Next Article in Journal
Pathophysiology of Inflammatory Bowel Disease: Innate Immune System
Next Article in Special Issue
Treg Therapy for the Induction of Immune Tolerance in Transplantation—Not Lost in Translation?
Previous Article in Journal
Anti-Aging Effects of Anthocyanin Extracts of Sambucus canadensis Caused by Targeting Mitochondrial-Induced Oxidative Stress
Previous Article in Special Issue
Healthy-like CD4+ Regulatory and CD4+ Conventional T-Cell Receptor Repertoires Predict Protection from GVHD Following Donor Lymphocyte Infusion
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Regulatory T Cells: Understanding the Role of Tregs in Select Autoimmune Skin Diseases and Post-Transplant Nonmelanoma Skin Cancers

by
Nicole Chizara Oparaugo
1,2,
Kelsey Ouyang
3,
Nam Phuong N. Nguyen
4,
Amanda M. Nelson
5 and
George W. Agak
2,*
1
David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
2
Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
3
Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
4
Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
5
Department of Dermatology, Penn State University College of Medicine, Hershey, PA 17033, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1527; https://doi.org/10.3390/ijms24021527
Submission received: 15 November 2022 / Revised: 4 January 2023 / Accepted: 9 January 2023 / Published: 12 January 2023
(This article belongs to the Special Issue T-regulatory Cells in Autoimmunity and Transplantation)

Abstract

:
Regulatory T cells (Tregs) play an important role in maintaining immune tolerance and homeostasis by modulating how the immune system is activated. Several studies have documented the critical role of Tregs in suppressing the functions of effector T cells and antigen-presenting cells. Under certain conditions, Tregs can lose their suppressive capability, leading to a compromised immune system. For example, mutations in the Treg transcription factor, Forkhead box P3 (FOXP3), can drive the development of autoimmune diseases in multiple organs within the body. Furthermore, mutations leading to a reduction in the numbers of Tregs or a change in their function facilitate autoimmunity, whereas an overabundance can inhibit anti-tumor and anti-pathogen immunity. This review discusses the characteristics of Tregs and their mechanism of action in select autoimmune skin diseases, transplantation, and skin cancer. We also examine the potential of Tregs-based cellular therapies in autoimmunity.

1. Introduction

Immune system homeostasis is tightly regulated by the proper functioning of regulatory T cells (Tregs), a T cell subpopulation that utilizes various suppressive mechanisms to modulate the activity of other immune cells [1]. The interaction between Tregs and other cells in the immune system is a necessary step in the maintenance of self-tolerance. From a functional perspective, Tregs are mainly responsible for suppressing the activation, proliferation, and cytokine production of CD4+ and CD8+ T cells and are also believed to suppress B cells and dendritic cells [2,3,4]. A notable genetic marker of Tregs is the transcriptional factor, Forkhead box P3 (FOXP3), which serves as a master regulator for Treg development and function [1,5]. A missense loss-of-function mutation within the FOXP3 locus can result in self-reactive lymphocytes that can lead to the development of severe autoimmunity in scurfy mice or cause a rare, but severe, disease IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked) syndrome in humans [1,2,3]. Therefore, the importance of Tregs is evident in that these cells are crucial in orchestrating immune suppression and helping to prevent autoimmune disease.
The functional stability of Tregs is required to control inflammation. Using FOXP3 fate reporter mice, Rubtsov et al. demonstrated that Tregs are a highly stable lineage [6]. However, in certain disease states, Treg lineage instability has been reported. In such instances, previously FOXP3-positive Tregs lose FOXP3 expression and demonstrate an effector T cell (Teff) phenotype [7,8,9]. The factors and tissue-specific cues that cause a total loss of Treg identity and partial shift towards Teff phenotype, yet still maintain the suppressive function, are yet to be elucidated. One proposed explanation for the loss of Treg identity is that a proportion of FOXP3+ Tregs only transiently express FOXP3 and lack the complete epigenetic Treg-cell program [10,11].
In healthy humans, circulating Tregs represent a highly heterogeneous population of phenotypes and gene expression profiles, with some FOXP3+ cells demonstrating close similarity to conventional T cells (Tconv). This means that FOXP3 is a highly specific, but not an absolute, Treg marker. As such, this indicates the importance of understanding the heterogeneity of FOXP3+ T cells in different clinical settings, including autoimmune skin diseases, transplantation, and cancer. Tconv broadly requires IL-7, whereas Tregs express the IL-2 receptor α-chain (CD25) and are dependent on IL-2 [12]. In all, Tregs circulating in human blood can be divided into different fractions and isolated using markers based on CD25+ FOXP3+ expression and several other markers that are beyond the scope of this review [13,14,15,16]. We first describe Tregs classification, followed by the proposed mechanisms of action and function in select disease settings. Lastly, we discuss the potential of Tregs-based therapies in autoimmunity.

2. Classification of Tregs

In response to environmental antigens and cues, Tregs either develop from autoreactive thymocytes in the thymus or naïve CD4+ T cells in the periphery [17]. Thymic-derived Tregs (tTregs) mature directly from CD4 and CD8 double-positive T cells within the thymus and are sometimes referred to as naturally occurring Tregs (nTregs). In this pathway, thymic T cells recognize self-antigen–MHC complexes expressed on thymic epithelial cells with relatively high avidity, leading to the development of a T cell receptor (TCR) repertoire with a self-bias necessary for autoimmune prevention [18,19,20]. On the other hand, peripheral Tregs (pTregs) develop from differentiated naïve T cells in the periphery upon stimulation with IL-2 and transforming growth factor-β (TGF-β) in vivo. The term inducible Tregs (iTregs) refers to Tregs generated with IL-2 and TGF-β in vitro. Both tTregs and pTregs differ in ontogeny, regulation, and function, ensuring their complementary role in maintaining immune tolerance and homeostasis [21,22]. TGF-β, which is predominately secreted by CD103+ dendritic cells (DCs), is essential for the generation of both tTregs and pTregs [23]. Several commensal bacteria can also induce TGF-β secretion by DCs [24,25]. Overall, tTregs and pTregs can regulate inflammation in different tissues throughout the body.

3. Tissue-Specific Tregs

Tregs are highly complex immune cells that carry out various functions specific to the peripheral tissues in which they reside [26]. Tissue-specific Tregs are found within the gastrointestinal tract (GI), visceral adipose tissue (VAT), and the skin, where they act as specialized suppressors of inflammation [27,28]. For example, within the GI tract IL-33-responsive GATA3+ Helios+ colonic tTregs ameliorate tissue damage during colitis [29]. In contrast, GATA3+ Helios+ colonic pTreg cells and RORγt+ Helios Tregs are induced by intestinal microbiota, and the loss of RORγt expression results in severe intestinal inflammation [30,31,32]. Additionally, within the epithelial layer of the small intestines, food antigens have been shown to induce the RORγt Helios Tregs subset that prevents allergic responses to food antigens [33]. Tregs are also present in healthy skeletal muscle and increase in numbers following muscle injury. In this case, the increased frequency of Tregs not only helps to suppress inflammation, as would be expected, but also produce factors, such as amphiregulin, which enhance muscle regeneration and repair [34]. Tregs in VAT sites express the transcriptional factor, PPARγ (peroxisome proliferator-activated receptor gamma), which is associated with the differentiation of adipocytes. VAT-residing Tregs control inflammatory states of adipose tissue, and their depletion abrogates metabolic parameters, such as insulin sensitivity [16,28].
Skin-resident Tregs have features of memory Tregs (mTregs) due to their ability to maintain homeostasis and residency within the cutaneous environment long after initial antigen exposure [35,36]. Phenotypically, human cutaneous Tregs express classic memory T cell markers, such as CD45RO [36]. In steady-state conditions, mTregs localized in the hair follicle stem cell niche are non-migratory and nearly unresponsive. These skin-resident Tregs produce Jagged1, a Notch ligand that is associated with hair regrowth and the differentiation of stem cells within the hair follicle [37]. In inflammatory microenvironments, mTregs in psoriatic lesions are rapidly proliferative, secreting low levels of proinflammatory cytokines, such as IL-17. In addition to attenuating inflammatory responses, Tregs in the skin aid in skin repair [38], maintain tolerance with commensal skin microbes, and assist in hair follicle regeneration [37,39].

4. Mechanism of Action

Tregs are a functionally heterogeneous population that can suppress immune responses through various versatile and complementary mechanisms [40]. Often this suppression is highly specific to certain types of immune responses. Although the molecular mechanisms by which Tregs exert their suppressor activity are not clearly defined, we discuss some proposed mechanisms of immune suppression (Figure 1).

4.1. Starving T Cells of IL-2

IL-2 secreted by thymic cells, DCs, and activated T cells is consumed in an autocrine/paracrine manner by cells that harbor IL-2R. Tregs express IL-2R and are physiologically primed to proliferate constitutively in the presence of IL-2. In fact, proper maturation of Tregs in the thymus depends heavily on IL-2R signaling [47,48,49]. Murine studies have shown that mice lacking components of the IL-2/IL-2R signaling axis develop autoimmune diseases [50,51], suggesting that IL-2 is critical in generating functional Tregs (Figure 1). The expression of the high affinity IL-2R allows Tregs to lower the IL-2 concentrations within the microenvironment, thereby decreasing the proliferative signal available to other T cell subsets [52].

4.2. Induction of T Cell Apoptosis

Tregs can inhibit Teff activity by inducing apoptosis via the release of cytotoxic molecules, perforin, and granzyme B (GzmB) [53]. Gondek et al. reported less effective Treg activity in GzmB-deficient mice, demonstrating the importance of GzmB in Treg-mediated suppression [54]. In addition to GzmB, the expression of TNF-related apoptosis-induced ligand (TRAIL), a suppressor that acts via interaction with death receptor 5 on CD4+ T and other effector cells, is pivotal. TRAIL activation results in caspase-8-mediated apoptosis [55,56].

4.3. Production of Inhibitory Cytokines

Tregs secrete high amounts of cytokines that have immunosuppressive actions, such as TGF-β, IL-10, and IL-35 (Figure 1). These cytokines have non-specific suppressive activity and can target effector B and T cells [57,58,59]. IL-10 and TGF-β can inhibit antigen presentation by DCs and enable the induction of pTreg populations, such as Th3 and Tr1 cells. Such Tregs have been observed at sites of chronic inflammation and in transplanted tissues [60,61,62]. Moreover, Tregs can induce other cell types to express IL-10 [63].

4.4. Inhibiting Immunostimulatory Signals and Metabolic Activity of APCs

Treg recognition of antigens presented via MHC-II molecules can lead to the generation of non-functional antigen-presenting cells (APCs) that cannot present antigens. The methods of Treg suppression are diverse and can include the binding of costimulatory molecules CD80/86 on APCs by CTLA-4 [64], removal of Ag-MHC-II complexes from the APC surface through trans-endocytosis [65], decreased indoleamine 2,3-deoxygenase (IDO) expression on the APC surface leading to reduced tryptophan levels, and the subsequent loss of proliferative capacity [66]. In this way, IDO acts as a rate-limiting enzyme that aids in sustaining Treg-mediated immune tolerance. When expressed on DCs, the enzyme drives the differentiation of naïve CD4+ T cells towards a FOXP3+ phenotype, enhances the Treg suppression of Teffs [67,68], and prevents the conversion (“reprogramming”) of Tregs into pro-inflammatory cells [69]. These processes disrupt the ability of APCs to process and present antigens, subsequently leading to T cell anergy. In addition, because of its immunoregulatory capacity, it has been proposed that IDO expression can suppress T cell responses and promote immune tolerance in mammalian pregnancy, autoimmunity, and allergic inflammation [70].
The disruption of Treg activity augments critical memory processes and immune system homeostasis. Understanding the role of cutaneous mTregs may open the door to tremendous breakthroughs in therapeutics. In this review, we focus on the role of Tregs in select autoimmune skin conditions and post-transplant cancers (Table 1).

5. Role of Tregs in Select Autoimmune Skin Diseases

5.1. Psoriasis

Cutaneous psoriasis is a prevalent disease that affects roughly 2–5% of the world’s population [98]. Patients with this condition may report debilitating symptoms that impact their physical and mental well-being. Psoriatic lesion sites contain an infiltration of immune cells and abnormal proliferation of keratinocytes within the dermis and epidermis [99].
Though not fully understood, the pathogenesis of psoriasis is complex and multifactorial, influenced by both the environment and genetic factors [100,101]. Environmental triggers include tobacco, infections, stress, and physical trauma [71,102]. Twin studies conducted by Brandrup et al. provide additional insight into the genetic component of psoriasis. Here, monozygotic twins were found to have a 40% concordance rate of psoriasis compared to a dizygotic twin concordance rate of 10% [103]. Furthermore, 35% of psoriasis patients reported a positive family history of psoriasis [104,105]. HLA genetic studies have identified increased MHC class I antigens in individuals with psoriasis [106]. A greater understanding of psoriatic triggers is needed and as our understanding grows, so do the opportunities for developing novel treatment options.
In psoriasis, Tregs lose their ability to effectively suppress the excessive expansion of Th17 cells [107]. The increased IL-17 levels are proposed to intensify T bet expression and IFN-γ production by further downregulating FOXP3 and TGF-β expression [108]. Because FOXP3 is vital to Treg development in the thymus, such an environment results in poorly developed Tregs, thereby exacerbating inflammation rather than suppression [109,110,111,112]. Rather than dampen these immune responses via FOXP3 repression of RORγt, Tregs that have undergone plasticity are reported to worsen inflammation by releasing IL-17, IFN-γ, and TNF-α via the phosphorylation of STAT3 [112]. In vitro, STAT3 phosphorylation occurs in the presence of IL-6, IL-21, and IL-23. These cytokines likely play a role in Treg dysfunction and transformation into Th1/Th17 cells [112,113,114]. This unchecked inflammatory environment where Tregs have lost their immunosuppressive ability and gained expression of inflammatory cytokines is critical for the progression of psoriasis.

5.2. Vitiligo

Vitiligo is an autoimmune disease characterized by the destruction of pigment-producing melanocytes by CD8+ T cells. Vitiligo affects approximately 0.5–2% of people worldwide [115,116]. The disease presents as white patches on the skin, hair, and mucous membranes. The development of vitiligo is attributed to oxidative stress that leads to the activation of the immune system and the attack of melanocytes [117]. Stress proteins, heat shock proteins (Hsp) 70i, and several chemokines, including CXCL9, CXCL10, and CXCL11, play a crucial role in the recruitment of cytotoxic CD8+ T cells to the skin. The extent of melanocyte destruction has been directly correlated with the amount of CD8+ T cell infiltration [117,118,119,120,121,122]. In addition, CD8+ T cells with cytotoxic activity against autologous melanocytes localize at the dermal/epidermal junction [121,123,124]. This loss of self-tolerance suggests that Tregs may be involved in the pathogenesis of vitiligo.
Discrepancies have been reported in the number of Tregs present in vitiligo patients and healthy controls. Depending on the study and study population, patients with non-segmental vitiligo were found to have decreased, unaltered, and increased levels of circulating Tregs [125,126,127,128,129,130,131]. Further inspection of these studies revealed differences in the antibody markers used to quantify Tregs. Using FOXP3+ or CD25+ alone as a marker for Tregs in human skin provides limited information, since activated CD4+ T cells can also express these two markers [128,132]. The small sample size and variations of Treg markers used by these studies could explain the variation in the observed results of circulating Tregs in generalized, non-segmental vitiligo. Identifying a specific marker for Tregs will help resolve this issue, and in doing so, our ability to monitor the development of vitiligo will improve.
Chemokines are a family of small, highly conserved cytokines that mediate critical biological processes, such as chemotaxis, hematopoiesis, and angiogenesis. Several subfamilies of chemokines (e.g., CXC, CC, C, and CX3C) have been defined by the positions of sequentially conserved cysteine residues [133]. Because of their significant involvement in various pathologies, chemokines and their receptors have been the focus of therapeutic discovery for clinical investigations in vitiligo. Several studies demonstrate reduced levels of CCL5/CCR4, CCL22, CCL21, and CCR6 in vitiligo skin, which could explain the failure of circulating Tregs to localize to the skin [126,129,134,135,136,137], thus suggesting that the modulating expression of these chemokines may be potential therapeutic targets in vitiligo. Therefore, it is envisaged that after modulation, these chemokines may re-establish proper immune regulation and self-tolerance by increasing the frequency of Treg migration and skin homing into vitiligo lesions [116,136].
Some Tregs respond to specific environmental cues, such as nutrients, metabolites, and cytokines (Figure 1). In such cases, Treg stability may be altered, leading to plasticity that impacts their suppressive functions [41]. Chen et al. demonstrated that normal Tregs could transition into Th1-like T-bet+IFN-γ+ Tregs in vitiligo patients, and serum from vitiligo patients caused normal Tregs from healthy control subjects to transition into Th1-like Tregs [138,139]. These Th1-like Tregs had attenuated suppressive activity against CD8+ T cells, which was consistent with previous findings [127,130]. Recently, studies have demonstrated that Th1-like Tregs promote tissue-resident memory CD8+ T cells, which have been shown to play a role in vitiligo relapse [140,141,142,143]. Understanding the mechanisms that regulate Treg conversion into Th1-like cells will provide new insights into immune homeostasis and disease pathogenesis, with important therapeutic implications for vitiligo patients.

5.3. Systemic Sclerosis

Affecting approximately 1 in every 10,000 individuals globally, systemic sclerosis (SSc) is a rare autoimmune disease that results in the fibrosis of connective tissues and various organs [144]. The disease commonly begins with Raynaud’s phenomenon, followed by gastro-esophageal reflux [145,146]. Although not yet fully understood, reports suggest that the age of onset may influence prognosis and disease severity as SSc remains a rheumatic disease with the highest mortality rate [147,148].
SSc is multifactorial driven by both genetic [149] and environmental [150] influence. The pathology of this autoimmune disorder is characterized by a dysregulated immune system, consisting of a Th17/Treg imbalance as reported by several studies that noted decreased levels of Tregs and increased levels of Th17 cells in both peripheral blood and skin lesions of SSc patients [89,151,152,153]. The accumulation of circulating pTregs and IL-17-producing T cells may be driven by Treg plasticity towards the Th17 phenotype, leading to the Th17/Treg imbalance. Although the mechanism is largely still unclear, available data suggests a correlation between increases in Th17-associated cytokines (IL-17, IL-21, and IL-22) and SSc severity [89,154]. Some studies propose that Th17 cells induce collagen secretion to promote fibrosis [89,155], while others suggest that Th17 cells promote inflammation and fibrosis [90,156,157,158]. Further, reduced levels of IL-10 present in the serum of SSc patients may be secondary to impaired Treg activity [159,160]. Taken together, Treg plasticity towards Th17-like expression and suppressive function implies an essential role for Tregs in SSc.
IL-4, IL-13, and IL-33 may contribute to the inflammatory phase, but the cellular origin of these cytokines in SSc is unclear [161,162,163]. Evidence suggests that IL-33 can induce increased numbers of Th2-like Tregs cells in the skin of patients with SSc [91,164]. Although the mechanism is not yet fully understood, Slobodin et al. reported a positive correlation between CD4+CD25brightFoxp3+ Tregs and SSc disease severity [165]. Further research into the mechanism by which IL-33 regulates Treg transdifferentiation and plasticity are, therefore, warranted.
Additional studies observed clinical improvement in SSc patients treated with Fresolimumab, an antibody that targets TGF-β-producing Tregs [166]. In aggregate, studies support the premise that Tregs fail to produce inhibitory cytokines or suppress Teffs in SSc, but the underlying mechanisms of how this occurs are unclear. Future work needs to address how the failure of Treg-mediated immune suppression contributes to SSc.

6. The Role of Tregs in Transplantation and Skin Cancer

Following transplantation, host immune defenses can recognize allografts as foreign and mount an immune attack against the graft [167]. The excess secretion of cytokines, chemokines, and other effector molecules further amplifies alloimmune responses, resulting in graft rejection [168]. To promote tolerance, immunosuppression regimens have been integrated into transplant care and management, resulting in drastic improvements in postoperative outcomes [169,170]. Immunosuppressants help re-educate the immune system during transplantation by suppressing immune attacks against the foreign graft [171]. The suppression of the immune system, while essential for transplant success, increases the likelihood of various complications [172,173]. By inhibiting Teffs and enhancing Tregs activation, immunosuppressive therapies can dampen the immune response to various pathogens and tumor cells [174].
The ratio of Tregs versus CD8+ T cells affects the success of transplantation, with higher Treg/CD8+ T cell ratios associated with improved transplant tolerance [12,175,176]. Given the role of Tregs in maintaining tolerance to self and foreign antigens, extensive work has been directed toward the utilization of Treg-based cellular therapies to optimize transplant survival. For example, multiple studies have considered the potential use of pharmacologically induced tolerogenic Tregs in post-transplant care [177]. Interestingly, the same mechanisms by which the increased ratio of Tregs can dampen immune responses are also the ones that may contribute to worse clinical outcomes following transplantation, such as the increased risk of cancer [178].
Immunosuppressed patients are at an increased risk for developing numerous malignancies, with skin cancer accounting for nearly 40% of malignancies in organ transplant recipients [178]. Prior studies have identified an increased presence of Tregs surrounding the tumor and lymphoid tissues that drain the tumor [179,180]. Furthermore, an increased Treg/CD8+ T cell ratio has been suggested to increase the likelihood of tumor evasion [181,182,183]. By secreting a wide array of anti-inflammatory cytokines, including IL-10, IL-35, and TGF- β , Tregs can prevent pro-inflammatory and anti-tumor responses [176,184]. Consequently, increased T cells predict worse prognoses for various cancer patients [180,185]. Below, we review the two most commonly reported skin cancers following transplantation, namely squamous cell carcinoma (SCC) and basal cell carcinoma (BCC), focusing specifically on the involvement of Tregs.

6.1. Squamous Cell Carcinoma

Cutaneous squamous cell carcinoma (cSCC) accounts for the highest proportion of post-transplant skin cancers [186]. Immunosuppression not only increases the risk of cSCC development but also contributes to worsened cSCC severity. Observations of immunosuppressed individuals demonstrate a more aggressive cSCC phenotype when compared to immunocompetent patients [187]. Transplant patients have an estimated 65 to 108 times higher risk of cSCC development than the general population [188]. Additionally, organ transplant recipients with cSCC have a 60 to 250 times greater risk of mortality from SCCs than immunocompetent individuals [189,190,191,192,193].
The high numbers of Tregs are associated with the development of cSCC in renal transplant patients [178]. In a follow-up study, analysis of the Treg-specific demethylated regions confirmed that kidney transplant recipients with a prior history of developing a cSCC had a higher proportion of Tregs than cytotoxic T cells in the immune microenvironment [194]. Using single cell TCR sequencing, Frazzette et al. demonstrated that immunosuppressed patients had a lower proportions of cytotoxic T cells in comparison to immunocompetent patients [195]. In addition, a distinct population of CD8+FOXP3+ T cells that had not previously been observed in cSCC were also described [195]. These unique Treg subpopulations expressed cytotoxic molecules, such as perforin [196,197]. Further studies are needed to elucidate the role of these CD8+ T cells as they may offer a unique novel immunotherapeutic avenue for the treatment of cSCC in transplant recipients.

6.2. Basal Cell Carcinoma

BCC is the second most common malignancy in solid organ transplant recipients (SOTRs) with SOTRs having a 10-fold higher risk compared to immunocompetent individuals [191]. Interestingly, a clinicopathologic study of 176 cases revealed that BCCs in SOTRs have distinct clinical characteristics compared to immunocompetent patients [198]. For example, even though BCCs in immunocompetent individuals and organ transplant recipients are typically seen on the head and neck, there is a higher percentage of SOTRs with BCCs that are localized in sun-protected sites, such as genitalia and the axilla, which are absent in immunocompetent individuals [198].
A closer histological examination of BCCs indicated that the peritumoral inflammatory cell infiltrates were significantly lower in SOTRs compared immunocompetent patients. Additionally, peritumoral skin showed a higher concentration of Tregs, while normal, non-UV-exposed buttock skin lacked Treg expression [198]. In a study by Omland et al., Tregs expressing CCL17, CCL18, and CCL22 were found accumulated in BCC tumors [199]. These chemokines are involved in the recruitment of Tregs in solid cancers [199]. Additionally, genome-wide association analyses (GWA) and functional interaction network analyses have revealed the enrichment of risk variants that function in an immunosuppressive regulatory network, which can impair immune surveillance and effective antitumor immunity [200]. In addition, the GWA data also revealed a global enrichment of genes linked to Treg-cell biology, underlining the importance of Tregs for BCC development.

7. Regulatory T Cell Therapies

Despite the many complications, immunosuppressive drugs remain the cornerstone of transplant medicine and many autoimmune conditions [201,202]. Non-specific actions of these drugs lead to unwanted side effects, such as increased risk of infection and malignancy [203,204,205]. Technological advances have helped define and characterize Tregs, increasing our understanding of the balance between Treg plasticity and Treg instability. In recent years, advances in cellular Treg-based therapies have sought to harness Treg’s unique ability to induce immune tolerance. Here, we summarize several promising Treg-targeted therapies and approaches that are currently under trial.

7.1. IL-2 Based Approaches: Low Dose IL-2 and IL-2 Complexes

IL-2 was first described by its ability to mediate T cell growth [48] and was later found to be involved in several other immune pathways involving natural killer cells and CD8+ T cell cytolytic activity [206], CD4+ T cell differentiation, and Treg expansion [207]. The role of IL-2 has proven to be critical in Treg proliferation and survival [48], and modifications to the amount of IL-2 present in the microenvironment can alter the Treg:Teff ratio, impacting immune tolerance. Low dose IL-2 approaches are based on the expression of the IL-2 receptor on Treg cells and allow for the preferential expansion of Tregs with low levels of IL-2 [208,209,210]. Further, anti-IL-2 monoclonal antibodies (mAb) can be designed to selectively expand Treg expression [211] and IL-2 fusion proteins (IL-2 bound to the Fc portion of IgG) work by slowing the IL-2 excretion rate [212]. In patients with alopecia areata, low-dose IL-2 treatment expanded Treg cells in the blood and hair follicles, leading to improved hair growth [213]. Klatzmann et al. further demonstrated the potential of low dose IL-2 strategy in the treatment of several other autoimmune diseases, including psoriasis [209]. These outcomes certainly highlight the potential of IL-2 based approaches in targeting autoimmune and inflammatory conditions [214,215].
Early findings from case reports showed promising results of IL-2 agents for psoriasis. For example, use of basiliximab, a chimeric IL-2 mAb, was effective for the treatment of severe psoriasis [216,217]. Additionally, a clinical trial demonstrated a reduction in the psoriasis area and severity index (PASI) by 30% at 8 weeks with use of daclizumab, a humanized IgG1 mAb that blocks the IL-2 receptor by binding to CD25 on T cells [218]. Despite early positive results, α-IL-2 agents have fallen out of favor for treatment of psoriasis due to relatively high toxicity and intermediate treatment response compared to other biologics for psoriasis.
However, the development of α-IL-2 therapeutics remains of interest for other conditions [219] as findings from a recent 24-week study by Yu et al. suggest that IL-2 may be used for SSc without obvious adverse effects. The group confirmed previous findings showing that SSc typically has an imbalance of T cells and showed that low-dose IL-2 therapy can restore the balance of the Th17 to Treg cell ratio, leading to reduced disease activity in SSc patients [220]. Furthermore, many centers have incorporated IL-2 antagonists in post-transplant immunosuppressive regimens [221]. Still, the results are unclear as to whether the use of IL-2 receptor antagonists are associated with improved post-transplant outcomes [221,222]. For example, a meta-analysis by Ali et al., demonstrated that IL-2R antibody induction therapy did not improve the rate of rejection or graft survival for renal transplant recipients on tacrolimus maintenance therapy. Therefore, further investigations and development of randomized controlled trials to examine the use of IL-2 immunosuppressive therapies are needed.

7.2. Adoptive Treg Therapy: Polyclonal and Antigen-Specific Tregs

When compared to conventional immunosuppressive agents, ex vivo generated Tregs have been associated with fewer adverse events when used in the setting of autoimmune diseases and transplant rejection [223,224,225,226]. Trzonkowski et al. were the first to use in vitro expanded Tregs in humans and reported symptomatic relief in patients with graft versus host disease (GvHD). In 2004, this treatment was later used in type 1 diabetes, where it demonstrated efficacy, minimized serious side effects, and showed the reversal of the disease [227]. In more recent years, studies have demonstrated the therapeutic potential of adoptive Treg therapy in cutaneous autoimmune conditions, such as vitiligo and SSc. In 2014, Chatterjee et al. reported disease remission in vitiligo mice models that received adoptive transfer of purified Tregs [228] and phase l/ll clinical trials are currently underway for the use of adoptive Treg therapy in the treatment of SSc (NCT05214014).
The challenge with polyclonal is the large quantity of Tregs needed to reach therapeutic levels. An alternative to polyclonal Tregs is the use of antigen-specific Tregs, a form of adoptive Treg therapy that offers a more targeted solution and requires the injection of fewer cells. The classic approach for designing antigen-specific Tregs utilizes either APCs and distinct antigens or engineered T cell receptors (TCRs) [229]. Although both may be reasonable options, limitations exist in the rate of Treg expansion when using APCs and MHC restrictions when engineering Tregs with TCRs. Thus, chimeric antigen receptor (CAR) technology is a preferred method for engineering antigen-specific Tregs that are non-MHC-restricted [230]. CARs bind specified antigens with high affinity using their extracellular antigen-binding domain comprised of a single chain variable fragment (scFv) [230]. The concept of CARs was first described over 25 years ago by Gross et al. and has been improved upon over time [231]. Today, the technique is being studied in various autoimmune connective tissue conditions [232], holding promise for the potential treatment of SSc [233]. Furthermore, the use of CAR-T-cell (CAR-T) therapy in BCC resulted in partial tumor regression [234]. A number of case reports have also documented the use of CAR-T therapy in post-transplant lymphoproliferative disorders in SOTRs [235,236,237,238,239,240,241,242,243]. As our understanding of CAR-T therapy improves, its diverse applications continue to grow.

8. Future Directions

The profound effects that Treg cell disruption has on immune system homeostasis has led to novel clinical investigations. Current human clinical trials are examining the effect of depleting Tregs as a cancer therapy. Furthermore, investigators are exploring the converse: expanding and transplanting Tregs for treating autoimmune diseases. However, despite significant progress, gaps in our understanding of Treg function within the skin and other organs still exist. Recent advances in single cell RNA sequencing, notably the possibility to analyze the Treg-transcriptome from different sites (tumor, diverse tissues, circulation, and the tumor microenvironment) at the single cell level, will provide unprecedented detail on the heterogeneity of these cells that may be exploited in future therapies. Overall, complete elucidation of the mechanism driving Treg plasticity would allow for the successful development of targeted Treg-based therapies.

Author Contributions

Conceptualization, G.W.A.; formal analysis, N.C.O., K.O. and N.P.N.N.; investigation N.C.O., K.O., N.P.N.N., A.M.N. and G.W.A.; resources, N.C.O., K.O., N.P.N.N. and G.W.A.; data curation, N.C.O., K.O., N.P.N.N. and G.W.A.; writing—original draft preparation, N.C.O., K.O., N.P.N.N., A.M.N. and G.W.A.; writing—review and editing, N.C.O., K.O., N.P.N.N., A.M.N. and G.W.A.; visualization, N.C.O., K.O. and N.P.N.N.; supervision, G.W.A.; project administration, N.C.O.; funding acquisition, G.W.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by NIH K01AR071479 and R01AR081337.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Hori, S.; Nomura, T.; Sakaguchi, S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003, 299, 1057–1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Martin, B.; Banz, A.; Bienvenu, B.; Cordier, C.; Dautigny, N.; Becourt, C.; Lucas, B. Suppression of CD4+ T lymphocyte effector functions by CD4+CD25+ cells in vivo. J. Immunol. 2004, 172, 3391–3398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Stassen, M.; Jonuleit, H.; Muller, C.; Klein, M.; Richter, C.; Bopp, T.; Schmitt, S.; Schmitt, E. Differential regulatory capacity of CD25+ T regulatory cells and preactivated CD25+ T regulatory cells on development, functional activation, and proliferation of Th2 cells. J. Immunol. 2004, 173, 267–274. [Google Scholar] [CrossRef] [Green Version]
  4. Oldenhove, G.; de Heusch, M.; Urbain-Vansanten, G.; Urbain, J.; Maliszewski, C.; Leo, O.; Moser, M. CD4+ CD25+ regulatory T cells control T helper cell type 1 responses to foreign antigens induced by mature dendritic cells in vivo. J. Exp. Med. 2003, 198, 259–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Fontenot, J.D.; Gavin, M.A.; Rudensky, A.Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 2003, 4, 330–336. [Google Scholar] [CrossRef]
  6. Rubtsov, Y.P.; Niec, R.E.; Josefowicz, S.; Li, L.; Darce, J.; Mathis, D.; Benoist, C.; Rudensky, A.Y. Stability of the regulatory T cell lineage in vivo. Science 2010, 329, 1667–1671. [Google Scholar] [CrossRef] [Green Version]
  7. Bailey-Bucktrout, S.L.; Martinez-Llordella, M.; Zhou, X.; Anthony, B.; Rosenthal, W.; Luche, H.; Fehling, H.J.; Bluestone, J.A. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity 2013, 39, 949–962. [Google Scholar] [CrossRef] [Green Version]
  8. Komatsu, N.; Okamoto, K.; Sawa, S.; Nakashima, T.; Oh-hora, M.; Kodama, T.; Tanaka, S.; Bluestone, J.A.; Takayanagi, H. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 2014, 20, 62–68. [Google Scholar] [CrossRef]
  9. Zhou, X.; Bailey-Bucktrout, S.; Jeker, L.T.; Bluestone, J.A. Plasticity of CD4(+) FoxP3(+) T cells. Curr. Opin. Immunol. 2009, 21, 281–285. [Google Scholar] [CrossRef] [Green Version]
  10. Hori, S. Lineage stability and phenotypic plasticity of Foxp3(+) regulatory T cells. Immunol. Rev. 2014, 259, 159–172. [Google Scholar] [CrossRef]
  11. Ohkura, N.; Hamaguchi, M.; Morikawa, H.; Sugimura, K.; Tanaka, A.; Ito, Y.; Osaki, M.; Tanaka, Y.; Yamashita, R.; Nakano, N.; et al. T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development. Immunity 2012, 37, 785–799. [Google Scholar] [CrossRef] [Green Version]
  12. Sakaguchi, S.; Sakaguchi, N.; Asano, M.; Itoh, M.; Toda, M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J. Immunol. 1995, 155, 1151–1164. [Google Scholar] [CrossRef]
  13. Miyara, M.; Yoshioka, Y.; Kitoh, A.; Shima, T.; Wing, K.; Niwa, A.; Parizot, C.; Taflin, C.; Heike, T.; Valeyre, D.; et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009, 30, 899–911. [Google Scholar] [CrossRef] [Green Version]
  14. Seddiki, N.; Santner-Nanan, B.; Tangye, S.G.; Alexander, S.I.; Solomon, M.; Lee, S.; Nanan, R.; Fazekas de Saint Groth, B. Persistence of naive CD45RA+ regulatory T cells in adult life. Blood 2006, 107, 2830–2838. [Google Scholar] [CrossRef] [Green Version]
  15. Valmori, D.; Merlo, A.; Souleimanian, N.E.; Hesdorffer, C.S.; Ayyoub, M. A peripheral circulating compartment of natural naive CD4 Tregs. J. Clin. Investig. 2005, 115, 1953–1962. [Google Scholar] [CrossRef]
  16. Wing, J.B.; Tanaka, A.; Sakaguchi, S. Human FOXP3(+) Regulatory T Cell Heterogeneity and Function in Autoimmunity and Cancer. Immunity 2019, 50, 302–316. [Google Scholar] [CrossRef] [Green Version]
  17. Tan, J.; Taitz, J.; Sun, S.M.; Langford, L.; Ni, D.; Macia, L. Your Regulatory T Cells Are What You Eat: How Diet and Gut Microbiota Affect Regulatory T Cell Development. Front. Nutr. 2022, 9, 878382. [Google Scholar] [CrossRef]
  18. Kanamori, M.; Nakatsukasa, H.; Okada, M.; Lu, Q.; Yoshimura, A. Induced Regulatory T Cells: Their Development, Stability, and Applications. Trends Immunol. 2016, 37, 803–811. [Google Scholar] [CrossRef]
  19. Takimoto, T.; Wakabayashi, Y.; Sekiya, T.; Inoue, N.; Morita, R.; Ichiyama, K.; Takahashi, R.; Asakawa, M.; Muto, G.; Mori, T.; et al. Smad2 and Smad3 are redundantly essential for the TGF-beta-mediated regulation of regulatory T plasticity and Th1 development. J. Immunol. 2010, 185, 842–855. [Google Scholar] [CrossRef] [Green Version]
  20. Sakai, R.; Komai, K.; Iizuka-Koga, M.; Yoshimura, A.; Ito, M. Regulatory T Cells: Pathophysiological Roles and Clinical Applications. Keio J. Med. 2020, 69, 1–15. [Google Scholar] [CrossRef]
  21. Yadav, M.; Louvet, C.; Davini, D.; Gardner, J.M.; Martinez-Llordella, M.; Bailey-Bucktrout, S.; Anthony, B.A.; Sverdrup, F.M.; Head, R.; Kuster, D.J.; et al. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J. Exp. Med. 2012, 209, 1713–1722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Huang, H.; Ma, Y.; Dawicki, W.; Zhang, X.; Gordon, J.R. Comparison of induced versus natural regulatory T cells of the same TCR specificity for induction of tolerance to an environmental antigen. J. Immunol. 2013, 191, 1136–1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Yoshimura, A.; Wakabayashi, Y.; Mori, T. Cellular and molecular basis for the regulation of inflammation by TGF-beta. J. Biochem. 2010, 147, 781–792. [Google Scholar] [CrossRef]
  24. Kashiwagi, I.; Morita, R.; Schichita, T.; Komai, K.; Saeki, K.; Matsumoto, M.; Takeda, K.; Nomura, M.; Hayashi, A.; Kanai, T.; et al. Smad2 and Smad3 Inversely Regulate TGF-beta Autoinduction in Clostridium butyricum-Activated Dendritic Cells. Immunity 2015, 43, 65–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kurebayashi, Y.; Baba, Y.; Minowa, A.; Nadya, N.A.; Azuma, M.; Yoshimura, A.; Koyasu, S.; Nagai, S. TGF-beta-induced phosphorylation of Akt and Foxo transcription factors negatively regulates induced regulatory T cell differentiation. Biochem. Biophys. Res. Commun. 2016, 480, 114–119. [Google Scholar] [CrossRef]
  26. Kretschmer, K.; Apostolou, I.; Hawiger, D.; Khazaie, K.; Nussenzweig, M.C.; von Boehmer, H. Inducing and expanding regulatory T cell populations by foreign antigen. Nat. Immunol 2005, 6, 1219–1227. [Google Scholar] [CrossRef]
  27. Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–341. [Google Scholar] [CrossRef] [Green Version]
  28. Cipolletta, D.; Feuerer, M.; Li, A.; Kamei, N.; Lee, J.; Shoelson, S.E.; Benoist, C.; Mathis, D. PPAR-gamma is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 2012, 486, 549–553. [Google Scholar] [CrossRef] [Green Version]
  29. Schiering, C.; Krausgruber, T.; Chomka, A.; Frohlich, A.; Adelmann, K.; Wohlfert, E.A.; Pott, J.; Griseri, T.; Bollrath, J.; Hegazy, A.N.; et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014, 513, 564–568. [Google Scholar] [CrossRef] [Green Version]
  30. Bovenschen, H.J.; van de Kerkhof, P.C.; van Erp, P.E.; Woestenenk, R.; Joosten, I.; Koenen, H.J. Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J. Investig. Dermatol. 2011, 131, 1853–1860. [Google Scholar] [CrossRef]
  31. Ohnmacht, C.; Park, J.H.; Cording, S.; Wing, J.B.; Atarashi, K.; Obata, Y.; Gaboriau-Routhiau, V.; Marques, R.; Dulauroy, S.; Fedoseeva, M.; et al. Mucosal immunology. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science 2015, 349, 989–993. [Google Scholar] [CrossRef]
  32. Sefik, E.; Geva-Zatorsky, N.; Oh, S.; Konnikova, L.; Zemmour, D.; McGuire, A.M.; Burzyn, D.; Ortiz-Lopez, A.; Lobera, M.; Yang, J.; et al. MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science 2015, 349, 993–997. [Google Scholar] [CrossRef] [Green Version]
  33. Kim, K.S.; Hong, S.W.; Han, D.; Yi, J.; Jung, J.; Yang, B.G.; Lee, J.Y.; Lee, M.; Surh, C.D. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science 2016, 351, 858–863. [Google Scholar] [CrossRef]
  34. Burzyn, D.; Kuswanto, W.; Kolodin, D.; Shadrach, J.L.; Cerletti, M.; Jang, Y.; Sefik, E.; Tan, T.G.; Wagers, A.J.; Benoist, C.; et al. A special population of regulatory T cells potentiates muscle repair. Cell 2013, 155, 1282–1295. [Google Scholar] [CrossRef] [Green Version]
  35. Rosenblum, M.D.; Gratz, I.K.; Paw, J.S.; Lee, K.; Marshak-Rothstein, A.; Abbas, A.K. Response to self antigen imprints regulatory memory in tissues. Nature 2011, 480, 538–542. [Google Scholar] [CrossRef] [Green Version]
  36. Sanchez Rodriguez, R.; Pauli, M.L.; Neuhaus, I.M.; Yu, S.S.; Arron, S.T.; Harris, H.W.; Yang, S.H.; Anthony, B.A.; Sverdrup, F.M.; Krow-Lucal, E.; et al. Memory regulatory T cells reside in human skin. J. Clin. Investig. 2014, 124, 1027–1036. [Google Scholar] [CrossRef] [Green Version]
  37. Ali, N.; Zirak, B.; Rodriguez, R.S.; Pauli, M.L.; Truong, H.A.; Lai, K.; Ahn, R.; Corbin, K.; Lowe, M.M.; Scharschmidt, T.C.; et al. Regulatory T Cells in Skin Facilitate Epithelial Stem Cell Differentiation. Cell 2017, 169, 1119–1129.e1111. [Google Scholar] [CrossRef] [Green Version]
  38. Nosbaum, A.; Prevel, N.; Truong, H.A.; Mehta, P.; Ettinger, M.; Scharschmidt, T.C.; Ali, N.H.; Pauli, M.L.; Abbas, A.K.; Rosenblum, M.D. Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing. J. Immunol. 2016, 196, 2010–2014. [Google Scholar] [CrossRef] [Green Version]
  39. Semjen, A.; Garcia-Colera, A.; Requin, J. On controlling force and time in rhythmic movement sequences: The effect of stress location. Ann. N. Y. Acad. Sci. 1984, 423, 168–182. [Google Scholar] [CrossRef]
  40. Shevyrev, D.; Tereshchenko, V. Treg Heterogeneity, Function, and Homeostasis. Front. Immunol. 2019, 10, 3100. [Google Scholar] [CrossRef]
  41. Shi, H.; Chi, H. Metabolic Control of Treg Cell Stability, Plasticity, and Tissue-Specific Heterogeneity. Front. Immunol. 2019, 10, 2716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Rueda, C.M.; Jackson, C.M.; Chougnet, C.A. Regulatory T-Cell-Mediated Suppression of Conventional T-Cells and Dendritic Cells by Different cAMP Intracellular Pathways. Front. Immunol. 2016, 7, 216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hope, C.M.; Coates, P.T.; Carroll, R.P. Immune profiling and cancer post transplantation. World J. Nephrol. 2015, 4, 41–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Welzl, K.; Weinberger, B.; Kronbichler, A.; Sturm, G.; Kern, G.; Mayer, G.; Grubeck-Loebenstein, B.; Koppelstaetter, C. How immunosuppressive therapy affects T cells from kidney transplanted patients of different age: The role of latent cytomegalovirus infection. Clin. Exp. Immunol. 2014, 176, 112–119. [Google Scholar] [CrossRef] [PubMed]
  45. Liberman, A.C.; Budzinski, M.L.; Sokn, C.; Gobbini, R.P.; Steininger, A.; Arzt, E. Regulatory and Mechanistic Actions of Glucocorticoids on T and Inflammatory Cells. Front. Endocrinol. (Lausanne) 2018, 9, 235. [Google Scholar] [CrossRef] [PubMed]
  46. Campbell, D.J.; Koch, M.A. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat. Rev. Immunol. 2011, 11, 119–130. [Google Scholar] [CrossRef] [Green Version]
  47. Barron, L.; Dooms, H.; Hoyer, K.K.; Kuswanto, W.; Hofmann, J.; O’Gorman, W.E.; Abbas, A.K. Cutting edge: Mechanisms of IL-2-dependent maintenance of functional regulatory T cells. J. Immunol. 2010, 185, 6426–6430. [Google Scholar] [CrossRef] [Green Version]
  48. Boyman, O.; Sprent, J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat. Rev. Immunol. 2012, 12, 180–190. [Google Scholar] [CrossRef]
  49. Yu, A.; Malek, T.R. Selective availability of IL-2 is a major determinant controlling the production of CD4+CD25+Foxp3+ T regulatory cells. J. Immunol. 2006, 177, 5115–5121. [Google Scholar] [CrossRef] [Green Version]
  50. Sadlack, B.; Merz, H.; Schorle, H.; Schimpl, A.; Feller, A.C.; Horak, I. Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene. Cell 1993, 75, 253–261. [Google Scholar] [CrossRef]
  51. Willerford, D.M.; Chen, J.; Ferry, J.A.; Davidson, L.; Ma, A.; Alt, F.W. Interleukin-2 receptor alpha chain regulates the size and content of the peripheral lymphoid compartment. Immunity 1995, 3, 521–530. [Google Scholar] [CrossRef] [Green Version]
  52. Chinen, T.; Kannan, A.K.; Levine, A.G.; Fan, X.; Klein, U.; Zheng, Y.; Gasteiger, G.; Feng, Y.; Fontenot, J.D.; Rudensky, A.Y. An essential role for the IL-2 receptor in Treg cell function. Nat. Immunol. 2016, 17, 1322–1333. [Google Scholar] [CrossRef]
  53. Grossman, W.J.; Verbsky, J.W.; Barchet, W.; Colonna, M.; Atkinson, J.P.; Ley, T.J. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 2004, 21, 589–601. [Google Scholar] [CrossRef] [Green Version]
  54. Gondek, D.C.; Lu, L.F.; Quezada, S.A.; Sakaguchi, S.; Noelle, R.J. Cutting edge: Contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J. Immunol. 2005, 174, 1783–1786. [Google Scholar] [CrossRef] [Green Version]
  55. Ren, X.; Ye, F.; Jiang, Z.; Chu, Y.; Xiong, S.; Wang, Y. Involvement of cellular death in TRAIL/DR5-dependent suppression induced by CD4(+)CD25(+) regulatory T cells. Cell Death Differ. 2007, 14, 2076–2084. [Google Scholar] [CrossRef] [Green Version]
  56. Bodmer, J.L.; Holler, N.; Reynard, S.; Vinciguerra, P.; Schneider, P.; Juo, P.; Blenis, J.; Tschopp, J. TRAIL receptor-2 signals apoptosis through FADD and caspase-8. Nat. Cell Biol. 2000, 2, 241–243. [Google Scholar] [CrossRef]
  57. Schmidt, A.; Oberle, N.; Krammer, P.H. Molecular mechanisms of treg-mediated T cell suppression. Front. Immunol. 2012, 3, 51. [Google Scholar] [CrossRef] [Green Version]
  58. Boks, M.A.; Kager-Groenland, J.R.; Haasjes, M.S.; Zwaginga, J.J.; van Ham, S.M.; ten Brinke, A. IL-10-generated tolerogenic dendritic cells are optimal for functional regulatory T cell induction—A comparative study of human clinical-applicable DC. Clin. Immunol. 2012, 142, 332–342. [Google Scholar] [CrossRef]
  59. Strobl, H.; Knapp, W. TGF-beta1 regulation of dendritic cells. Microbes Infect. 1999, 1, 1283–1290. [Google Scholar] [CrossRef]
  60. Wallet, M.A.; Sen, P.; Tisch, R. Immunoregulation of dendritic cells. Clin. Med. Res. 2005, 3, 166–175. [Google Scholar] [CrossRef]
  61. Safinia, N.; Scotta, C.; Vaikunthanathan, T.; Lechler, R.I.; Lombardi, G. Regulatory T Cells: Serious Contenders in the Promise for Immunological Tolerance in Transplantation. Front. Immunol. 2015, 6, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Thepmalee, C.; Panya, A.; Junking, M.; Chieochansin, T.; Yenchitsomanus, P.T. Inhibition of IL-10 and TGF-beta receptors on dendritic cells enhances activation of effector T-cells to kill cholangiocarcinoma cells. Hum. Vaccin. Immunother. 2018, 14, 1423–1431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Kearley, J.; Barker, J.E.; Robinson, D.S.; Lloyd, C.M. Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4+CD25+ regulatory T cells is interleukin 10 dependent. J. Exp. Med. 2005, 202, 1539–1547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Onishi, Y.; Fehervari, Z.; Yamaguchi, T.; Sakaguchi, S. Foxp3+ natural regulatory T cells preferentially form aggregates on dendritic cells in vitro and actively inhibit their maturation. Proc. Natl. Acad. Sci. USA 2008, 105, 10113–10118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Akkaya, B.; Oya, Y.; Akkaya, M.; Al Souz, J.; Holstein, A.H.; Kamenyeva, O.; Kabat, J.; Matsumura, R.; Dorward, D.W.; Glass, D.D.; et al. Regulatory T cells mediate specific suppression by depleting peptide-MHC class II from dendritic cells. Nat. Immunol. 2019, 20, 218–231. [Google Scholar] [CrossRef]
  66. Fallarino, F.; Grohmann, U.; Hwang, K.W.; Orabona, C.; Vacca, C.; Bianchi, R.; Belladonna, M.L.; Fioretti, M.C.; Alegre, M.L.; Puccetti, P. Modulation of tryptophan catabolism by regulatory T cells. Nat. Immunol. 2003, 4, 1206–1212. [Google Scholar] [CrossRef]
  67. Fallarino, F.; Grohmann, U.; You, S.; McGrath, B.C.; Cavener, D.R.; Vacca, C.; Orabona, C.; Bianchi, R.; Belladonna, M.L.; Volpi, C.; et al. The combined effects of tryptophan starvation and tryptophan catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J. Immunol. 2006, 176, 6752–6761. [Google Scholar] [CrossRef] [Green Version]
  68. Sharma, M.D.; Baban, B.; Chandler, P.; Hou, D.Y.; Singh, N.; Yagita, H.; Azuma, M.; Blazar, B.R.; Mellor, A.L.; Munn, D.H. Plasmacytoid dendritic cells from mouse tumor-draining lymph nodes directly activate mature Tregs via indoleamine 2,3-dioxygenase. J. Clin. Investig. 2007, 117, 2570–2582. [Google Scholar] [CrossRef] [Green Version]
  69. Sharma, M.D.; Hou, D.Y.; Liu, Y.; Koni, P.A.; Metz, R.; Chandler, P.; Mellor, A.L.; He, Y.; Munn, D.H. Indoleamine 2,3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood 2009, 113, 6102–6111. [Google Scholar] [CrossRef] [Green Version]
  70. Puccetti, P.; Grohmann, U. IDO and regulatory T cells: A role for reverse signalling and non-canonical NF-kappaB activation. Nat. Rev. Immunol. 2007, 7, 817–823. [Google Scholar] [CrossRef]
  71. Tagami, H. Triggering factors. Clin. Dermatol. 1997, 15, 677–685. [Google Scholar] [CrossRef]
  72. Ho, P.; Bruce, I.N.; Silman, A.; Symmons, D.; Newman, B.; Young, H.; Griffiths, C.E.; John, S.; Worthington, J.; Barton, A. Evidence for common genetic control in pathways of inflammation for Crohn’s disease and psoriatic arthritis. Arthritis Rheum. 2005, 52, 3596–3602. [Google Scholar] [CrossRef]
  73. Conrad, C.; Boyman, O.; Tonel, G.; Tun-Kyi, A.; Laggner, U.; de Fougerolles, A.; Kotelianski, V.; Gardner, H.; Nestle, F.O. Alpha1beta1 integrin is crucial for accumulation of epidermal T cells and the development of psoriasis. Nat. Med. 2007, 13, 836–842. [Google Scholar] [CrossRef]
  74. Love, T.J.; Zhu, Y.; Zhang, Y.; Wall-Burns, L.; Ogdie, A.; Gelfand, J.M.; Choi, H.K. Obesity and the risk of psoriatic arthritis: A population-based study. Ann. Rheum. Dis. 2012, 71, 1273–1277. [Google Scholar] [CrossRef] [Green Version]
  75. El-Boghdady, N.A.; Ismail, M.F.; Abd-Alhameed, M.F.; Ahmed, A.S.; Ahmed, H.H. Bidirectional Association Between Psoriasis and Obesity: Benefits and Risks. J. Interferon Cytokine Res. 2018, 38, 12–19. [Google Scholar] [CrossRef]
  76. Ogawa, E.; Sato, Y.; Minagawa, A.; Okuyama, R. Pathogenesis of psoriasis and development of treatment. J. Dermatol. 2018, 45, 264–272. [Google Scholar] [CrossRef] [Green Version]
  77. Nguyen, U.D.T.; Zhang, Y.; Lu, N.; Louie-Gao, Q.; Niu, J.; Ogdie, A.; Gelfand, J.M.; LaValley, M.P.; Dubreuil, M.; Sparks, J.A.; et al. Smoking paradox in the development of psoriatic arthritis among patients with psoriasis: A population-based study. Ann. Rheum. Dis. 2018, 77, 119–123. [Google Scholar] [CrossRef]
  78. Gouirand, V.; Habrylo, I.; Rosenblum, M.D. Regulatory T Cells and Inflammatory Mediators in Autoimmune Disease. J. Investig. Dermatol. 2022, 142, 774–780. [Google Scholar] [CrossRef]
  79. Marie, J.; Kovacs, D.; Pain, C.; Jouary, T.; Cota, C.; Vergier, B.; Picardo, M.; Taieb, A.; Ezzedine, K.; Cario-Andre, M. Inflammasome activation and vitiligo/nonsegmental vitiligo progression. Br. J. Dermatol. 2014, 170, 816–823. [Google Scholar] [CrossRef]
  80. Sushama, S.; Dixit, N.; Gautam, R.K.; Arora, P.; Khurana, A.; Anubhuti, A. Cytokine profile (IL-2, IL-6, IL-17, IL-22, and TNF-alpha) in vitiligo-New insight into pathogenesis of disease. J. Cosmet. Dermatol. 2019, 18, 337–341. [Google Scholar] [CrossRef]
  81. Dong, J.; An, X.; Zhong, H.; Wang, Y.; Shang, J.; Zhou, J. Interleukin-22 participates in the inflammatory process of vitiligo. Oncotarget 2017, 8, 109161–109174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Chen, X.; Guo, W.; Chang, Y.; Chen, J.; Kang, P.; Yi, X.; Cui, T.; Guo, S.; Xiao, Q.; Jian, Z.; et al. Oxidative stress-induced IL-15 trans-presentation in keratinocytes contributes to CD8(+) T cells activation via JAK-STAT pathway in vitiligo. Free Radic. Biol. Med. 2019, 139, 80–91. [Google Scholar] [CrossRef] [PubMed]
  83. Tomaszewska, K.; Kozlowska, M.; Kaszuba, A.; Lesiak, A.; Narbutt, J.; Zalewska-Janowska, A. Increased Serum Levels of IFN-gamma, IL-1beta, and IL-6 in Patients with Alopecia Areata and Nonsegmental Vitiligo. Oxid. Med. Cell. Longev. 2020, 2020, 5693572. [Google Scholar] [CrossRef] [PubMed]
  84. Atwa, M.A.; Ali, S.M.M.; Youssef, N.; Mahmoud Marie, R.E. Elevated serum level of interleukin-15 in vitiligo patients and its correlation with disease severity but not activity. J. Cosmet Dermatol. 2021, 20, 2640–2644. [Google Scholar] [CrossRef]
  85. Custurone, P.; Di Bartolomeo, L.; Irrera, N.; Borgia, F.; Altavilla, D.; Bitto, A.; Pallio, G.; Squadrito, F.; Vaccaro, M. Role of Cytokines in Vitiligo: Pathogenesis and Possible Targets for Old and New Treatments. Int. J. Mol. Sci. 2021, 22, 11429. [Google Scholar] [CrossRef]
  86. Bhatia, S.; Khaitan, B.K.; Gupta, V.; Khandpur, S.; Sahni, K.; Sreenivas, V. Efficacy of NB-UVB in Progressive Versus Non-Progressive Non-Segmental Vitiligo: A Prospective Comparative Study. Indian Dermatol. Online J. 2021, 12, 701–705. [Google Scholar] [CrossRef]
  87. Kim, S.R.; Heaton, H.; Liu, L.Y.; King, B.A. Rapid Repigmentation of Vitiligo Using Tofacitinib Plus Low-Dose, Narrowband UV-B Phototherapy. JAMA Dermatol. 2018, 154, 370–371. [Google Scholar] [CrossRef]
  88. Krasimirova, E.; Velikova, T.; Ivanova-Todorova, E.; Tumangelova-Yuzeir, K.; Kalinova, D.; Boyadzhieva, V.; Stoilov, N.; Yoneva, T.; Rashkov, R.; Kyurkchiev, D. Treg/Th17 cell balance and phytohaemagglutinin activation of T lymphocytes in peripheral blood of systemic sclerosis patients. World J. Exp. Med. 2017, 7, 84–96. [Google Scholar] [CrossRef]
  89. Yang, X.; Yang, J.; Xing, X.; Wan, L.; Li, M. Increased frequency of Th17 cells in systemic sclerosis is related to disease activity and collagen overproduction. Arthritis Res. Ther. 2014, 16, R4. [Google Scholar] [CrossRef] [Green Version]
  90. Xing, X.; Li, A.; Tan, H.; Zhou, Y. IFN-gamma(+) IL-17(+) Th17 cells regulate fibrosis through secreting IL-21 in systemic scleroderma. J. Cell. Mol. Med. 2020, 24, 13600–13608. [Google Scholar] [CrossRef]
  91. MacDonald, K.G.; Dawson, N.A.J.; Huang, Q.; Dunne, J.V.; Levings, M.K.; Broady, R. Regulatory T cells produce profibrotic cytokines in the skin of patients with systemic sclerosis. J. Allergy Clin. Immunol. 2015, 135, 946–955.e949. [Google Scholar] [CrossRef]
  92. Deegan, A.E. Bacterial endocarditis. J. Am. Dent. Assoc. 1997, 128, 1628. [Google Scholar] [CrossRef]
  93. Frantz, C.; Cauvet, A.; Durand, A.; Gonzalez, V.; Pierre, R.; Do Cruzeiro, M.; Bailly, K.; Andrieu, M.; Orvain, C.; Avouac, J.; et al. Driving Role of Interleukin-2-Related Regulatory CD4+ T Cell Deficiency in the Development of Lung Fibrosis and Vascular Remodeling in a Mouse Model of Systemic Sclerosis. Arthritis Rheumatol. 2022, 74, 1387–1398. [Google Scholar] [CrossRef]
  94. Ingegnoli, F.; Ughi, N.; Mihai, C. Update on the epidemiology, risk factors, and disease outcomes of systemic sclerosis. Best Pract. Res. Clin. Rheumatol. 2018, 32, 223–240. [Google Scholar] [CrossRef]
  95. Bukiri, H.; Volkmann, E.R. Current advances in the treatment of systemic sclerosis. Curr. Opin. Pharmacol. 2022, 64, 102211. [Google Scholar] [CrossRef]
  96. Higuchi, T.; Kawaguchi, Y.; Takagi, K.; Tochimoto, A.; Ota, Y.; Katsumata, Y.; Ichida, H.; Hanaoka, M.; Kawasumi, H.; Tochihara, M.; et al. Sildenafil attenuates the fibrotic phenotype of skin fibroblasts in patients with systemic sclerosis. Clin. Immunol. 2015, 161, 333–338. [Google Scholar] [CrossRef]
  97. Parisi, S.; Bruzzone, M.; Centanaro Di Vittorio, C.; Lagana, A.; Peroni, C.L.; Fusaro, E. Efficacy of bosentan in the treatment of Raynaud’s phenomenon in patients with systemic sclerosis never treated with prostanoids. Reumatismo 2014, 65, 286–291. [Google Scholar] [CrossRef] [Green Version]
  98. Christophers, E. Psoriasis--epidemiology and clinical spectrum. Clin. Exp. Dermatol. 2001, 26, 314–320. [Google Scholar] [CrossRef]
  99. Ni, X.; Lai, Y. Keratinocyte: A trigger or an executor of psoriasis? J. Leukoc. Biol. 2020, 108, 485–491. [Google Scholar] [CrossRef]
  100. Farber, E.M.; Nall, M.L. The natural history of psoriasis in 5600 patients. Dermatologica 1974, 148, 1–18. [Google Scholar] [CrossRef]
  101. Chandran, V.; Raychaudhuri, S.P. Geoepidemiology and environmental factors of psoriasis and psoriatic arthritis. J. Autoimmun. 2010, 34, J314–J321. [Google Scholar] [CrossRef]
  102. Ghei, O.K.; Kay, W.W. Regulation of C4-dicarboxylic acid transport in Bacillus subtilis. Can. J. Microbiol. 1975, 21, 527–536. [Google Scholar] [CrossRef] [PubMed]
  103. Brandrup, F.; Holm, N.; Grunnet, N.; Henningsen, K.; Hansen, H.E. Psoriasis in monozygotic twins: Variations in expression in individuals with identical genetic constitution. Acta Derm. Venereol. 1982, 62, 229–236. [Google Scholar] [PubMed]
  104. Swanbeck, G.; Inerot, A.; Martinsson, T.; Wahlstrom, J. A population genetic study of psoriasis. Br. J. Dermatol. 1994, 131, 32–39. [Google Scholar] [CrossRef] [PubMed]
  105. Kimberling, W.; Dobson, R.L. The inheritance of psoriasis. J. Investig. Dermatol. 1973, 60, 538–540. [Google Scholar] [CrossRef] [Green Version]
  106. Harden, J.L.; Krueger, J.G.; Bowcock, A.M. The immunogenetics of Psoriasis: A comprehensive review. J. Autoimmun. 2015, 64, 66–73. [Google Scholar] [CrossRef] [Green Version]
  107. Sugiyama, H.; Gyulai, R.; Toichi, E.; Garaczi, E.; Shimada, S.; Stevens, S.R.; McCormick, T.S.; Cooper, K.D. Dysfunctional blood and target tissue CD4+CD25high regulatory T cells in psoriasis: Mechanism underlying unrestrained pathogenic effector T cell proliferation. J. Immunol. 2005, 174, 164–173. [Google Scholar] [CrossRef] [Green Version]
  108. Liu, Y.; Zhang, C.; Li, B.; Yu, C.; Bai, X.; Xiao, C.; Wang, L.; Dang, E.; Yang, L.; Wang, G. A novel role of IL-17A in contributing to the impaired suppressive function of Tregs in psoriasis. J. Dermatol. Sci. 2021, 101, 84–92. [Google Scholar] [CrossRef]
  109. Kim, C.H. FOXP3 and its role in the immune system. Adv. Exp. Med. Biol. 2009, 665, 17–29. [Google Scholar] [CrossRef]
  110. Georgiev, P.; Charbonnier, L.M.; Chatila, T.A. Regulatory T Cells: The Many Faces of Foxp3. J. Clin. Immunol. 2019, 39, 623–640. [Google Scholar] [CrossRef]
  111. von Knethen, A.; Heinicke, U.; Weigert, A.; Zacharowski, K.; Brune, B. Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells. Int. J. Mol. Sci. 2020, 21, 2356. [Google Scholar] [CrossRef] [Green Version]
  112. Yang, L.; Li, B.; Dang, E.; Jin, L.; Fan, X.; Wang, G. Impaired function of regulatory T cells in patients with psoriasis is mediated by phosphorylation of STAT3. J. Dermatol. Sci. 2016, 81, 85–92. [Google Scholar] [CrossRef]
  113. Goodman, W.A.; Levine, A.D.; Massari, J.V.; Sugiyama, H.; McCormick, T.S.; Cooper, K.D. IL-6 signaling in psoriasis prevents immune suppression by regulatory T cells. J. Immunol. 2009, 183, 3170–3176. [Google Scholar] [CrossRef] [Green Version]
  114. Peluso, I.; Fantini, M.C.; Fina, D.; Caruso, R.; Boirivant, M.; MacDonald, T.T.; Pallone, F.; Monteleone, G. IL-21 counteracts the regulatory T cell-mediated suppression of human CD4+ T lymphocytes. J. Immunol. 2007, 178, 732–739. [Google Scholar] [CrossRef] [Green Version]
  115. Wang, Y.; Li, S.; Li, C. Clinical Features, Immunopathogenesis, and Therapeutic Strategies in Vitiligo. Clin. Rev. Allergy Immunol. 2021, 61, 299–323. [Google Scholar] [CrossRef]
  116. Gellatly, K.J.; Strassner, J.P.; Essien, K.; Refat, M.A.; Murphy, R.L.; Coffin-Schmitt, A.; Pandya, A.G.; Tovar-Garza, A.; Frisoli, M.L.; Fan, X.; et al. scRNA-seq of human vitiligo reveals complex networks of subclinical immune activation and a role for CCR5 in Treg function. Sci. Transl. Med. 2021, 13, eabd8995. [Google Scholar] [CrossRef]
  117. Mosenson, J.A.; Zloza, A.; Klarquist, J.; Barfuss, A.J.; Guevara-Patino, J.A.; Poole, I.C. HSP70i is a critical component of the immune response leading to vitiligo. Pigment Cell Melanoma Res. 2012, 25, 88–98. [Google Scholar] [CrossRef] [Green Version]
  118. Ahn, Y.; Seo, J.; Lee, E.J.; Kim, J.Y.; Park, M.Y.; Hwang, S.; Almurayshid, A.; Lim, B.J.; Yu, J.W.; Oh, S.H. ATP-P2X7-Induced Inflammasome Activation Contributes to Melanocyte Death and CD8(+) T-Cell Trafficking to the Skin in Vitiligo. J. Investig. Dermatol. 2020, 140, 1794–1804.e4. [Google Scholar] [CrossRef]
  119. Richmond, J.M.; Bangari, D.S.; Essien, K.I.; Currimbhoy, S.D.; Groom, J.R.; Pandya, A.G.; Youd, M.E.; Luster, A.D.; Harris, J.E. Keratinocyte-Derived Chemokines Orchestrate T-Cell Positioning in the Epidermis during Vitiligo and May Serve as Biomarkers of Disease. J. Investig. Dermatol. 2017, 137, 350–358. [Google Scholar] [CrossRef] [Green Version]
  120. Tulic, M.K.; Cavazza, E.; Cheli, Y.; Jacquel, A.; Luci, C.; Cardot-Leccia, N.; Hadhiri-Bzioueche, H.; Abbe, P.; Gesson, M.; Sormani, L.; et al. Innate lymphocyte-induced CXCR3B-mediated melanocyte apoptosis is a potential initiator of T-cell autoreactivity in vitiligo. Nat. Commun. 2019, 10, 2178. [Google Scholar] [CrossRef]
  121. van den Boorn, J.G.; Konijnenberg, D.; Dellemijn, T.A.; van der Veen, J.P.; Bos, J.D.; Melief, C.J.; Vyth-Dreese, F.A.; Luiten, R.M. Autoimmune destruction of skin melanocytes by perilesional T cells from vitiligo patients. J. Investig. Dermatol. 2009, 129, 2220–2232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Li, S.; Zhu, G.; Yang, Y.; Jian, Z.; Guo, S.; Dai, W.; Shi, Q.; Ge, R.; Ma, J.; Liu, L.; et al. Oxidative stress drives CD8(+) T-cell skin trafficking in patients with vitiligo through CXCL16 upregulation by activating the unfolded protein response in keratinocytes. J. Allergy Clin. Immunol. 2017, 140, 177–189.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. van den Wijngaard, R.; Wankowicz-Kalinska, A.; Le Poole, C.; Tigges, B.; Westerhof, W.; Das, P. Local immune response in skin of generalized vitiligo patients. Destruction of melanocytes is associated with the prominent presence of CLA+ T cells at the perilesional site. Lab. Investig. 2000, 80, 1299–1309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mandelcorn-Monson, R.L.; Shear, N.H.; Yau, E.; Sambhara, S.; Barber, B.H.; Spaner, D.; DeBenedette, M.A. Cytotoxic T lymphocyte reactivity to gp100, MelanA/MART-1, and tyrosinase, in HLA-A2-positive vitiligo patients. J. Investig. Dermatol. 2003, 121, 550–556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hegab, D.S.; Attia, M.A. Decreased Circulating T Regulatory Cells in Egyptian Patients with Nonsegmental Vitiligo: Correlation with Disease Activity. Dermatol. Res. Pract. 2015, 2015, 145409. [Google Scholar] [CrossRef]
  126. Eby, J.M.; Kang, H.K.; Tully, S.T.; Bindeman, W.E.; Peiffer, D.S.; Chatterjee, S.; Mehrotra, S.; Le Poole, I.C. CCL22 to Activate Treg Migration and Suppress Depigmentation in Vitiligo. J. Investig. Dermatol. 2015, 135, 1574–1580. [Google Scholar] [CrossRef] [Green Version]
  127. Lili, Y.; Yi, W.; Ji, Y.; Yue, S.; Weimin, S.; Ming, L. Global activation of CD8+ cytotoxic T lymphocytes correlates with an impairment in regulatory T cells in patients with generalized vitiligo. PLoS ONE 2012, 7, e37513. [Google Scholar] [CrossRef] [Green Version]
  128. Zhou, L.; Li, K.; Shi, Y.L.; Hamzavi, I.; Gao, T.W.; Henderson, M.; Huggins, R.H.; Agbai, O.; Mahmoud, B.; Mi, X.; et al. Systemic analyses of immunophenotypes of peripheral T cells in non-segmental vitiligo: Implication of defective natural killer T cells. Pigment Cell Melanoma Res. 2012, 25, 602–611. [Google Scholar] [CrossRef] [Green Version]
  129. Klarquist, J.; Denman, C.J.; Hernandez, C.; Wainwright, D.A.; Strickland, F.M.; Overbeck, A.; Mehrotra, S.; Nishimura, M.I.; Le Poole, I.C. Reduced skin homing by functional Treg in vitiligo. Pigment Cell Melanoma Res. 2010, 23, 276–286. [Google Scholar] [CrossRef]
  130. Ben Ahmed, M.; Zaraa, I.; Rekik, R.; Elbeldi-Ferchiou, A.; Kourda, N.; Belhadj Hmida, N.; Abdeladhim, M.; Karoui, O.; Ben Osman, A.; Mokni, M.; et al. Functional defects of peripheral regulatory T lymphocytes in patients with progressive vitiligo. Pigment Cell Melanoma Res. 2012, 25, 99–109. [Google Scholar] [CrossRef]
  131. Moftah, N.H.; El-Barbary, R.A.; Ismail, M.A.; Ali, N.A. Effect of narrow band-ultraviolet B on CD4(+) CD25(high) FoxP3(+) T-lymphocytes in the peripheral blood of vitiligo patients. Photodermatol. Photoimmunol. Photomed. 2014, 30, 254–261. [Google Scholar] [CrossRef]
  132. Ujiie, H. Regulatory T cells in autoimmune skin diseases. Exp. Dermatol. 2019, 28, 642–646. [Google Scholar] [CrossRef] [Green Version]
  133. Miller, M.C.; Mayo, K.H. Chemokines from a Structural Perspective. Int. J. Mol. Sci. 2017, 18, 88. [Google Scholar] [CrossRef] [Green Version]
  134. Le Poole, I.C.; Mehrotra, S. Replenishing Regulatory T Cells to Halt Depigmentation in Vitiligo. J. Investig. Dermatol. Symp. Proc. 2017, 18, S38–S45. [Google Scholar] [CrossRef] [Green Version]
  135. Klarquist, J.; Tobin, K.; Farhangi Oskuei, P.; Henning, S.W.; Fernandez, M.F.; Dellacecca, E.R.; Navarro, F.C.; Eby, J.M.; Chatterjee, S.; Mehrotra, S.; et al. Ccl22 Diverts T Regulatory Cells and Controls the Growth of Melanoma. Cancer Res. 2016, 76, 6230–6240. [Google Scholar] [CrossRef] [Green Version]
  136. Tembhre, M.K.; Parihar, A.S.; Sharma, V.K.; Sharma, A.; Chattopadhyay, P.; Gupta, S. Alteration in regulatory T cells and programmed cell death 1-expressing regulatory T cells in active generalized vitiligo and their clinical correlation. Br. J. Dermatol. 2015, 172, 940–950. [Google Scholar] [CrossRef]
  137. Essien, K.I.; Katz, E.L.; Strassner, J.P.; Harris, J.E. Regulatory T Cells Require CCR6 for Skin Migration and Local Suppression of Vitiligo. J. Investig. Dermatol. 2022, 142, 3158–3166. [Google Scholar] [CrossRef]
  138. Chen, J.; Wang, X.; Cui, T.; Ni, Q.; Zhang, Q.; Zou, D.; He, K.; Wu, W.; Ma, J.; Wang, Y.; et al. Th1-like Treg in vitiligo: An incompetent regulator in immune tolerance. J. Autoimmun. 2022, 131, 102859. [Google Scholar] [CrossRef]
  139. Wankowicz-Kalinska, A.; van den Wijngaard, R.M.; Tigges, B.J.; Westerhof, W.; Ogg, G.S.; Cerundolo, V.; Storkus, W.J.; Das, P.K. Immunopolarization of CD4+ and CD8+ T cells to Type-1-like is associated with melanocyte loss in human vitiligo. Lab. Investig. 2003, 83, 683–695. [Google Scholar] [CrossRef] [Green Version]
  140. Ferreira, C.; Barros, L.; Baptista, M.; Blankenhaus, B.; Barros, A.; Figueiredo-Campos, P.; Konjar, S.; Laine, A.; Kamenjarin, N.; Stojanovic, A.; et al. Type 1 Treg cells promote the generation of CD8(+) tissue-resident memory T cells. Nat. Immunol. 2020, 21, 766–776. [Google Scholar] [CrossRef]
  141. Richmond, J.M.; Strassner, J.P.; Rashighi, M.; Agarwal, P.; Garg, M.; Essien, K.I.; Pell, L.S.; Harris, J.E. Resident Memory and Recirculating Memory T Cells Cooperate to Maintain Disease in a Mouse Model of Vitiligo. J. Investig. Dermatol. 2019, 139, 769–778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Riding, R.L.; Harris, J.E. The Role of Memory CD8(+) T Cells in Vitiligo. J. Immunol. 2019, 203, 11–19. [Google Scholar] [CrossRef] [PubMed]
  143. Boniface, K.; Jacquemin, C.; Darrigade, A.S.; Dessarthe, B.; Martins, C.; Boukhedouni, N.; Vernisse, C.; Grasseau, A.; Thiolat, D.; Rambert, J.; et al. Vitiligo Skin Is Imprinted with Resident Memory CD8 T Cells Expressing CXCR3. J. Investig. Dermatol. 2018, 138, 355–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Fett, N. Scleroderma: Nomenclature, etiology, pathogenesis, prognosis, and treatments: Facts and controversies. Clin. Dermatol. 2013, 31, 432–437. [Google Scholar] [CrossRef] [PubMed]
  145. Cutolo, M.; Soldano, S.; Smith, V. Pathophysiology of systemic sclerosis: Current understanding and new insights. Expert Rev. Clin. Immunol. 2019, 15, 753–764. [Google Scholar] [CrossRef] [Green Version]
  146. Rongioletti, F.; Kaiser, F.; Cinotti, E.; Metze, D.; Battistella, M.; Calzavara-Pinton, P.G.; Damevska, K.; Girolomoni, G.; Andre, J.; Perrot, J.L.; et al. Scleredema. A multicentre study of characteristics, comorbidities, course and therapy in 44 patients. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 2399–2404. [Google Scholar] [CrossRef]
  147. Nihtyanova, S.I.; Tang, E.C.; Coghlan, J.G.; Wells, A.U.; Black, C.M.; Denton, C.P. Improved survival in systemic sclerosis is associated with better ascertainment of internal organ disease: A retrospective cohort study. QJM 2010, 103, 109–115. [Google Scholar] [CrossRef]
  148. Tyndall, A.J.; Bannert, B.; Vonk, M.; Airo, P.; Cozzi, F.; Carreira, P.E.; Bancel, D.F.; Allanore, Y.; Muller-Ladner, U.; Distler, O.; et al. Causes and risk factors for death in systemic sclerosis: A study from the EULAR Scleroderma Trials and Research (EUSTAR) database. Ann. Rheum. Dis. 2010, 69, 1809–1815. [Google Scholar] [CrossRef] [Green Version]
  149. Romano, E.; Manetti, M.; Guiducci, S.; Ceccarelli, C.; Allanore, Y.; Matucci-Cerinic, M. The genetics of systemic sclerosis: An update. Clin. Exp. Rheumatol. 2011, 29, S75–S86. [Google Scholar]
  150. Katsumoto, T.R.; Whitfield, M.L.; Connolly, M.K. The pathogenesis of systemic sclerosis. Annu. Rev. Pathol. 2011, 6, 509–537. [Google Scholar] [CrossRef]
  151. Radstake, T.R.; van Bon, L.; Broen, J.; Wenink, M.; Santegoets, K.; Deng, Y.; Hussaini, A.; Simms, R.; Cruikshank, W.W.; Lafyatis, R. Increased frequency and compromised function of T regulatory cells in systemic sclerosis (SSc) is related to a diminished CD69 and TGFbeta expression. PLoS ONE 2009, 4, e5981. [Google Scholar] [CrossRef]
  152. Fenoglio, D.; Battaglia, F.; Parodi, A.; Stringara, S.; Negrini, S.; Panico, N.; Rizzi, M.; Kalli, F.; Conteduca, G.; Ghio, M.; et al. Alteration of Th17 and Treg cell subpopulations co-exist in patients affected with systemic sclerosis. Clin. Immunol. 2011, 139, 249–257. [Google Scholar] [CrossRef]
  153. Antiga, E.; Quaglino, P.; Bellandi, S.; Volpi, W.; Del Bianco, E.; Comessatti, A.; Osella-Abate, S.; De Simone, C.; Marzano, A.; Bernengo, M.G.; et al. Regulatory T cells in the skin lesions and blood of patients with systemic sclerosis and morphoea. Br. J. Dermatol. 2010, 162, 1056–1063. [Google Scholar] [CrossRef]
  154. Zhou, Y.; Hou, W.; Xu, K.; Han, D.; Jiang, C.; Mou, K.; Li, Y.; Meng, L.; Lu, S. The elevated expression of Th17-related cytokines and receptors is associated with skin lesion severity in early systemic sclerosis. Hum. Immunol. 2015, 76, 22–29. [Google Scholar] [CrossRef]
  155. Nakashima, T.; Jinnin, M.; Yamane, K.; Honda, N.; Kajihara, I.; Makino, T.; Masuguchi, S.; Fukushima, S.; Okamoto, Y.; Hasegawa, M.; et al. Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts. J. Immunol. 2012, 188, 3573–3583. [Google Scholar] [CrossRef] [Green Version]
  156. Brembilla, N.C.; Montanari, E.; Truchetet, M.E.; Raschi, E.; Meroni, P.; Chizzolini, C. Th17 cells favor inflammatory responses while inhibiting type I collagen deposition by dermal fibroblasts: Differential effects in healthy and systemic sclerosis fibroblasts. Arthritis Res. Ther. 2013, 15, R151. [Google Scholar] [CrossRef] [Green Version]
  157. Lei, L.; Zhao, C.; Qin, F.; He, Z.Y.; Wang, X.; Zhong, X.N. Th17 cells and IL-17 promote the skin and lung inflammation and fibrosis process in a bleomycin-induced murine model of systemic sclerosis. Clin. Exp. Rheumatol. 2016, 34 (Suppl. S100), 14–22. [Google Scholar]
  158. Carvalheiro, T.; Affandi, A.J.; Malvar-Fernandez, B.; Dullemond, I.; Cossu, M.; Ottria, A.; Mertens, J.S.; Giovannone, B.; Bonte-Mineur, F.; Kok, M.R.; et al. Induction of Inflammation and Fibrosis by Semaphorin 4A in Systemic Sclerosis. Arthritis Rheumatol. 2019, 71, 1711–1722. [Google Scholar] [CrossRef] [Green Version]
  159. Almanzar, G.; Klein, M.; Schmalzing, M.; Hilligardt, D.; El Hajj, N.; Kneitz, H.; Wild, V.; Rosenwald, A.; Benoit, S.; Hamm, H.; et al. Disease Manifestation and Inflammatory Activity as Modulators of Th17/Treg Balance and RORC/FoxP3 Methylation in Systemic Sclerosis. Int. Arch. Allergy Immunol. 2016, 171, 141–154. [Google Scholar] [CrossRef]
  160. Ugor, E.; Simon, D.; Almanzar, G.; Pap, R.; Najbauer, J.; Nemeth, P.; Balogh, P.; Prelog, M.; Czirjak, L.; Berki, T. Increased proportions of functionally impaired regulatory T cell subsets in systemic sclerosis. Clin. Immunol. 2017, 184, 54–62. [Google Scholar] [CrossRef]
  161. Hamilton, R.F., Jr.; Parsley, E.; Holian, A. Alveolar macrophages from systemic sclerosis patients: Evidence for IL-4-mediated phenotype changes. Am. J. Physiol. Lung Cell Mol. Physiol. 2004, 286, L1202–L1209. [Google Scholar] [CrossRef] [PubMed]
  162. Fuschiotti, P.; Medsger, T.A., Jr.; Morel, P.A. Effector CD8+ T cells in systemic sclerosis patients produce abnormally high levels of interleukin-13 associated with increased skin fibrosis. Arthritis Rheum. 2009, 60, 1119–1128. [Google Scholar] [CrossRef] [PubMed]
  163. Gasparini, G.; Cozzani, E.; Parodi, A. Interleukin-4 and interleukin-13 as possible therapeutic targets in systemic sclerosis. Cytokine 2020, 125, 154799. [Google Scholar] [CrossRef] [PubMed]
  164. Saigusa, R.; Asano, Y.; Taniguchi, T.; Hirabayashi, M.; Nakamura, K.; Miura, S.; Yamashita, T.; Takahashi, T.; Ichimura, Y.; Toyama, T.; et al. Fli1-haploinsufficient dermal fibroblasts promote skin-localized transdifferentiation of Th2-like regulatory T cells. Arthritis Res. Ther. 2018, 20, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Slobodin, G.; Ahmad, M.S.; Rosner, I.; Peri, R.; Rozenbaum, M.; Kessel, A.; Toubi, E.; Odeh, M. Regulatory T cells (CD4(+)CD25(bright)FoxP3(+)) expansion in systemic sclerosis correlates with disease activity and severity. Cell Immunol. 2010, 261, 77–80. [Google Scholar] [CrossRef]
  166. Rice, L.M.; Padilla, C.M.; McLaughlin, S.R.; Mathes, A.; Ziemek, J.; Goummih, S.; Nakerakanti, S.; York, M.; Farina, G.; Whitfield, M.L.; et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J. Clin. Investig. 2015, 125, 2795–2807. [Google Scholar] [CrossRef]
  167. Marino, J.; Paster, J.; Benichou, G. Allorecognition by T Lymphocytes and Allograft Rejection. Front. Immunol. 2016, 7, 582. [Google Scholar] [CrossRef] [Green Version]
  168. Ingulli, E. Mechanism of cellular rejection in transplantation. Pediatr. Nephrol. 2010, 25, 61–74. [Google Scholar] [CrossRef] [Green Version]
  169. Starzl, T.E. History of clinical transplantation. World J. Surg. 2000, 24, 759–782. [Google Scholar] [CrossRef] [Green Version]
  170. Pilch, N.A.; Bowman, L.J.; Taber, D.J. Immunosuppression trends in solid organ transplantation: The future of individualization, monitoring, and management. Pharmacotherapy 2021, 41, 119–131. [Google Scholar] [CrossRef]
  171. Zdanowicz, M.M. The pharmacology of immunosuppression. Am. J. Pharm. Educ. 2009, 73, 144. [Google Scholar] [CrossRef] [Green Version]
  172. Dantal, J.; Soulillou, J.P. Immunosuppressive drugs and the risk of cancer after organ transplantation. N. Engl. J. Med. 2005, 352, 1371–1373. [Google Scholar] [CrossRef]
  173. Busnach, G.; Piselli, P.; Arbustini, E.; Baccarani, U.; Burra, P.; Carrieri, M.P.; Citterio, F.; De Juli, E.; Bellelli, S.; Pradier, C.; et al. Immunosuppression and cancer: A comparison of risks in recipients of organ transplants and in HIV-positive individuals. Transplant. Proc. 2006, 38, 3533–3535. [Google Scholar] [CrossRef]
  174. Furukawa, A.; Wisel, S.A.; Tang, Q. Impact of Immune-Modulatory Drugs on Regulatory T Cell. Transplantation 2016, 100, 2288–2300. [Google Scholar] [CrossRef] [Green Version]
  175. Bernaldo-de-Quiros, E.; Lopez-Abente, J.; Camino, M.; Gil, N.; Panadero, E.; Lopez-Esteban, R.; Martinez-Bonet, M.; Pion, M.; Correa-Rocha, R. The Presence of a Marked Imbalance Between Regulatory T Cells and Effector T Cells Reveals That Tolerance Mechanisms Could Be Compromised in Heart Transplant Children. Transplant. Direct 2021, 7, e693. [Google Scholar] [CrossRef]
  176. Okeke, E.B.; Uzonna, J.E. The Pivotal Role of Regulatory T Cells in the Regulation of Innate Immune Cells. Front. Immunol. 2019, 10, 680. [Google Scholar] [CrossRef] [Green Version]
  177. Shaban, E.; Bayliss, G.; Malhotra, D.K.; Shemin, D.; Wang, L.J.; Gohh, R.; Dworkin, L.D.; Gong, R. Targeting Regulatory T Cells for Transplant Tolerance: New Insights and Future Perspectives. Kidney Dis. (Basel) 2018, 4, 205–213. [Google Scholar] [CrossRef]
  178. Carroll, R.P.; Segundo, D.S.; Hollowood, K.; Marafioti, T.; Clark, T.G.; Harden, P.N.; Wood, K.J. Immune phenotype predicts risk for posttransplantation squamous cell carcinoma. J. Am. Soc. Nephrol. 2010, 21, 713–722. [Google Scholar] [CrossRef] [Green Version]
  179. Shimizu, J.; Yamazaki, S.; Sakaguchi, S. Induction of tumor immunity by removing CD25+CD4+ T cells: A common basis between tumor immunity and autoimmunity. J. Immunol. 1999, 163, 5211–5218. [Google Scholar] [CrossRef]
  180. Gliwinski, M.; Piotrowska, M.; Iwaszkiewicz-Grzes, D.; Urban-Wojciuk, Z.; Trzonkowski, P. Therapy with CD4(+)CD25(+) T regulatory cells—Should we be afraid of cancer? Contemp. Oncol. (Pozn) 2019, 23, 1–6. [Google Scholar] [CrossRef]
  181. Biller, B.J.; Guth, A.; Burton, J.H.; Dow, S.W. Decreased ratio of CD8+ T cells to regulatory T cells associated with decreased survival in dogs with osteosarcoma. J. Vet. Intern. Med. 2010, 24, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  182. Facciabene, A.; Motz, G.T.; Coukos, G. T-regulatory cells: Key players in tumor immune escape and angiogenesis. Cancer Res. 2012, 72, 2162–2171. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, Y.L.; Fang, J.H.; Lai, M.D.; Shan, Y.S. Depletion of CD4(+)CD25(+) regulatory T cells can promote local immunity to suppress tumor growth in benzo[a]pyrene-induced forestomach carcinoma. World J. Gastroenterol. 2008, 14, 5797–5809. [Google Scholar] [CrossRef] [PubMed]
  184. Tang, Q.; Bluestone, J.A. Regulatory T-cell therapy in transplantation: Moving to the clinic. Cold Spring Harb. Perspect. Med. 2013, 3, a015552. [Google Scholar] [CrossRef] [PubMed]
  185. Shang, B.; Liu, Y.; Jiang, S.J.; Liu, Y. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: A systematic review and meta-analysis. Sci. Rep. 2015, 5, 15179. [Google Scholar] [CrossRef] [Green Version]
  186. Garrett, G.L.; Blanc, P.D.; Boscardin, J.; Lloyd, A.A.; Ahmed, R.L.; Anthony, T.; Bibee, K.; Breithaupt, A.; Cannon, J.; Chen, A.; et al. Incidence of and Risk Factors for Skin Cancer in Organ Transplant Recipients in the United States. JAMA Dermatol. 2017, 153, 296–303. [Google Scholar] [CrossRef] [Green Version]
  187. Tam, S.; Yao, C.; Amit, M.; Gajera, M.; Luo, X.; Treistman, R.; Khanna, A.; Aashiq, M.; Nagarajan, P.; Bell, D.; et al. Association of Immunosuppression With Outcomes of Patients With Cutaneous Squamous Cell Carcinoma of the Head and Neck. JAMA Otolaryngol. Head Neck Surg. 2020, 146, 128–135. [Google Scholar] [CrossRef]
  188. Blue, E.D.; Freeman, S.C.; Lobl, M.B.; Clarey, D.D.; Fredrick, R.L.; Wysong, A.; Whitley, M.J. Cutaneous Squamous Cell Carcinoma Arising in Immunosuppressed Patients: A Systematic Review of Tumor Profiling Studies. JID Innov. 2022, 2, 100126. [Google Scholar] [CrossRef]
  189. Euvrard, S.; Kanitakis, J.; Claudy, A. Skin cancers after organ transplantation. N. Engl. J. Med 2003, 348, 1681–1691. [Google Scholar] [CrossRef] [Green Version]
  190. Bibee, K.; Swartz, A.; Sridharan, S.; Kurten, C.H.L.; Wessel, C.B.; Skinner, H.; Zandberg, D.P. Cutaneous squamous cell carcinoma in the organ transplant recipient. Oral Oncol. 2020, 103, 104562. [Google Scholar] [CrossRef]
  191. Lindelof, B.; Sigurgeirsson, B.; Gabel, H.; Stern, R.S. Incidence of skin cancer in 5356 patients following organ transplantation. Br. J. Dermatol. 2000, 143, 513–519. [Google Scholar]
  192. Tessari, G.; Naldi, L.; Boschiero, L.; Nacchia, F.; Fior, F.; Forni, A.; Rugiu, C.; Faggian, G.; Sassi, F.; Gotti, E.; et al. Incidence and clinical predictors of a subsequent nonmelanoma skin cancer in solid organ transplant recipients with a first nonmelanoma skin cancer: A multicenter cohort study. Arch. Dermatol. 2010, 146, 294–299. [Google Scholar] [CrossRef] [Green Version]
  193. Moloney, F.J.; Comber, H.; O’Lorcain, P.; O’Kelly, P.; Conlon, P.J.; Murphy, G.M. A population-based study of skin cancer incidence and prevalence in renal transplant recipients. Br. J. Dermatol. 2006, 154, 498–504. [Google Scholar] [CrossRef]
  194. Sherston, S.N.; Vogt, K.; Schlickeiser, S.; Sawitzki, B.; Harden, P.N.; Wood, K.J. Demethylation of the TSDR is a marker of squamous cell carcinoma in transplant recipients. Am. J. Transplant. 2014, 14, 2617–2622. [Google Scholar] [CrossRef] [Green Version]
  195. Frazzette, N.; Khodadadi-Jamayran, A.; Doudican, N.; Santana, A.; Felsen, D.; Pavlick, A.C.; Tsirigos, A.; Carucci, J.A. Decreased cytotoxic T cells and TCR clonality in organ transplant recipients with squamous cell carcinoma. NPJ Precis. Oncol. 2020, 4, 13. [Google Scholar] [CrossRef]
  196. Kim, H.J.; Verbinnen, B.; Tang, X.; Lu, L.; Cantor, H. Inhibition of follicular T-helper cells by CD8(+) regulatory T cells is essential for self tolerance. Nature 2010, 467, 328–332. [Google Scholar] [CrossRef] [Green Version]
  197. Saligrama, N.; Zhao, F.; Sikora, M.J.; Serratelli, W.S.; Fernandes, R.A.; Louis, D.M.; Yao, W.; Ji, X.; Idoyaga, J.; Mahajan, V.B.; et al. Opposing T cell responses in experimental autoimmune encephalomyelitis. Nature 2019, 572, 481–487. [Google Scholar] [CrossRef]
  198. Kanitakis, J.; Alhaj-Ibrahim, L.; Euvrard, S.; Claudy, A. Basal cell carcinomas developing in solid organ transplant recipients: Clinicopathologic study of 176 cases. Arch. Dermatol. 2003, 139, 1133–1137. [Google Scholar] [CrossRef] [Green Version]
  199. Omland, S.H.; Nielsen, P.S.; Gjerdrum, L.M.; Gniadecki, R. Immunosuppressive Environment in Basal Cell Carcinoma: The Role of Regulatory T Cells. Acta Derm. Venereol. 2016, 96, 917–921. [Google Scholar] [CrossRef] [Green Version]
  200. Adolphe, C.; Xue, A.; Fard, A.T.; Genovesi, L.A.; Yang, J.; Wainwright, B.J. Genetic and functional interaction network analysis reveals global enrichment of regulatory T cell genes influencing basal cell carcinoma susceptibility. Genome Med. 2021, 13, 19. [Google Scholar] [CrossRef]
  201. Anaya, J.M.; Shoenfeld, Y.; Rojas-Villarraga, A.; Levy, R.A.; Cervera, R. (Eds.) Autoimmunity: From Bench to Bedside; El Rosario University Press: Bogota, Colombia, 2013. [Google Scholar]
  202. Vesely, M.D. Getting under the Skin: Targeting Cutaneous Autoimmune Disease. Yale J. Biol. Med. 2020, 93, 197–206. [Google Scholar] [PubMed]
  203. Fishman, J.A.; Rubin, R.H. Infection in organ-transplant recipients. N. Engl. J. Med. 1998, 338, 1741–1751. [Google Scholar] [CrossRef] [PubMed]
  204. Gutierrez-Dalmau, A.; Campistol, J.M. Immunosuppressive therapy and malignancy in organ transplant recipients: A systematic review. Drugs 2007, 67, 1167–1198. [Google Scholar] [CrossRef] [PubMed]
  205. Penn, I.; Starzl, T.E. Proceedings: The effect of immunosuppression on cancer. Proc. Natl. Cancer Conf. 1972, 7, 425–436. [Google Scholar] [PubMed]
  206. Siegel, J.P.; Sharon, M.; Smith, P.L.; Leonard, W.J. The Il-2 Receptor Beta-Chain (P70)—Role in Mediating Signals for Lak, Nk, and Proliferative Activities. Science 1987, 238, 75–78. [Google Scholar] [CrossRef]
  207. Shevach, E.M. Mechanisms of Foxp3(+) T Regulatory Cell-Mediated Suppression. Immunity 2009, 30, 636–645. [Google Scholar] [CrossRef] [Green Version]
  208. Saadoun, D.; Rosenzwajg, M.; Joly, F.; Six, A.; Carrat, F.; Thibault, V.; Sene, D.; Cacoub, P.; Klatzmann, D. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N. Engl. J. Med. 2011, 365, 2067–2077. [Google Scholar] [CrossRef]
  209. Klatzmann, D.; Abbas, A.K. The promise of low-dose interleukin-2 therapy for autoimmune and inflammatory diseases. Nat. Rev. Immunol. 2015, 15, 283–294. [Google Scholar] [CrossRef]
  210. Yu, A.; Zhu, L.; Altman, N.H.; Malek, T.R. A low interleukin-2 receptor signaling threshold supports the development and homeostasis of T regulatory cells. Immunity 2009, 30, 204–217. [Google Scholar] [CrossRef] [Green Version]
  211. Boyman, O.; Kovar, M.; Rubinstein, M.P.; Surh, C.D.; Sprent, J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science 2006, 311, 1924–1927. [Google Scholar] [CrossRef] [Green Version]
  212. Pilat, N.; Sprent, J. Treg Therapies Revisited: Tolerance Beyond Deletion. Front. Immunol. 2020, 11, 622810. [Google Scholar] [CrossRef]
  213. Castela, E.; Le Duff, F.; Butori, C.; Ticchioni, M.; Hofman, P.; Bahadoran, P.; Lacour, J.P.; Passeron, T. Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in alopecia areata. JAMA Dermatol. 2014, 150, 748–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Rosenzwajg, M.; Lorenzon, R.; Cacoub, P.; Pham, H.P.; Pitoiset, F.; El Soufi, K.; Ribet, C.; Bernard, C.; Aractingi, S.; Banneville, B.; et al. Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a single, open clinical trial. Ann. Rheum. Dis. 2019, 78, 209–217. [Google Scholar] [CrossRef]
  215. Goschl, L.; Scheinecker, C.; Bonelli, M. Treg cells in autoimmunity: From identification to Treg-based therapies. Semin. Immunopathol. 2019, 41, 301–314. [Google Scholar] [CrossRef]
  216. Owen, C.M.; Harrison, P.V. Successful treatment of severe psoriasis with basiliximab, an interleukin-2 receptor monoclonal antibody. Clin. Exp. Dermatol. 2000, 25, 195–197. [Google Scholar] [CrossRef]
  217. Salim, A.; Emerson, R.M.; Dalziel, K.L. Successful treatment of severe generalized pustular psoriasis with basiliximab (interleukin-2 receptor blocker). Br. J. Dermatol. 2000, 143, 1121–1122. [Google Scholar] [CrossRef]
  218. Krueger, J.G.; Walters, I.B.; Miyazawa, M.; Gilleaudeau, P.; Hakimi, J.; Light, S.; Sherr, A.; Gottlieb, A.B. Successful in vivo blockade of CD25 (high-affinity interleukin 2 receptor) on T cells by administration of humanized anti-Tac antibody to patients with psoriasis. J. Am. Acad. Dermatol. 2000, 43, 448–458. [Google Scholar] [CrossRef]
  219. Tahvildari, M.; Dana, R. Low-Dose IL-2 Therapy in Transplantation, Autoimmunity, and Inflammatory Diseases. J. Immunol. 2019, 203, 2749–2755. [Google Scholar] [CrossRef]
  220. Yu, Z.; Cheng, H.; Ding, T.; Liang, Y.; Yan, C.; Gao, C.; Wen, H. Absolute decrease in regulatory T cells and low-dose interleukin-2 therapy: Restoring and expanding regulatory T cells to treat systemic sclerosis: A 24-week study. Clin. Exp. Dermatol. 2022, 47, 2188–2195. [Google Scholar] [CrossRef] [PubMed]
  221. Ali, H.; Mohiuddin, A.; Sharma, A.; Shaheen, I.; Kim, J.J.; El Kosi, M.; Halawa, A. Implication of interleukin-2 receptor antibody induction therapy in standard risk renal transplant in the tacrolimus era: A meta-analysis. Clin. Kidney J. 2019, 12, 592–599. [Google Scholar] [CrossRef]
  222. Evans, R.D.R.; Lan, J.H.; Kadatz, M.; Brar, S.; Chang, D.T.; McMichael, L.; Gill, J.; Gill, J.S. Use and Outcomes of Induction Therapy in Well-Matched Kidney Transplant Recipients. Clin. J. Am. Soc. Nephrol. 2022, 17, 271–279. [Google Scholar] [CrossRef] [PubMed]
  223. Trzonkowski, P.; Bieniaszewska, M.; Juscinska, J.; Dobyszuk, A.; Krzystyniak, A.; Marek, N.; Mysliwska, J.; Hellmann, A. First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127- T regulatory cells. Clin. Immunol. 2009, 133, 22–26. [Google Scholar] [CrossRef]
  224. Di Ianni, M.; Falzetti, F.; Carotti, A.; Terenzi, A.; Castellino, F.; Bonifacio, E.; Del Papa, B.; Zei, T.; Ostini, R.I.; Cecchini, D.; et al. Tregs prevent GVHD and promote immune reconstitution in HLA-haploidentical transplantation. Blood 2011, 117, 3921–3928. [Google Scholar] [CrossRef]
  225. Brunstein, C.G.; Miller, J.S.; Cao, Q.; McKenna, D.H.; Hippen, K.L.; Curtsinger, J.; Defor, T.; Levine, B.L.; June, C.H.; Rubinstein, P.; et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: Safety profile and detection kinetics. Blood 2011, 117, 1061–1070. [Google Scholar] [CrossRef] [PubMed]
  226. Theil, A.; Tuve, S.; Oelschlagel, U.; Maiwald, A.; Dohler, D.; Ossmann, D.; Zenkel, A.; Wilhelm, C.; Middeke, J.M.; Shayegi, N.; et al. Adoptive transfer of allogeneic regulatory T cells into patients with chronic graft-versus-host disease. Cytotherapy 2015, 17, 473–486. [Google Scholar] [CrossRef] [PubMed]
  227. Bluestone, J.A.; Buckner, J.H.; Fitch, M.; Gitelman, S.E.; Gupta, S.; Hellerstein, M.K.; Herold, K.C.; Lares, A.; Lee, M.R.; Li, K.; et al. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci. Transl. Med. 2015, 7, 315ra189. [Google Scholar] [CrossRef] [Green Version]
  228. Chatterjee, S.; Eby, J.M.; Al-Khami, A.A.; Soloshchenko, M.; Kang, H.K.; Kaur, N.; Naga, O.S.; Murali, A.; Nishimura, M.I.; Caroline Le Poole, I.; et al. A quantitative increase in regulatory T cells controls development of vitiligo. J. Investig. Dermatol. 2014, 134, 1285–1294. [Google Scholar] [CrossRef] [Green Version]
  229. Zhang, Q.; Lu, W.; Liang, C.L.; Chen, Y.; Liu, H.; Qiu, F.; Dai, Z. Chimeric Antigen Receptor (CAR) Treg: A Promising Approach to Inducing Immunological Tolerance. Front. Immunol. 2018, 9, 2359. [Google Scholar] [CrossRef] [Green Version]
  230. Boardman, D.; Maher, J.; Lechler, R.; Smyth, L.; Lombardi, G. Antigen-specificity using chimeric antigen receptors: The future of regulatory T-cell therapy? Biochem. Soc. Trans. 2016, 44, 342–348. [Google Scholar] [CrossRef]
  231. Gross, G.; Waks, T.; Eshhar, Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl. Acad. Sci. USA 1989, 86, 10024–10028. [Google Scholar] [CrossRef] [Green Version]
  232. Horton, H.; McMorrow, I.; Burke, B. Independent expression and assembly properties of heterologous lamins A and C in murine embryonal carcinomas. Eur. J. Cell Biol. 1992, 57, 172–183. [Google Scholar]
  233. Xue, E.; Minniti, A.; Alexander, T.; Del Papa, N.; Greco, R.; Autoimmune Diseases Working Party (ADWP) of the European Society for Blood; Marrow Transplantation (EBMT). Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells 2022, 11, 3346. [Google Scholar] [CrossRef]
  234. Hu, G.; Li, G.; Wen, W.; Ding, W.; Zhou, Z.; Zheng, Y.; Huang, T.; Ren, J.; Chen, R.; Zhu, D.; et al. Case report: B7-H3 CAR-T therapy partially controls tumor growth in a basal cell carcinoma patient. Front. Oncol. 2022, 12, 956593. [Google Scholar] [CrossRef]
  235. de Nattes, T.; Camus, V.; Francois, A.; Dallet, G.; Ferrand, C.; Guerrot, D.; Lemoine, M.; Morin, F.; Thieblemont, C.; Veresezan, E.L.; et al. Kidney Transplant T Cell-Mediated Rejection Occurring After Anti-CD19 CAR T-Cell Therapy for Refractory Aggressive Burkitt-like Lymphoma With 11q Aberration: A Case Report. Am. J. Kidney Dis. 2022, 79, 760–764. [Google Scholar] [CrossRef]
  236. Feng, G.; Li, Q.; Zhu, H.; Jiang, Y.; Yuan, J.; Fu, Y.; Deng, Q. Safety and Efficacy of Anti-CD19-Chimeric Antigen Receptor T Cell Combined With Programmed Cell Death 1 Inhibitor Therapy in a Patient With Refractory Post-Transplant Lymphoproliferative Disease: Case Report and Literature Review. Front. Oncol. 2021, 11, 726134. [Google Scholar] [CrossRef] [PubMed]
  237. Hernani, R.; Sancho, A.; Amat, P.; Hernandez-Boluda, J.C.; Perez, A.; Pinana, J.L.; Carretero, C.; Goterris, R.; Gomez, M.; Saus, A.; et al. CAR-T therapy in solid transplant recipients with post-transplant lymphoproliferative disease: Case report and literature review. Curr. Res. Transl. Med. 2021, 69, 103304. [Google Scholar] [CrossRef] [PubMed]
  238. Krishnamoorthy, S.; Ghobadi, A.; Santos, R.D.; Schilling, J.D.; Malone, A.F.; Murad, H.; Bartlett, N.L.; Alhamad, T. CAR-T therapy in solid organ transplant recipients with treatment refractory posttransplant lymphoproliferative disorder. Am. J. Transplant. 2021, 21, 809–814. [Google Scholar] [CrossRef] [PubMed]
  239. Luttwak, E.; Hagin, D.; Perry, C.; Wolach, O.; Itchaki, G.; Amit, O.; Bar-On, Y.; Freund, T.; Kay, S.; Eshel, R.; et al. Anti-CD19 CAR-T therapy for EBV-negative posttransplantation lymphoproliferative disease-a single center case series. Bone Marrow Transplant. 2021, 56, 1031–1037. [Google Scholar] [CrossRef]
  240. Mamlouk, O.; Nair, R.; Iyer, S.P.; Edwards, A.; Neelapu, S.S.; Steiner, R.E.; Adkins, S.A.; Hawkins, M.; Saini, N.; Devashish, K.; et al. Safety of CAR T-cell therapy in kidney transplant recipients. Blood 2021, 137, 2558–2562. [Google Scholar] [CrossRef]
  241. Melilli, E.; Mussetti, A.; Linares, G.S.; Ruella, M.; La Salette, C.; Savchenko, A.; Taco, M.D.R.; Montero, N.; Grinyo, J.; Fava, A.; et al. Acute Kidney Injury Following Chimeric Antigen Receptor T-Cell Therapy for B-Cell Lymphoma in a Kidney Transplant Recipient. Kidney Med. 2021, 3, 665–668. [Google Scholar] [CrossRef]
  242. Rosler, W.; Bink, A.; Bissig, M.; Imbach, L.; Marques Maggio, E.; Manz, M.G.; Muller, T.; Roth, P.; Rushing, E.; Widmer, C.; et al. CAR T-cell Infusion Following Checkpoint Inhibition Can Induce Remission in Chemorefractory Post-transplant Lymphoproliferative Disorder of the CNS. Hemasphere 2022, 6, e733. [Google Scholar] [CrossRef]
  243. Portuguese, A.J.; Gauthier, J.; Tykodi, S.S.; Hall, E.T.; Hirayama, A.V.; Yeung, C.C.S.; Blosser, C.D. CD19 CAR-T therapy in solid organ transplant recipients: Case report and systematic review. Bone Marrow Transplant. 2022, 1–7. [Google Scholar] [CrossRef]
Figure 1. Tregs homeostasis and dysregulation. Tregs interact with effector T cells (Teffs) to modulate the immune responses and maintain self-tolerance. Tregs act by suppressing Teffs through various mechanisms that, when disrupted, can lead to several autoimmune conditions and malignancies. (A) Mechanisms that drive autoimmunity can occur via: (1) Loss of the ability of Tregs to inhibit the activity of Teffs, leading to an overactivation of the immune system. (2) Loss of high affinity IL-2R on the Treg cell surface leads to increased levels of IL-2 in the environment that is available for other T cell subsets to utilize for their proliferation. (3) Treg plasticity leads to an unstable phenotype that impacts their suppressive functions [41]. Notably, Treg plasticity towards Teffs activity can enhance inflammation when cytokines, such as IL-17, IFN-γ, and TNF-α, are released into the microenvironment. (B) Mechanisms that promote a healthy state involve: (1) Treg sequestration of IL-2 by high affinity IL-2Rαβγ, which decreases IL-2 availability for Teffs, indirectly inhibiting their survival. (2) Treg release of adenosine and cAMP impairs Teff metabolism and promotes homeostasis [42]. (3) Teff apoptosis can occur secondary to perforin and granzyme release by Tregs via FasL–Fas interactions. (4) Tregs have the ability to obstruct co-stimulation on APCs through CTLA-4, preventing Teff binding via CD28. (5) Treg release of anti-inflammatory cytokines IL-10, TGF-β, and IL-35 suppresses Teff activity. (C) Post-transplant mechanisms driving tumor development. Although it is still unclear how immunosuppressive therapies affect immune subtypes, evidence suggests that immunosuppressants affect clonal expansion and immune cell functionality by various mechanisms, such as decreased production of IL-2 and IFN-γ [43,44,45]. Immunosuppressants that favor increased ratio of Tregs versus Teffs have been associated with decreased incidence of graft rejection [46]. Conversely, an immunosuppressed environment can promote decreased immune surveillance and antitumoral responses, such as the inhibition of cytolytic responses against the tumor by Teffs, leading to tumor development [43]. Created with Biorender.com.
Figure 1. Tregs homeostasis and dysregulation. Tregs interact with effector T cells (Teffs) to modulate the immune responses and maintain self-tolerance. Tregs act by suppressing Teffs through various mechanisms that, when disrupted, can lead to several autoimmune conditions and malignancies. (A) Mechanisms that drive autoimmunity can occur via: (1) Loss of the ability of Tregs to inhibit the activity of Teffs, leading to an overactivation of the immune system. (2) Loss of high affinity IL-2R on the Treg cell surface leads to increased levels of IL-2 in the environment that is available for other T cell subsets to utilize for their proliferation. (3) Treg plasticity leads to an unstable phenotype that impacts their suppressive functions [41]. Notably, Treg plasticity towards Teffs activity can enhance inflammation when cytokines, such as IL-17, IFN-γ, and TNF-α, are released into the microenvironment. (B) Mechanisms that promote a healthy state involve: (1) Treg sequestration of IL-2 by high affinity IL-2Rαβγ, which decreases IL-2 availability for Teffs, indirectly inhibiting their survival. (2) Treg release of adenosine and cAMP impairs Teff metabolism and promotes homeostasis [42]. (3) Teff apoptosis can occur secondary to perforin and granzyme release by Tregs via FasL–Fas interactions. (4) Tregs have the ability to obstruct co-stimulation on APCs through CTLA-4, preventing Teff binding via CD28. (5) Treg release of anti-inflammatory cytokines IL-10, TGF-β, and IL-35 suppresses Teff activity. (C) Post-transplant mechanisms driving tumor development. Although it is still unclear how immunosuppressive therapies affect immune subtypes, evidence suggests that immunosuppressants affect clonal expansion and immune cell functionality by various mechanisms, such as decreased production of IL-2 and IFN-γ [43,44,45]. Immunosuppressants that favor increased ratio of Tregs versus Teffs have been associated with decreased incidence of graft rejection [46]. Conversely, an immunosuppressed environment can promote decreased immune surveillance and antitumoral responses, such as the inhibition of cytolytic responses against the tumor by Teffs, leading to tumor development [43]. Created with Biorender.com.
Ijms 24 01527 g001
Table 1. Table summarizes the key effector cells, cytokines, reported and proposed treatments, and risk factors of select cutaneous autoimmune diseases. IL, interleukin; Th, T helper; Treg, T regulatory; TNF-α, tumor necrosis factor alpha; IFN-γ, interferon gamma; PDE-5 = phosphodiesterase 5; JAK, Janus kinase; and NB-UVB, narrowband ultraviolet B.2.3.
Table 1. Table summarizes the key effector cells, cytokines, reported and proposed treatments, and risk factors of select cutaneous autoimmune diseases. IL, interleukin; Th, T helper; Treg, T regulatory; TNF-α, tumor necrosis factor alpha; IFN-γ, interferon gamma; PDE-5 = phosphodiesterase 5; JAK, Janus kinase; and NB-UVB, narrowband ultraviolet B.2.3.
DiseaseRisk FactorsTeff CellsCytokinesTreatmentsReferences
PsoriasisObesity, infection, traumaTh1, Treg-Th17IL-2, IL-17, IL-22, IL-23, IL-26, TNF-α, IFN-γAnti-TNF-α inhibitors, T-cell-targeted therapies[71,72,73,74,75,76,77,78]
VitiligoGenetics, traumaTreg-Th1IL-1β, IL-6, IL-15, IL-22, IL-33, TNF-α, IFN-γJAK inhibitor, NB-UVB therapy, anti-TNF-α inhibitors[79,80,81,82,83,84,85,86,87]
Systemic sclerosisGenetics, silica, solvents, heavy metalTreg-Th2, Treg-Th17IL-4, IL-13, IL-17, IL-21, IL-22, IL-33Low-dose IL-2 therapy, PDE-5 inhibitors, calcium channel blocker, bosentan[88,89,90,91,92,93,94,95,96,97]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oparaugo, N.C.; Ouyang, K.; Nguyen, N.P.N.; Nelson, A.M.; Agak, G.W. Human Regulatory T Cells: Understanding the Role of Tregs in Select Autoimmune Skin Diseases and Post-Transplant Nonmelanoma Skin Cancers. Int. J. Mol. Sci. 2023, 24, 1527. https://doi.org/10.3390/ijms24021527

AMA Style

Oparaugo NC, Ouyang K, Nguyen NPN, Nelson AM, Agak GW. Human Regulatory T Cells: Understanding the Role of Tregs in Select Autoimmune Skin Diseases and Post-Transplant Nonmelanoma Skin Cancers. International Journal of Molecular Sciences. 2023; 24(2):1527. https://doi.org/10.3390/ijms24021527

Chicago/Turabian Style

Oparaugo, Nicole Chizara, Kelsey Ouyang, Nam Phuong N. Nguyen, Amanda M. Nelson, and George W. Agak. 2023. "Human Regulatory T Cells: Understanding the Role of Tregs in Select Autoimmune Skin Diseases and Post-Transplant Nonmelanoma Skin Cancers" International Journal of Molecular Sciences 24, no. 2: 1527. https://doi.org/10.3390/ijms24021527

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop