Skip to main content

Tipping the balance in autoimmunity: are regulatory t cells the cause, the cure, or both?

Abstract

Regulatory T cells (Tregs) are a specialized subgroup of T-cell lymphocytes that is crucial for maintaining immune homeostasis and preventing excessive immune responses. Depending on their differentiation route, Tregs can be subdivided into thymically derived Tregs (tTregs) and peripherally induced Tregs (pTregs), which originate from conventional T cells after extrathymic differentiation at peripheral sites. Although the regulatory attributes of tTregs and pTregs partially overlap, their modes of action, protein expression profiles, and functional stability exhibit specific characteristics unique to each subset. Over the last few years, our knowledge of Treg differentiation, maturation, plasticity, and correlations between their phenotypes and functions has increased. Genetic and functional studies in patients with numeric and functional Treg deficiencies have contributed to our mechanistic understanding of immune dysregulation and autoimmune pathologies. This review provides an overview of our current knowledge of Treg biology, discusses monogenetic Treg pathologies and explores the role of Tregs in various other autoimmune disorders. Additionally, we discuss novel approaches that explore Tregs as targets or agents of innovative treatment options.

Background

The immune system is a complex network of cells and molecules that defends the body against harmful pathogens while maintaining self-tolerance. This delicate balance is orchestrated by various cell types, including T cells, which play a central role in adaptive immunity. Regulatory T cells (Tregs) are a specialized subset of T lymphocytes that play a pivotal role in maintaining immune system homeostasis and preventing excessive immune responses such as autoimmune diseases. More than four decades ago, Tregs emerged as a cornerstone of immunological research. Tregs encompass a heterogeneous population of cells with varying origins and functions. Functionally, Tregs constitute the physiological counterplayers of conventional or cytotoxic T cells and crucially contribute to the maintenance of peripheral immune tolerance [1]. Since their identification by Sakaguchi et al. in 1995, regulatory T cells have grown into a large and complex family of regulatory cell populations. Among these, thymically derived Tregs (tTregs), which develop within the thymus before being released into the periphery, represent the majority of peripheral FoxP3 + Tregs. In contrast, peripherally induced Tregs (pTregs) develop from mature conventional FoxP3CD4+ T cells upon continuous antigen stimulation in peripheral tissues. During this process, conventional T cells acquire regulatory properties directed by multiple factors, including the presence of certain cytokines and the formation of cellular synapses between various immune cells. This duality highlights the dynamic nature of Treg development and its adaptability to various immunological contexts. Within this review we will summarize biology and functions of Tregs and present the current understand of Tregs deficiencies in monogenetic immunodeficiency and multifactorial autoimmune diseases. Additionally, we will discuss novel therapeutic approaches using Tregs as target or agent to overcome currently unmet medical needs.

Treg biology and function

The functional characteristics of tTregs and pTregs overlap but differ in terms of their stability. pTregs show a high plasticity and exert regulatory functions only temporarily by transient expression of FoxP3 and additional regulatory elements, which induce the formation of regulatory cytokines [2, 3]. In contrast, tTregs express high levels of FoxP3 [4] and IL2R alpha chain CD25, but low levels of IL-7 receptor CD127. These characteristic elements are critical for the development, function, and homeostasis of tTregs and are tightly linked to their regulatory stability irrespective of the immunologic milieu [5, 6]. However, despite substantial efforts and the discussion of various promising candidates, a phenotypic marker or marker combination that is uniquely expressed by tTregs or allows the discrimination between tTregs and pTregs has not yet been identified [7].

In humans, Tregs constitute only 3–10% of the naïve peripheral CD4+ T-cell population. During embryogenesis, Tregs are present within the thymus at 12 gestational weeks and remain stable throughout pregnancy and infancy [8]. Fetal tTregs already express FoxP3 and other markers characteristically linked to their early established immunosuppressive phenotype, e. g. the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and glucocorticoid-induced TNFR-related protein (GITR) [8, 9]. To protect the human body from autoimmunity, tTregs possess a T-cell receptor (TCR) with a specific affinity for autoantigens [10]. TCR-dependent maturation is mediated by the thymic selection process of tTregs, which focuses on self-protection through the presentation of autoantigens. The presentation of various self-peptides, the so-called tissue-specific antigens (TSA), in medullary thymic epithelial cells (mTECs) is regulated by the transcription factor AIRE (Autoimmune Regulator) and the zinc finger protein Fezf2 [11, 12]. T-cell selection and maturation in a TSA-rich environment ensures immunological self-tolerance. Only T cells bearing TCRs with an intermediate affinity for self-peptides differentiate into tTregs. In contrast, T cells are deleted if they recognize self-peptides with a high-affinity TCR or differentiate into naïve CD4+ T cells if self-peptides are recognized with low-affinity TCRs [7, 13,14,15,16]. This leads to the effect that pTreg TCRs have a low affinity towards self-antigens but a high affinity for foreign antigens, for example, microbial structures. Sequencing analysis of tTregs, pTregs, and conventional T-cell populations revealed that clonal overlap between these populations is particularly low [17,18,19,20]. The close link between thymic maturation and stable expression of FoxP3 has been demonstrated to result from a unique pattern of DNA demethylation within an enhancer element of the FoxP3 promoter region (regulatory T-cell–specific demethylated region [TSDR]) and activation of histone modifications [21,22,23,24]. However, even before FoxP3 is functionally expressed, Treg-specific super-enhancers and additional tTreg signature genes, including CTLA-4, IL2RA, which encodes the IL-2 receptor CD25, and IKZF2, encoding the transcription factor Helios and IFZF4, encoding Eos, are activated in Treg progenitor cells [25]. During this process, Satb1, a genome stabilizer, binds to specific genomic sites and supports the opening of chromatin and activation of super-enhancers. Together with the histone lysine methyltransferase MLL4 [26], Satb1 binds to conserved enhancer regions within the FoxP3 promoter, such as CNS0, CNS3, and CNS2, and induces stable activation of these enhancers, as well as the FoxP3 promoter itself [25, 27, 28]. After thymic release, tTregs reside within lymph nodes and peripheral blood. Continuous recognition of self-antigens maintains tTregs in a highly proliferative state and mediates physiological immune homeostasis. Expression of CCR7 and CD62L in naive tTregs and enables Treg homing to the secondary lymphoid organs [29]. There, tTregs—similar to other T-cell subsets—undergo peripheral maturation from naïve (TN CD45RA+CCR7+) to central memory (TCM, CD45RACCR7+), effector memory (TEM, CD45RACCR7) and finally CD45RA expressing terminally differentiated effector memory cells (TEMRA, CD45RA+CCR7) [30,31,32,33,34]. Tregs further employ a broad range of chemokine receptors and adhesion molecules for recruitment to inflammatory sites. The release of attractive chemokines at these sites induces Treg migration along a chemotactic gradient. CCR2, CCR4, and CCR5, and particularly CXCR3, CCR6, and CCR8, support recruitment towards sites dominated by Th1, Th2, and Th17 inflammation [35, 36]. The inflammatory response and recruitment of Tregs are further supported by other T-cell subsets and macrophages through the release of IL-2, IL-35, or TGF-β. These not only enhance the recruitment, function, and survival of Tregs, but also support the polarization of naïve CD4+ T cells towards pTregs [2]. Conversely, immunosuppressive molecules, including IL-10, IL-35, and TGF-β, are induced in Tregs, which themselves promote crucial survival signals to sustain Tregs in peripheral tissues and mediate non-specific anti-inflammatory signals [37].

Both tTregs and pTregs act as inhibitory immunomodulators through several cell–cell contact-dependent and -independent mechanisms, including inhibition of effector cell proliferation, targeted T-cell cytolysis, ATP consumption (metabolic disruption), and alteration of antigen presentation by macrophages and dendritic cells Fig. 1. Although cytotoxicity is characteristically attributed to conventional CD8+ T cells, Tregs have been observed to use Granzyme B and Perforin-mediated cytolysis of target T cells as additional MHC/TCR-independent mechanisms to control inflammation [38,39,40]. Activation induced expression of CTLA-4 orchestrates broad antigen-specific suppressive functions of Tregs in a contact-dependent fashion [37, 41]. CTLA-4 competitively binds to CD80/CD86 on APCs, including B cells and dendritic cells [37, 42] and thereby reducing the CD28-mediated co-stimulation of conventional T-cells (Tconv). CLTA-4 binding to DCs further reduces the density of antigen-specific MHC-II and CD80/86 on DC via trans-endocytosis [43,44,45,46]. Thus, DC lose their capability for MHC-II-mediated antigen-specific activation and T-cell co-stimulation. Antigen presentation on DCs is further reduced by CTLA-4 mediated induction of indoleamine 2,3-dioxygenase (IDO) in DCs. IDO leads to tryptophan depletion and accumulation of the tryptophan metabolite kynurenine, which (i) mediates suppression of proliferation and activation of effector T cells and (ii) promotes differentiation of other immune cells towards a regulatory phenotype [47]. Accordingly, monocytic differentiation is shifted towards anti-inflammatory M2 macrophages, while differentiation to pro-inflammatory M1 macrophages and Th17-cell expansion is inhibited [48,49,50]. The expression of additional inhibitory surface molecules further contributes to the contact-dependent inhibition of T cells, B-cell and DCs. Among these, PD-1/PD-1L interaction induces phosphorylation of ITSM, which downmodulates intracellular cascades including TCR-, PI3K/AKT-, and Ras/MEK/ERK-signaling and represses T- and B-cell proliferation [51,52,53,54]. Additional metabolic pathways used by Tregs not only induce peripheral Treg differentiation and proliferation but also deprive effector T cells of key nutrients, including the degradation of ATP to adenosine by CD39/CD73 and the competitive consumption of key nutrients such as glucose or amino acids [55, 56]. While all T cells express dimeric IL-2 receptors consisting of the γ-chain CD132 and IL2Rβ subunit CD122 with an intermediate IL-2 affinity, Tregs are characterized by the additional and constitutive expression of the high-affinity IL2Rα subunit CD25. This trimeric receptor is characterized by an ca. 1000-fold higher affinity for IL-2. Thus, the expression of CD25 confers a selection advantage during thymic Treg differentiation and induces FoxP3 expression early during Treg development. The phenotypic characteristics of particularly high CD25 expression further correlate with the exertion of regulatory functions. In the periphery, Treg survival and proliferation are strictly dependent on IL-2 produced by activated conventional T cells. Intracellularly, IL-2R signaling in Tregs has recently been shown to be crucially involved in the JAK-STAT5 pathway. Activated transcription factor STAT5 translocates to the nucleus and mediates the induction of CD25 and FoxP3. In addition to the crucial role of CD25 in Treg differentiation and maintenance, extracellular CD25 mediated consumption of IL-2 adds to the key repertoire of Treg mechanisms for suppressing CD8+ T-cell proliferation [57]. In contrast, inhibition of CD4+ T cells has recently been shown to require IL2R mediated activation of Tregs but to occur independently of extracellular IL-2 deprivation [57]. The situation in pTregs however is different as pTregs may modulate their phenotype and function in response to the microenvironment. In a Th1-mediated milieu, which is characterized by the release of IFN-γ, the expression of the transcription factor T-bet is induced in Tregs. Expression of T-bet (T-box expressed in T cells, also called T-box transcription factor TBX21) promotes the expression of the chemokine receptor CXCR3 and accumulation of Tregs at Th1-cell rich inflammatory sites [58]. In contrast, in a Th-2-rich environment the transcription factor GATA-3 is upregulated and maintains high levels of FoxP3 expression in pTregs [59]. This enables pTregs to contain excessive pro-inflammatory polarization and promote the accumulation of pTregs at Th-2 inflammatory sites [59, 60].. Further, although the expression of the transcription factors IRF-4 [61] and STAT3 [62] is typically assigned to Th-2 and Th-17 cells, respectively, both can be induced in Tregs and enhance their immunomodulatory potential when Th17-cells dominate inflammation [62,63,64]. In summary, the micromilieu substantially influences how pTregs can modulate their phenotypic and functional phenotype, while tTregs are characterized by an exceptionally high stability.

Fig. 1
figure 1

Suppressive Mechanisms of Tregs. Tregs-mediated immune regulation occurs by various cell-contact dependent and contact-independent mechanisms. Anti-inflammatory cytokines, such as TGF-β, IL35, and IL-10, modulate conventional T cells and monocytes towards immunotolerant states. Secretion of Perforin and Granzyme directly targets effector cells and induces apoptosis. By contact-dependent interaction via PD-1/PD-1L, MHC-II/TCR and CTLA4/CD80/CD86 Tregs prevent costimulatory signals in conventional T cells and downregulate the expression of MHC-II and CD80/86 on APCs. Similarly, binding of CTLA-4 to CD80/86 on APCs induces the expression of IDO, converts tryptophan to kynurenine and thereby further suppresses the activation of conventional T cells and promotes regulatory the exertion function in various cell types. Characteristically, high expression of CD25 on Tregs enables Tregs to sequester IL-2 from the environment, thereby limiting IL-2-mediated activation of conventional T cells. Metabolic deprivation is further enhanced by the expression of CD39 on Tregs, which converts ATP to AMP, reducing T-cell proliferation. Tregs may further reduce monocyte differentiation toward pro-inflammatory monocytes and promote M2 macrophage development

Monogenetic treg deficiencies

Numeric or functional Treg deficiencies due to monogenetic variations in the human genome lead to the clinical phenotype of primary immunodeficiency, with predominant signs of polytopic immune dysregulation. Although the clinical phenotype overlaps for various Tregs deficiencies, understanding the underlying molecular defect is important in order to tailor and select appropriate disease-specific therapeutic approaches.

IPEX-syndrome due to FoxP3 deficiency

The clinical picture of the most profound monogenetic Treg deficiency, named X-linked immune dysregulation, polyendocrinopathy and enteropathy (IPEX) syndrome was first described by Powell et al. in 1982. The authors reported a family with 19 affected males from one large family, of whom only two survived the first decade. All affected individuals suffered from severe eczema, enteropathy, thyroiditis, type I diabetes, autoimmune cytopenia and immunodeficiency [65]. Later reports added antibody-mediated intestinal villous atrophy, eosinophilia, high IgE levels, lymphoid hyperplasia, islet-cell hypo- or aplasia, arthritis, kidney involvement, and severe immunodeficiency [66,67,68,69,70,71,72]. The scurfymouse model mimics X-linked disease with a clinical phenotype similar to that of patients with IPEX, such as scaly skin, runting, diarrhea, lymphadenopathy, hepatosplenomegaly, and progressive anemia. Even before the molecular cause of IPEX syndrome was identified, this mouse line served as a helpful model for the pathophysiological understanding of IPEX syndrome [73,74,75]. At the cellular level, CD4+ lymphoproliferation, lymphocytic tissue infiltrates, and elevated proinflammatory cytokine levels characterize the disease [73,74,75]. However, it was not until the early 2000s that the genetic defect, localized in the centromeric region of the X chromosome (Xp11.23-Xq13.3) was identified [71] and closed the pathophysiological loop between the clinical presentation of IPEX disease, the crucial role of FoxP3 signaling and regulatory T cells functions [4, 6, 76]. Since then, IPEX syndrome is also named according to the molecular defect as FoxP3 deficiency. Independent of the genetic mutation, the clinical presentation of patients with IPEX varies substantially. In a cohort of 96 IPEX patients, 39 had neonatal onset of enteropathy, type I diabetes, and eczema, and in less than half of the patients, nephropathy, autoimmune cytopenia, hepatitis, or thyroiditis. Only a few patients present with arthritis, alopecia, lymphadenopathy, or neutropenia [77]. Vice versa type I diabetes may be the only clinical sign at time of diagnosis [78, 79].

Meanwhile, numerous additional monogenetic diseases that lead to clinically relevant Treg deficiency have been described. These diseases share mutations in individual genes encoding proteins that crucially support regulatory T-cell functions. In the last update in 2022, the International Union of Immunological Societies (IUIS) classified these pathologies as diseases of immune dysregulation, summarized in the IUIS group IV. Most of these diseases result from monogenetic loss-of-function mutations (LOF), e. g. in FOXP3, CD25, LRBA, CTLA4, AIRE, IL-10, IL-10R, STAT5B, or BACH2. Additionally, gain-of-function mutations (GOF) in STAT3, may lead to a similar phenotype. However, as the genetic and phenotypic heterogeneity of Treg deficiencies makes the clinical diagnosis difficult, the term “Tregopathies” has been aptly established [80]. Laboratory findings in patients with numeric or functional Treg deficiencies are only indicative and require specific Treg staining because lymphocyte distribution frequently shows largely normal results. Furthermore, despite the shared clinical characteristics of genetically different Treg pathologies, understanding the underlying functional pathomechanisms to direct target-specific therapies is crucial.

CTLA4-deficiency

As a member of the immunoglobulin superfamily and a costimulatory molecule that transmits inhibitory signals to T cells, CTLA-4 is a key element in Treg-mediated immune regulation [4, 81]. The phenotypic overlap between FoxP3, CTLA-4 and also TGF-β deficiency was first described in mice [82,83,84,85]. Tregs are severely impaired in patients with CTLA4 haploinsufficiency. The clinical phenotype is dominated by immune dysregulation with autoimmunity, immunodeficiency, and lymphoproliferation (IDAIL) but shows a highly variable presentation [86,87,88,89,90]. Based on its genotypic background, this syndrome is also referred to as CHAI (CTLA-4 Haploinsufficiency with Autoimmune Infiltration). Although late disease onset is possible, most patients report their initial clinical signs during early childhood. In a cohort of 133 patients from 54 unrelated families with genetically confirmed CTLA4 deficiency initial symptoms included autoimmune cytopenia (33%), respiratory manifestations (21%), enteropathy (17%), type 1 diabetes (8%), neurologic symptoms (seizures and headache (6%)), thyroid disease (5%), arthritis (3%), growth retardation, fever or night sweats, atopic dermatitis, or alopecia [87]. The pattern of lymphocytic organ infiltration was found to be heterogeneous, even among members of the same family [86,87,88,89]. Susceptibility to infection in CHAI reflects the characteristics of combined immunodeficiency with infections predominantly caused by Hemophilus influenzae, pneumococci, Salmonella enteritidis, and fungal species. Both EBV and CMV infections may affect multiple organs and typically recur [87]. While the frequency of circulating FoxP3 + Tregs may be normal or even increased, the expression of both CLTA-4 and FoxP3 is substantially reduced and must be specifically requested during immunological workup. Insufficient CTLA-4-mediated T-cell inhibition results in increased T-cell activation and the loss of naive CD45RA + CD4+ T cells. Dysregulated activation of the T-cell compartment contributes to autoimmune manifestations and uncontrolled lymphoproliferation. Without sufficient control of non-specific T-cell activation, pathogen-specific immune responses and lymphocytic maturation are compromised. Altered T-/B-cellular interactions may result in hypogammaglobulinemia and the formation of autoreactive antibodies [87, 88, 90]. Similar to Foxp3 deficiency, pathogenic mutations in DEF6 may lead to a secondary CTLA-4 deficiency [91]. The guanine nucleotide exchange factor DEF6, also known as IRF-4 binding protein (IBP), interacts downstream of TCR with the GTPase RAB11 and is crucially involved not only in CTLA-4 availability and trafficking, but also in multiple processes of the innate and adaptive immune system, particularly T-cell differentiation, expansion, and maturation [91, 92].

LRBA deficiency

CTLA4 recycling to the cell surface is further dependent on lipopolysaccharide-responsive and beige-like anchor protein LRBA. LRBA deficiency, which is typically associated with autoantibodies, regulatory T (Treg) cell defects, autoimmune infiltration, and enteropathy (called LATAIE) was first described in 2012 [93, 94]. Although the initial clinical manifestation within the first four years of life is dominated by autoimmunity including antibody-mediated cytopenia or endocrinopathy (42%), chronic diarrhea (27%) recurrent infections particularly of the respiratory tract (16%) and lymphoproliferation (5%), other signs of manifestation should not be missed [95]. The latter may include asthma and allergies, fever or unspecific failure to thrive. Later, autoimmunity (82%), enteropathy (63%), splenomegaly (57%) and pneumonia (49%) dominate the clinical course of disease [95],, but are accompanied by lymphadenopathy and lymphocytic tissue infiltration [96,97,98,99]. Homozygous mutations in the LRBA gene result in the loss of LRBA and impaired cellular signalling, dysfunctional vesicular trafficking, and lysosomal degradation of CTLA-4. Tregs in LRBA deficient patients are both numerically and functionally reduced with decreased expression of the most important Treg markers FoxP3, CD25 and CTLA4 [97]. The suppressive capacity of LRBA-deficient Tregs is significantly impaired, although IL-10 production seems to increase compared to healthy controls [97]. Uncontrolled T-cell activation leads to a loss of naivety and enhanced T-cell turnover. Dysregulated T follicular helper cells and defective B-cell activation result in peripheral B-cell lymphopenia and early-onset hypogammaglobulinemia, which seems to be the primary cause for recurrent respiratory tract infections [100]. At the same time functional Treg deficiency promotes the development of autoantibodies [97, 100].

IL-2 signaling deficiencies

Signaling events within the IL-2 pathway crucially involve the IL-2 receptor CD25 and the transcription factors STAT3, STAT5B and FoxP3. CD25 deficiency due to homozygous mutations in the IL-2 receptor alpha chain (IL2RA) was first described in 1997 by Sharfe et al. in a child with increased susceptibility to viral, bacterial, and fungal infections, lymphoproliferation and chronic lung disease [101]. As CD25 is critically involved in global T-cell activation, CD25 deficiency not only results in reduced and functionally impaired Tregs and autoimmunity but also in profound T-cell proliferation deficits and nonspecific lymphoproliferation [101,102,103,104]. In the absence of functional CD25, IL-2-mediated Treg differentiation and maturation is hampered, leading to substantially reduced and dysfunctional Tregs and increased numbers of autoreactive T cells [104]. Although the clinical picture of CD25 deficiency varies with respect to the sites of manifestation and severity, the triad comprising immune dysregulation, autoimmunity, and severe susceptibility to viral, bacterial, and fungal infections has been described almost all patients [101,102,103,104,105]. The profoundness of this disorder is not only indicated by the manifestation severity but also by the early onset of the disease (mean 1.25 months) [106]. As the disease progresses, autoimmune-mediated cytopenia, hepatitis, pneumopathy and small vessel pulmonary vasculitis may occur [102, 105, 107]. Closely related, impaired IL-2 signalling may be caused by a homozygous mutation in the IL2RB (CD122) gene [108]. Affected individuals present with life-threatening immune dysregulation, including nonspecific but severe lymphoproliferation, enteropathy, eczema, and susceptibility to viral infections, particularly CMV, during early infancy [108, 109]. The immunological phenotype further includes impaired NK cell differentiation despite increased peripheral NK cell frequencies and combined immunodeficiency with hypergammaglobulinemia and autoantibody formation [108, 109].

IL-10/IL-10R deficiencies

As IL-2 responsiveness is important for IL-10 induction in CD4+ T cells, patients with CD25 deficiency share severe and early onset inflammatory bowel disease with patients suffering from IL-10 or IL-10R (IL-10RA or IL-10-RB) deficiency [102, 110,111,112]. IL-10 is involved in the development and maintenance of Tregs and supports Treg-mediated suppression of pathogenic Th-17 cells in a STAT3 dependent manner [62, 113]. In in a cohort of 286 patients with IL-10/IL-10R deficiency, gastrointestinal disorders occurred in all patients with perianal manifestations (92%), fistulae (60%), and colitis (50%) being the most prominent signs, Interestingly, perianal abscesses (57%) and complications beyond the gastrointestinal tract including failure to thrive (58%), susceptibility to infections (≤ 23%), lymphoproliferation (≤ 12%), dermatologic manifestations (49%), or rheumatologic disorders (13%) were strictly linked to IL-10R deficiences and did not occur in IL-10 deficient patients [114].

STAT3 and STAT5B signaling deficiencies

Downstream of the IL-2 and IL-10 receptor STAT3 acts as transcription factor that orchestrates multiple cellular functions. Furthermore, STAT3 acts as a key regulator in multiple signaling cascades downstream of e. g. receptors that involve the common gamma chain (IL-2, IL-4, IL-9, IL-15, IL-21) [115,116,117,118,119], receptors of the interferon family [120] and hormone receptors. Upon receptor activation STAT3 is phosphorylated in a JAK-dependent fashion and subsequently translocates to the nucleus where it binds as homo- or heterodimer to responsive elements triggering the transcription of cytokine responsive genes [121]. Loss of function mutations result in early-onset eczema, bacterial and fungal infections particularly of the skin and the lung, facial dysmorphism and joint hyperextensibility accompanies by elevated serum IgE levels [122]; but autoimmune phenomena are very rare [123]. In contrast, gain-of-function mutations in STAT3, which follow an autosomal inheritance, usually manifest during early childhood as a poly-autoimmune disease with lymphoproliferation, polyendocrinopathy, enteropathy, cytopenia, and severe interstitial lung disease. Further, increased susceptibility to infections, eczema, and short stature has been reported in these patients [124,125,126,127]. The functional details of enhanced STAT3 are only partially understood and the cellular phenotype in STAT3 GOF patients seems heterogeneous, but absolute T-cell and B-cell numbers are characteristically reduced, and frequently associated with hypogammaglobulinema and impaired antigen-specific B-cell maturation and hypogammaglobulinemia [124,125,126]. At the molecular level, activated STAT3 induces the expression of SOCS3, an inhibitor of STAT5 [128]. Accordingly, STAT3 GOF mutations result in secondary STAT5b deficiency [128]. As STAT5 itself is a crucial transcription factor for the expression of FOXP3, reduced FoxP3 expression can be observed in most patients with STAT3 GOF, and Tregs are functionally impaired [129,130,131,132]. This further explains why GOF mutations in STAT3 and LOF mutations in STAT5B share not only multiple aspects of the immunological IPEX-like phenotype including severe immune dysregulation but is also characterized by short stature due to (partial) growth hormone insensitivity [128, 133,134,135,136,137]. The clinical picture caused by pathogenic STAT5B mutations further includes severe pneumopathy, variable immunodeficiencies associated with susceptibility to severe sinopulmonary, dermal, and gastrointestinal infections. Initially described to follow an autosomal-recessive (AR) inheritance [134], STAT5B LOF mutations were identified to also occur as autosomal-dominant (AD) negative pathogenic variants in 2018 [137]. While growth retardation and eczema frequently occur in both AR and AD disease, additional clinical features occur not only in milder manifestations but also in less than 10% of patients with AD disease [106, 138]. The latter might explain why only few cases have been reported so far and suggests a high number of unrecognized cases. In summary, although defects in the IL-2 signalling pathway share many clinical features, depending on the defective molecule clinical phenomena differ in terms of the severity and frequency. For example, while eczema is with a prevalence of > 50% similarly frequent in patients with LOF mutations in FOXP3, CD25, STAT5B or GOF mutations in STAT3, autoimmune phenomena including cytopenia, thyroiditis and hepatitis as well as lymphoproliferations are characteristic for CD25 deficiency but occur in only less than 10% of patients with STAT5 deficiency [106, 129, 135, 139].

CARMIL2 deficiency

Early onset skin lesions, including eczematous dermatitis associated with chronic mucocutaneous candidiasis, molluscum contagiosum bacterial abscesses, warts, inflammatory plaques, and hyperkeratosis, are characteristic hallmarks of CARMIL2 deficiency. Early reports on patients with homozygous CARMIL2 mutations further described a particular susceptibility to EBV infections, although recurrent and severe infections by other viruses, bacteria, mycobacteria, and fungi may occur [140,141,142,143]. Secondary to EBV infection, affected individuals show an increased risk of EBV-associated tumors [142, 144]. The CARMIL2 gene is located on chromosome 16q22.1, and encodes the cytosolic protein CARMIL2 (Capping Protein Arp 2/3, Myosin-I Linker), also known as RLTPR [145], which acts as a scaffold bridging the CD28 to CARD11 and NFκB signaling cascades [146]. Functional analysis of CD4+ and CD8+ T-cell responses confirmed deficient CD3/CD28 costimulation in CARMIL2 deficient individuals [143]. While peripheral T-, B-, and NK cell counts are typically normal, Tregs are profoundly reduced. Due to deficient T-cell maturation, both CD4+ and CD8+ T-cell subsets are skewed towards naïve forms [140,141,142,143]. Within the B-cell compartment, class-switched B cells and plasmablasts may be reduced and show impaired immunoglobulin formation [141, 142]. Clinically, this results in a combined immunodeficiency syndrome with profound, early onset skin and inflammatory bowel disease and susceptibility to infections [147,148,149].

BACH2 deficiency

The transcription factor BACH2, a highly conserved basic leucine zipper protein, is a key modulator of multiple immune processes, including T- and B-cell differentiation and maturation [150,151,152]. The BACH2 locus contains a T-cell super-enhancer that regulates the expression of multiple pro-inflammatory cytokines and cytokine receptors [153, 154] and thereby reducing effector T-cell differentiation. In Tregs, BACH2 induces high FoxP3 expression, thereby promoting Treg development, maturation, and survival [151, 155]. BACH2 haploinsufficiency causes low Treg frequency and function, while differentiation of Th1-cells, which express the intestinal homing receptors CCR9 and ITGB7, is enhanced [97]. Similarly, the lack of BACH2 mediated repression of Th-2 differentiation results in increased Th-2 cytokine formation, promoting both airway and bowel inflammation [156]. As the effects of BACH2 deficiency manifest at every level of B-cell development, B-cell maturation and IgG class switch are profoundly impaired, resulting in increased transitional B-cell numbers, low immunoglobulins, and inability to generate appropriate antibody responses to specific antigens of vaccines. Accordingly, the clinical picture of BACH2-related immunodeficiency and autoimmunity (BRIDA syndrome) syndrome is dominated by sinopulmonary infections and autoimmune gastrointestinal diseases, which may present early in life [154].

Tregs in autoimmune diseases

Unlike monogenetic Treg disorders polygenetic or multifactorial Treg deficiencies involve a complex interplay of multiple genes and environmental factors. Due to the multifactorial and polygenetic nature of these diseases understanding the interwoven factors contributing to the specific pathophysiology remains challenging. As mentioned earlier, we now know that Tregs represent a diverse subpopulation characterized by distinct transcriptional repertoires influenced by tissue- or context-specific transcription factors. For example, Tregs residing in adipose tissue express the transcription factor PPARγ, whereas those critical for driving Th1-type responses increase Tbet [58, 157, 158]. However, our current challenge is to use this knowledge to identify biomarkers that indicate Treg function in clinical settings. The broad spectrum of Treg functions makes the selection of a single marker or in vitro functional assay challenging, particularly in the context of a particular disease. The difficulties become even greater when assessing Treg activities in humans, primarily because of the obstacles associated with isolating Tregs from tissues other than blood.

Tregs in type 1 diabetes

Type 1 diabetes (T1D) is the best-characterized autoimmune disease, and is often referred to as Juvenile Diabetes. It is a persistent autoimmune ailment characterized by a targeted immune response driven by both T- and B-cells, culminating in the destruction of insulin-producing β-cells nestled in the pancreatic islets [159]. T1D is one of the most common chronic metabolic diseases, affecting approximately 1.5 million people under 20 years of age [160]. It is one of the most frequent chronic metabolic diseases in childhood and adolescence, with a global increase in the incidence rate of 3–4% per year and strong regional differences [161]. Many autoimmune disorders, including T1D, frequently share disruptions in the control of effector cell populations as a fundamental contributing element [162, 163], and this aberration might stem from irregularities in the suppressive functions governed by Tregs.

A significant number of studies have indicated no disparities in peripheral blood Treg frequencies among T1D patients [164]. Nonetheless, anomalies in Treg phenotype and their suppressive potential have been documented [165, 166]. As described above, the challenge of obtaining healthy human tissue is particularly daunting when studying the role and function of Tregs in T1D, as pancreatic samples can only be obtained postmortem. Unfortunately, the unavailability of pancreatic samples from T1D patients has primarily confined data collection to peripheral blood, obscuring whether Tregs actively mitigate β-cell destruction or exhibit modified traits within islets during disease progression. Consequently, animal models such as mice have been harnessed to scrutinize disease advancement within the islet microenvironment.

Therefore, non-obese diabetic (NOD) mice are an essential model for T1D research. NOD mice spontaneously develop autoimmune diabetes, typically commencing at approximately 12 weeks in females, with the incidence increasing until approximately 25 weeks [167]. Male NOD mice experience delayed onset and progression of diabetes. The incidence is approximately 70% in females and 30% in males, a difference potentially rooted in gender-based variances in the gut microbiome and hormonal fluctuations [168]. Environmental factors, including housing conditions and diet, have been implicated in autoimmune diabetes onset [167]. Genomic investigations have identified susceptibility loci termed as insulin-dependent diabetes (IDD) loci in NOD mice. A plethora of over 40 IDD loci has been cataloged, with the major histocompatibility complex (MHC) exhibiting the most substantial link to T1D incidence [167, 169, 170]. Although the NOD mouse manifests several similarities with human T1D, some distinctions persist. Nevertheless, NOD mice have emerged as valuable tools for elucidating the role of Tregs in autoimmune diabetes [167].

Undoubtedly, genetic susceptibility constitutes a fundamental cornerstone in the evolution of T1D, with a significant proportion of susceptible single nucleotide polymorphisms (SNPs) being closely linked to immune-related genes, thereby underscoring immune dysregulation. Particularly noteworthy is the robust correlation observed with genes that have considerable influence over Treg function, most prominently IL2RA, IL-2, PTPN2, CTLA4, and IL-10 [171, 172]. However, translation of these SNPs into functional outcomes has only been achieved in a few studies. Additionally, because numerous pivotal genes serve both effector T cell and Treg functions, deciphering the relative impact of allelic variants on regulatory and effector T cells poses a formidable challenge. Several studies have reported SNP-associated impairments in Treg function, with a particular emphasis on IL-2 signaling [173,174,175]. These findings, coupled with analogous findings in NOD mice highlighting a deficiency in IL-2 signaling within Tregs, have galvanized efforts to harness this pathway for therapeutic intervention [175].

A regrettable adverse facet of low-dose IL-2 therapy, which effectively amplified Tregs, was the simultaneous escalation of eosinophils and natural killer cells, coupled with a reduction in C-peptide levels [176]. However, recent studies on Treg-specific IL-2 administration hold promise for overcoming off-target effects [177,178,179]. Furthermore, novel methodologies geared towards manipulating the pharmacokinetics of IL-2 therapy are expected to enhance its efficacy. One notable study employed the administration of low-dose IL-2/CD25 fusion protein, forestalling diabetes onset and even managing overt diabetes in the NOD mouse model of T1D. The augmented half-life of this IL-2 analog facilitates prolonged interaction with CD25-expressing Tregs, thereby amplifying IL-10 production and encouraging its migration to the pancreas [180].

More recently, a study harnessed T-cell population-specific epigenetic analysis to precisely locate susceptible SNPs within enhancer regions pivotal for Treg function in autoimmunity [181]. Comparative epigenetic evaluations across Treg and conventional T-cell populations revealed that autoimmune-associated SNPs were enriched in naïve Treg-specific demethylated regions and, to a lesser extent, in activated Treg-demethylated regions. These insights suggest that autoimmune-linked SNPs exert a more profound influence on thymus-derived Treg development and function than on aberrant activation of autoimmune effector T cells.

Several pathways critical for Treg development, function, and lineage stability are perturbed in T1D, potentially resulting in Treg dysfunction. Although studies employing Tregs derived from peripheral blood have provided evidence of altered Treg function in T1D patients [182, 183], the extent to which peripheral blood can accurately reflect Treg function at the tissue site remains ambiguous. Mouse models, particularly the NOD model, have provided invaluable insights into the mechanisms underlying the Treg suppression of islet autoimmunity. These investigations have illuminated the notion that certain deficiencies in Treg function are exacerbated at the tissue site, with Treg deficits not always conspicuous in in vitro assays [184, 185]. It is plausible that an amalgamation of chronic inflammatory mediators, anomalies in the IL-2 signaling pathway, and diminished TCR diversity, among other factors, converge within the pancreatic tissue, collectively weakening Treg function [184,185,186,187]. An optimal therapeutic strategy tailored to Tregs should be meticulously devised to address this combination of defects by stabilizing FOXP3 [188, 189].

Tregs in autoimmune hepatitis

The incidence of autoimmune hepatitis (AIH) is similar. Due to the much smaller number of patients, the data were more uncertain. A pioneering genome-wide association study (GWAS) identified mainly genes of the HLA complex as risk factors, such as in T1D, but none were directly related to Tregs [190]. In relation to AIH, research has shown that Tregs may play a significant role in this context. A couple of studies from Vergani et al. have observed that patients with AIH may have reduced Treg numbers in the peripheral blood compared to healthy individuals [191,192,193]. The reduction in Tregs may contribute to an uncontrolled immune response against endogenous liver cells, ultimately leading to inflammation and liver damage. In particular, in pediatric AIH, decreased Treg, characterized as CD3+CD4+CD25+ numbers and impaired Treg function have been documented. Nonetheless, in a more recent study that included FOXP3 in Treg characterization (CD3+CD4+CD25highFOXP3+), the opposite was described as patients with AIH had increased Treg numbers in the blood [194]. This is consistent with the intrahepatic observation that the number of Tregs increases during active disease [195]. Notably, the same group also observed the opposite result in untreated pediatric patients with AIH [196]. In addition, the more pronounced effect of standard steroid therapy on decreasing Tregs over T effector cells was striking, leading to an increase in the apoptosis of Tregs [195, 197].

In mice, knockout of genes related to Treg development or function, such as aire or pd-1, leads to fatal autoimmunity in the liver and other organs, accompanied by decreased or absent numbers of Tregs [198,199,200]. In contrast, in animal models of AIH that resemble different aspects of human disease, an intrahepatic increase in Tregs in active AIH has also been observed [201,202,203,204].

Tregs in colitis

The role of Tregs in colitis is closely linked to the balance between the inflammatory and regulatory immune responses in the gut. Studies have shown that Tregs play a key role in preventing excessive inflammatory processes in the gut. Patients with colitis, particularly Crohn's disease (CD) and ulcerative colitis, have been observed to have deficiencies in the number and function of Tregs [205,206,207,208]. This, in turn, promotes an unbridled and chronic inflammatory response that can damage intestinal tissue.

Previously conducted genome-wide association studies have identified over 100 separate genetic loci that contribute to either susceptibility or defense against the development of inflammatory bowel disease (IBD), with a considerable portion of these loci being shared between the two conditions [209, 210]. Administration of NOD2 ligands, including peptides or muramyl dipeptides, has demonstrated the potential to alleviate colitis induced by 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sulfate sodium (DSS) in normal mice [211, 212]. In a TNBS-induced colitis model, treatment with Lactobacillus peptidoglycan increased the number of Tregs in mesenteric lymph nodes and elevated IL-10 expression in the colonic mucosa, implying that NOD2 activity within the intestinal mucosa fosters a milieu conducive to immune tolerance. Furthermore, receptors associated with T cell and Treg migration, such as CD62L, C–C chemokine receptor (CCR)4, CCR5, CCR7, CCR9, αEβ7 integrin, and α4β7 integrin, also contribute to the pathogenesis of IBD [213,214,215,216,217,218,219]. The presence of these receptors on Treg cells plays a pivotal role in maintaining intestinal immunological equilibrium, and their compromised expression has been linked to the development of IBD, owing to the impaired migration of Treg cells into the intestinal tract. For example, the absence of CCR7 impedes Treg cell functionality in an experimental colitis model [214].

Tregs as therapeutic agent or target

Among genetic and multifactorial autoimmune diseases, IBD is the most promising disease for polyclonal Treg transfer therapies. It has been shown, that Tr1 cells inhibit the proliferation of antigen-specific T cells through an IL-10-dependent mechanism and exhibit protective effects in the adoptive transfer model of colitis involving naïve T cells, as in SCID patients with colitis [220]. Although both FOXP3 + Treg cells and Tr1 cells generate IL-10, Tr1 cells appear to play a crucial role in upholding tolerance towards commensal bacteria. Nonetheless, Battaglia et al. demonstrated the necessity of FOXP3 + Treg cells next to Tr1 cells to persist for the initial induction of tolerance in autoinflammatory diseases [221,222,223]. We have highlighted the importance of Tregs to home to the gut and expand into the lamina propria to regain immunological tolerance [224]. As observed in SCID patients, the transfer of polyspecific Tregs is sufficient to treat autoimmune diseases that lack functional Tregs, such as SCID or APS-1 patients. Later, this was shown in other studies in the corresponding aire-deficient mouse model [199, 225,226,227,228]. To ensure the safety and efficacy of poly- or ovalbumin-specific autologous Tregs in the treatment of CD, many clinical trials have been conducted, including NCT03185000 (TRIBUTE) [229, 230], Eudract no. 2006–004712-44 [231], NCT02327221, NCT05566977, NCT03011021, NCT02932826, and NCT02691247. In addition, also antigen-specific Tregs bearing a chimeric antigen receptor (CAR) against model antigens were very successful in controlling colitis in animal models [232,233,234,235].

In T1D, the situation is very different, as polyspecific Tregs show no positive effects in mouse models or patients. It has been shown that antigen-specific Tregs are required to control diabetes and prevent its induction. Considering that Treg insufficiency potentially fuels T1D and autoimmune diabetes, bolstering the Treg count in circulation could serve as a strategy to counter this inadequacy. Notably, recurring adoptive Treg transfers into neonatal NOD mice have demonstrated the ability to postpone the onset of autoimmune diabetes, implying that Treg number or functionality might wane in NOD mice over time, necessitating supplementation [236]. Many T1D studies use BDC-2.5 mice, which are genetically modified NOD mice, carrying a transgenic TCR that recognizes a pancreatic antigen in NOD mice. These T cells destroy the insulin-producing cells in the pancreas. Therefore, another convincing strategy involves the adoptive transfer of a small quantity of DC-expanded BDC2.5 TCR-tg Tregs into pre-diabetic NOD mice. The transfer successfully prevented diabetes development and even salvaged mice with manifest diabetes [237]. When pre-diabetic NOD splenocytes or BDC2.5 TCR-tg Teff cells are transplanted into immunodeficient NOD mice, autoimmune diabetes typically emerges approximately 14 d post-transfer. Interestingly, co-transplantation with over a million polyclonal Tregs or a few thousand BDC2.5 TCR-tg Tregs can prevent the disease [238]. While a minimal number of antigen-specific Tregs have the capacity to reverse autoimmune diabetes, adopting ten-fold more polyclonal Tregs was not as effective in the therapeutic treatment of NOD mice, underscoring the critical significance of specificity for β-cell antigens in optimizing Treg functionality [239,240,241,242,243].

Clinically, in vitro-expanded polyclonal Tregs are being evaluated as a promising avenue that diverges from pharmacologically based treatments. Early phase clinical trials encompassing both pediatric and adult participants with autologous, polyspecific Tregs have been conducted, reflecting no immediate safety concerns, such as ISRCTN06128462, NCT01210664, NCT02932826, and NCT02772679 [244,245,246,247]. Notably, in children, potential efficacy has been assessed based on C-peptide levels at 4–5 weeks post-treatment. However, while initial elevations in C-peptide levels were evident at the one- and two-year follow-ups, they gradually diminished over time. Intriguingly, nearly 25% of transferred Tregs, characterized by a naïve/memory-like profile, persisted in patients at the one-year follow-up based on deuterium incorporation. A parallel trial conducted in Poland has yielded encouraging outcomes. In a cohort of 12 children with T1D, a one-year follow-up revealed augmented C-peptide levels and reduced insulin usage in 8 of 12 patients, resulting in complete insulin independence in 2 of the 12 patients [244,245,246,247]. Whether these encouraging observations endure and can be replicated in phase 2 clinical trials remains to be ascertained.

The potential for a more robust success may rely on combination therapy. Potential synergies with Tregs have been explored to optimize their therapeutic response in different autoimmune diseases. One effective strategy could involve bolstering the Treg population through the infusion of ex vivo expanded Tregs, while concurrently reducing the Teff population using agents such as anti-CD3 monoclonal antibody (NCT00129259) [248] or LFA3-Immunglobulin (Ig) (NCT00965458) [249, 250], which have shown promise in initial trials for new-onset T1D, followed by more than 30 other trials with similar results.

Another avenue to consider is the coupling of Treg cell infusion with interleukin-2 (IL-2). In vivo, IL-2 at low doses plays a pivotal role in the growth and survival of Tregs [251, 252], and constitute a critical component of Treg expansion protocols. Either Tregs might be directly equipped with IL-2 signals [253] or employing low-dose IL-2 has been effective in enhancing endogenous Tregs, leading to diabetes prevention and reversal in the NOD mouse model [184, 254]. Preliminary clinical investigations involving low-dose IL-2 have demonstrated selective increases in Tregs, yielding positive clinical outcomes in many (auto-)inflammatory conditions [255,256,257,258,259]. While a clinical trial combining IL-2 with rapamycin showed transient deterioration in beta cell function, potentially due to the relatively higher IL-2 dose or the influence of rapamycin, which led to significant increases in natural killer cells and eosinophils, early studies on lower IL-2 doses in T1D have shown no acute degradation in beta cell function [260, 261]. Several ongoing studies are aimed at evaluating the safety and efficacy of this approach. In an upcoming phase I trial involving autologous ex vivo expanded Tregs followed by low-dose IL-2, we will assess whether low-dose IL-2 enhances in vivo survival and functionality of infused Tregs.

Tregs administered in the initial clinical trials on T1D did not exhibit TCRs or CARs that specifically targeted diabetes antigens. There are several possible explanations for the selection of polyclonal Tregs for such an administration. The safety aspect was the most important, as all studies validated the safety of polyclonal cells, which is a primary consideration in devising a clinical research protocol. Thus, the effectiveness of polyspecific Tregs has been demonstrated in animal models, and data from animal studies involving exogenous IL-2, which augments Treg numbers, suggest the efficacy of polyclonal cells. Unlike other cellular processes, Treg-mediated suppression lacks antigen specificity; hence, polyclonal T cells should be capable of regulating cells that possess specificities for diabetes antigens.

Preclinical investigations have proposed that antigen-specific Tregs are more efficient in controlling autoimmune-mediated beta cell destruction than polyclonal Tregs [239,240,241,242,243]. Although target antigens for T1D have been identified, a significant challenge lies in isolating these less common cells from peripheral circulation and subsequently expanding them for clinical application [262]. Hence, it may be advantageous to generate antigen-specific de novo Tregs. Engineered Tregs featuring CARs have exhibited success in pre-clinical T1D [263, 264] and could potentially be viable for clinical deployment, albeit the specific antigen profile might need customization for each individual patient.

Polyspecific Tregs were also used in a clinical trials for AIH (NCT02704338) [265], Pemphigus Vulgaris NCT03239470, and Lupus Erythematosus NCT02428309 proving safety. However, polyspecific Treg transfer was not very effective in AIH mouse models [201, 203]. Therefore, we and others have successfully used optimization protocols for Tregs to determine their therapeutic responses in different AIH models [266,267,268]. Nonetheless, data on combination therapy or antigen-specific CAR-Tregs are lacking.

Notably, the significance of antigen-specific CAR-Treg cells in preventing genetic resilience against organ-specific autoimmunity and inhibiting autoimmune tissue damage has been well documented in various disease models, including multiple sclerosis [269] and Alzheimer’s disease [270].

Conclusion

Numeric and functional Treg deficiencies may be due to monogenetic, inborn errors of immunity or occur by a more complex multifactorial processes. While congenital global Treg deficiencies result in polytopic diseases and usually affect multiple organ systems, genetically undefined autoimmune diseases, such as T1D or AIH, are characterized by the lack of particular antigen-specific Treg subsets but normal Tregs numbers. With the rapidly growing understanding of the genetic and functional Treg biology, new therapeutic approaches are currently being developed to overcome unmet medical needs in Treg diseases. The adoptive transfer of ex vivo expanded or even genetically modified Treg products is in the focus of upcoming clinical trials. The success of these approaches will depend on both overcoming technical and regulatory hurdles and ensuring product safety and efficacy in larger patient cohorts. Undoubtedly, the possibility to tailor highly personalized Treg products that address patient or disease specific needs has certainly opened a new dimension of target specific treatment approaches.

Availability of data and materials

Not applicable.

Abbreviations

AIRE:

Autoimmune Regulator

AIH:

Autoimmune Hepatitis

APS-1:

Autoimmune Polyendocrine Syndrome Type 1

BACH2:

BTB Domain and CNC Homolog 2 (a transcription factor)

CAR:

Chimeric Antigen Receptor

CCR:

C–C Chemokine Receptor

CD:

Crohn's Disease

CD25:

IL-2 Receptor Alpha Chain (also known as IL2RA)

CTLA-4:

Cytotoxic T-lymphocyte-associated Protein 4

CXCR3:

Chemokine Receptor

DC:

Dendritic Cell

EBV:

Epstein-Barr Virus

FOXP3:

Forkhead Box P3 (a transcription factor crucial for Treg development and function)

GITR:

Glucocorticoid-Induced TNFR-Related Protein

GWAS:

Genome-Wide Association Study

IDD:

Insulin-dependent diabetes

Ig:

Immunoglobulin

IDO:

Indoleamine 2,3-Dioxygenase

IFN-γ:

Interferon-gamma

IL-2:

Interleukin-2

IL-10:

Interleukin-10

IL-10R:

IL-10 Receptor

IL2RA:

IL-2 Receptor Alpha Chain

IL-35:

Interleukin-35

IPEX Syndrome:

X-linked Immune Dysregulation, Polyendocrinopathy, and Enteropathy

IUIS:

International Union of Immunological Societies

mTECs:

Medullary Thymic Epithelial Cells

NOD:

Non-obese Diabetic (mouse model)

PD-1/PD-1L:

Programmed Cell Death Protein 1/Programmed Cell Death 1 Ligand 1

SNPs:

Single Nucleotide Polymorphisms

STAT3:

Signal Transducer and Activator of Transcription 3

STAT5:

Signal Transducer and Activator of Transcription 5

STAT5B:

Signal Transducer and Activator of Transcription 5B

T1D:

Type 1 Diabetes

TCR:

T-Cell Receptor

Teff cells:

Effector T cells

Tregs:

Regulatory T cells

Tr1 cells:

Type 1 regulatory T cells

TSA:

Tissue-Specific Antigens

Tregopathies:

Diseases Causing Regulatory T-cell Deficiencies

TNBS:

2,4,6-Trinitrobenzenesulfonic Acid

tTregs:

Thymically derived Tregs

pTregs:

Peripherally induced Tregs

References

  1. Caramalho Í, Nunes-Cabaço H, Foxall RB, Sousa AE (2015) Regulatory T-cell development in the human thymus. Front Immunol 6:395. https://doi.org/10.3389/fimmu.2015.00395

  2. Chen WJ, Jin W, Hardegen N et al (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(12):1875–1886

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Apostolou I, Von Boehmer H (2004) In Vivo Instruction of Suppressor Commitment in Naive T Cells. J Exp Med 199(10):1401–1408

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hori S, Nomura T, Sakaguchi S (2003) Control of regulatory T cell development by the transcription factor Foxp3. Science 299(5609):1057–1061

    Article  CAS  PubMed  Google Scholar 

  5. Liu W, Putnam AL, Xu-yu Z et al (2006) CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 203(7):1701–1711

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Fontenot JD, Gavin MA, Rudensky AY (2003) Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 4(4):330–336

    Article  CAS  PubMed  Google Scholar 

  7. Povoleri GAM, Scottà C, Nova-Lamperti EA et al (2013) Thymic versus induced regulatory T cells - who regulates the regulators? Front Immunol 4:169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Darrasse-Jèze G, Marodon G, Salomon BL, Catala M, Klatzmann D (2005) Ontogeny of CD4+CD25+ regulatory/suppressor T cells in human fetuses. Blood 105(12):4715–4721

    Article  PubMed  Google Scholar 

  9. Cupedo T, Nagasawa M, Weijer K, Blom B, Spits H (2005) Development and activation of regulatory T cells in the human fetus. Eur J Immunol 35(2):383–390

    Article  CAS  PubMed  Google Scholar 

  10. Mazerolles F, Stolzenberg M-C, Pelle O et al (2018) Autoimmune Lymphoproliferative Syndrome-FAS Patients Have an Abnormal Regulatory T Cell (Treg) Phenotype but Display Normal Natural Treg-Suppressive Function on T Cell Proliferation. Front Immunol 9:718

    Article  PubMed  PubMed Central  Google Scholar 

  11. Qi Y, Zhang R, Lu Y, Zou X, Yang W (2022) Aire and Fezf2, two regulators in medullary thymic epithelial cells, control autoimmune diseases by regulating TSAs: Partner or complementer?. Front Immunol 13:948259. https://doi.org/10.3389/fimmu.2022.948259

  12. Su MA, Anderson MS (2016) Aire expands: new roles in immune tolerance and beyond. Nat Rev Immunol 16(4):247–258

    Article  PubMed  PubMed Central  Google Scholar 

  13. Hsieh C-S, Lee H-M, Lio C-WJ (2012) Selection of regulatory T cells in the thymus. Nat. Rev. Immunol 12(3):157–167

    Article  CAS  PubMed  Google Scholar 

  14. Jordan MS, Boesteanu A, Reed AJ et al (2001) Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat Immunol 2(4):301–306

    Article  CAS  PubMed  Google Scholar 

  15. Moran AE, Holzapfel KL, Xing Y et al (2011) T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J Exp Med 208(6):1279–1289

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kieback E, Hilgenberg E, Stervbo U et al (2016) Thymus-Derived Regulatory T Cells Are Positively Selected on Natural Self-Antigen through Cognate Interactions of High Functional Avidity. Immunity 44(5):1114–1126

    Article  CAS  PubMed  Google Scholar 

  17. Pacholczyk R, Ignatowicz H, Kraj P, Ignatowicz L (2006) Origin and T Cell Receptor Diversity of Foxp3+CD4+CD25+ T Cells. Immunity 25(2):249–259

    Article  CAS  PubMed  Google Scholar 

  18. Lei H, Kuchenbecker L, Streitz M et al (2015) Human CD45RA- FoxP3hi Memory-Type Regulatory T Cells Show Distinct TCR Repertoires with Conventional T Cells and Play an Important Role in Controlling Early Immune Activation. Am J Transplant 15(10):2625–2635

    Article  CAS  PubMed  Google Scholar 

  19. Wyss L, Stadinski BD, King CG et al (2016) Affinity for self antigen selects Treg cells with distinct functional properties. Nat Immunol 17(9):1093–1101

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yadav M, Stephan S, Bluestone JA (2013) Peripherally induced tregs - role in immune homeostasis and autoimmunity. Front Immunol 4:232

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chorro L, Suzuki M, Chin SS et al (2018) Interleukin 2 modulates thymic-derived regulatory T cell epigenetic landscape. Nat Commun 9(1):5368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Polansky JK, Schreiber L, Thelemann C et al (2010) Methylation matters: binding of Ets-1 to the demethylated Foxp3 gene contributes to the stabilization of Foxp3 expression in regulatory T cells. J Mol Med Berl Ger 88(10):1029–1040

    Article  CAS  Google Scholar 

  23. Polansky JK, Kretschmer K, Freyer J et al (2008) DNA methylation controls Foxp3 gene expression. Eur J Immunol 38(6):1654–1663

    Article  CAS  PubMed  Google Scholar 

  24. Toker A, Engelbert D, Garg G et al (2013) Active demethylation of the Foxp3 locus leads to the generation of stable regulatory T cells within the thymus. J. Immunol. Baltim. Md 1950 190(7):3180–8

    CAS  Google Scholar 

  25. Kitagawa Y, Ohkura N, Kidani Y et al (2017) Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat Immunol 18(2):173–183

    Article  CAS  PubMed  Google Scholar 

  26. Zhao D-M, Xue H-H (2017) MLL4 keeps Foxp3 in the loop. Nat Immunol 18(9):957–958

    Article  CAS  PubMed  Google Scholar 

  27. Kondo M, Tanaka Y, Kuwabara T et al (2016) SATB1 Plays a Critical Role in Establishment of Immune Tolerance. J Immunol 196(2):563–572

    Article  CAS  PubMed  Google Scholar 

  28. Beyer M, Thabet Y, Müller R-U et al (2011) Repression of SATB1 in regulatory T cells is required for suppressive function and inhibition of effector differentiation. Nat Immunol 12(9):898–907

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sallusto F, Kremmer E, Palermo B et al (1999) Switch in chemokine receptor expression upon TCR stimulation reveals novel homing potential for recently activated T cells. Eur J Immunol 29(6):2037–2045

    Article  CAS  PubMed  Google Scholar 

  30. Sallusto F, Lenig D, Förster R, Lipp M, Lanzavecchia A (1999) Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401(6754):708–712

    Article  CAS  PubMed  Google Scholar 

  31. Gattinoni L, Lugli E, Ji Y et al (2011) A human memory T cell subset with stem cell-like properties. Nat Med 17(10):1290–1297

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schmueck-Henneresse M, Sharaf R, Vogt K et al (2015) Peripheral Blood-Derived Virus-Specific Memory Stem T Cells Mature to Functional Effector Memory Subsets with Self-Renewal Potency. J Immunol 194(11):5559–5567

    Article  CAS  PubMed  Google Scholar 

  33. Berger C, Jensen MC, Lansdorp PM et al (2008) Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invesitgation 118(1):294–305

    Article  CAS  Google Scholar 

  34. Kumar BV, Connors T, Farber DL (2018) Human T cell development, localization, and function throughout life. Immunity 48(2):202–213

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Campbell DJ, Koch MA (2011) Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol 11(2):119–130

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Menning A, Höpken UE, Siegmund K et al (2007) Distinctive role of CCR7 in migration and functional activity of naive- and effector/memory-like Treg subsets. Eur J Immunol 37(6):1575–1583

    Article  CAS  PubMed  Google Scholar 

  37. Wing K, Onishi Y, Prieto-Martin P et al (2008) CTLA-4 Control over Foxp3+ Regulatory T Cell Function. Science 322(5899):271–275

    Article  CAS  PubMed  Google Scholar 

  38. Grossman WJ, Verbsky JW, Barchet W et al (2004) Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 21(4):589–601

    Article  CAS  PubMed  Google Scholar 

  39. Karreci ES, Eskandari SK, Dotiwala F et al (2017) Human regulatory T cells undergo self-inflicted damage via granzyme pathways upon activation. JCI Insight 2(21):e91599. https://doi.org/10.1172/jci

  40. Cao X, Cai SF, Fehniger TA et al (2007) Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 27(4):635–646

    Article  CAS  PubMed  Google Scholar 

  41. Onishi Y, Fehervari Z, Yamaguchi T, Sakaguchi S (2008) Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. Proc Natl Acad Sci U S A 105(29):10113–10118

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sage PT, Paterson AM, Lovitch SB, Sharpe AH (2014) The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 41(6):1026–1039

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Akkaya B, Akkaya M, Kamenyeva O et al (2017) Unique interaction dynamics and peptide-MHC class II (pMHC II) transendocytosis lead to antigen-specific T regulatory cell (Treg)-mediated suppression. Immunol 198(1_Supplement):80.8. https://doi.org/10.4049/jimmunol.198.Supp.80.8

  44. Ovcinnikovs V, Ross EM, Petersone L et al (2019) CTLA-4-mediated transendocytosis of costimulatory molecules primarily targets migratory dendritic cells. Sci. Immunol 4(35):eaaw0902

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Kennedy A, Waters E, Rowshanravan B et al (2022) Differences in CD80 and CD86 transendocytosis reveal CD86 as a key target for CTLA-4 immune regulation. Nat Immunol 23(9):1365–1378

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Qureshi OS, Zheng Y, Nakamura K et al (2011) Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 332(6029):600–603

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mellor AL, Lemos H, Huang L (2017) Indoleamine 2,3-Dioxygenase and Tolerance: Where Are We Now? Front Immunol 8:1360

    Article  PubMed  PubMed Central  Google Scholar 

  48. Romano M, Fanelli G, Tan N et al (2018) Expanded Regulatory T Cells Induce Alternatively Activated Monocytes With a Reduced Capacity to Expand T Helper-17 Cells. Front Immunol 9:1625

    Article  PubMed  PubMed Central  Google Scholar 

  49. Murray PJ, Wynn TA (2011) Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 11(11):723–737

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Romano M, Fanelli G, Albany CJ, Giganti G, Lombardi G (2019) Past, Present, and Future of Regulatory T Cell Therapy in Transplantation and Autoimmunity. Front Immunol 10:43. https://doi.org/10.3389/fimmu.2019.00043

  51. Okazaki T, Maeda A, Nishimura H, Kurosaki T, Honjo T (2001) PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc Natl Acad Sci U S A 98(24):13866–13871

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Patsoukis N, Brown J, Petkova V et al (2012) Selective effects of PD-1 on Akt and Ras pathways regulate molecular components of the cell cycle and inhibit T cell proliferation. Sci. Signal 5(230):ra46

    Article  PubMed  PubMed Central  Google Scholar 

  53. Hofmeyer KA, Jeon H, Zang X (2011) The PD-1/PD-L1 (B7–H1) pathway in chronic infection-induced cytotoxic T lymphocyte exhaustion. J Biomed Biotechnol 2011:451694

    Article  PubMed  PubMed Central  Google Scholar 

  54. Jiang S, Park SE, Yadav P, Paoletti LC, Wessels MR (2012) Regulation and function of pilus island 1 in group B streptococcus. J Bacteriol 194(10):2479–2490

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Priyadharshini B, Loschi M, Newton RH et al (2018) Cutting Edge: TGF-β and Phosphatidylinositol 3-Kinase Signals Modulate Distinct Metabolism of Regulatory T Cell Subsets. J. Immunol. Baltim. Md 1950 201(8):2215–2219

    CAS  Google Scholar 

  56. Gerriets VA, Kishton RJ, Johnson MO et al (2016) Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat Immunol 17(12):1459–1466

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chinen T, Kannan AK, Levine AG et al (2016) An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 17(11):1322–1333

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Koch MA, Tucker-Heard G, Perdue NR et al (2009) The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol 10(6):595–602

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wang Y, Su MA, Wan YY (2011) An essential role of the transcription factor GATA-3 for the function of regulatory T cells. Immunity 35(3):337–348

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wohlfert EA, Grainger JR, Bouladoux N et al (2011) GATA3 controls Foxp3+ regulatory T cell fate during inflammation in mice. J Clin Invest 121(11):4503–4515

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zheng Y, Chaudhry A, Kas A et al (2009) Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 458(7236):351–356

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Chaudhry A, Rudra D, Treuting P et al (2009) CD4+ regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 326(5955):986–991

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kluger MA, Melderis S, Nosko A et al (2016) Treg17 cells are programmed by Stat3 to suppress Th17 responses in systemic lupus. Kidney Int 89(1):158–166

    Article  CAS  PubMed  Google Scholar 

  64. Kluger MA, Luig M, Wegscheid C et al (2014) Stat3 Programs Th17-Specific Regulatory T Cells to Control GN. J Am Soc Nephrol JASN 25(6):1291–1302

    Article  CAS  PubMed  Google Scholar 

  65. Powell BR, Buist NRM, Stenzel P (1982) An X-linked syndrome of diarrhea, polyendocrinopathy, and fatal infection in infancy. J Pediatr 100(5):731–737

    Article  CAS  PubMed  Google Scholar 

  66. Satake N, Nakanishi M, Okano M et al (1993) A Japanese family of X-linked auto-immune enteropathy with haemolytic anaemia and polyendocrinopathy. Eur J Pediatr 152(4):313–315

    Article  CAS  PubMed  Google Scholar 

  67. Peake JE, McCrossin RB, Byrne G, Shepherd R (1996) X-linked immune dysregulation, neonatal insulin dependent diabetes, and intractable diarrhoea. Arch Dis Child Fetal Neonatal Ed 74(3):F195–199

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Goulet OJ, Brousse N, Canioni D et al (1998) Syndrome of Intractable Diarrhoea with Persistent Villous Atrophy in Early Childhood: A Clinicopathological Survey of 47 Cases. J Pediatr Gastroenterol Nutr 26(2):151

    CAS  PubMed  Google Scholar 

  69. Roberts J, Searle J (1995) Neonatal Diabetes Mellitus Associated with Severe Diarrhea, Hyperimmunoglobulin E Syndrome, and Absence of Islets of Langerhans. Pediatr Pathol Lab Med 15(3):477–483

    Article  CAS  PubMed  Google Scholar 

  70. Rocco MD, Marta R (1996) X linked immune dysregulation, neonatal insulin dependent diabetes, and intractable diarrhoea. Arch. Dis. Child. - Fetal Neonatal Ed 75(2):F144

    Article  PubMed  PubMed Central  Google Scholar 

  71. Ferguson PJ, Blanton SH, Saulsbury FT et al (2000) Manifestations and linkage analysis in X-linked autoimmunity-immunodeficiency syndrome. Am J Med Genet 90(5):390–397

    Article  CAS  PubMed  Google Scholar 

  72. Gambineri E, Ciullini Mannurita S, Hagin D et al (2018) Clinical, Immunological, and Molecular Heterogeneity of 173 Patients With the Phenotype of Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked (IPEX) Syndrome. Front Immunol 9:2411

    Article  PubMed  PubMed Central  Google Scholar 

  73. Clark LB, Appleby MW, Brunkow ME et al (1999) Cellular and molecular characterization of the scurfy mouse mutant. J. Immunol. Baltim. Md 1950 162(5):2546–2554

    CAS  Google Scholar 

  74. Lyon MF, Peters J, Glenister PH, Ball S, Wright E (1990) The scurfy mouse mutant has previously unrecognized hematological abnormalities and resembles Wiskott-Aldrich syndrome. Proc Natl Acad Sci 87(7):2433–2437

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Ramsdell F, Ziegler SF (2014) FOXP3 and scurfy: how it all began. Nat Rev Immunol 14(5):343–349

    Article  CAS  PubMed  Google Scholar 

  76. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM (2009) A census of human transcription factors: function, expression and evolution. Nat Rev Genet 10(4):252–263

    Article  CAS  PubMed  Google Scholar 

  77. Barzaghi F, Amaya Hernandez LC, Neven B et al (2018) Long-term follow-up of IPEX syndrome patients after different therapeutic strategies: An international multicenter retrospective study. J Allergy Clin Immunol 141(3):1036–1049.e5

    Article  PubMed  Google Scholar 

  78. Hwang JL, Park S-Y, Ye H et al (2018) FOXP3 mutations causing early-onset insulin-requiring diabetes but without other features of immune dysregulation, polyendocrinopathy, enteropathy. X-linked syndrome Pediatr Diabetes 19(3):388–392

    Article  CAS  PubMed  Google Scholar 

  79. De Benedetti F, Insalaco A, Diamanti A et al (2006) Mechanistic Associations of a Mild Phenotype of Immunodysregulation, Polyendocrinopathy, Enteropathy. X-Linked Syndrome Clin Gastroenterol Hepatol 4(5):653–659

    Article  PubMed  Google Scholar 

  80. Cepika A-M, Sato Y, Liu JM-H et al (2018) Tregopathies: Monogenic diseases resulting in regulatory T-cell deficiency. J. Allergy Clin. Immunol 142(6):1679–1695

    Article  CAS  PubMed  Google Scholar 

  81. Magistrelli G, Jeannin P, Herbault N et al (1999) A soluble form of CTLA-4 generated by alternative splicing is expressed by nonstimulated human T cells. Eur J Immunol 29(11):3596–3602

    Article  CAS  PubMed  Google Scholar 

  82. Tivol EA, Borriello F, Schweitzer AN et al (1995) Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3(5):541–547

    Article  CAS  PubMed  Google Scholar 

  83. Waterhouse P, Penninger JM, Timms E et al (1995) Lymphoproliferative Disorders with Early Lethality in Mice Deficient in Ctla-4. Science 270(5238):985–988

    Article  CAS  PubMed  Google Scholar 

  84. Brunkow ME, Jeffery EW, Hjerrild KA et al (2001) Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet 27(1):68–73

    Article  CAS  PubMed  Google Scholar 

  85. Kulkarni AB, Huh CG, Becker D et al (1993) Transforming growth factor beta 1 null mutation in mice causes excessive inflammatory response and early death. Proc Natl Acad Sci 90(2):770–774

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Kuehn HS, Ouyang W, Lo B et al (2014) Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345(6204):1623–1627

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Schwab C, Gabrysch A, Olbrich P et al (2018) Phenotype, penetrance, and treatment of 133 cytotoxic T-lymphocyte antigen 4-insufficient subjects. J Allergy Clin Immunol 142(6):1932–1946

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Schubert D, Bode C, Kenefeck R et al (2014) Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med 20(12):1410–1416

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. López-Nevado M, González-Granado LI, Ruiz-García R et al (2021) Primary Immune Regulatory Disorders With an Autoimmune Lymphoproliferative Syndrome-Like Phenotype: Immunologic Evaluation. Early Diagnosis and Management Front Immunol 12:671755

    Article  PubMed  Google Scholar 

  90. Lo B, Fritz JM, Su HC et al (2016) CHAI and LATAIE: new genetic diseases of CTLA-4 checkpoint insufficiency. Blood 128(8):1037–1042

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Serwas NK, Hoeger B, Ardy RC et al (2019) Human DEF6 deficiency underlies an immunodeficiency syndrome with systemic autoimmunity and aberrant CTLA-4 homeostasis. Nat Commun 10(1):3106

    Article  PubMed  PubMed Central  Google Scholar 

  92. Fournier B, Tusseau M, Villard M et al (2021) DEF6 deficiency, a mendelian susceptibility to EBV infection, lymphoma, and autoimmunity. J Allergy Clin Immunol 147(2):740–743.e9

    Article  CAS  PubMed  Google Scholar 

  93. Alangari A, Alsultan A, Adly N et al (2012) LPS-responsive beige-like anchor (LRBA) gene mutation in a family with inflammatory bowel disease and combined immunodeficiency. J Allergy Clin Immunol 130(2):481–488.e2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Burns SO, Zenner HL, Plagnol V et al (2012) LRBA gene deletion in a patient presenting with autoimmunity without hypogammaglobulinemia. J Allergy Clin Immunol 130(6):1428–1432

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Habibi S, Zaki-Dizaji M, Rafiemanesh H et al (2019) Clinical, Immunologic, and Molecular Spectrum of Patients with LPS-Responsive Beige-Like Anchor Protein Deficiency: A Systematic Review. J Allergy Clin Immunol Pract 7(7):2379–2386.e5

    Article  CAS  PubMed  Google Scholar 

  96. Revel-Vilk S, Fischer U, Keller B et al (2015) Autoimmune lymphoproliferative syndrome-like disease in patients with LRBA mutation. Clin Immunol 159(1):84–92

    Article  CAS  PubMed  Google Scholar 

  97. Charbonnier L-M, Janssen E, Chou J et al (2015) Regulatory T-cell deficiency and immune dysregulation, polyendocrinopathy, enteropathy, X-linked-like disorder caused by loss-of-function mutations in LRBA. J Allergy Clin Immunol 135(1):217–227

    Article  CAS  PubMed  Google Scholar 

  98. Alkhairy OK, Abolhassani H, Rezaei N et al (2016) Spectrum of Phenotypes Associated with Mutations in LRBA. J Clin Immunol 36(1):33–45

    Article  CAS  PubMed  Google Scholar 

  99. Jamee M, Hosseinzadeh S, Sharifinejad N et al (2021) Comprehensive comparison between 222 CTLA-4 haploinsufficiency and 212 LRBA deficiency patients: a systematic review. Clin Exp Immunol 205(1):28–43

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Lopez-Herrera G, Tampella G, Pan-Hammarström Q et al (2012) Deleterious Mutations in LRBA Are Associated with a Syndrome of Immune Deficiency and Autoimmunity. Am J Hum Genet 90(6):986–1001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Sharfe N, Dadi HK, Shahar M, Roifman CM (1997) Human immune disorder arising from mutation of the alpha chain of the interleukin-2 receptor. Proc Natl Acad Sci U S A 94(7):3168–3171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Caudy AA, Reddy ST, Chatila T, Atkinson JP, Verbsky JW (2007) CD25 deficiency causes an immune dysregulation, polyendocrinopathy, enteropathy, X-linked–like syndrome, and defective IL-10 expression from CD4 lymphocytes. J Allergy Clin Immunol 119(2):482–487

    Article  CAS  PubMed  Google Scholar 

  103. Goudy K, Aydin D, Barzaghi F et al (2013) Human IL2RA null mutation mediates immunodeficiency with lymphoproliferation and autoimmunity. Clin Immunol 146(3):248–261

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Roifman CM (2000) Human IL-2 Receptor α Chain Deficiency. Pediatr Res 48(1):6–11

    Article  CAS  PubMed  Google Scholar 

  105. Bezrodnik L, Caldirola MS, Seminario AG, Moreira I, Gaillard MI (2014) Follicular bronchiolitis as phenotype associated with CD25 deficiency. Clin Exp Immunol 175(2):227–234

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Consonni F, Favre C, Gambineri E (2021) IL-2 Signaling Axis Defects: How Many Faces?. Front Pediatr 9:669298. https://doi.org/10.3389/fped.2021.669298

  107. Al Sukaiti N, Al Sinani A, Al Ismaily S, Shaikh S, Al AS (2014) Pulmonary hemorrhage in a case of CD25 deficiency. LymphoSign J 01(01):39–43

    Article  Google Scholar 

  108. Fernandez IZ, Baxter RM, Garcia-Perez JE et al (2019) A novel human IL2RB mutation results in T and NK cell–driven immune dysregulation. J Exp Med 216(6):1255–1267

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Zhang Z, Gothe F, Pennamen P et al (2019) Human interleukin-2 receptor β mutations associated with defects in immunity and peripheral tolerance. J Exp Med 216(6):1311–1327

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Engelhardt KR, Shah N, Faizura-Yeop I et al (2013) Clinical outcome in IL-10– and IL-10 receptor–deficient patients with or without hematopoietic stem cell transplantation. J Allergy Clin Immunol 131(3):825–830.e9

    Article  CAS  PubMed  Google Scholar 

  111. Shah N, Kammermeier J, Elawad M, Glocker E-O (2012) Interleukin-10 and interleukin-10-receptor defects in inflammatory bowel disease. Curr Allergy Asthma Rep 12(5):373–379

    Article  CAS  PubMed  Google Scholar 

  112. Zhou X, Wang J, Shi W et al (2010) Isolation of purified and live Foxp3+ regulatory T cells using FACS sorting on scatter plot. J Mol Cell Biol 2(3):164–169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Chaudhry A, Samstein RM, Treuting P et al (2011) Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 34(4):566–578

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Sharifinejad N, Zaki-Dizaji M, Sepahvandi R et al (2022) The clinical, molecular, and therapeutic features of patients with IL10/IL10R deficiency: a systematic review. Clin Exp Immunol 208(3):281–291

    Article  PubMed  PubMed Central  Google Scholar 

  115. Russell SM, Keegan AD, Harada N et al (1993) Interleukin-2 receptor gamma chain: a functional component of the interleukin-4 receptor. Science 262(5141):1880–1883

    Article  CAS  PubMed  Google Scholar 

  116. Noguchi M, Nakamura Y, Russell SM et al (1993) Interleukin-2 receptor gamma chain: a functional component of the interleukin-7 receptor. Science 262(5141):1877–1880

    Article  CAS  PubMed  Google Scholar 

  117. Giri JG, Kumaki S, Ahdieh M et al (1995) Identification and cloning of a novel IL-15 binding protein that is structurally related to the alpha chain of the IL-2 receptor. EMBO J 14(15):3654–3663

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Avery DT, Deenick EK, Ma CS et al (2010) B cell–intrinsic signaling through IL-21 receptor and STAT3 is required for establishing long-lived antibody responses in humans. J Exp Med 207(1):155–171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Waldmann TA (2015) The shared and contrasting roles of interleukin-2 (IL-2) and IL-15 in the life and death of normal and neoplastic lymphocytes: implications for cancer therapy. Cancer Immunol Res 3(3):219–227

    Article  MathSciNet  CAS  PubMed  PubMed Central  Google Scholar 

  120. Platanias LC (2005) Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol 5(5):375–386

    Article  CAS  PubMed  Google Scholar 

  121. Hillmer EJ, Zhang H, Li HS, Watowich SS (2016) STAT3 signaling in immunity. Cytokine Growth Factor Rev 31:1–15

    Article  PubMed  PubMed Central  Google Scholar 

  122. Tsilifis C, Freeman AF, Gennery AR (2021) STAT3 Hyper-IgE Syndrome—an Update and Unanswered Questions. J Clin Immunol 41(5):864–880

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Oikonomopoulou C, Goussetis E (2020) Autosomal dominant hyper-IgE syndrome: When hematopoietic stem cell transplantation should be considered? Pediatr. Transplant 24(5):e13699

    CAS  Google Scholar 

  124. Flanagan SE, Haapaniemi E, Russell MA et al (2014) Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 46(8):812–814

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Haapaniemi EM, Kaustio M, Rajala HLM et al (2015) Autoimmunity, hypogammaglobulinemia, lymphoproliferation, and mycobacterial disease in patients with activating mutations in STAT3. Blood 125(4):639–648

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Milner JD, Vogel TP, Forbes L et al (2015) Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations. Blood 125(4):591–599

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Khoury T, Molho-Pessach V, Ramot Y et al (2017) Tocilizumab Promotes Regulatory T-cell Alleviation in STAT3 Gain-of-function-associated Multi-organ Autoimmune Syndrome. Clin Ther 39(2):444–449

    Article  CAS  PubMed  Google Scholar 

  128. Gutiérrez M, Scaglia P, Keselman A et al (2018) Partial growth hormone insensitivity and dysregulatory immune disease associated with de novo germline activating STAT3 mutations. Mol Cell Endocrinol 473:166–177

    Article  PubMed  PubMed Central  Google Scholar 

  129. Cohen AC, Nadeau KC, Tu W et al (2006) Cutting Edge: Decreased Accumulation and Regulatory Function of CD4+CD25high T Cells in Human STAT5b Deficiency1. J Immunol 177(5):2770–2774

    Article  CAS  PubMed  Google Scholar 

  130. Pelham SJ, Caldirola MS, Avery DT et al (2022) STAT5B restrains human B-cell differentiation to maintain humoral immune homeostasis. J Allergy Clin Immunol 150(4):931–946

    Article  CAS  PubMed  Google Scholar 

  131. Passerini L, Allan SE, Battaglia M et al (2008) STAT5-signaling cytokines regulate the expression of FOXP3 in CD4+CD25+ regulatory T cells and CD4+CD25- effector T cells. Int Immunol 20(3):421–431

    Article  CAS  PubMed  Google Scholar 

  132. Burchill MA, Yang J, Vogtenhuber C, Blazar BR, Farrar MA (2007) IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3+ regulatory T cells. J. Immunol. Baltim. Md 1950 178(1):280–290

    CAS  Google Scholar 

  133. Gutiérrez M (2020) Activating mutations of STAT3: Impact on human growth. Mol Cell Endocrinol 518:110979

    Article  PubMed  Google Scholar 

  134. Kofoed EM, Hwa V, Little B et al (2003) Growth Hormone Insensitivity Associated with a STAT5b Mutation. N Engl J Med 349(12):1139–1147

    Article  CAS  PubMed  Google Scholar 

  135. Bernasconi A, Marino R, Ribas A et al (2006) Characterization of Immunodeficiency in a Patient With Growth Hormone Insensitivity Secondary to a Novel STAT5b Gene Mutation. Pediatrics 118(5):e1584–e1592

    Article  PubMed  Google Scholar 

  136. Hwa V, Camacho-Hübner C, Little BM et al (2007) Growth Hormone Insensitivity and Severe Short Stature in Siblings: A Novel Mutation at the Exon 13-Intron 13 Junction of the STAT5b Gene. Horm Res 68(5):218–224

    CAS  PubMed  Google Scholar 

  137. Klammt J, Neumann D, Gevers EF et al (2018) Dominant-negative STAT5B mutations cause growth hormone insensitivity with short stature and mild immune dysregulation. Nat Commun 9(1):2105

    Article  PubMed  PubMed Central  Google Scholar 

  138. Smith MR, Satter LRF, Vargas-Herna´ ndez A (2023) STAT5b: A master regulator of key biological pathways. Front Immunol 13:1025373. https://doi.org/10.3389/fimmu.2022.1025373

  139. Kanai T, Jenks J, Nadeau KC (2012) The STAT5b Pathway Defect and Autoimmunity. Front Immunol 3:234

    Article  PubMed  PubMed Central  Google Scholar 

  140. Wang Y, Ma CS, Ling Y et al (2016) Dual T cell– and B cell–intrinsic deficiency in humans with biallelic RLTPR mutations. J Exp Med 213(11):2413–2435

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Sorte HS, Osnes LT, Fevang B et al (2016) A potential founder variant in CARMIL2/RLTPR in three Norwegian families with warts, molluscum contagiosum, and T-cell dysfunction. Mol Genet Genomic Med 4(6):604–616

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Schober T, Magg T, Laschinger M et al (2017) A human immunodeficiency syndrome caused by mutations in CARMIL2. Nat Commun 8(1):14209

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Alazami AM, Al-Helale M, Alhissi S, Al-Saud B, Alajlan H, Monies D et al (2018) Novel CARMIL2 Mutations in Patients with Variable Clinical Dermatitis, Infections, and Combined Immunodeficiency. Front Immunol 9:203. https://doi.org/10.3389/fimmu.2018.00203

  144. Yonkof JR, Gupta A, Rueda CM, Mangray S, Prince BT, Rangarajan HG et al (2020) A Novel Pathogenic Variant in CARMIL2 (RLTPR) Causing CARMIL2 Deficiency and EBV-Associated Smooth Muscle Tumors. Front Immunol 11:884. https://doi.org/10.3389/fimmu.2020.00884

  145. Matsuzaka Y, Okamoto K, Mabuchi T et al (2004) Identification, expression analysis and polymorphism of a novel RLTPR gene encoding a RGD motif, tropomodulin domain and proline/leucine-rich regions. Gene 343(2):291–304

    Article  CAS  PubMed  Google Scholar 

  146. Roncagalli R, Cucchetti M, Jarmuzynski N et al (2016) The scaffolding function of the RLTPR protein explains its essential role for CD28 co-stimulation in mouse and human T cells. J Exp Med 213(11):2437–2457

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Magg T, Shcherbina A, Arslan D et al (2019) CARMIL2 Deficiency Presenting as Very Early Onset Inflammatory Bowel Disease. Inflamm Bowel Dis 25(11):1788–1795

    Article  PubMed  PubMed Central  Google Scholar 

  148. Lévy R, Gothe F, Momenilandi M et al (2022) Human CARMIL2 deficiency underlies a broader immunological and clinical phenotype than CD28 deficiency. J Exp Med 220(2):e20220275

    Article  PubMed  PubMed Central  Google Scholar 

  149. Kolukisa B, Baser D, Akcam B et al (2022) Evolution and long-term outcomes of combined immunodeficiency due to CARMIL2 deficiency. Allergy 77(3):1004–1019

    Article  CAS  PubMed  Google Scholar 

  150. Igarashi K, Ochiai K, Itoh-Nakadai A, Muto A (2014) Orchestration of plasma cell differentiation by Bach2 and its gene regulatory network. Immunol Rev 261(1):116–125

    Article  CAS  PubMed  Google Scholar 

  151. Roychoudhuri R, Hirahara K, Mousavi K et al (2013) BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature 498(7455):506–510

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Tsukumo S, Unno M, Muto A et al (2013) Bach2 maintains T cells in a naive state by suppressing effector memory-related genes. Proc Natl Acad Sci 110(26):10735–10740

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Vahedi G, Kanno Y, Furumoto Y et al (2015) Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520(7548):558–562

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Afzali B, Grönholm J, Vandrovcova J et al (2017) BACH2 immunodeficiency illustrates an association between super-enhancers and haploinsufficiency. Nat Immunol 18(7):813–823

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Kim EH, Gasper DJ, Lee SH et al (2014) Bach2 Regulates Homeostasis of Foxp3+ Regulatory T Cells and Protects against Fatal Lung Disease in Mice. J Immunol 192(3):985–995

    Article  CAS  PubMed  Google Scholar 

  156. Liu G, Liu F (2022) Bach2: A Key Regulator in Th2-Related Immune Cells and Th2 Immune Response. J Immunol Res 2022:2814510

    Article  PubMed  PubMed Central  Google Scholar 

  157. Cipolletta D, Feuerer M, Li A et al (2012) PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 486(7404):549–553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Tan TG, Mathis D, Benoist C (2016) Singular role for T-BET+CXCR3+ regulatory T cells in protection from autoimmune diabetes. Proc Natl Acad Sci U A 113(49):14103–14108

    Article  CAS  Google Scholar 

  159. Herold KC, Vignali DA, Cooke A, Bluestone JA (2013) Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol 13(4):243–256

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Gregory GA, Robinson TIG, Linklater SE et al (2022) Global incidence, prevalence, and mortality of type 1 diabetes in 2021 with projection to 2040: a modelling study. Lancet Diabetes Endocrinol 10(10):741–760

    Article  PubMed  Google Scholar 

  161. Tuomilehto J, Ogle GD, Lund-Blix NA, Stene LC (2020) Update on Worldwide Trends in Occurrence of Childhood Type 1 Diabetes in 2020. Pediatr Endocrinol Rev 17(Suppl 1):198–209

    PubMed  Google Scholar 

  162. Yaciuk JC, Pan Y, Schwarz K et al (2015) Defective selection of thymic regulatory T cells accompanies autoimmunity and pulmonary infiltrates in Tcra-deficient mice double transgenic for human La/Sjogren’s syndrome-B and human La-specific TCR. J Immunol 194(4):1514–1522

    Article  CAS  PubMed  Google Scholar 

  163. Long SA, Buckner JH (2011) CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. J Immunol 187(5):2061–2066

    Article  CAS  PubMed  Google Scholar 

  164. Brusko TM, Hulme MA, Myhr CB, Haller MJ, Atkinson MA (2007) Assessing the in vitro suppressive capacity of regulatory T cells. Immunol Invest 36(5–6):607–628

    Article  CAS  PubMed  Google Scholar 

  165. Viisanen T, Gazali AM, Ihantola EL et al (2019) FOXP3+ Regulatory T Cell Compartment Is Altered in Children With Newly Diagnosed Type 1 Diabetes but Not in Autoantibody-Positive at-Risk Children. Front Immunol 10:19

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Shapiro MR, Dong X, Perry DJ et al (2023) Human immune phenotyping reveals accelerated aging in type 1 diabetes. JCI Insight 8(17):e170767. https://doi.org/10.1172/jci.insight.170767

  167. Anderson MS, Bluestone JA (2005) The NOD mouse: a model of immune dysregulation. Annu Rev Immunol 23:447–485

    Article  CAS  PubMed  Google Scholar 

  168. Markle JG, Frank DN, Mortin-Toth S et al (2013) Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339(6123):1084–1088

    Article  CAS  PubMed  Google Scholar 

  169. Driver JP, Serreze DV, Chen YG (2011) Mouse models for the study of autoimmune type 1 diabetes: a NOD to similarities and differences to human disease. Semin Immunopathol 33(1):67–87

    Article  CAS  PubMed  Google Scholar 

  170. Driver JP, Chen YG, Zhang W, Asrat S, Serreze DV (2011) Unmasking genes in a type 1 diabetes-resistant mouse strain that enhances pathogenic CD8 T-cell responses. Diabetes 60(4):1354–1359

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Barrett JC, Clayton DG, Concannon P et al (2009) Genome-wide association study and meta-analysis find that over 40 loci affect risk of type 1 diabetes. Nat Genet 41(6):703–707

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Todd JA, Walker NM, Cooper JD et al (2007) Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes. Nat Genet 39(7):857–864

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Long SA, Cerosaletti K, Wan JY et al (2011) An autoimmune-associated variant in PTPN2 reveals an impairment of IL-2R signaling in CD4(+) T cells. Genes Immun 12(2):116–125

    Article  CAS  PubMed  Google Scholar 

  174. Yang JH, Cutler AJ, Ferreira RC et al (2015) Natural Variation in Interleukin-2 Sensitivity Influences Regulatory T-Cell Frequency and Function in Individuals With Long-standing Type 1 Diabetes. Diabetes 64(11):3891–3902

    Article  CAS  PubMed  Google Scholar 

  175. Dendrou CA, Wicker LS (2008) The IL-2/CD25 pathway determines susceptibility to T1D in humans and NOD mice. J Clin Immunol 28(6):685–696

    Article  CAS  PubMed  Google Scholar 

  176. Long SA, Rieck M, Sanda S et al (2012) Rapamycin/IL-2 combination therapy in patients with type 1 diabetes augments Tregs yet transiently impairs beta-cell function. Diabetes 61(9):2340–2348

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Trotta E, Bessette PH, Silveria SL et al (2018) A human anti-IL-2 antibody that potentiates regulatory T cells by a structure-based mechanism. Nat Med 24(7):1005–1014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Abbas AK, Trotta E, R. Simeonov D, Marson A, Bluestone JA (2018) Revisiting IL-2: Biology and therapeutic prospects. Revisiting IL-2: Biology and therapeutic prospects. Sci Immunol 3:eaat1482. https://doi.org/10.1126/sciimmunol.aat1482

  179. Sockolosky JT, Trotta E, Parisi G et al (2018) Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science 359(6379):1037–1042

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Ward NC, Lui JB, Hernandez R et al (2020) Persistent IL-2 Receptor Signaling by IL-2/CD25 Fusion Protein Controls Diabetes in NOD Mice by Multiple Mechanisms. Diabetes 69(11):2400–2413

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Ohkura N, Yasumizu Y, Kitagawa Y et al (2020) Regulatory T Cell-Specific Epigenomic Region Variants Are a Key Determinant of Susceptibility to Common Autoimmune Diseases. Immunity 52(6):1119–1132 e4

    Article  CAS  PubMed  Google Scholar 

  182. Hull CM, Peakman M, Tree TIM (2017) Regulatory T cell dysfunction in type 1 diabetes: what’s broken and how can we fix it? Diabetologia 60(10):1839–1850

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Lindley S, Dayan CM, Bishop A et al (2005) Defective suppressor function in CD4(+)CD25(+) T-cells from patients with type 1 diabetes. Diabetes 54(1):92–99

    Article  CAS  PubMed  Google Scholar 

  184. Tang Q, Adams JY, Penaranda C et al (2008) Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 28(5):687–697

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Bettini ML, Pan F, Bettini M et al (2012) Loss of epigenetic modification driven by the Foxp3 transcription factor leads to regulatory T cell insufficiency. Immunity 36(5):717–730

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Baker RL, Jamison BL, Wiles TA et al (2018) CD4 T Cells Reactive to Hybrid Insulin Peptides Are Indicators of Disease Activity in the NOD Mouse. Diabetes 67(9):1836–1846

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Zhou X, Bailey-Bucktrout SL, Jeker LT et al (2009) Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol 10(9):1000–1007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. McGovern J, Holler A, Thomas S, Stauss HJ (2022) Forced Fox-P3 expression can improve the safety and antigen-specific function of engineered regulatory T cells. J Autoimmun 132:102888

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Henschel P, Landwehr-Kenzel S, Engels N et al (2023) Supraphysiological FOXP3 expression in human CAR-Tregs results in improved stability, efficacy, and safety of CAR-Treg products for clinical application. J Autoimmun 138:103057

    Article  CAS  PubMed  Google Scholar 

  190. de Boer YS, van Gerven NM, Zwiers A et al (2014) Genome-wide association study identifies variants associated with autoimmune hepatitis type 1. Gastroenterology 147(2):443–52 (e5)

    Article  PubMed  Google Scholar 

  191. Longhi MS, Hussain MJ, Mitry RR et al (2006) Functional study of CD4+CD25+ regulatory T cells in health and autoimmune hepatitis. J Immunol 176(7):4484–4491

    Article  CAS  PubMed  Google Scholar 

  192. Ferri S, Longhi MS, De Molo C et al (2010) A multifaceted imbalance of T cells with regulatory function characterizes type 1 autoimmune hepatitis. Hepatology 52(3):999–1007

    Article  CAS  PubMed  Google Scholar 

  193. Liberal R, Grant CR, Holder BS et al (2012) The impaired immune regulation of autoimmune hepatitis is linked to a defective galectin-9/tim-3 pathway. Hepatology 56(2):677–686

    Article  CAS  PubMed  Google Scholar 

  194. Peiseler M, Sebode M, Franke B et al (2012) FOXP3+ regulatory T cells in autoimmune hepatitis are fully functional and not reduced in frequency. J Hepatol 57(1):125–132

    Article  CAS  PubMed  Google Scholar 

  195. Taubert R, Hardtke-Wolenski M, Noyan F et al (2014) Intrahepatic regulatory T cells in autoimmune hepatitis are associated with treatment response and depleted with current therapies. J Hepatol 61(5):1106–1114

    Article  CAS  PubMed  Google Scholar 

  196. Diestelhorst J, Junge N, Schlue J et al (2017) Pediatric autoimmune hepatitis shows a disproportionate decline of regulatory T cells in the liver and of IL-2 in the blood of patients undergoing therapy. PLoS ONE 12(7):e0181107

    Article  PubMed  PubMed Central  Google Scholar 

  197. John K, Hardtke-Wolenski M, Jaeckel E et al (2017) Increased apoptosis of regulatory T cells in patients with active autoimmune hepatitis. Cell Death Dis 8(12):3219

    Article  PubMed  PubMed Central  Google Scholar 

  198. Kido M, Watanabe N, Okazaki T et al (2008) Fatal autoimmune hepatitis induced by concurrent loss of naturally arising regulatory T cells and PD-1-mediated signaling. Gastroenterology 135(4):1333–1343

    Article  CAS  PubMed  Google Scholar 

  199. Hardtke-Wolenski M, Taubert R, Noyan F et al (2015) Autoimmune hepatitis in a murine autoimmune polyendocrine syndrome type 1 model is directed against multiple autoantigens. Hepatology 61(4):1295–1305

    Article  CAS  PubMed  Google Scholar 

  200. Bonito AJ, Aloman C, Fiel MI et al (2013) Medullary thymic epithelial cell depletion leads to autoimmune hepatitis. J Clin Invest 123(8):3510–3524

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Hardtke-Wolenski M, Fischer K, Noyan F et al (2013) Genetic predisposition and environmental danger signals initiate chronic autoimmune hepatitis driven by CD4+ T cells. Hepatology 58(2):718–728

    Article  CAS  PubMed  Google Scholar 

  202. Holdener M, Hintermann E, Bayer M et al (2008) Breaking tolerance to the natural human liver autoantigen cytochrome P450 2D6 by virus infection. J Exp Med 205(6):1409–1422

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Lapierre P, Beland K, Yang R, Alvarez F (2013) Adoptive transfer of ex vivo expanded regulatory T cells in an autoimmune hepatitis murine model restores peripheral tolerance. Hepatology 57(1):217–227

    Article  CAS  PubMed  Google Scholar 

  204. Zierden M, Kuhnen E, Odenthal M, Dienes HP (2010) Effects and regulation of autoreactive CD8+ T cells in a transgenic mouse model of autoimmune hepatitis. Gastroenterology 139(3):975–86 (986 e1–3)

    Article  CAS  PubMed  Google Scholar 

  205. Veltkamp C, Anstaett M, Wahl K et al (2011) Apoptosis of regulatory T lymphocytes is increased in chronic inflammatory bowel disease and reversed by anti-TNFalpha treatment. Gut 60(10):1345–1353

    Article  CAS  PubMed  Google Scholar 

  206. Mohammadnia-Afrouzi M, Zavaran Hosseini A, Khalili A et al (2015) Decrease of CD4(+) CD25(+) CD127(low) FoxP3(+) regulatory T cells with impaired suppressive function in untreated ulcerative colitis patients. Autoimmunity 48(8):556–561

    Article  CAS  PubMed  Google Scholar 

  207. Sznurkowska K, Luty J, Bryl E et al (2020) Enhancement of Circulating and Intestinal T Regulatory Cells and Their Expression of Helios and Neuropilin-1 in Children with Inflammatory Bowel Disease. J Inflamm Res 13:995–1005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Fantini MC, Rizzo A, Fina D et al (2009) Smad7 controls resistance of colitogenic T cells to regulatory T cell-mediated suppression. Gastroenterology 136(4):1308–16 (e1–3)

    Article  CAS  PubMed  Google Scholar 

  209. Jostins L, Ripke S, Weersma RK et al (2012) Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491(7422):119–124

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Luo Y, de Lange KM, Jostins L et al (2017) Exploring the genetic architecture of inflammatory bowel disease by whole-genome sequencing identifies association at ADCY7. Nat Genet 49(2):186–192

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Watanabe T, Asano N, Murray PJ et al (2008) Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis. J Clin Invest 118(2):545–559

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Macho Fernandez E, Valenti V, Rockel C et al (2011) Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut 60(8):1050–1059

    Article  PubMed  Google Scholar 

  213. Venturi GM, Conway RM, Steeber DA, Tedder TF (2007) CD25+CD4+ regulatory T cell migration requires L-selectin expression: L-selectin transcriptional regulation balances constitutive receptor turnover. J Immunol 178(1):291–300

    Article  CAS  PubMed  Google Scholar 

  214. Suffia I, Reckling SK, Salay G, Belkaid Y (2005) A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J Immunol 174(9):5444–5455

    Article  CAS  PubMed  Google Scholar 

  215. Belkaid Y, Rouse BT (2005) Natural regulatory T cells in infectious disease. Nat Immunol 6(4):353–360

    Article  CAS  PubMed  Google Scholar 

  216. Denning TL, Kim G, Kronenberg M (2005) Cutting edge: CD4+CD25+ regulatory T cells impaired for intestinal homing can prevent colitis. J Immunol 174(12):7487–7491

    Article  CAS  PubMed  Google Scholar 

  217. Pedros C, Gaud G, Bernard I et al (2015) An Epistatic Interaction between Themis1 and Vav1 Modulates Regulatory T Cell Function and Inflammatory Bowel Disease Development. J Immunol 195(4):1608–1616

    Article  CAS  PubMed  Google Scholar 

  218. Yuan Q, Bromley SK, Means TK et al (2007) CCR4-dependent regulatory T cell function in inflammatory bowel disease. J Exp Med 204(6):1327–1334

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Schneider MA, Meingassner JG, Lipp M, Moore HD, Rot A (2007) CCR7 is required for the in vivo function of CD4+ CD25+ regulatory T cells. J Exp Med 204(4):735–745

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Bacchetta R, Bigler M, Touraine JL et al (1994) High levels of interleukin 10 production in vivo are associated with tolerance in SCID patients transplanted with HLA mismatched hematopoietic stem cells. J Exp Med 179(2):493–502

    Article  CAS  PubMed  Google Scholar 

  221. Battaglia M, Stabilini A, Migliavacca B et al (2006) Rapamycin promotes expansion of functional CD4+CD25+FOXP3+ regulatory T cells of both healthy subjects and type 1 diabetic patients. J Immunol 177(12):8338–8347

    Article  CAS  PubMed  Google Scholar 

  222. Battaglia M, Stabilini A, Draghici E et al (2006) Induction of tolerance in type 1 diabetes via both CD4+CD25+ T regulatory cells and T regulatory type 1 cells. Diabetes 55(6):1571–1580

    Article  CAS  PubMed  Google Scholar 

  223. Roncarolo MG, Gregori S, Bacchetta R, Battaglia M, Gagliani N (2018) The Biology of T Regulatory Type 1 Cells and Their Therapeutic Application in Immune-Mediated Diseases. Immunity 49(6):1004–1019

    Article  CAS  PubMed  Google Scholar 

  224. Hadis U, Wahl B, Schulz O et al (2011) Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity 34(2):237–246

    Article  CAS  PubMed  Google Scholar 

  225. Bansal K, Yoshida H, Benoist C, Mathis D (2017) The transcriptional regulator Aire binds to and activates super-enhancers. Nat Immunol 18(3):263–273

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Fujikado N, Mann AO, Bansal K et al (2016) Aire Inhibits the Generation of a Perinatal Population of Interleukin-17A-Producing gammadelta T Cells to Promote Immunologic Tolerance. Immunity 45(5):999–1012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Devoss JJ, Shum AK, Johannes KP et al (2008) Effector mechanisms of the autoimmune syndrome in the murine model of autoimmune polyglandular syndrome type 1. J Immunol 181(6):4072–4079

    Article  CAS  PubMed  Google Scholar 

  228. Anderson MS, Venanzi ES, Klein L et al (2002) Projection of an immunological self shadow within the thymus by the aire protein. Science 298(5597):1395–1401

    Article  CAS  PubMed  Google Scholar 

  229. Goldberg R, Scotta C, Cooper D et al (2019) Correction of Defective T-Regulatory Cells From Patients With Crohn’s Disease by Ex Vivo Ligation of Retinoic Acid Receptor-alpha. Gastroenterology 156(6):1775–1787

    Article  CAS  PubMed  Google Scholar 

  230. Canavan JB, Scotta C, Vossenkamper A et al (2016) Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn’s disease. Gut 65(4):584–594

    Article  CAS  PubMed  Google Scholar 

  231. Desreumaux P, Foussat A, Allez M et al (2012) Safety and efficacy of antigen-specific regulatory T-cell therapy for patients with refractory Crohn’s disease. Gastroenterology 143(5):1207–1217 (e2)

    Article  CAS  PubMed  Google Scholar 

  232. Elinav E, Adam N, Waks T, Eshhar Z (2009) Amelioration of colitis by genetically engineered murine regulatory T cells redirected by antigen-specific chimeric receptor. Gastroenterology 136(5):1721–1731

    Article  CAS  PubMed  Google Scholar 

  233. Blat D, Zigmond E, Alteber Z, Waks T, Eshhar Z (2014) Suppression of murine colitis and its associated cancer by carcinoembryonic antigen-specific regulatory T cells. Mol Ther 22(5):1018–1028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  234. Maliar A, Servais C, Waks T et al (2012) Redirected T cells that target pancreatic adenocarcinoma antigens eliminate tumors and metastases in mice. Gastroenterology 143(5):1375–1384 e5

    Article  CAS  PubMed  Google Scholar 

  235. Elinav E, Waks T, Eshhar Z (2008) Redirection of regulatory T cells with predetermined specificity for the treatment of experimental colitis in mice. Gastroenterology 134(7):2014–2024

    Article  PubMed  Google Scholar 

  236. Wu AJ, Hua H, Munson SH, McDevitt HO (2002) Tumor necrosis factor-alpha regulation of CD4+CD25+ T cell levels in NOD mice. Proc Natl Acad Sci U A 99(19):12287–12292

    Article  CAS  Google Scholar 

  237. Tarbell KV, Petit L, Zuo X et al (2007) Dendritic cell-expanded, islet-specific CD4+ CD25+ CD62L+ regulatory T cells restore normoglycemia in diabetic NOD mice. J Exp Med 204(1):191–201

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Petzold C, Riewaldt J, Watts D et al (2013) Foxp3(+) regulatory T cells in mouse models of type 1 diabetes. J Diabetes Res 2013:940710

    Article  PubMed  PubMed Central  Google Scholar 

  239. Tarbell KV, Yamazaki S, Olson K, Toy P, Steinman RM (2004) CD25+ CD4+ T cells, expanded with dendritic cells presenting a single autoantigenic peptide, suppress autoimmune diabetes. J Exp Med 199(11):1467–1477

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Jaeckel E, Lipes MA, von Boehmer H (2004) Recessive tolerance to preproinsulin 2 reduces but does not abolish type 1 diabetes. Nat Immunol 5(10):1028–1035

    Article  CAS  PubMed  Google Scholar 

  241. Tang Q, Henriksen KJ, Bi M et al (2004) In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. J Exp Med 199(11):1455–1465

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Masteller EL, Warner MR, Tang Q et al (2005) Expansion of functional endogenous antigen-specific CD4+CD25+ regulatory T cells from nonobese diabetic mice. J Immunol 175(5):3053–3059

    Article  CAS  PubMed  Google Scholar 

  243. Jaeckel E, Klein L, Martin-Orozco N, von Boehmer H (2003) Normal incidence of diabetes in NOD mice tolerant to glutamic acid decarboxylase. J Exp Med 197(12):1635–1644

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Marek-Trzonkowska N, Mysliwiec M, Dobyszuk A et al (2014) Therapy of type 1 diabetes with CD4(+)CD25(high)CD127-regulatory T cells prolongs survival of pancreatic islets - results of one year follow-up. Clin Immunol 153(1):23–30

    Article  CAS  PubMed  Google Scholar 

  245. Marek-Trzonkowska N, Mysliwiec M, Dobyszuk A et al (2012) Administration of CD4+CD25highCD127- regulatory T cells preserves β-cell function in type 1 diabetes in children. Diabetes Care 35(9):1817–1820

    Article  PubMed  PubMed Central  Google Scholar 

  246. Bluestone JA, Buckner JH, Fitch M et al (2015) Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci. Transl. Med 7(315):315ra189

    Article  PubMed  PubMed Central  Google Scholar 

  247. Marek-Trzonkowska N, Mysliwiec M, Iwaszkiewicz-Grzes D et al (2016) Factors affecting long-term efficacy of T regulatory cell-based therapy in type 1 diabetes. J Transl Med 14(1):332

    Article  PubMed  PubMed Central  Google Scholar 

  248. Herold KC, Gitelman SE, Ehlers MR et al (2013) Teplizumab (anti-CD3 mAb) treatment preserves C-peptide responses in patients with new-onset type 1 diabetes in a randomized controlled trial: metabolic and immunologic features at baseline identify a subgroup of responders. Diabetes 62(11):3766–3774

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Rigby MR, Harris KM, Pinckney A et al (2015) Alefacept provides sustained clinical and immunological effects in new-onset type 1 diabetes patients. J Clin Invest 125(8):3285–3296

    Article  PubMed  PubMed Central  Google Scholar 

  250. Rigby MR, DiMeglio LA, Rendell MS et al (2013) Targeting of memory T cells with alefacept in new-onset type 1 diabetes (T1DAL study): 12 month results of a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Diabetes Endocrinol 1(4):284–294

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Malek TR (2008) The biology of interleukin-2. Annu Rev Immunol 26:453–479

    Article  CAS  PubMed  Google Scholar 

  252. Wu R, Li N, Zhao X, Ding T, Xue H, Gao C, Li X, Wang C (2020) Low-dose Interleukin-2: Biology and therapeutic prospects in rheumatoid arthritis. Autoimmun Rev 19(10):102645. https://doi.org/10.1016/j.autrev.2020.102645

  253. Kremer J, Henschel P, Simon D et al (2022) Membrane-bound IL-2 improves the expansion, survival, and phenotype of CAR Tregs and confers resistance to calcineurin inhibitors. Front Immunol 13:1005582

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Grinberg-Bleyer Y, Baeyens A, You S et al (2010) IL-2 reverses established type 1 diabetes in NOD mice by a local effect on pancreatic regulatory T cells. J Exp Med 207(9):1871–1878

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  255. Rosenzwajg M, Salet R, Lorenzon R et al (2020) Low-dose IL-2 in children with recently diagnosed type 1 diabetes: a Phase I/II randomised, double-blind, placebo-controlled, dose-finding study. Diabetologia 63(9):1808–1821

    Article  CAS  PubMed  Google Scholar 

  256. Rosenzwajg M, Lorenzon R, Cacoub P 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(2):209–217

    Article  CAS  PubMed  Google Scholar 

  257. Saadoun D, Rosenzwajg M, Joly F et al (2011) Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 365(22):2067–2077

    Article  CAS  PubMed  Google Scholar 

  258. Koreth J, Kim HT, Jones KT et al (2017) Efficacy, durability, and response predictors of low-dose interleukin-2 therapy for chronic graft-versus-host disease. Blood 128(1):130–138

    Article  Google Scholar 

  259. Castela E, Duff FL, Butori C et al (2014) Effects of Low-Dose Recombinant Interleukin 2 to Promote T-Regulatory Cells in Alopecia Areata. JAMA Dermatol 150(7):748–751

    Article  CAS  PubMed  Google Scholar 

  260. Rosenzwajg M, Churlaud G, Mallone R et al (2015) Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun 58:48–58

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Hartemann A, Bensimon G, Payan CA et al (2013) Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 1(4):295–305

    Article  CAS  PubMed  Google Scholar 

  262. Roep BO, Peakman M (2012) Antigen targets of type 1 diabetes autoimmunity. Cold Spring Harb Perspect Med 2(4):a007781

    Article  PubMed  PubMed Central  Google Scholar 

  263. Pieper T, Roth KDR, Glaser V, Riet T, Buitrago-Molina LE, Hagedorn M, Lieber M, Hust M, Noyan F, Jaeckel E et al (2023) Generation of Chimeric Antigen Receptors against Tetraspanin 7. Cells 12(11):1453. https://doi.org/10.3390/cells12111453

  264. Tenspolde M, Zimmermann K, Weber LC et al (2019) Regulatory T cells engineered with a novel insulin-specific chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes. J Autoimmun 103:102289

    Article  CAS  PubMed  Google Scholar 

  265. Chung YY, Rahim MN, Heneghan MA (2022) Autoimmune hepatitis and pregnancy: considerations for the clinician. Expert Rev Clin Immunol 18(4):325–333

    Article  CAS  PubMed  Google Scholar 

  266. Buitrago-Molina LE, Pietrek J, Noyan F et al (2021) Treg-specific IL-2 therapy can reestablish intrahepatic immune regulation in autoimmune hepatitis. J Autoimmun 117:102591

    Article  CAS  PubMed  Google Scholar 

  267. Buitrago-Molina LE, Dywicki J, Noyan F, Schepergerdes L, Pietrek J, Lieber M, Schlue J, Manns MP, Wedemeyer H, Jaeckel E et al (2021) Anti-CD20 Therapy Alters the Protein Signature in Experimental Murine AIH, but Not Exclusively towards Regeneration. Cells 10(6):1471. https://doi.org/10.3390/cells10061471

  268. Marceau G, Yang R, Lapierre P, Beland K, Alvarez F (2015) Low-dose anti-CD3 antibody induces remission of active autoimmune hepatitis in xenoimmunized mice. Liver Int 35(1):275–284

    Article  CAS  PubMed  Google Scholar 

  269. Fransson M, Piras E, Burman J et al (2012) CAR/FoxP3-engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery. J Neuroinflammation 9:112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Saetzler V, Riet T, Schienke A, Henschel P, Freitag K, Haake A, Heppner FL, Buitrago-Molina LE, Noyan F, Jaeckel E et al (2023) Development of Beta-Amyloid-Specific CAR-Tregs for the Treatment of Alzheimer’s Disease. Cells 12(16):2115. https://doi.org/10.3390/cells12162115

Download references

Funding

Open Access funding enabled and organized by Projekt DEAL. This work was supported by grants from the German Research Foundation (DFG) (HA 6880/2–3) and the European Union’s Horizon 2020 Research and Innovation Program (825392; RESHAPE consortium). SLK was competitively selected for “CORE100Pilot”, which is an advanced clinician-scientist program co-funded by the Else Kröner Fresenius Foundation and the Ministry for Science and Culture of the State of Lower Saxony.

Author information

Authors and Affiliations

Authors

Contributions

MHW and SLK conceptualized, wrote, and revised the manuscript. Both authors contributed equally and approved the final manuscript.

Corresponding author

Correspondence to Sybille Landwehr-Kenzel.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hardtke-Wolenski, M., Landwehr-Kenzel, S. Tipping the balance in autoimmunity: are regulatory t cells the cause, the cure, or both?. Mol Cell Pediatr 11, 3 (2024). https://doi.org/10.1186/s40348-024-00176-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40348-024-00176-8

Keywords