Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 4565 2023-04-26 11:19:32 |
2 layout & references -6 word(s) 4559 2023-04-27 03:04:44 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Pietraforte, I.; Frasca, L. Tregs in Psoriasis. Encyclopedia. Available online: https://encyclopedia.pub/entry/43513 (accessed on 29 April 2024).
Pietraforte I, Frasca L. Tregs in Psoriasis. Encyclopedia. Available at: https://encyclopedia.pub/entry/43513. Accessed April 29, 2024.
Pietraforte, Immacolata, Loredana Frasca. "Tregs in Psoriasis" Encyclopedia, https://encyclopedia.pub/entry/43513 (accessed April 29, 2024).
Pietraforte, I., & Frasca, L. (2023, April 26). Tregs in Psoriasis. In Encyclopedia. https://encyclopedia.pub/entry/43513
Pietraforte, Immacolata and Loredana Frasca. "Tregs in Psoriasis." Encyclopedia. Web. 26 April, 2023.
Tregs in Psoriasis
Edit

Psoriasis is a chronic inflammatory skin disease, which affects 2–4% of the population worldwide. Psoriasis has a strong genetic component, but environmental factors are also important. T-cell derived factors such as Psoriasis is characteristic, with thickening and scaling of the epidermisdue to hyper-proliferation of keratinocytes (acanthosis). CD4 and CD8 T-cell infiltrate characterizes the skin in psoriasis, a T-cell-driven disease, but other cell types are also present in the lesions, including neutrophils, macrophages, NK cells, dendritic cells. It has been long known that the pathogenesis of psoriasis is driven by T-cell derived factors, produced by subsets including T-helper (Th) 1 cells, Th17, Th22 and regulatory T cells (Tregs). 

autoreactivity Tregs psoriasis

1. Introduction

There is evidence in animal models that Tregs could play a role in ameliorating psoriasis [1]. However, these Tregs are considered dysfunctional, as they are altered in frequency, phenotype, and function [2]. Tregs are of various types: they can act through secretion of regulatory/suppressive cytokines like IL-10, TGF-β, IL-35 or act by contact. Tregs that express Foxp3 can lose their capacity to suppress in certain circumstances. For instance, Tregs can convert to IL-17-like cells under inflammatory conditions, especially in the presence of IL-6, which is a cytokine highly up-regulated in psoriasis [3]. Th17 and Th1 cytokines or cytokines such as IL-23, which favors Th17-expansion/differentiation, dominate in the disease. Along with the assumption that psoriasis is a T-cell-driven disease, Tregs have been studied extensively over the years, both in the skin and in circulation. Here, researchers discuss how T reg increase in psoriasis but are impared in their regulatory/suppressive function and put particular emphasis on therapies that seem to counteract this conversion. Finally, by our preliminary experimental data, analyzing T-cells specific for the autoantigen LL37 in a healthy subject, researchers found that a shared specificity may exist between Tregs and autoreactive responder T-cells.

2. Tregs Subsets in Health and Disease

Work with animal models has shown that more Tregs subsets exist, which parallel the T-helper cell subsets [4]. Most of these studies were in mice, as analogous subsets in humans are more difficult to study because of lack of accurate identification markers [5][6].
Tregs expressing the Th1 factor T-bet exist, which also express chemokine receptor CXCR3 [7]. The deletion of Foxp3 in T-bet-expressing cells was shown to determine uncontrolled Th1 immune responses at steady state [8]. In addition, Th2-like Tregs have been identified, for instance, in non-lymphoid organs. The Th2-like Tregs can express GATA-3 and have been detected at mucosal surfaces: skin, gut, and lungs [9][10]. Indeed, studies demonstrated that, in mice, GATA-3 expression is required for Tregs stability and maintenance of Foxp3 expression [11]. A pathogenic reprogramming of T-cells can cause disease, and there are several examples of this. For instance, Th2 Tregs can play a role in food allergies [12]. Relevant to psoriasis, Tregs can also express the canonical Th17 transcription factor, RORγt. Cells of these types have been detected in the gut [13][14], but they have been also described in patients with psoriasis [15], as well as in inflammatory bowel diseases (IBD), and arthritis; in some cases, they convert into IL-17 secreting cells [16][17][18][19].
Single cell RNA sequence analysis shows the existence of distinct T-cell subsets in relapsing psoriasis for tissue-resident T-cells. Th17 cells and NF-kB signaling pathways evidenced include Pellino-1 (PELI1). Mice with systemic and conditional depletion of PELI1 were protected from psoriasiform dermatitis and showed reduced IL-17A production and NFkB activation in Th17 cells. The inhibition of PELI1 significantly ameliorated murine psoriasiform dermatitis by reducing IL-17A production. PELI1 belongs to a member of the E3 ubiquitin ligases, mediating immune receptor signaling cascades involving, among others, the NFkB pathway; it also promotes intrinsic activation of skin resident Th17 cells in psoriasis. This points out that this gene inhibition could be a promising therapeutic strategy for psoriasis, limiting skin-resident Th17 cell responses. Both the T-cell receptor (TCR) and IL-23 stimulation upregulate PELI1 expression in Th17 cells from lesional skin. However, it is currently unknown whether PELI1 directly modulates the suppressive function of Tregs [20].
Recently, the regulatory counterpart of follicular helper T-cells (Tfh) cells, which promote germinal center (GC) responses and antibody production, has been described. These cells express TCF1 and LEF1, required for expression of B-cell lymphoma 6 (Bcl-6) [4].

3. Tregs in the Skin and in Psoriasis

It has been shown that Tregs are part of the TRM (Tissue Resident Memory cells) pool, and it is postulated that these cells can re-circulate. Human psoriatic plaques contain abundant numbers of IL-17A-producing T-cells with a CD69+ and/or CD103+ TRM phenotype. TRM can play a role in chronic inflammatory diseases of the skin, such as vitiligo and psoriasis, and are likely involved in recurrent lesions. A greater number of CD8 TRM cells infiltrate psoriatic lesional and non-lesional skin, as compared to normal skin. These are mainly infiltrating CD8+IL-17A+ TRM cells and correlate with disease duration. Apparently, healed skin contains CD8 TRM cells, which mostly localize in the epidermis (as they express CD103), while CD4 TRM cells localize in the dermis. Moreover, a large pool of TRM cells express IL-17A alone, IL-17A and IL-22, or IL-22 alone, and persist in post-lesional psoriasis skin. Cytokine production is not exclusively oriented toward a Th17 phenotype as TRM also express IFN-γ, showing a Th1 phenotype, which could be induced by IL-15 stimulation [21].
Tregs residing in the skin seem primary localized, in mouse and humans, in the hair follicles (HFs) [18][22][23]; this is due to HF epithelial cells production of CCL20, which recruits CCR6-expressing Tregs into the skin [22]. In human and mice skin, Tregs constitute about 20–40% of the CD4 T-cells [18]. These Tregs can express GATA-3 [11]. Their role is to maintain the cutaneous immune homeostasis, promote wound healing, and repair tissues. Interestingly, they express ST2 [24], the receptor for IL-33, which is released in situation of tissue damage [25]. IL-33 is an alarmin, which can regulate Foxp3 in the mucosal tissue [26]. IL-33 was originally described as a cytokine that activates Th2 cells, but it can affect various cell types, including Th1 cells or innate lymphoid cells (ILC) and CD8 T-cells [27]. Its exact role in psoriasis is not completely clear due to these pleiotropic effects.
In support of the idea that Tregs increased because of tissue damage stimuli, a recent study showed that Tregs in the skin expand following UVB irradiation [28]. After injury, Tregs express the AREG receptor, EGFR, suggesting an AREG autocrine role in tissue repair [29]. The peripheral blood of healthy humans contains Tregs, which express cutaneous lymphocyte antigen (CLA) and other skin homing receptors [30], suggesting that Tregs migrate to the skin using these unique receptors. Injury and microbial invasion influence this migration. Similar to human Tregs, mouse Tregs express some homing receptors, which facilitate their recruitment into inflamed skin [31].
Mouse and human studies indicate that, in psoriasis, the IL-23/IL-17 axis of inflammation, together with Tregs dysfunction, determines the Th17/Tregs imbalance implicated in the disease [2][32], although the link between Tregs and disease severity is debated [33]. The conflicting results may depend on several factors: the site of biopsies, psoriasis subtypes, and different disease status. Although Tregs and Th17 cells increase in adult and pediatric psoriasis patients, Tregs were shown to be unable to suppress Th17 cells activation, and their effector T-cell responses and proliferation [2][33].
Several papers support Tregs dysfunction in psoriasis. The Tregs suppressive function seem impaired due to the pro-inflammatory cytokine milieu. For example, the exposure to high level of IL-6 decreases Tregs activity [3][34]. Dysfunctional Tregs in peripheral blood of patients with psoriasis have been reported showing that they have a phosphorylation and an aberrant activation of STAT3, which is due to the effects of pro-inflammatory cytokines, not only IL-6, but also IL-21 and IL-23 [35]. Additionally, inhibition of Foxp3 by up regulation of microRNA (mir-210), in CD4 T-cells, results in decreased levels of IL-10 and TGF-β by these cells, accompanied by increased levels of pro-inflammatory cytokines release, such as TNF-α and IL-17A, as described by Zhao et al. [36].
Tregs can express CD39 and CD73, so they can use the adenosine signaling for exerting suppressive effects. Yan et al. reported that Tregs from patients with psoriasis have a reduced expression of CD73 and a CD73/AMPK pathway deactivation [37]. The adenosine pathway (CD39 and CD73) is widely known to play a crucial role for the Tregs immunosuppressive function. CD39 removes extracellular ATP by hydrolyzing ATP/UTP and ADP/UDP into AMP. Successively, AMP is rapidly degraded into adenosine in the presence of CD73. Then, adenosine, which binds to the A2AR receptor on Tregs, triggers the accumulation of intracellular cAMP, activation of AMPK and inactivation of mTOR, blocking IL-17 and IFN-γ production. This also blocks the differentiation of Tregs into IL-17 secreting Tregs, which have a decreased immune suppressive function. Moreover, adenosine binding to A2AR of the T effector cells, achieves local repression of immune response and down regulation of INF-γ production.
Inefficient recruitment of Tregs to inflamed skin can also concur to the inefficiency in restraining inflammation in individuals with psoriasis [38]. Finally, although microbial infection in the skin is a contributing factor in psoriasis, a connection by skin microbiota and Tregs in psoriasis can be hypothesized [39]. All these data in the scientific literature show that the suppressive function of circulating and in skin-resident Tregs is important in psoriasis and support an impairment of Tregs in the psoriatic disease.

4. Tregs Plasticity in Psoriasis

Tregs and Th17 cells require TGF-β to develop from a common precursor, indicating a constant competition between the two cell types [40], highlighting a special relationship in the development and function of Th17 and Tregs. Several studies revealed that a subset of Tregs in the skin might differentiate into Th17 cells [15][41]. It seems that CD27 and OX40 expression on Tregs plays a role in suppressing Tregs differentiation toward Th17 phenotype, whereas lack of expression of these molecules induces the expression of high levels of IL-17A and the transcription factor RORα [42][43]. Tregs from patients with psoriasis are able to differentiate into IL-17A producing cells after stimulation ex vivo. Tregs may also differentiate into IL-17A producing cells upon activation of the histone deacetylase 1 (HDAC-1), which is elevated in psoriatic lesions. Moreover, the presence of IL-17A+Foxp3+CD4 T-cells was observed in psoriatic skin [15]. All these data show not only a Tregs dysfunction but also a phenotypic alteration of these cells in psoriasis.

5. Treatments That Can Affect Tregs in Psoriasis

Many treatments are in use for psoriasis, and others are being explored. An overview on how these treatments appear to influence expansion or functionality of Tregs and the Th17/Tregs balance is reported below. The therapeutic approach to psoriasis comprises two major categories of drugs: biological agents, and immunosuppressive drugs (methotrexate, cyclosporine), and treatments sub-groups are evidenced.

5.1. Biological Agents

TNF-α Antagonists

Among biological agents, TNF-α antagonists have been extensively used. These include infliximab (a chimeric monoclonal antibody composed of a human IgG1 constant region and a murine variable region), etanercept (a soluble TNFR, made of two extracellular domains of the human TNFR2 fused to the Fc fragment of human IgG1), and adalimumab (a humanized monoclonal antibody). Different studies in humans refer that anti TNF-α agents increase Tregs and decrease the Th17 cells frequency in peripheral blood of psoriasis patients [44][45][46]. Etanercept regimen showed a more significant modification of the T-cell subsets, as compared to the other two drugs [47][48][49]. Moreover, Diluvio et al. reported that infliximab treatment induces a polyclonal expansion of Tregs, sorted from peripheral blood of patients, showing a diverse TCR repertoire [50]. In all of these studies Tregs suppressive function was not addressed and the modification of the subset of Tregs in the skin has not been analyzed; the data refer only to peripheral blood cells. Of note, in a murine psoriasiform model, some data are conflicting, and not confirmed in humans [51].

IL-17 and IL-23 Antagonists

Classes of biologic agents targeting either IL-17 or IL-23 demonstrated higher rates of response and superiority compared to previous biologic agents. Among these new therapeutic agents are an anti-IL-17A (secukinumab), an anti-IL-23p19, called guselkumab, and an anti-p40, called ustekinumab. In a model of imiquimod-induced psoriasis in mice, it was described an increase of Foxp3+ Tregs in the skin and restoration of their suppressive function following use of IL-17 or IL-23 blocking antibodies but not with an anti-TNF-α treatment [51]. Of note, Kanman et al. described a principal role of IL-23 in regulation of Tregs plasticity and conversion into a Th17 like phenotype [52]. These data are supported by observations in humans that IL-23 inhibitors act as potent disease modifying drug, more than IL-17 antagonists [53][54][55]. Clinical trials testing IL-23 inhibitors showed long-lasting maintenance of the therapeutic response following treatment discontinuation, compared to IL-17 inhibitors [54][56][57]. Another clinical trial, comparing guselkumab to secukinumab, provided relevant insights about the skin compartment. During a 24-week treatment with guselkumab or secukinumab, the number of CD4 and CD8 TRM cells decreased in psoriatic lesions of both treatment arms, but guselkumab reduced memory T-cells, maintaining Tregs whereas the opposite was observed for secukinumab treatment [53]. Secukinumab treatment decreased the number of Tregs in a more pronounced way than guselkumab. Moreover, a greater decrease of LC (Langerhans cells), infiltrating post-lesional skin, was observed after IL-23 blockade. These findings suggest a successful response to either IL-23 or IL-17 inhibitors, with an increased Tregs/CD8 TRM ratio. A superior long-term control of skin inflammation was achieved by inhibiting IL-23 with a reset of the pathogenic inflammatory T-cells and an increase of Tregs. IL-23 acts on the T-cell compartment and stimulates the expression of RORγt, and the production of IL-17A, IL-1F, and IL-22. Moreover, IL-23 drives and maintains the differentiation of Th17 cells. In contrast, IL-17A is an effectors cytokine that induces skin inflammation. It is also expressed by neutrophil and mast cells and produces indirect effect on the T-cell compartment. This may explain why IL-17 inhibitors have lower modulator ability than IL-23 inhibitors.

IL-6 Antagonists

IL-6 is another important cytokine involved in psoriasis. Although IL-6 plays a role in the maturation of Th17 cells, search of the literature and clinical trials in websites did not reveal psoriasis studies with anti-IL-6/IL-6 receptors and role on Tregs frequency and Th1/Tregs balance [58]. A study in vitro reported that IL-6 was necessary and sufficient to reverse human T-cell suppression by Tregs in in vitro models using activated DCs as a source of IL-6 [3]. Although IL-6 may be another potential target for psoriasis treatment, data in the literature show that attempts to treat psoriasis with tocilizumab (TCZ), a humanized anti-interleukin-6 (IL-6) receptor antibody licensed for the treatment of rheumatoid arthritis (RA), have been unsuccessful [59]. On the other hand, the use of new IL-6 inhibitors such as clazakizumab, a monoclonal antibody with high affinity and specificity for IL-6, could be more promising for psoriatic arthritis (PsA) [53][60][61][62].

5.2. Photo Therapy

Many photo therapeutic approaches can treat psoriasis: natural phototherapy, broadband UVB, narrowband UVB, selective UV phototherapy, Xenon chloride excimer laser, Xenon chloride excimer lamp, UVB light emitting diode, flat-type fluorescent UVB lamp, UVA, Mixed UVB/UVA, Psoralen+ UVA photochemotherapy (PUVA), bath water delivery of 8-methoxypsoralen and subsequent UVA-irradiation (bath-PUVA therapy), UVA-1 phototherapy, Pulsed Dye laser, and others [63][64]. The principal immunomodulatory effect of phototherapy is promoting the death of effector cells, such as T-cells, and keratocytes, and inhibition of LC, macrophages, neutrophils and NK cell function. Effect of bath-PUVA therapy was reported on three distinct Foxp3+ subsets: activated Tregs (aTregs), resting Tregs (rTregs), and cytokine-secreting non-suppressive T-cells from peripheral blood of psoriasis patients and healthy controls. Bath-PUVA therapy increased Tregs and restored dysfunctional Tregs activity in patients: in particular aTregs were significantly increased in the early bath-PUVA therapy sessions, and then diminished. RTregs, which were lower in patients than healthy controls, increased during therapy [64]. Takuya Furuhashi et al. confirmed these data by functional assays. CD4 CD25 cells separated from PBMCs of psoriasis patients treated with PUVA and activated with anti-CD3/CD28-bound beads, were cultured with or without CD4 CD25+ T-cells. The ability of Tregs to suppress CD4 CD25 T-cells was calculated by comparing the proliferation rates of CD4 CD25 T-cells in the presence/absence of CD4 CD25+ T-cells [65]. The same conclusions derive from an UV (B) treatment in psoriasis patients with polymorphic light eruption (PLE), in which UV increased the number of Tregs. This might be a compensatory mechanism to counteract the susceptibility to PLE [66]. Tregs from patients with PLE lacked any capacity to suppress effector T-cell proliferation but this capacity improved after therapy, as demonstrated by regulatory T-cell suppression assay. Moreover, after UVB treatment, keratinocytes upregulated the expression of receptor of activated nuclear factor-B ligand (RANKL). This receptor interacts with RANK on DCs, making DCs able to expand the number of Tregs [67][68]. These data were confirmed also in a mouse model of psoriasis [69].

5.3. Vitamins

Vitamin A

Vitamin A derivatives, retinoids, are also of common use to treat psoriasis. Retinoids, such as etretinate or acitretin, are absorbed in the small intestine and then are metabolized in other organs to the active acid form of retinoid acids (RAs), which interact with retinoid X receptors (RXRs). This heterodimer binds the RA response element on CNS1 of Foxp3, inducing Foxp3 expression and the generation of peripheral Tregs from naive T-cells [70]. Of note, retinoids not only promote Tregs generation but also regulate TGF-β, capable of inhibiting the IL-6. IL-6 is driving activation of pro-inflammatory Th17 cells, acting on RORγt [71][72].

Vitamin D

Vitamin D seems to regulate Tregs. Vitamin D status correlates with circulating Tregs in patients affected by psoriasis; a correlation with the severity of the disease, evaluated with Psoriasis Area Severity Index (PASI) score is present. In a clinical study, patients were analyzed for PASI-score, serum levels vitamin D and regulatory T-cells percentage. Using non parametric Spearman coefficient test to assess correlation between serum levels of vitamin D and the single variables of disease, this study found a positive association between vitamin D and Tregs population (p < 0.001), and an inverse correlation between vitamin D and PASI-score (p = 0.04) [73]. The effects of maxacalcitol, a vitamin D3 analogue, and betamethasone valerate (BV) steroid lotion, confirmed the effects of vitamin D on the differentiation of T-cells with suppressive phenotypes in an imiquimod (IMQ)-induced psoriasiform skin inflammation animal model. The authors report that maxacalcitol and BV reduced the MHC Class II+ inflammatory cell infiltrate and down-regulated IL-17A, IL-17F, IL-22, IL-12p40, TNF-α and IL-6 mRNA expression levels in the inflamed mouse ski. Maxacalcitol alone downregulated IL-23p19 expression, and increased Foxp3+ T-cell infiltrations and IL-10 expression. Of note, adoptive transfer of Tregs from maxacalcitol-treated donor mice improved IMQ-induced inflammation more than of Tregs from a BV-treated donor group [74]. Many data in the literature, from humans and mouse models, report that vitamin D induces myeloid dendritic cells with a tolerogenic phenotype responsible for the differentiation of CD4 CD25+ Tregs from naive T cells [75][76][77].

5.4. Topical Therapies

Topical therapies based on glucocorticoids (GC) and calcipotriol are usually sufficient to manage mild and moderate psoriasis [78]. GCs produce anti-inflammatory effects through GC receptors (GR) and by acting on specific target genes, inhibiting several cytokines [79]. Calcipotriol exerts its effect by binding to the nuclear vitamin D3 receptor [80]. The anti-inflammatory effects of calcipotriol are inferior compared with those of GCs, but an incremented effect is seen with a combinatory therapy [81]. Keijsers and co-workers showed that topical calcipotriol/betamethasone treatment for eight weeks decreased the number of Tregs in psoriatic lesions and the expression of Foxp3 in the skin and PBMCs [82]. Minna E. Kubin et al. confirmed that a combination therapy down-regulated the expression of TNF-α, IL-23, IL-17A, S100A7, CCL20 and interferon-γ in the skin and TNF-α, IL-6, IL-23A, T-bet and IFN-γ in PBMCs. Calcipotriol/betamethasone, but not betamethasone alone, down-regulated expression of Foxp3 in both skin and PBMCs [83].

5.5. Oral Small Molecules

Dimethyl Fumarate (DMF)

The European Medicines Agency [84] approves this drug for the treatment of psoriasis patients as of 2017, as an oral formulation. Studies in vitro show that DMF promotes oxidative stress reducing vitality of conventional T-cells but not Tregs. An increased expression on Tregs of cell surface-reduced thiols or thioredoxin-1 [85], protect Tregs from oxidative stress, mediated by DMF. The anti-psoriatic effect of DMF favors Tregs survival but not Th17 expansion [86]. In psoriasis patients, DMF treatment increased Tregs frequency and decreased Th17 cells, confirming in vitro data [87].

Sotrastaurin

A clinical study in psoriasis patients, using the pan-protein kinase C (PKC) inhibitor sotrastaurin (AEB071), showed a reduction of psoriasis clinical severity [88]. Currently, sotrastaurin is in phase II clinical trial studies for psoriasis [89]. Sotrastaurin blocks more than one PKC isoform. The latter belongs to a sub family of PKC calcium-independent and is most abundant in T-cells [90]. The activation of T-cells by CD28 and TCR promotes PKC-theta activation and translocation into the membrane at site of immunological synapse (IS), leading to the activation of NF-kB. Inhibition of PKC-theta restored activity of defective Tregs from RA patients and enhanced protection of mice from inflammatory colitis [91]. Moreover, studies in PKC-knockout mice have shown that PKC-theta is required for productive Th2 [92] and Th17 [93] responses but not for Th1 responses. In particular, Xuehui He et al. confirmed that sotrastaurin prevented TCR/CD28-induced T-cell activation and pro-inflammatory cytokine production, and enhanced Tregs response [94].

Janus Kinase (JAK) Inhibitors

This class of inhibitors is apparently restoring Tregs activity in psoriasis. The binding of cytokines to their receptors enables the activation of the JAK/STAT signaling pathways. This happens for IL-6, IFN-γ, IL-22, and IL-21, all involved in psoriasis. JAK inhibitors may thus suppress the effects of inflammatory cytokines involved in the disease [95]. The same (JAK) inhibitors are indicated for treatment of PsA, as the Food and Drug Administration approved the inhibitor tofacitinib, whereas the JAK1 inhibitor upadacitinib is approved in Japan. In a model of hepatitis in mice, induced by concanavalinA (ConA), tofacitinib increased the ratio of Tregs/Th17 cells as detected not only in the mouse liver but also in the spleen, which is representative of the situation in the peripheral regions [96]. An in vitro study reported that tofacitinib suppresses T-effector functions but preserves activity of CD4 CD25bright Tregs. This may explain its capacity to increase the Tregs/Th17 ratio [97].

Methrotrexate (MTX)

MTX, a folic acid analogue, is another treatment for psoriasis that is able to inhibit the activation of lymphocytes and macrophages, thus modulating cytokines, and inhibiting neutrophil chemotaxis [98]. MTX monotherapy determined, after 15 weeks of treatment, an increase in the percentages of Th2/Treg cells and a concomitant decrease of Th1 and Th17 cells [99]. In a study by K. Yan et al., Tregs and effector T-cells were isolated from blood of patients with psoriasis and healthy controls. In psoriasis patients, Tregs had a decreased immune suppressive function and a reduced expression of CD73, as compared to the healthy controls. Both IL-17 and IFN-γ were significantly upregulated in psoriasis, implying that T effector cells in the tissues possessed an aberrant secretion capacity of Th1/Th17 cytokines. The authors observed that, in patients, MTX treatment induced a significant growth inhibition of T effector cells. The production of IL-17 and IFN-γ by Tregs was also reduced, suggesting that MTX restores the function of Tregs and restrains the proliferation of T effectors in psoriasis patients. The authors analyzed CD73 expression by flow cytometry, and the phosphorylation of AMPK and mTOR by western blot. In all patients, MTX treatment reversed down-regulation of CD73, activated AMPK and inactivated mTOR [37]. In contrast to conventional resting T-cells, Tregs were found to express both CD39 and CD73 at high levels. These surface nucleosidases enzymatically active possess immunosuppressive properties on effector T-cells by negative feedback responses via the adenosine receptor (A2AR) but also via low-affinity receptors (like the A2B-adenosine receptor, A2BR). A2AR is ubiquitously expressed in a wide variety of immune cells including T-cells, B cells, NK cells, NKT cells, macrophages, dendritic cells, and granulocytes; A2BR plays a distinctive role in controlling inflammation, for example, via the induction of a tolerogenic antigen presenting cells (APC), via an alternative activation. Upon interaction with A2AR, adenosine is responsible for the inhibition of T-cell activation. Moreover, immunosuppressive activity may be further enhanced by adenosine, which induces Tregs, promoting tolerogenic antigen-presenting cells (APC) and myeloid-derived suppressor cells (MDSCctivities [100].
Other potential and promising targets, which can be useful in the regulation of Tregs, or to reset the imbalance of T-helper/Tregs in psoriasis, are under investigation; among them, IL-2 at low dose, histone deacetylase inhibitors sodium butyrate, STAT3 inhibitors, probiotics and T-cell based therapies [101][102][103][104][105][106].

6. Tregs Specific for the Autoantigen LL37 Can Be Present in Humans

The specificity of psoriasis T-cells driving inflammation in the skin has been elusive for a long time. Previous studies demonstrated that specificity was directed towards keratins [107]. Researchers discovered that the antimicrobial peptide (AMP) cathelicidin LL37 is an autoantigen  in psoriasis [46], and another study found that ADAMTSL5 is also an autoantigen in psoriasis [108]. In the past, researchers have identified LL37- specific T-cells by using peptide-MHC-tetramers. This approach, together with epitope mapping and cloning of the T-cells, identified the most immunogenic parts of LL37 and the restriction molecules for presentation to T-cells (there were several HLA-class I and class II alleles involved in the recognition by CD4 and CD8 T-cells, among which were HLA-DR7, HLA-DR11, HLA-DR4, and HLA-Cw6) [46][109]. Usually, healthy donors (HD) do not respond to LL37 or LL37-derived shorter antigenic peptides in T-cell proliferation assays [46][109]. Occasionally, low and rare proliferation can be detected in HD, which is not significant as compared to the psoriasis group. With this in mind, researchers tried to check, in HD with a low LL37 T-cell proliferative response, whether LL37-specific T-cells existed and whether these cells belong to the Tregs compartment. Our own preliminary experimental findings suggest that autoantigen specific, in this case LL37 specific T-cells, can be part of the physiological Tregs pool in HD. A person with T-cells responding to LL37 may have Tregs that are also specific for LL37 (as shown by using peptide-MHC-tetramers specific for LL37-epitopes). After expansion [110], such T-cells, which were able to act as regulatory cells in vitro by suppressing cytokine production by their own reactive LL37-specific T-cells, could be natural Tregs or induced Tregs. The evidence that LL37 is expressed not only in various organs but also in the thymus [111] could support the hypothesis that natural Tregs, specific for LL37, are present physiologically in HD. Expression of LL37 in the thymus may allow the deletion of LL37-specific T-cells by negative selection, or the selection of T-cells, which such specificity, endowed with regulatory/suppressive activity, which regulate the immune responses [112].

7. Conclusion

Tregs certainly play an important role in psoriasis, a disease in which they are dysfunctional. Many psoriasis treatments seem to exert an effects on Tregs, which in some cases acquire again their lost regulatory functions, as shown by the review of the literature. The presence of Tregs specific for LL37 and other T-cell autoantigens in psoriasis can be addressed by using peptide-MHC-tetramers. The mechanism of immune-suppression could be also addressed. The experiments are not easy to conduct, as one should perform HLA-typing of different HD and use several peptide -MHC-tetramers to stain the Tregs and identify the correct cells. One issue to address, using improved protocols, is whether antigen-specific Tregs are altered in psoriasis and whether effectors autoreactive T-cells are derived from existing Tregs or are newly formed. Similar experiments can be useful to address whether the therapies used for psoriasis, mentioned here, can induce recovering of dysfunctional T-cells or imply a de novo induction of Tregs, or elimination of responder autoreactive T-cells.

References

  1. Stockenhuber, K.; Hegazy, A.N.; West, N.R.; Ilott, N.E.; Stockenhuber, A.; Bullers, S.J.; Thornton, E.E.; Arnold, I.C.; Tucci, A.; Waldmann, H.; et al. Foxp3+ T reg cells control psoriasiform inflammation by restraining an IFN-I-driven CD8+ T cell response. J. Exp. Med. 2018, 215, 1987–1998.
  2. 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.
  3. 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.
  4. Sjaastad, L.E.; Owen, D.L.; Tracy, S.I.; Farrar, M.A. Phenotypic and Functional Diversity in Regulatory T Cells. Front. Cell Dev. Biol. 2021, 9, 715901.
  5. Mohr, A.; Malhotra, R.; Mayer, G.; Gorochov, G.; Miyara, M. Human FOXP3(+) T regulatory cell heterogeneity. Clin. Transl. Immunol. 2018, 7, e1005.
  6. Wing, J.B.; Tanaka, A.; Sakaguchi, S. Human FOXP3(+) regulatoryT cell heterogeneity and function in autoimmunity and cancer. Immunity 2019, 50, 302–316.
  7. Koch, M.A.; Tucker-Heard, G.; Perdue, N.R.; Killebrew, J.R.; Urdahl, K.B.; Campbell, D.J. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat. Immunol. 2009, 10, 595–602.
  8. Levine, A.G.; Mendoza, A.; Hemmers, S.; Moltedo, B.; Niec, R.E.; Schizas, M.; Hoyos, B.E.; Putintseva, E.V.; Chaudhry, A.; Dikiy, S.; et al. Stability and function of regulatory T cells expressing the transcription factor T-bet. Nature 2017, 546, 421–425.
  9. Wohlfert, E.A.; Grainger, J.R.; Bouladoux, N.; Konkel, J.E.; Oldenhove, G.; Ribeiro, C.H.; Hall, J.A.; Yagi, R.; Naik, S.; Bhairavabhotla, R.; et al. GATA3 controls Foxp3? regulatory T cell fate during inflammation in mice. J. Clin. Investig. 2011, 121, 4503–4515.
  10. Harrison, O.J.; Linehan, J.L.; Shih, H.Y.; Bouladoux, N.; Han, S.J.; Smelkinson, M.; Sen, S.K.; Byrd, A.L.; Enamorado, M.; Yao, C.; et al. Commensal-specific T cell plasticity promotes rapid tissue adaptation to injury. Science 2019, 363, eaat6280.
  11. Wang, Y.; Su, M.A.; Wan, Y.Y. An essential role of the transcriptionfactor GATA-3 for the function of regulatory T cells. Immunity 2011, 35, 337–348.
  12. Noval Rivas, M.; Burton, O.T.; Wise, P.; Charbonnier, L.M.; Georgiev, P.; Oettgen, H.C.; Rachid, R.; Chatila, T.A. Regulatory T cell reprogramming toward a Th2-cell like lineage impairs oral tolerance and promotes food allergy. Immunity 2015, 42, 512–523.
  13. 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. Individual intestinal symbionts induce a distinct population of RORgamma(+) regulatory T cells. Science 2015, 349, 993–997.
  14. 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.
  15. 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.
  16. Hovhannisyan, Z.; Treatman, J.; Littman, D.R.; Mayer, L. Characterization of interleukin-17-producing regulatory T cells in infame intestinal mucosa from patients with inflammatory bowel diseases. Gastroenterology 2011, 140, 957–965.
  17. 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.
  18. 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.
  19. Massoud, A.H.; Charbonnier, L.M.; Lopez, D.; Pellegrini, M.; Phipatanakul, W.; Chatila, T.A. An asthma-associated IL4R variant exacerbates airway inflammation by promoting conversion of regulatory T cells to TH17- like cells. Nat. Med. 2016, 22, 1013–1022.
  20. Kim, S.H.; Oh, J.; Roh, W.S.; Park, J.; Chung, K.B.; Lee, G.H.; Lee, Y.S.; Kim, J.H.; Lee, H.K.; Lee, H.; et al. Pellino-1 promotes intrinsic activation of skin-resident IL-17A-producing T cells in psoriasis. J. Allergy Clin. Immunol. 2023, in press.
  21. Samat, A.A.K.; van der Geest, J.; Vastert, S.J.; van Loosdregt, J.; van Wijk, F. Tissue–Resident Memory T Cells in Chronic Inflammation—Local Cells with Systemic Effects? Cells 2021, 10, 409.
  22. Scharschmidt, T.C.; Vasquez, K.S.; Pauli, M.L.; Leitner, E.G.; Chu, K.; Truong, H.A.; Lowe, M.M.; Sanchez Rodriguez, R.; Ali, N.; Laszik, Z.G.; et al. Commensal microbes and hair follicle morphogenesis coordinately drive Treg migration into neonatal skin. Cell Host Microbe 2017, 21, 467–477.e5.
  23. Furue, K.; Ito, T.; Tsuji, G.; Nakahara, T.; Furue, M. The CCL20 and CCR6 axis in psoriasis. Scand. J. Immunol. 2020, 91, e12846.
  24. Hemmers, S.; Schizas, M.; Rudensky, A.Y. T reg cell-intrinsic requirements for ST2 signaling in health and neuroinflammation. J. Exp. Med. 2021, 218, e20201234.
  25. Cayrol, C.; Girard, J.P. IL-33: An alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr. Opin. Immunol. 2014, 31, 31–37.
  26. Alvarez, F.; Istomine, R.; Shourian, M.; Pavey, N.; Al-Aubodah, T.A.; Qureshi, S.; Fritz, J.H.; Piccirillo, C.A. The alarmins IL-1 and IL-33 differentially regulate the functional specialisation of Foxp3(+) regulatory T cells during mucosal inflammation. Mucosal. Immunol. 2019, 12, 746–760.
  27. Liew, F.Y.; Girard, J.P.; Turnquist, H.R. Interleukin-33 in health and disease. Nat. Rev. Immunol. 2016, 16, 676–689.
  28. Shime, H.; Odanaka, M.; Tsuiji, M.; Matoba, T.; Imai, M.; Yasumizu, Y.; Uraki, R.; Minohara, K.; Watanabe, M.; Bonito, A.J.; et al. Proenkephalin(+) regulatory T cells expanded by ultraviolet B exposure maintain skin homeostasis with a healing function. Proc. Natl. Acad. Sci. USA 2020, 117, 20696–20705.
  29. 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.
  30. Hirahara, K.; Liu, L.; Clark, R.A.; Yamanaka, K.; Fuhlbrigge, R.C.; Kupper, T.S. The majority of human peripheral blood CD4+CD25highFoxp3+ regulatory T cells bear functional skin homing receptors. J. Immunol. 2006, 177, 4488.
  31. Tietz, W.; Allemand, Y.; Borges, E.; Von Laer, D.; Hallmann, R.; Vestweber, D.; Hamann, A. CD4+ T cells migrate into inflamed skin only if they express ligands for E- and P-selectin. J. Immunol. 1998, 161, 963.
  32. Van der Fits, L.; Mourits, S.; Voerman, J.S.; Kant, M.; Boon, L.; Laman, J.D.; Cornelissen, F.; Mus, A.M.; Florencia, E.; Prens, E.P.; et al. Imiquimod induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J. Immunol. 2009, 182, 5836.
  33. Zhang, L.; Li, Y.; Yang, X.; Wei, J.; Zhou, S.; Zhao, Z.; Cheng, J.; Duan, H.; Jia, T.; Lei, Q.; et al. Characterization of Th17 and FoxP3 (+) Treg cells in pediatric psoriasis patients. Scand. J. Immunol. 2016, 83, 174–180.
  34. Nussbaum, L.; Chen, Y.L.; Ogg, G.S. Role of regulatory T cells in psoriasis pathogenesis and treatment. Br. J. Dermatol. 2021, 184, 14–24.
  35. 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.
  36. Zhao, M.; Wang, L.T.; Liang, G.P.; Zhang, P.; Deng, X.J.; Tang, Q.; Zhai, H.Y.; Chang, C.C.; Su, Y.W.; Lu, Q.J. Up-regulation of microRNA-210 induces immune dysfunction via targeting FOXP3 in CD4(+) T cells of psoriasis vulgaris. Clin. Immunol. 2014, 150, 22–30.
  37. Yan, K.; Xu, W.; Huang, Y.; Zhang, Z.; Huang, Q.; Xin, K.Z.; Ma, Y.; Han, L. Methotrexate restores the function of peripheral blood regulatory T cells in psoriasis vulgaris via the CD73/AMPK/ mTOR pathway. Br. J. Dermatol. 2018, 179, 896–905.
  38. Soler, D.C.; Sugiyama, H.; Young, A.B.; Massari, J.V.; McCormick, T.S.; Cooper, K.D. Psoriasis patients exhibit impairment of the high potency CCR5(+) T regulatory cell subset. Clin. Immunol. 2013, 149, 111–118.
  39. Fry, L.; Baker, B.S. Triggering psoriasis: The role of infections and medications. Clin. Dermatol. 2007, 25, 606–615.
  40. Lochner, M.; Wang, Z.; Sparwasser, T. The special relationship in the development and function of T helper 17 and regulatory Tt cells. Prog. Mol. Biol. Transl. Sci. 2015, 136, 99–129.
  41. Koenen, H.J.; Smeets, R.L.; Vink, P.M.; van Rijssen, E.; Boots, A.M.; Joosten, I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood 2008, 112, 2340.
  42. Korn, T.; Bettelli, E.; Oukka, M.; Kuchroo, V.K. IL-17 and Th17 Cells. Annu. Rev. Immunol. 2009, 27, 485–517.
  43. Remedios, K.A.; Zirak, B.; Sandoval, P.M.; Lowe, M.M.; Boda, D.; Henley, E.; Bhattrai, S.; Scharschmidt, T.C.; Liao, W.; Naik, H.B. The TNFRSF members CD27 and OX40 coordinately limit TH17 differentiation in regulatory T cells. Sci. Immunol. 2018, 3, eaau2042.
  44. Richetta, A.G.; Mattozzi, C.; Salvini, M.; Giancristofaro, S.; D’ Epiro, S.; Milana, B.; Carboni, V.; Zampetti, M.; Calvieri, S.; Morrone, S. CD4+ CD25+ T-regulatory cells in psoriasis. Correlation between their numbers and biologics-induced clinical improvement. Eur. J. Dermatol. 2011, 21, 344–348.
  45. Zhang, L.; Yang, X.Q.; Cheng, J.; Hui, R.S.; Gao, T.W. Increased T17 cells are accompanied by FoxP3(+) Treg cell accumulation and correlated with psoriasis disease severity. Clin. Immunol. 2010, 135, 108–117.
  46. Lande, R.; Botti, E.; Jandus, C.D.; Dojcinovic, G.; Fanelli, C.; Conrad, G.; Chamilos, L.; Feldmeyer, B.; Marinari, S.; Chon, L.; et al. The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat. Commun. 2014, 5, 5621.
  47. Quaglino, P.; Ortoncelli, M.; Comessatti, A.; Ponti, R.; Novelli, M.; Bergallo, M.; Costa, C.; Cicchelli, S.; Savoia, P.; Bernengo, M.G. Circulating CD4+CD25 bright FOXP3+ T cells are up-regulated by biological therapies and correlate with the clinical response in psoriasis patients. Dermatology 2009, 250–258.
  48. Quaglino, P.; Bergallo, M.; Ponti, R.; Barberio, E.; Cicchelli, S.; Buffa, E.; Comessatti, A.; Costa, C.; Terlizzi, M.E.; Astegiano, S.; et al. Th1,Th2, Th17 and Regulatory T Cell Pattern in Psoriatic Patients: Modulation of Cytokines and Gene Targets Induced by Etanercept Treatment and Correlation with Clinical Response. Dermatology 2011, 223, 57–67.
  49. Cordiali-Fei, P.; Bianchi, L.; Bonifati, C.; Trento, E.; Ruzzetti, M.; Francesconi, F.; Bultrini, S.; D’Agosto, G.; Bordignon, V.; Francavilla, V.; et al. Immunologic biomarkers for clinical and therapeutic management of psoriasis. Mediat. Inflamm. 2014, 2014, 236060.
  50. Diluvio, L.; Romiti, M.L.; Angelini, F.; Campione, E.; Rossi, P.; Prinz, J.C.; Chimenti, S.; Lamioni, A. Infliximab therapy induces increased polyclonality of CD4+CD25+ regulatory T cells in psoriasis. Br. J. Dermatol. 2010, 162, 895–899.
  51. Shimizu, T.; Kamata, M.; Fukaya, S.; Hayashi, K.; Fukuyasu, A.; Tanaka, T.; Ishikawa, T.; Ohnishi, T.; Tada, Y. Anti-IL-17A and IL-23p19 antibodies but not anti-TNFα antibody induce expansion of regulatory T cells and restoration of their suppressive function in imiquimod-induced psoriasiform dermatitis. J. Dermatol. Sci. 2019, 95, 90–98.
  52. Kannan, A.K.; Su, Z.; Gauvin, D.M.; Paulsboe, S.E.; Duggan, R.; Lasko, L.M.; Honore, P.; Kort, M.E.; McGaraughty, S.P.; Scott, V.E.; et al. IL-23 induces regulatory T cell plasticity with implications for inflammatory skin diseases. Sci. Rep. 2019, 9, 17675.
  53. Mehta, H.; Mashiko, S.; Angsana, J.; Rubio, M.; Hsieh, Y.C.M.; Maari, C.; Reich, K.; Blauvelt, A.; Bissonnette, R.; Muñoz-Elías, E.J.; et al. Differential Changes in Inflammatory Mononuclear Phagocyte and T-Cell Profiles within Psoriatic Skin during Treatment with Guselkumab vs. Secukinumab. J. Investig. Dermatol. 2021, 141, 1707–1718.e9.
  54. Puig, L.; Costanzo, A.; Muñoz-Elías, E.J.; Jazra, M.; Wegner, S.; Paul, C.F.; Conrad, C. The biological basis of disease recurrence in psoriasis: A historical perspective and current models. Br. J. Dermatol. 2022, 186, 773–781.
  55. López-Sánchez, C.; Puig, L. Guselkumab in the treatment of moderate-to-severe plaque psoriasis. Immunotherapy 2020, 12, 355–371.
  56. Blauvelt, A.; Leonardi, C.L.; Gooderham, M.; Papp, K.A.; Philipp, S.; Wu, J.J.; Igarashi, A.; Flack, M.; Geng, Z.; Wu, T.; et al. Efficacy and Safety of Continuous Risankizumab Therapy vs Treatment Withdrawal in Patients with Moderate to Severe Plaque Psoriasis. JAMA Dermatol. 2021, 156, 649–658.
  57. Warren, R.; Carrascosa, J.; Fumero, E.; Schoenenberger, A.; Lebwohl, M.; Szepietowski, J.; Reich, K. Time to relapse after tildrakizumab withdrawal in patients with moderate-to-severe psoriasis who were responders at week 28: Post hoc analysis through 64 weeks from reSURFACE 1 trial. J. Eur. Acad. Dermatol. Venereol. 2020, 35, 919–927.
  58. Tsai, Y.C.; Tsai, T.F. Anti-interleukin and interleukin therapies for psoriasis: Current evidence and clinical usefulness. Ther. Adv. Musculoskel. Dis. 2017, 9, 277–294.
  59. Ogata, A.; Umegaki, N.; Katayama, I.; Kumanogoh, A.; Tanaka, T. Psoriatic arthritis in two patients with an inadequate response to treatment with tocilizumab. Jt. Bone Spine 2012, 79, 85–87.
  60. Wu, D.; Yue, J.; Tam, L.S. Efficacy and safety of biologics targeting interleukin 6, -12/23 and -17 pathways for peripheral psoriatic arthritis: A network meta-analysis. Rheumatology 2018, 57, 563571.
  61. Mease, P.J.; Gottlieb, A.B.; Berman, A.; Drescher, E.; Xing, J.; Wong, R.; Banerjee, S. The Efficacy and Safety of Clazakizumab, an Anti-Interleukin-6 Monoclonal Antibody, in a Phase IIb Study of Adults with Active Psoriatic Arthritis. Arthritis Rheumatol. 2016, 68, 2163–2173.
  62. Bilal, J.; Riaz, I.B.; Kamal, M.U.; Elyan, M.; Sudano, D.; Khan, M.A. A Systematic Review and Meta-analysis of Novel Interleukin Inhibitors’ Efficacy and Safety in the Treatment of Psoriatic Arthritis. J. Clin. Rheumatol. 2018, 1, 6–13.
  63. Kemény, L.; Varga, E.; Novak, Z. Advances in phototherapy for psoriasis and atopic dermatitis. Expert Rev. Clin. Immunol. 2019, 15, 1205–1214.
  64. Kubo, R.; Muramatsu, S.; Sagawa, Y.; Saito, C.; Kasuya, S.; Nishioka, A.; Nishida, E.; Yamazaki, S.; Morita, A. Bath–PUVA therapy improves impaired resting regulatory T cells and increases activated regulatory T cells in psoriasis. J. Dermatol. Sci. 2017, 86, 46–53.
  65. Furuhashi, T.; Saito, C.; Torii, K.; Nishida, E.; Yamazaki, S.; Morita, A. Photo (chemo)therapy Reduces Circulating Th17 Cells and Restores Circulating Regulatory T Cells in Psoriasis. PLoS ONE 2013, 8, e54895.
  66. Schweintzger, N.; Gruber-Wackernagel, A.; Reginato, E.; Bambach, I.; Quehenberger, F.; Byrne, S.N.; Wolf, P. Levels and function of regulatory T cells in patients with polymorphic light eruption: Relation to photohardening. Br. J. Dermatol. 2015, 173, 519–526.
  67. Loser, K.; Mehling, A.; Loeser, S.; Apelt, J.; Kuhn, A.; Grabbe, S.; Schwarz, T.; Penninger, J.; Beissert, S. Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat. Med. 2006, 12, 1372–1379.
  68. Akiyama, T.; Shinzawa, M.; Akiyama, N. RANKL-RANK interaction in immune regulatory systems. World J. Orthop. 2012, 3, 142–150.
  69. Kripke, M.L.; Morison, W.L.; Parrish, J.A. Systemic Suppression of Contact Hypersensitivity in Mice by Psoralen Plus UV A Radiation (PUV A). J. Investig. Dermatol. 1983, 81, 87–92.
  70. Khalil, S.; Bardawil, T.; Stephan, C.; Darwiche, N.; Abbas, O.; Kibbi, A.G.; Nemer, G.; Kurban, M. Retinoids: A journey from the molecular structures and mechanisms of action to clinical uses in dermatology and adverse effects. J. Dermatol. Treat. 2017, 28, 684–696.
  71. Mucida, D.; Park, Y.; Kim, G.; Turovskaya, O.; Scott, I.; Kronenberg, M.; Cheroutre, H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science 2007, 317, 256–260.
  72. Elias, K.M.; Laurence, A.; Davidson, T.S.; Stephens, G.; Kanno, Y.; Shevach, E.M.; O’Shea, J.J. Retinoic acid inhibits Th17 polarization and enhances FoxP3 expression through a Stat-3/Stat5 independent signaling pathway. Blood 2008, 111, 1013–1022.
  73. Mattozzi, C.; Paolino, G.; Salvi, M.; Macaluso, L.; Luci, C.; Morrone, S.; Calvieri, S.; Richetta, A.G. Peripheral blood regulatory T cell measurements correlate with serum vitamin D level in patients with psoriasis. Eur. Rev. Med. Pharmacol. Sci. 2016, 9, 1675–1679.
  74. Hau, C.S.; Shimizu, T.; Tada, Y.; Kamata, M.; Takeoka, S.; Shibata, S.; Mitsui, A.; Asano, Y.; Sugaya, M.; Kadono, T.; et al. The vitamin D3 analog, maxacalcitol, reduces psoriasiform skin inflammation by inducing regulatory T cells and downregulating IL-23 and IL-17 production. J. Dermatol. Sci. 2018, 92, 117–126.
  75. Penna, G.; Amuchastegui, S.; Giarratana, N.; Daniel, K.C.; Vulcano, M.; Sozzani, S.; Adorini, L. 1,25-dihydroxyvitamin D3 selectively modulates tolerogenic properties in myeloid but not plasmacytoid dendritic cells. J. Immunol. 2007, 178, 145–153.
  76. Piemonti, L.; Monti, P.; Sironi, M.; Fraticelli, P.; Leone, B.E.; Dal Cin, E.; Allavena, P.; Di Carlo, V. Vitamin D3 affects differentiation, maturation, and function of human monocyte-derived dendritic cells. J. Immunol. 2000, 164, 4443–4451.
  77. Gregori, S.; Casorati, M.; Amuchastegui, S.; Smiroldo, S.; Davalli, A.M.; Adorini, L. Regulatory T cells induced by 1 alpha,25-dihydroxyvitamin D3 and mycophenolate mofetil treatment mediate transplantation tolerance. J. Immunol. 2001, 167, 1945–1953.
  78. Van de Kerkhof, P.C. An update on topical therapies for mild/moderate psoriasis. Dermatol. Clin. 2015, 33, 73–77.
  79. Leung, D.Y.; Bloom, J.W. Update on glucocorticoid action and resistance. J. Allergy Clin. Immunol. 2003, 111, 3–22.
  80. O’Neill, J.L.; Feldman, S.R. Vitamine D analogue-based therapies for psoriasis. Drugs Today 2010, 46, 351–360.
  81. van der Velden, H.M.; Pasch, M.C.; van Erp, P.E.; van Lingen, R.G.; Otero, M.E.; de Boer-van Huizen, R.T.; van de Kerkhof, P.C. Treatment of plaque psoriasis with the two-compound product calcipotriol/betamethasone dipropionate versus both monotherapies: An immunohistochemical study. J. Dermatolog. Treat. 2010, 21, 13.
  82. Keijsers, R.R.; Joosten, I.; Hendriks, A.G.; Koenen, H.J.; van Erp, P.E.; van de Kerkhof, P.C. Balance of Treg versus T-effector cells during systemic treatment with adalimumab and topical treatment with calcipotriol-betamethasone dipropionate ointment. Exp. Dermatol. 2015, 24, 65–67.
  83. Kubin, M.E.; Kokkonen, N.; Palatsi, R.; Hägg, P.M.; Väyrynen, J.P.; Glumoff, V.; Haapasaari, K.M.; Hurskainen, T.; Tasanen, K. Clinical Efficiency of Topical Calcipotriol/Betamethasone Treatment in Psoriasis Relies on Suppression of the Inflammatory TNFα—IL-23—IL-17 Axis. Acta Derm. Venereol. 2017, 97, 449–455.
  84. Mrowietz, U.; Van De Kerkhof, P.; Schoenenberger, A.; Ryzhkova, A.; Pau-Charles, I.; Llamas-Velasco, M.; Daudén, E.; Carrascosa, J.M.; De La Cueva, P.; Salgado-Boquete, L.; et al. Efficacy of dimethyl fumarate treatment for moderate-to-severe plaque psoriasis: Presentation extracts from the 29th EADV virtual congress, 29–31 October 2020. Expert Rev. Clin. Immunol. 2021, 17, 1–11.
  85. Mougiakakos, D.; Johansson, C.C.; Jitschin, R.; Böttcher, M.; Kiessling, R. Increased thioredoxin-1 production in human naturally occurring regulatory T cells confers enhanced tolerance to oxidative stress. Blood 2011, 117, 857–861.
  86. Mougiakakos, D.; Johansson, C.C.; Kiessling, R. Naturally occurring regulatory T cells show reduced sensitivity toward oxidative stress–induced cell death. Blood 2009, 113, 3542–3545.
  87. Sulaimani, J.; Cluxton, D.; Clowry, J.; Petrasca, A.; Molloy, O.; Moran, B.; Sweeney, C.; Malara, A.; McNicholas, N.; McGuigan, C.; et al. Dimethyl fumarate modulates the Treg–Th17 cell axis in patients with psoriasis. Br. J. Dermatol. 2021, 184, 495–503.
  88. Skvara, H.; Dawid, M.; Kleyn, E.; Wolff, B.; Meingassner, J.G.; Knight, H.; Dumortier, T.; Kopp, T.; Fallahi, N.; Stary, G.; et al. The PKC inhibitor AEB071 may be a therapeutic option for psoriasis. J. Clin. Investig. 2008, 118, 3151–3159.
  89. Wagner, J.; von Matt, P.; Faller, B.; Cooke, N.G.; Albert, R.; Sedrani, R.; Wiegand, H.; Jean, C.; Beerli, C.; Weckbecker, G.; et al. Structure-activity relationship and pharmacokinetic studies of sotrastaurin (AEB071), a promising novel medicine for prevention of graft rejection and treatment of psoriasis. J. Med. Chem. 2011, 54, 6028–6039.
  90. Evenou, J.P.; Wagner, J.; Zenke, G.; Brinkmann, V.; Wagner, K.; Kovarik, J.; Welzenbach, K.A.; Weitz-Schmidt, G.; Guntermann, C.; Towbin, H.; et al. The potent protein kinase C-selective inhibitor AEB071 (sotrastaurin) represents a new class of immunosuppressive agents affecting early T-cell activation. J. Pharmacol. Exp. Ther. 2009, 330, 792–801.
  91. Zanin-Zhorov, A.; Ding, Y.; Kumari, S.; Attur, M.; Hippen, K.L.; Brown, M.; Blazar, B.R.; Abramson, S.B.; Lafaille, J.J.; Dustin, M.L. Protein kinase C-theta mediates negative feedback on regulatory T cell function. Science 2010, 328, 372–376.
  92. Marsland, B.J.; Kopf, M. T-cell fate and function: PKC and beyond. Trends Immunol. 2008, 29, 179–185.
  93. Tan, S.L.; Zhao, J.; Bi, C.; Chen, X.C.; Hepburn, D.L.; Wang, J.; Sedgwick, J.D.; Chintalacharuvu, S.R.; Na, S. Resistance to experimental autoimmune encephalomyelitis and impaired IL-17 production in protein kinase C theta-deficient mice. J. Immunol. 2006, 176, 2872–2879.
  94. He, X.; Koenen, H.J.P.M.; Smeets, R.L.; Keijsers, R.; van Rijssen, E.; Koerber, A.; van de Kerkhof, P.C.; Boots, A.M.H.; Joosten, I. Targeting PKC in human T cells using sotrastaurin (AEB071) preserves regulatory T cells and prevents IL-17 production. J. Investig. Dermatol. 2014, 134, 975–983.
  95. Virtanen, A.T.; Haikarainen, T.; Raivola, J.; Silvennoinen, O. Selective JAKinibs: Prospects in Inflammatory and Autoimmune Diseases. BioDrugs 2019, 33, 15–32.
  96. Wang, H.; Feng, X.; Han, P.; Lei, Y.; Xia, Y.; Tian, D.; Yan, W. The JAK inhibitor tofacitinib ameliorates immune-mediated liver injury in mice. Mol. Med. Rep. 2019, 20, 4883–4892.
  97. Sewgobind, V.D.K.D.; Quaedackers, M.E.; van der Laan, L.; Kraaijeveld, R.; Korevaar, S.S.; Chan, G.; Weimar, W.; Baan, C.C. The Jak Inhibitor CP-690,550 Preserves the Function of CD4+CD25brightFoxP3+ Regulatory T Cells and Inhibits Effector T Cells. Arab. Archaeol. Epigr. 2010, 10, 1785–1795.
  98. Seitz, M.; Loetscher, P.; Dewald, B.; Towbin, H.; Rordorf, C.; Gallati, H.; Baggiolini, M.; Gerber, N.J. Methotrexate action in rheumatoid arthritis: Stimulation of cytokine inhibitor and inhibition of chemokine production by peripheral blood mononuclear cells. Br. J. Rheumatol. 1995, 34, 602–609.
  99. Priyadarssini, M.; Chandrashekar, L.; Rajappa, M. Effect of methotrexate monotherapy on T-cell subsets in the peripheral circulation in psoriasis. Clin. Exp. Dermatol. 2019, 44, 491–497.
  100. Ohta, A.; Kini, R.; Ohta, A.; Subramanian, M.; Madasu, M.; Sitkovsky, M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012, 3, 190.
  101. 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.
  102. Von Knethen, A.; Heinicke, U.; Weigert, A.; Zacharowski, K.; Brüne, B. Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells. Int. J. Mol. Sci. 2020, 21, 2356.
  103. Schwarz, A.; Philippsen, R.; Schwarz, T. Induction of regulatory T cells and correction of cytokine dysbalance by short chain fatty acids—Implications for the therapy of psoriasis. J. Investig. Dermatol. 2020, 141, 95.e2–104.e2.
  104. Miyoshi, K.; Takaishi, M.; Nakajima, K.; Ikeda, M.; Kanda, T.; Tarutani, M.; Iiyama, T.; Asao, N.; DiGiovanni, J.; Sano, S. Stat3 as a Therapeutic Target for the Treatment of Psoriasis: A Clinical Feasibility Study with STA-21, a Stat3 Inhibitor. J. Investig. Dermatol. 2011, 131, 108–117.
  105. Alzahrani, Y.A.; Alesa, D.I.; Alshamrani, H.M.; Alamssi, D.N.; Alzahrani, N.S.; Almohammadi, M.E. The role of gut microbiome in the pathogenesis of psoriasis and the therapeutic effects of probiotics. J. Fam. Med. Prim. Care. 2019, 8, 3496–3503.
  106. Bluestone, J.A.; Tang, Q. Treg cells—The next frontier of cell therapy. Science 2018, 362, 154–155.
  107. Johnston, A.; Gudjónsson, J.E.; Sigmundsdóttir, H.; Löve, T.H.; Valdimarsson, H. Peripheral blood T-cell responses to keratin peptides that share sequences with M proteins are largely restricted to skin-homing CD8+ T cells. Scand. J. Immunol. 2008, 138, 83–93.
  108. Arakawa, A.; Siewert, K.; Stöhr, J.; Besgen, P.; Kim, S.M.; Rühl, G.; Nickel, J.; Vollmer, S.; Thomas, P.; Krebs, S.; et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J. Exp. Med. 2015, 212, 2203–2212.
  109. Lande, R.; Palazzo, R.; Gestermann, N.; Jandus, C.; Falchi, M.; Spadaro, F.; Riccieri, V.; James, E.A.; Butera, A.; Boirivant, M.; et al. Native/citrullinated LL37-specific T-cells help autoantibody production in Systemic Lupus Erythematosus. Sci. Rep. 2020, 10, 5851.
  110. Sagoo, P.; Ali, N.; Garg, G.; Nestle, F.O.; Lechler, R.I.; Lombardi, G. Human regulatory T cells with alloantigen specificity are more potent inhibitors of alloimmune skin graft damage than polyclonal regulatory T cells. Sci. Transl. Med. 2011, 3, 83ra42.
  111. Bals, R.; Wang, X.; Zasloff, M.; Wilson, J.M. The peptide antibiotic LL-37/hCAP-18 is expressed in epithelia of the human lung where it has broad antimicrobial activity at the airway surface. Proc. Natl. Acad. Sci. USA 1998, 95, 9541–9546.
  112. Takaba, H.; Takayanagi, H. The Mechanisms of T Cell Selection in the Thymus. Trends Immunol. 2017, 38, 805–816.
More
Information
Subjects: Dermatology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 221
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 27 Apr 2023
1000/1000