1135–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Akimova T, Zhang T, Negorev D et al Human lung tumor FOXP3+ Tregs upregulate four ‘Treg‐locking’ transcription factors. JCI Insight 2017; 2:pii 94075. 10.1172/jci.insight.94075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Toh BH, van Driel IR, Gleeson PA. Pernicious anemia. N Engl J Med 1997; 337:1441–8. [DOI] [PubMed] [Google Scholar]
- 48. Shevach EM. Foxp3(+) T regulatory cells: still many unanswered questions – perspective after 20 years of study. Front Immunol 2018; 9:1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Grant CR, Liberal R, Mieli‐Vergani G, Vergani D, Longhi MS. Regulatory T‐cells in autoimmune diseases: challenges, controversies and‐yet‐unanswered questions. Autoimmun Rev 2015; 14:105–16. [DOI] [PubMed] [Google Scholar]
- 50. Wildin RS, Ramsdell F, Peake J et al X‐linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet 2001; 27:18–20. [DOI] [PubMed] [Google Scholar]
- 51. Bennett CL, Christie J, Ramsdell F et al The immune dysregulation, polyendocrinopathy, enteropathy, X‐linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001; 27:20–1. [DOI] [PubMed] [Google Scholar]
- 52. Brunkow ME, Jeffery EW, Hjerrild KA et al Disruption of a new forkhead/winged‐helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 2001; 27:68–73. [DOI] [PubMed] [Google Scholar]
- 53. Kim J, Lahl K, Hori S et al Cutting edge: depletion of Foxp3+ cells leads to induction of autoimmunity by specific ablation of regulatory T cells in genetically targeted mice. J Immunol 2009; 183:7631–4. [DOI] [PubMed] [Google Scholar]
- 54. Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol 2007; 8:191–7. [DOI] [PubMed] [Google Scholar]
- 55. Dittmar M, Kahaly GJ. Polyglandular autoimmune syndromes: immunogenetics and long‐term follow‐up. J Clin Endocrinol Metab 2003; 88:2983–92. [DOI] [PubMed] [Google Scholar]
- 56. Liberal R, Grant CR, Longhi MS, Mieli‐Vergani G, Vergani D. Regulatory T cells: Mechanisms of suppression and impairment in autoimmune liver disease. IUBMB Life 2015; 67:88–97. [DOI] [PubMed] [Google Scholar]
- 57. Shevach EM. Mechanisms of foxp3+ T regulatory cell‐mediated suppression. Immunity 2009; 30:636–45. [DOI] [PubMed] [Google Scholar]
- 58. Wing K, Onishi Y, Prieto‐Martin P et al CTLA‐4 control over Foxp3+ regulatory T cell function. Science 2008; 322:271–5. [DOI] [PubMed] [Google Scholar]
- 59. Kuehn HS, Ouyang W, Lo B et al Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 2014; 345:1623–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Schubert D, Bode C, Kenefeck R et al Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 2014; 20:1410–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Jago CB, Yates J, Camara NO, Lechler RI, Lombardi G. Differential expression of CTLA‐4 among T cell subsets. Clin Exp Immunol 2004; 136:463–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Mitsuiki N, Schwab C, Grimbacher B. What did we learn from CTLA‐4 insufficiency on the human immune system? Immunol Rev 2019; 287:33–49. [DOI] [PubMed] [Google Scholar]
- 63. Lo B, Zhang K, Lu W et al Patients with LRBA deficiency show CTLA4 loss and immune dysregulation responsive to abatacept therapy. Science 2015; 349:436–40. [DOI] [PubMed] [Google Scholar]
- 64. Alkhairy OK, Abolhassani H, Rezaei N et al Spectrum of phenotypes associated with mutations in LRBA. J Clin Immunol 2016; 36:33–45. [DOI] [PubMed] [Google Scholar]
- 65. Ohkura N, Hamaguchi M, Morikawa H et al T cell receptor stimulation‐induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 2012; 37:785–99. [DOI] [PubMed] [Google Scholar]
- 66. Cribbs AP, Kennedy A, Penn H, et al Treg cell function in rheumatoid arthritis is compromised by CTLA‐4 promoter methylation resulting in a failure to activate the indoleamine 2,3‐dioxygenase pathway. Arthritis Rheumatol 2014; 66:2344–54. [DOI] [PubMed] [Google Scholar]
- 67. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med 2011; 365:2205–19. [DOI] [PubMed] [Google Scholar]
- 68. Miyara M, Sakaguchi S. Natural regulatory T cells: mechanisms of suppression. Trends Mol Med 2007; 13:108–16. [DOI] [PubMed] [Google Scholar]
- 69. Viglietta V, Baecher‐Allan C, Weiner HL, Hafler DA. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med 2004; 199:971–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Pandiyan P, Zheng L, Ishihara S, Reed J, Lenardo MJ. CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation‐mediated apoptosis of effector CD4+ T cells. Nat Immunol 2007; 8:1353–62. [DOI] [PubMed] [Google Scholar]
- 71. Chinen T, Kannan AK, Levine AG et al An essential role for the IL‐2 receptor in Treg cell function. Nat Immunol 2016; 17:1322–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Hafler DA, Compston A, Sawcer S et al Risk alleles for multiple sclerosis identified by a genomewide study. N Engl J Med 2007; 357:851–62. [DOI] [PubMed] [Google Scholar]
- 73. Komatsu N, Okamoto K, Sawa S et al Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat Med 2014; 20:62–8. [DOI] [PubMed] [Google Scholar]
- 74. Miyara M, Wing K, Sakaguchi S. Therapeutic approaches to allergy and autoimmunity based on FoxP3+ regulatory T‐cell activation and expansion. J Allergy Clin Immunol 2009; 123:749–55; quiz 56–7. [DOI] [PubMed] [Google Scholar]
- 75. Miyara M, Ito Y, Sakaguchi S. TREG‐cell therapies for autoimmune rheumatic diseases. Nat Rev Rheumatol 2014; 10:543–51. [DOI] [PubMed] [Google Scholar]
- 76. Arenas‐Ramirez N, Woytschak J, Boyman O. Interleukin‐2: biology, design and application. Trends Immunol 2015; 36:763–77. [DOI] [PubMed] [Google Scholar]
- 77. Van Gool F, Molofsky AB, Morar MM et al Interleukin‐5‐producing group 2 innate lymphoid cells control eosinophilia induced by interleukin‐2 therapy. Blood 2014; 124:3572–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Saadoun D, Rosenzwajg M, Joly F et al Regulatory T‐cell responses to low‐dose interleukin‐2 in HCV‐induced vasculitis. N Engl J Med 2011; 365:2067–77. [DOI] [PubMed] [Google Scholar]
- 79. Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T cell subsets with antibody–cytokine immune complexes. Science 2006; 311:1924–7. [DOI] [PubMed] [Google Scholar]
- 80. Dooms H, Abbas AK. Revisiting the role of IL‐2 in autoimmunity. Eur J Immunol 2010; 40:1538–40. [DOI] [PubMed] [Google Scholar]
- 81. Ward NC, Yu A, Moro A et al 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 2018; 201:2579–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Hoffmann P, Boeld TJ, Eder R et al Loss of FOXP3 expression in natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stimulation. Eur J Immunol 2009; 39:1088–97. [DOI] [PubMed] [Google Scholar]
- 83. Hoffmann P, Eder R, Boeld TJ et al Only the CD45RA+ subpopulation of CD4+CD25high T cells gives rise to homogeneous regulatory T‐cell lines upon in vitro expansion. Blood 2006; 108:4260–7. [DOI] [PubMed] [Google Scholar]
- 84. Polansky JK, Kretschmer K, Freyer J et al DNA methylation controls Foxp3 gene expression. Eur J Immunol 2008; 38:1654–63. [DOI] [PubMed] [Google Scholar]
- 85. Nikolouli E, Hardtke‐Wolenski M, Hapke M et al Alloantigen‐induced regulatory T cells generated in presence of vitamin C display enhanced stability of Foxp3 expression and promote skin allograft acceptance. Front Immunol 2017; 8:748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Yang XO, Nurieva R, Martinez GJ et al Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 2008; 29:44–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Koenen HJ, Smeets RL, Vink PM, van Rijssen E, Boots AM, Joosten I. Human CD25highFoxp3pos regulatory T cells differentiate into IL‐17‐producing cells. Blood 2008; 112:2340–52. [DOI] [PubMed] [Google Scholar]
- 88. Beriou G, Costantino CM, Ashley CW et al IL‐17‐producing human peripheral regulatory T cells retain suppressive function. Blood 2009; 113:4240–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Shevach EM, Thornton AM. tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev 2014; 259:88–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Brunstein CG, Miller JS, Cao Q et al Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2011; 117:1061–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Mizui M, Tsokos GC. Low‐dose IL‐2 in the treatment of lupus. Curr Rheumatol Rep 2016; 18:68. [DOI] [PubMed] [Google Scholar]
- 92. Bluestone JA, Buckner JH, Fitch M et al Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015; 7:315ra189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Di Ianni M, Falzetti F, Carotti A et al Tregs prevent GVHD and promote immune reconstitution in HLA‐haploidentical transplantation. Blood 2011; 117:3921–8. [DOI] [PubMed] [Google Scholar]
- 94. Trzonkowski P, Bieniaszewska M, Juscinska J et al First‐in‐man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127– T regulatory cells. Clin Immunol 2009; 133:22–6. [DOI] [PubMed] [Google Scholar]
- 95. Mathew JM, Jessica H, LeFever A et al A phase I clinical trial with ex vivo expanded recipient regulatory T cells in living donor kidney transplants. Sci Rep 2018; 8:7428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Tang Q, Vincenti F. Transplant trials with Tregs: perils and promises. J Clin Invest 2017; 127:2505–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Baecher‐Allan C, Viglietta V, Hafler DA. Inhibition of human CD4(+)CD25(+high) regulatory T cell function. J Immunol 2002; 169:6210–7. [DOI] [PubMed] [Google Scholar]
- 98. Long SA, Rieck M, Sanda S et al Rapamycin/IL‐2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs beta‐cell function. Diabetes 2012; 61:2340–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Cojocaru M, Cojocaru IM, Silosi I, Vrabie CD, Tanasescu R. Extra‐articular manifestations in rheumatoid arthritis. Maedica (Buchar) 2010; 5:286–91. [PMC free article] [PubMed] [Google Scholar]
- 100. Vavricka SR, Schoepfer A, Scharl M, Lakatos PL, Navarini A, Rogler G. Extraintestinal manifestations of inflammatory bowel disease. Inflamm Bowel Dis 2015; 21:1982–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
