Next Article in Journal
Modulators of MicroRNA Function in the Immune System
Next Article in Special Issue
The Role of Histone Acetylation and the Microbiome in Phytochemical Efficacy for Cardiovascular Diseases
Previous Article in Journal
Cytotoxicity of NiO and Ni(OH)2 Nanoparticles Is Mediated by Oxidative Stress-Induced Cell Death and Suppression of Cell Proliferation
Previous Article in Special Issue
In silico Design of Novel Histone Deacetylase 4 Inhibitors: Design Guidelines for Improved Binding Affinity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells

1
Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, 60590 Frankfurt, Germany
2
Fraunhofer—IME, Project Group Translational Medicine and Pharmacology (TMP), 60596 Frankfurt, Germany
3
Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt/Main, 60590 Frankfurt, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(7), 2356; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072356
Submission received: 13 February 2020 / Revised: 12 March 2020 / Accepted: 26 March 2020 / Published: 29 March 2020
(This article belongs to the Special Issue Histone Deacetylase Inhibitors in Health and Disease II)

Abstract

:
Regulatory T cells (Tregs) are important mediators of immunological self-tolerance and homeostasis. Being cluster of differentiation 4+Forkhead box protein3+ (CD4+FOXP3+), these cells are a subset of CD4+ T lymphocytes and can originate from the thymus (tTregs) or from the periphery (pTregs). The malfunction of CD4+ Tregs is associated with autoimmune responses such as rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), inflammatory bowel diseases (IBD), psoriasis, systemic lupus erythematosus (SLE), and transplant rejection. Recent evidence supports an opposed role in sepsis. Therefore, maintaining functional Tregs is considered as a therapy regimen to prevent autoimmunity and allograft rejection, whereas blocking Treg differentiation might be favorable in sepsis patients. It has been shown that Tregs can be generated from conventional naïve T cells, called iTregs, due to their induced differentiation. Moreover, Tregs can be effectively expanded in vitro based on blood-derived tTregs. Taking into consideration that the suppressive role of Tregs has been mainly attributed to the expression and function of the transcription factor Foxp3, modulating its expression and binding to the promoter regions of target genes by altering the chromatin histone acetylation state may turn out beneficial. Hence, we discuss the role of histone deacetylation inhibitors as epigenetic modulators of Tregs in this review in detail.

Graphical Abstract

1. Introduction

Regulatory T cells (Tregs) are important to guarantee immunological self-tolerance and homeostasis. Since their first description in 1995 [1], several subpopulations of Tregs have been described to fulfill these requirements [2]. First, Tregs can originate from the thymus. Accordingly, these Tregs are named tTregs. [3]. Second, Tregs can develop from effector T cells in the periphery and are thus designated as pTregs [4]. This usually happens upon the activation of post-naïve effectors with mainly oral antigens in the presences of specific cytokines. In the thymus, thymocytes are educated to self-antigenic peptides first in the cortex and then in the medulla with medium affinity, whereas thymocytes destined to become Tregs are educated to recognized self-antigenic peptides with high affinity, mainly in the cortical–medullary junction and the medulla, before being released to the periphery. To achieve this, self-reactive thymocytes are eliminated by negative selection [5]. However, although this mechanism is very effective, some self-reactive, and thus possibly autoimmunity-inducing T cells, escape this machinery [6,7]. Therefore, a system must exist that restricts activity of these cells. This was proven by the classical thymectomy experiment in neonate mice, which showed T cell-dependent autoimmunity when the thymus was removed at day three after birth but not at day one or day seven [8,9,10]. These tTregs, which migrate to the periphery after day three, are essential for self-tolerance. Recent evidence identified thymocyte apoptosis, occurring after birth [11], as leading to the intrathymic release of transforming growth factor (TGF)-β as reason for the delayed tTregs export compared to cluster of differentiation (CD)4+ single positive (SP) thymocytes [12]. TGF-β initiates Foxp3 expression and tTregs development [12]. However, earlier data have shown normal tTregs development in mice deficient for TGF-β 1 but significantly reduced pTregs [13]. The expression of the transcription factor Foxp3 is a marker of Tregs. The activation of the corresponding gene locus is a multistep process [14,15]. It requires a high affinity binding of major histocompatibility complex (MHC)-self peptide complexes from thymic antigen-presenting cells (APCs) to the T cell receptor (TCR) and costimulatory signals as well as cytokine environments (IL-2) [16,17]. Foxp3 provokes the expression of target genes, which are important to trigger and maintain the immune suppressive Tregs phenotype, as shown by genome-wide analyses in mice and humans [18,19]. In mice, Foxp3 binding results in both the activation and repression of its target genes. This was determined by chromatin immunoprecipitation (ChIP) against epigenetic markers such as acetylated H3K9/14 (AcH3), tri-methyl H3K4 (Me3K4), and tri-methyl H3K27 (Me3K27). These data identified the cell surface molecules Il2ra (CD25), Ctla4 (CD152), Nt5e (CD73), and Icos (CD278) as well as the transcription factor Ikzf2 (Helios) to be Foxp3-dependently upregulated based on the chromatin markers AcH3 and Me3K4. In contrast, the phosphodiesterase Pde3b showed tri-methylation at H3K27, mandatory for its inhibited expression [20]. For the latter one, it was recently shown that Foxp3 also induces the microRNA-142-5p, which as an intracellular cAMP sensor leads to the posttranscriptional repression of the cAMP hydrolyzing enzyme Pde3b [21]. In human Tregs, a similar expression profile was observed, demonstrating the selective gene expression of IKZF2 (HELIOS) [22], IL2RA (CD25), and cytotoxic T lymphocyte associated protein 4 (CTLA4) (CD152) [23]. Moreover, the expression of the T cell survival factor IL-2 is Foxp3-dependently downregulated [24]. Further proof for the significance of Foxp3 in Treg differentiation and function came from studies analyzing mutations associated with the nonfunctional expression of Foxp3 in humans, causing IPEX syndrome (immune dysregulation polyendocrinopathy and enteropathy), which requires bone marrow transplantation in early childhood [25]. In mice, a lack of Foxp3 expression, as observed in scurfy mice, induces a similar phenotype [26]. The role of Foxp3 has been further corroborated in mice where the experimental depletion of Foxp3+ Tregs in healthy adult mice has been found to provoke autoimmunity and death [26,27].
Considering this important role of Tregs in maintaining immune self-tolerance, treatment with in vitro generated Tregs may be a therapeutic approach towards autoimmune-mediated diseases. Tregs can be generated ex vivo from conventional naïve T cells after TCR stimulation in the presence of TGF-β and IL-2. These cells are named iTregs according to their induced differentiation [28]. However, the stability of Foxp3 expression in these cells is much lower compared to tTregs or pTregs [29]. Therefore, treatment regimens to prolong and stabilize Foxp3 expression in iTregs are a topic of current research. In contrast, the role of Tregs in sepsis patients seems to be deleterious [30,31,32,33,34]. In this case, reducing the pTreg or tTreg number and function might prevent immunosuppression, consequently improving survival. Thus, epigenetic modifications are an interesting approach to cope with these opposite tasks.

2. Role of Tregs in Disease

2.1. Autoimmune Diseases

Based on Tregs’ role on the induction and maintenance of peripheral tolerance, Treg dysfunction is associated with severe autoimmune conditions. Disease patterns such as systemic lupus erythematosus (SLE) [35] and organ-specific autoimmune diseases, e.g., type 1 diabetes (T1D) [36] and psoriasis [37], have been attributed to a reduced number of Tregs or the failure of their function. Considering this important role of Tregs, therapies have been developed to medicate autoimmune disorders (for reviews of single clinical trials, see [38,39]). Phase I clinical studies have been using autologous Tregs to treat SLE, pemphigus vulgaris, or T1D. Moreover, already published studies have supported an ameliorative impact of these treatment regimens for T1D, prolonging the survival of β -cells [40,41]. Beside these polyclonal Treg therapies, Treg-enhancing drugs are of interest. Among others, rapamycin-dependent mTOR inhibition was used to efficiently expand human Treg cells [42] and to treat SLE patients, with significant improvement of the clinical outcome [43]. Several other approaches have been shown to block mTOR activation in animal models and T cells derived from the blood of SLE patients, including the blocking of S1P receptors, antioxidants, and calmodulin kinase type II and type IV inhibitors [44,45,46,47]. Low-dose IL-2 treatment has also been identified for the treatment of patients with diseases associated with a decreased number of Tregs such as SLE [48,49,50]. Considering that IL-2 activates Tregs as well as Teff, a dose finding study was performed [51]. In this setting, clinical phase II trials are already running for the treatment of rheumatoid arthritis (RA), SLE, multiple sclerosis (MS), T1D, and amyotrophic lateral sclerosis (ALS).

2.2. Transplant Rejection

Patients with end-stage organ failure need organ transplantation as their therapy of choice. Moreover, an autologous bone marrow transfer is required in patients suffering from chemotherapeutic treatment or an allogenic transfer in patients with lymphoma or leukemia. As expected, Tregs are important mediators of graft tolerance induction following these transplantations. Based on their immunosuppressive function, an increased number of Tregs in the periphery and graft microenvironment has been attributed to confer graft tolerance, thus guaranteeing a long lasting life of solid organ transplants or transferred bone marrow [39,52,53]. Considering this important role of Tregs, in-man phase 1 and phase 2 clinical trials, as summarized in [53,54], have been performed [55,56,57] or registered. Briefly, liver transplantations were supported with Treg cell therapy, following Treg generation using autologous Tregs stimulated with irradiated donor PBMCs with inhibited costimulation, autologous donor antigen-expanded Tregs, or autologous, polyclonally-expanded Tregs. A similar setup was used to Treg-dependently assist liver transplantation. Additionally, in bone marrow transfer approaches, Tregs have been shown to suppress T cell alloreactions and to prevent graft-versus-host disease in mouse models [28,58,59,60] and in the human situation [61].

2.3. Sepsis

Sepsis is a syndrome where T cell depletion and, consequently, an inappropriate immune response to the recurring initial infection or acquired second infection is one characteristic [62]. Consequently, it is of interest to follow the fate of Tregs during sepsis initiation and progression. As shown recently by Carvelli et al. [32], the number of Tregs was decreased in patients with septic shock, which is in some discrepancy to previous reports that have shown an increased number of Treg cells in these patients as one reason for long-term immune-suppression [33,34]. Based on these contradictory data, the role of Tregs in sepsis needs further evaluation. Thus, the different stages, i.e., infection, organ dysfunction caused by an inappropriate immune response, septic shock, and finally sepsis survivors, must be carefully examined to draw any conclusion whether Tregs are important to block an overwhelming immune response or whether these cells are crucial for the resolution of inflammation. In both situations, excessive Tregs might be detrimental. Therefore, the pharmaceutical or immunological fine tuning of Tregs will be advantageous to intervene with the respective prevailing pro- vs. anti-inflammatory responses. This has also been shown for the role of Tregs in resolving lung injury [63]. In this animal approach, mice were treated with lipopolysaccharide (LPS) or recombinant high-mobility-group-protein B1 (HMGB1), which is a key mediator during inflammation to induce acute lung injury (ALI). Tregs were modulated with myeloid-specific β-catenin and phosphatase and tensin homologue (PTEN)-knockout mice. As shown in Table 1, several rodent studies have shown an altered number of Tregs following polymicrobial sepsis by cecal ligation and puncture (CLP). In these studies, various mouse (BALB/c, C57BL/6, FVB/N, ICR, NMRI) and rat strains (Fischer, Wistar, Wistar Hannover, Sprague Dawley) have been used, applying different severities of polymicrobial sepsis. This can be achieved by the diameter of the needle and the number of cecum perforations [64,65]. Most studies have demonstrated an increase of Tregs in spleen or blood, independently from the execution of the model. This is in line with the assumption that Tregs are generally involved in downregulating the immune response, thus contributing to an immunosuppressed phenotype during sepsis. Interestingly, our own data support this notion. We found that a prevention of CLP-dependent liver damage was associated with a decreased number of liver localized Tregs [66]. Thus, reducing the Tregs count might be a prerequisite for improving sepsis outcome by restoring a functional T cell response.

3. HAT and HDAC Activities in Treg Differentiation

Epigenetic, i.e., reversible modifications of chromatin that do not alter the DNA sequence, can be achieved by inhibiting histone deacetylase (HDACs) to maintain chromatin histone acetylation, consequently keeping genomic DNA accessible for the binding of transcription factors and the RNA polymerase. This finally allows for the increased expression of genes known to be involved in Treg differentiation and function.

3.1. Foxp3

Foxp3 is a member of the Forkhead box protein (Foxp) subfamily of transcription factors. Due to its function as a master regulator of tTreg and pTreg differentiation and immunosuppressive performance, understanding Foxp3’s transcriptional, translational, and post-translational regulation is important [14,98,99]. Therefore first, the gene structure is crucial [100,101]. The expression of the Foxp3 gene is mediated by five control elements. Starting 5′, the first conserved non-coding sequence (CNS) 0 [102], which is the binding site for the special AT-rich sequence-binding protein (SATB) 1, a super-enhancer that enables Treg-lineage-specific gene induction was recently identified [102,103]. Following CNS0, the promoter region [104,105] and three further CNS (CNS1–3) are located. CNS1, located in intron 1, is associated with TGF-β inducibility (TGF-β sensor) [105]. CNS2, also located in intron 1, is the so called Treg-cell-specific demethylation region (TSDR) [106,107] and CNS3, localized in intron 3, named the Foxp3 pioneer element, is known to confer the NF-κB inducibility of Foxp3 [108]. All these five elements are mainly characterized by the existence of CpG islands (promoter, CNS2) and histones, which can be acetylated (promoter, CNS0-3) or show permissive methylation (CNS3) [18,102]. Thus, these regulatory structures are targets for epigenetic modifications, altering the accessibility of the Foxp3 gene [109]. In naïve CD4+ T cells, CpGs are heavily methylated (Figure 1A), silencing the Foxp3 gene [18]. Especially, the promoter region is the target of protein inhibitor of activated STAT (signal transducer and activator of transcription) (PIAS1), a SUMO E3 ligase, which restricts Treg differentiation by recruiting DNA methyltransferases and heterochromatin protein 1 to the Foxp3 promoter [110]. Following DNA demethylation, histone acetylation, and permissive methylation, Foxp3 expression and, consequently, Treg differentiation are induced by the activation of transcription factors in response to T cell receptor (TCR) engagement, CD28 co-stimulation, IL-2 treatment, and TGF-β addition. Established transcription factors are activator protein 1 (AP-1) (promoter) [111], cAMP-responsive element binding protein (CREB) (CNS2) [105], Ets-1 (CNS2) [112,113], FoxO1 (promoter) [114], nuclear factor of activated T cells (NFAT) (promoter, CNS1) [18,111], nuclear receptor 4a (NR4a) (promoter) [115], c-Rel (CNS3) [108], retinoid x receptor/retinoid acid receptor (RXR/RAR) (promoter, CNS1) [116,117], Runt-related transcription factor 1 (RUNX) (promoter, CNS2) [118], STAT3/5 (promoter, CNS2) [119], and SMAD2/3/4 (CNS1) [120,121] (Figure 1B).
The Foxp3 protein contains four domains, including a repressor domain at the N-terminal end (responsible for transcriptional repression), a zinc finger domain with a so-far unclear function, a leucine zipper domain (important for dimerization), and, finally at its C-terminus, the Forkhead domain, which is important for DNA-binding (Figure 1C). It has been established that the repressor domain located at the N-terminus of Foxp3 is associated with the downregulation of the expression of HIF1α, RORγt, RORα, and Eos. Thus, among others, differentiation towards a Th17 phenotype is prevented [121]. When expressed, Foxp3 can form heterodimers with FoxO1, keeping Foxp3 in an inactive state. It can transiently homodimerize, which enables its regulation, or stably consequently leading to the expression or repression of target genes (Figure 1D). Moreover, transient Foxp3 homodimers may combine as clusters. Foxp3 can additionally bind roughly 700 different proteins, which is important to activate or repress the expression of target genes. Stable Foxp3 coiled-coil-mediated homodimerization is essential for Treg function [122]. As shown in Figure 2A, Foxp3 associates with histone acetylases (HATs) (e.g., p300 or HIV-Tat-interactive protein (TIP60) [123]), leading to Foxp3 (hyper)acetylation, which increases Foxp3 stability as well as HDACs (e.g., silent information regulator 1 (SIRT1) or HDAC5 [124,125]), which reciprocally deacetylate Foxp3, making it more susceptible for proteasomal degradation [126]. Foxp3 lysine residues identified to be affected are shown in Figure 2B. Foxp3 expression and function are also regulated by the histone H3K27 methyltransferase enhancer of zeste homolog 2 (EZH2), which is not expressed in naïve Tregs and is upregulated in CD28-activated Tregs, provoking a stable Treg phenotype by allowing for Foxp3 expression and stabilization [127,128]. Consequently, EZH2-specific inhibitors as well as specifically disrupting EZH2 in Tregs, reduced Foxp3 expression, and concomitantly attenuated the immune suppressive Treg phenotype [127]. However, Foxp3 binding to EZH2 seems to inactivate Foxp3 [129]. Correspondingly, histone demethylases are involved in Foxp3 regulation. There, Jumonji domain-containing 3 (Jmjd3) is the most prominent demethylase, responsible for H3K27me2 and H3K4me3 demethylation, provoking Foxp3 expression and, accordingly, promoting Treg differentiation [130]. Interestingly, in an acute lung injury (ALI) model in mice, the expression of JMJD3 is downregulated in Tregs isolated from the lungs [131]. These data support the notion of an organ and microenvironment specificity of Tregs.

3.2. Cytotoxic T Lymphocyte-Associated Protein 4 (CTLA4 or CD152)

Considering a connection of the suppressive effect conferred by Tregs and their CTLA4 expression [134,135], mechanistic insights into the regulation of CTLA4 expression are important. CTLA4 is a co-inhibitor that is generally upregulated upon antigen stimulation via the TCR to prevent an uncontrolled immune response [136,137], allowing for the fine tuning or consequently shutdown of the immune response as an immune checkpoint [138]. This is achieved by its higher affinity to the co-activators CD80/86 (B7-1/-2) expressed on antigen-presenting cells such as macrophages (MΦ) and dendritic cells (DC) compared to the co-activator CD28. Besides this role, CTLA4 is constitutively expressed on Tregs [139,140], contributing to the immunosuppressive phenotype of these cells [141]. Moreover, it has been shown that the transgenic expression of CTLA4 is one prerequisite of converting a conventional to a regulatory T cell [142]. When expressed on the T cell surface, CTLA4 binds to CD80/CD86 on antigen-presenting cells (APC) with a higher affinity than CD28, downregulating CD80/86 on DC to decrease the potency of APC to activate T cells [135]. From this data, it is obvious to assume that altering CTLA4 expression, i.e., to enhance or downregulate its expression, will be an appropriate treatment regime in autoimmune diseases to enhance Treg-dependent tolerance induction, e.g., to prevent cardiac allograft rejection [143], or to inhibit this immune-suppressive reaction to enhance anti-tumor immunity, e.g., by using CTLA4-neutralizing antibodies [144]. The HDAC canonical pan-inhibitor SAHA (suberoylanilide hydroxamic acid, Vorinostat), inhibiting HDAC1-9 with similar potency, has been shown to enhance CTLA4 expression in Tregs [143]. This upregulation was even enhanced when tacrolimus was added to inhibit calcineurin in parallel. Additionally, SAHA promoted selectively effector T cell apoptosis, which is consequently associated with an increased Treg proportion. This combined setting might be a therapeutic concept in preventing allograft rejection.

3.3. HDACs and HDACi as a Starting Point for Altering Treg Function

Thus far, 18 HDAC enzymes have been described. Eleven are Zn2+-dependent (HDAC1-11) and seven need NAD+ (Sirt1-7) for their activity. Though there have already been several clinical trials using HDAC inhibitors (HDACi) for treatment in oncology, none have been initiated for the therapy of autoimmune diseases. Based on the use of HDACi to change the epigenetic structure of Treg-lineage-dependent genes, experiments using pan-HDACi have been performed. It has been shown that the differentiation of human CD25highFoxp3+ Tregs into IL-17 producing cells can be prevented by the HDACi trichostatin A (TSA) [145]. TSA inhibits, similarly to SAHA, HDAC1-9 without any preference [146]. Taking this unspecific inhibition into consideration, it is difficult to provide any data on the role of a single HDAC, apart from mouse knockout studies or HDACs, where specific inhibitors already are at hand. As shown in Table 2, Tregs express class I HDACs 1, 2, 3, and 8 [147], class IIa HDACs 5, 7, and 9 [148], class IIb HDACs 6 and 10 [149], unrelated class III SIRTs 1, 2, 3, and 4 [150], and class IV HDAC 11 [151].
According to the diverse roles of HDACs in Treg immunology (see Graphical Abstract), corresponding HDAC inhibitors might be used to reduce or enhance Treg cell number and function. Obviously, the expression of Foxp3 is an essential element in Treg differentiation. Its transcription is inhibited by HDAC10 [167]. HDAC10 deletion in mice has been shown to enhance Foxp3 stability and increase H2K4Me3-activating marks on the Foxp3 promoter and CNS2 region [167]. Moreover, SIRT2 [171] and SIRT4 [173] downregulate protein Foxp3 expression by a so far unknown mechanism. Blocking these three HDACs will likely increase Foxp3 expression and concomitantly start and enhance Treg differentiation. HDAC7 associates with NR4a and Foxp3, being involved in the Foxp3-dependent repression of target genes. Following protein kinase D-dependent phosphorylation, HDAC7 is exported from the nucleus, consequently allowing for gene expression [174]. HDAC3, 6, 9, 11, and SIRT1 have been established to deacetylate Foxp3, which target it for proteasomal degradation [155,158,160,175]. Moreover, HDAC9 inhibits the expression of PPARgamma coactivator 1 alpha (PGC1α), an important factor in inducing proteins of the oxidative phosphorylation (OXPHOS)-system, important for the mitochondrial-dependent energy supply of the cells [172]. Finally, HDAC1 has been attributed to block the activity of the transcription factor RUNX, mandatory to maintain CD4+ T cell integrity [152,176,177].
In various models, the role of HDAC inhibition or deletion has been determined. Briefly, the inhibition of class I HDACs in models of cardiac allograft transplantation (CAT) or colitis has shown an enhanced Treg function following HDAC2 deletion, thus preventing HDAC2 association with Foxp3 [153,154], whereas the blockage of HDAC1, 3, and 8 has been shown to provoke an attenuated Treg number and function by destabilizing Foxp3 [152,153,155,156]. Blocking class IIa HDACs attenuates Treg function following HDAC5 and 7 inhibition in CAT and positive and negative selection in the thymus [157,158,159], whereas blocked HDAC9 enhances Treg function by Foxp3 stabilization in a colitis model [132,160,161]. Interestingly, class IIb HDACs are only involved Foxp3 destabilization, as shown in cystic fibrosis, collagen-induced arthritis, juvenile idiopathic arthritis, and lupus prone mice, as well as in cardiac allograft transplantation and colitis. Thus, their inhibition enhances and restores Treg-dependent effects [160,162,163,164,165,166,167,168,169]. Members of the sirtuin-family of HDACs (class III HDACs) are important regulators of the inflammatory stress response in immune and non-immune cells linking inflammation and metabolism [178,179]. Therefore, their role in Treg cell differentiation is mainly characterized by a Foxp3 destabilizing effect in murine sepsis (cecal-ligation and puncture), heterotrophic cardiac and orthotropic renal allograft transplantation, colitis [55,87,126,160,169,170,171,180], and by preventing Foxp3 expression in a mouse model of transient middle cerebral artery occlusion [173]. In contrast, SIRT3 is important for the metabolic adaption of Tregs, which makes it necessary for their function. Thus, SIRT3 inhibition is associated with a reduced number of Tregs in cardiac allograft transplantation [172]. Lastly, intervening with the function of HDAC11, the only class IV HDAC, has been shown to result in Foxp3 stabilization, enhancing Treg function in cardiac allograft transplantation [158].

4. Concluding Remarks

Considering these various effects of HDACs related to the epigenetic regulation of genes that are important for Treg differentiation and maintenance, the development of a therapy setting including HDAC-specific inhibitors is a promising task.
Based on the already established methods to generate and expand polyclonal, antigen-specific, or engineered Tregs ex vivo for adoptive cell therapy (for review see [52]), these can be treated with specific HDAC inhibitors to enhance Foxp3 expression, which will consequently induce and maintain an immunosuppressive Treg phenotype. After these Tregs have been infused back, the limiting factor is the half-life of transferred Tregs. This is especially important in patients with autoimmune diseases, where a permanent Treg-based immunosuppression is required. This holds true as well for patients following solid organ transplantation. Bone marrow transfer includes the risk of graft-versus-host disease, which exists temporarily and does not demand a very long Treg life. However, in sepsis patients, the situation is completely different. Because here Tregs are mainly deleterious and contribute to an immunosuppressive state that is linked to an inappropriate immune response that finally causes a fatal outcome, the number of these cells should be reduced or their immunosuppressive phenotype should be immediately mitigated. This possibly can be achieved by HDAC inhibition, provoking Foxp3 destabilization or reduced expression. However, it should be taken into account, that, if the HDAC inhibitor is applied in an unspecific formulation to the sepsis patient, it will operate in all cells that express the corresponding HDAC. Therefore, putative side effects have to be carefully proven before. Moreover, a Treg cell-specific HDAC-inhibitor formulation might be a chance to circumvent these side effect studies.
As discussed for autoimmune diseases, transplant rejection and sepsis, adequately altering the generation and number of Tregs, i.e., increasing or decreasing their count, is associated with an improved outcome. To achieve this successfully, clinical trials are mandatory in the near future to clarify the role of epigenetics, especially during sepsis initiation and progression. Moreover, the development of HDAC-specific inhibitors is important to allow for the fine tuning of chromatin histone acetylation. Considering the expression and activity of the transcription factor Foxp3 as the main mediator for Treg cell differentiation and function, its epigenetic modulation might be an appropriate target to reduce or enhance Treg function according to the disease state.

Author Contributions

This work was conceptualized and written by A.v.K., U.H., A.W., K.Z., and B.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by a grant from the Deutsche Forschungsgemeinschaft (KN493/13-1 and SFB815 TP3, TP8). The work was supported by the Else Kröner-Fresenius Foundation (EKFS), Research Training Group Translational Research Innovation-Pharma (TRIP) and the Landesoffensive zur Entwicklung wissenschaftlich-ökonomischer Exzellenz (LOEWE), Schwerpunkt Anwendungsorientierte Arzneimittel-forschung.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Ødiameter
Acacetylation
ALIacute lung injury
ALSamyolotrophic lateral sclerosis
AP1activator protein 1
APCantigen presenting cell
ASankylosis spondylitis
Ccolitis
CATcardiac allograft transplantation
CDcluster of differentiation
CFcystic fibrosis
ChIPchromatin immunoprecipitation
CIAcollagen-induced arthritis
CLPcecal ligation and puncture
CNSconserved non-coding region
CREBcAMP-responsive element binding protein
CTLA4cytotoxic T lymphocyte associated protein 4
ETSE26-AMV virus oncogene cellular homologue
ffemale
Foxp3Forkhead-box-protein P3
ggauge
Hhistone
HAThistone acetyltransferase
HDAChistone deacetylase
HIFhypoxia-inducible factor
HMGB1high-mobility-group-protein B1
HP1heterochromatin protein-1
iin vitro
IBDinflammatory bowel disease
ICOSinducible T-cell costimulatory
Ikzf2ICAROS family zinc finger 2
IPEXimmune dysregulation polyendocrinopathy and enteropathy
Klysine
LPSlipopolysaccharide
mmale
Memethylation
MHCmajor histocompatibility complex
MLNmesenteric lymph nodes
MSmultiple sclerosis
NFATnuclear factor of activated T cells
NRa4nuclear receptor 4a
Nt5eecto-5’-nucleotidase
OXPHOSoxidative phosphorylation
pperiphery
PCperitoneal cavity
PGC1αPPARgamma coactivator alpha
PIASprotein inhibitor of activated STAT
PSOpsoriasis
PTENphosphatase and tensin homologue
RArheumatoid arthritis
Pde3bphosphodiesterase 3b
RARretinoid acid receptor
RUNXRunt-related transcription factor 1
RXRretinoid X receptor
SAHAsuberoylanilide hydroxamic acid
Satb1 special AT-rich sequence binding protein
SIRTsilent information regulator
Smadsmall mothers against decapentaplegic
SPsingle positive
SScsystemic sclerosis
STAT signal transducer and activator of transcription
sumosmall ubiquitin-like modifier
tthymus
T1Dtype 1 diabetes
TCRT cell receptor
TIP60HIV-Tat-interactive protein
TGF-βtransforming growth factor-beta
TLRtoll-like receptor
Tregregulatory T cell
TSDRTreg-cell specific region

References

  1. 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] [PubMed]
  2. Abbas, A.K.; Benoist, C.; Bluestone, J.A.; Campbell, D.J.; Ghosh, S.; Hori, S.; Jiang, S.; Kuchroo, V.K.; Mathis, D.; Roncarolo, M.G.; et al. Regulatory T cells: Recommendations to simplify the nomenclature. Nat. Immunol. 2013, 14, 307–308. [Google Scholar] [CrossRef] [PubMed]
  3. Yang, J.; Zou, M.; Pezoldt, J.; Zhou, X.; Huehn, J. Thymus-derived Foxp3+ regulatory T cells upregulate RORγt expression under inflammatory conditions. J. Mol. Med. 2018, 96, 1387–1394. [Google Scholar] [CrossRef] [PubMed]
  4. Sakaguchi, S.; Ono, M.; Setoguchi, R.; Yagi, H.; Hori, S.; Fehervari, Z.; Shimizu, J.; Takahashi, T.; Nomura, T. Foxp3+ CD25+ CD4+ natural regulatory T cells in dominant self-tolerance and autoimmune disease. Immunol. Rev. 2006, 212, 8–27. [Google Scholar] [CrossRef] [PubMed]
  5. Palmer, E.; Naeher, D. Affinity threshold for thymic selection through a T-cell receptor-co-receptor zipper. Nat. Rev. Immunol. 2009, 9, 207–213. [Google Scholar] [CrossRef]
  6. Koehli, S.; Naeher, D.; Galati-Fournier, V.; Zehn, D.; Palmer, E. Optimal T-cell receptor affinity for inducing autoimmunity. Proc. Natl. Acad. Sci. USA 2014, 111, 17248–17253. [Google Scholar] [CrossRef] [Green Version]
  7. Zehn, D.; Bevan, M.J. T cells with low avidity for a tissue-restricted antigen routinely evade central and peripheral tolerance and cause autoimmunity. Immunity 2006, 25, 261–270. [Google Scholar] [CrossRef] [Green Version]
  8. Nishizuka, Y.; Sakakura, T. Thymus and reproduction: Sex-linked dysgenesia of the gonad after neonatal thymectomy in mice. Science 1969, 166, 753–755. [Google Scholar] [CrossRef]
  9. Asano, M.; Toda, M.; Sakaguchi, N.; Sakaguchi, S. Autoimmune disease as a consequence of developmental abnormality of a T cell subpopulation. J. Exp. Med. 1996, 184, 387–396. [Google Scholar] [CrossRef]
  10. Fontenot, J.D.; Dooley, J.L.; Farr, A.G.; Rudensky, A.Y. Developmental regulation of Foxp3 expression during ontogeny. J. Exp. Med. 2005, 202, 901–906. [Google Scholar] [CrossRef] [Green Version]
  11. Surh, C.D.; Sprent, J. T-cell apoptosis detected in situ during positive and negative selection in the thymus. Nature 1994, 372, 100–103. [Google Scholar] [CrossRef] [PubMed]
  12. Konkel, J.E.; Jin, W.; Abbatiello, B.; Grainger, J.R.; Chen, W. Thymocyte apoptosis drives the intrathymic generation of regulatory T cells. Proc. Natl. Acad. Sci. USA 2014, 111, E465–E473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Marie, J.C.; Letterio, J.J.; Gavin, M.; Rudensky, A.Y. TGF-beta1 maintains suppressor function and Foxp3 expression in CD4+CD25+ regulatory T cells. J. Exp. Med. 2005, 201, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
  14. 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] [PubMed]
  15. Ramoji, A.; Ryabchykov, O.; Galler, K.; Tannert, A.; Markwart, R.; Requardt, R.P.; Rubio, I.; Bauer, M.; Bocklitz, T.; Popp, J.; et al. Raman Spectroscopy Follows Time-Dependent Changes in T Lymphocytes Isolated from Spleen of Endotoxemic Mice. Immunohorizons 2019, 3, 45–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Apostolou, I.; Sarukhan, A.; Klein, L.; von Boehmer, H. Origin of regulatory T cells with known specificity for antigen. Nat. Immunol. 2002, 3, 756–763. [Google Scholar] [CrossRef] [PubMed]
  17. Jordan, M.S.; Boesteanu, A.; Reed, A.J.; Petrone, A.L.; Holenbeck, A.E.; Lerman, M.A.; Naji, A.; Caton, A.J. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat. Immunol. 2001, 2, 301–306. [Google Scholar] [CrossRef]
  18. Zheng, Y.; Josefowicz, S.; Chaudhry, A.; Peng, X.P.; Forbush, K.; Rudensky, A.Y. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 2010, 463, 808–812. [Google Scholar] [CrossRef] [Green Version]
  19. Bhairavabhotla, R.; Kim, Y.C.; Glass, D.D.; Escobar, T.M.; Patel, M.C.; Zahr, R.; Nguyen, C.K.; Kilaru, G.K.; Muljo, S.A.; Shevach, E.M. Transcriptome profiling of human FoxP3+ regulatory T cells. Hum. Immunol. 2016, 77, 201–213. [Google Scholar] [CrossRef] [Green Version]
  20. Zheng, Y.; Josefowicz, S.Z.; Kas, A.; Chu, T.-T.; Gavin, M.A.; Rudensky, A.Y. Genome-wide analysis of Foxp3 target genes in developing and mature regulatory T cells. Nature 2007, 445, 936–940. [Google Scholar] [CrossRef]
  21. Anandagoda, N.; Willis, J.C.; Hertweck, A.; Roberts, L.B.; Jackson, I.; Gökmen, M.R.; Jenner, R.G.; Howard, J.K.; Lord, G.M. microRNA-142-mediated repression of phosphodiesterase 3B critically regulates peripheral immune tolerance. J. Clin. Investig. 2019, 129, 1257–1271. [Google Scholar] [CrossRef] [PubMed]
  22. Getnet, D.; Grosso, J.F.; Goldberg, M.V.; Harris, T.J.; Yen, H.-R.; Bruno, T.C.; Durham, N.M.; Hipkiss, E.L.; Pyle, K.J.; Wada, S.; et al. A role for the transcription factor Helios in human CD4+CD25+ regulatory T cells. Mol. Immunol. 2010, 47, 1595–1600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Pfoertner, S.; Jeron, A.; Probst-Kepper, M.; Guzman, C.A.; Hansen, W.; Westendorf, A.M.; Toepfer, T.; Schrader, A.J.; Franzke, A.; Buer, J.; et al. Signatures of human regulatory T cells: An encounter with old friends and new players. Genome Biol. 2006, 7, R54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ono, M.; Yaguchi, H.; Ohkura, N.; Kitabayashi, I.; Nagamura, Y.; Nomura, T.; Miyachi, Y.; Tsukada, T.; Sakaguchi, S. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature 2007, 446, 685–689. [Google Scholar] [CrossRef] [PubMed]
  25. D’Hennezel, E.; Ben-Shoshan, M.; Ochs, H.D.; Torgerson, T.R.; Russell, L.J.; Lejtenyi, C.; Noya, F.J.; Jabado, N.; Mazer, B.; Piccirillo, C.A. FOXP3 Forkhead Domain Mutation and Regulatory T Cells in the IPEX Syndrome. N. Engl. J. Med. 2009, 361, 1710–1713. [Google Scholar] [CrossRef]
  26. Lahl, K.; Loddenkemper, C.; Drouin, C.; Freyer, J.; Arnason, J.; Eberl, G.; Hamann, A.; Wagner, H.; Huehn, J.; Sparwasser, T. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J. Exp. Med. 2007, 204, 57–63. [Google Scholar] [CrossRef] [Green Version]
  27. Kim, J.M.; Rasmussen, J.P.; Rudensky, A.Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 2007, 8, 191–197. [Google Scholar] [CrossRef]
  28. Hoffmann, P.; Eder, R.; Kunz-Schughart, L.A.; Andreesen, R.; Edinger, M. Large-scale in vitro expansion of polyclonal human CD4+CD25high regulatory T cells. Blood 2004, 104, 895–903. [Google Scholar] [CrossRef]
  29. Koenecke, C.; Czeloth, N.; Bubke, A.; Schmitz, S.; Kissenpfennig, A.; Malissen, B.; Huehn, J.; Ganser, A.; Förster, R.; Prinz, I. Alloantigen-specific de novo-induced Foxp3+ Treg revert in vivo and do not protect from experimental GVHD. Eur. J. Immunol. 2009, 39, 3091–3096. [Google Scholar] [CrossRef]
  30. Roquilly, A.; McWilliam, H.E.G.; Jacqueline, C.; Tian, Z.; Cinotti, R.; Rimbert, M.; Wakim, L.; Caminschi, I.; Lahoud, M.H.; Belz, G.T.; et al. Local Modulation of Antigen-Presenting Cell Development after Resolution of Pneumonia Induces Long-Term Susceptibility to Secondary Infections. Immunity 2017, 47, 135–147. [Google Scholar] [CrossRef] [Green Version]
  31. Gupta, D.L.; Bhoi, S.; Mohan, T.; Galwnkar, S.; Rao, D.N. Coexistence of Th1/Th2 and Th17/Treg imbalances in patients with post traumatic sepsis. Cytokine 2016, 88, 214–221. [Google Scholar] [CrossRef]
  32. Carvelli, J.; Piperoglou, C.; Bourenne, J.; Farnarier, C.; Banzet, N.; Demerlé, C.; Gainnier, M.; Vély, F. Imbalance of Circulating Innate Lymphoid Cell Subpopulations in Patients With Septic Shock. Front. Immunol. 2019, 10, 2179. [Google Scholar] [CrossRef] [PubMed]
  33. Venet, F.; Chung, C.-S.; Kherouf, H.; Geeraert, A.; Malcus, C.; Poitevin, F.; Bohé, J.; Lepape, A.; Ayala, A.; Monneret, G. Increased circulating regulatory T cells (CD4+CD25+CD127) contribute to lymphocyte anergy in septic shock patients. Intensive Care Med. 2009, 35, 678–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nascimento, D.C.; Melo, P.H.; Piñeros, A.R.; Ferreira, R.G.; Colón, D.F.; Donate, P.B.; Castanheira, F.V.; Gozzi, A.; Czaikoski, P.G.; Niedbala, W.; et al. IL-33 contributes to sepsis-induced long-term immunosuppression by expanding the regulatory T cell population. Nat. Commun. 2017, 8, 14919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mizui, M.; Tsokos, G.C. Targeting Regulatory T Cells to Treat Patients With Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 786. [Google Scholar] [CrossRef]
  36. Visperas, A.; Vignali, D.A.A. Are Regulatory T Cells Defective in Type 1 Diabetes and Can We Fix Them? J. Immunol. 2016, 197, 3762–3770. [Google Scholar] [CrossRef] [Green Version]
  37. Sabat, R.; Wolk, K.; Loyal, L.; Döcke, W.D.; Ghoreschi, K. T cell pathology in skin inflammation. Semin. Immunopathol. 2019, 41, 359–377. [Google Scholar] [CrossRef] [Green Version]
  38. Esensten, J.H.; Muller, Y.D.; Bluestone, J.A.; Tang, Q. Regulatory T-cell therapy for autoimmune and autoinflammatory diseases: The next frontier. J. Allergy Clin. Immunol. 2018, 142, 1710–1718. [Google Scholar] [CrossRef] [Green Version]
  39. Ferreira, L.M.R.; Muller, Y.D.; Bluestone, J.A.; Tang, Q. Next-generation regulatory T cell therapy. Nat. Rev. Drug Discov. 2019, 18, 749–769. [Google Scholar] [CrossRef] [Green Version]
  40. Marek-Trzonkowska, N.; Myśliwiec, M.; Dobyszuk, A.; Grabowska, M.; Derkowska, I.; Juścińska, J.; Owczuk, R.; Szadkowska, A.; Witkowski, P.; Młynarski, W.; et al. Therapy of type 1 diabetes with CD4+CD25highCD127-regulatory T cells prolongs survival of pancreatic islets—Results of one year follow-up. Clin. Immunol. 2014, 153, 23–30. [Google Scholar] [CrossRef]
  41. 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] [PubMed] [Green Version]
  42. Strauss, L.; Czystowska, M.; Szajnik, M.; Mandapathil, M.; Whiteside, T.L. Differential responses of human regulatory T cells (Treg) and effector T cells to rapamycin. PLoS ONE 2009, 4, e5994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Fernandez, D.; Bonilla, E.; Mirza, N.; Niland, B.; Perl, A. Rapamycin reduces disease activity and normalizes T cell activation-induced calcium fluxing in patients with systemic lupus erythematosus. Arthritis Rheum. 2006, 54, 2983–2988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Liu, G.; Yang, K.; Burns, S.; Shrestha, S.; Chi, H. The S1P1-mTOR axis directs the reciprocal differentiation of TH1 and Treg cells. Nat. Immunol. 2010, 11, 1047–1056. [Google Scholar] [CrossRef]
  45. Lai, Z.-W.; Hanczko, R.; Bonilla, E.; Caza, T.N.; Clair, B.; Bartos, A.; Miklossy, G.; Jimah, J.; Doherty, E.; Tily, H.; et al. N-acetylcysteine reduces disease activity by blocking mammalian target of rapamycin in T cells from systemic lupus erythematosus patients: A randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2012, 64, 2937–2946. [Google Scholar] [CrossRef] [Green Version]
  46. Yin, Y.; Choi, S.-C.; Xu, Z.; Perry, D.J.; Seay, H.; Croker, B.P.; Sobel, E.S.; Brusko, T.M.; Morel, L. Normalization of CD4+ T cell metabolism reverses lupus. Sci. Transl. Med. 2015, 7, 274ra18. [Google Scholar] [CrossRef] [Green Version]
  47. Koga, T.; Hedrich, C.M.; Mizui, M.; Yoshida, N.; Otomo, K.; Lieberman, L.A.; Rauen, T.; Crispín, J.C.; Tsokos, G.C. CaMK4-dependent activation of AKT/mTOR and CREM-α underlies autoimmunity-associated Th17 imbalance. J. Clin. Investig. 2014, 124, 2234–2245. [Google Scholar] [CrossRef] [Green Version]
  48. He, J.; Zhang, X.; Wei, Y.; Sun, X.; Chen, Y.; Deng, J.; Jin, Y.; Gan, Y.; Hu, X.; Jia, R.; et al. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat. Med. 2016, 22, 991–993. [Google Scholar] [CrossRef]
  49. Humrich, J.Y.; Spee-Mayer, C.; von Siegert, E.; Alexander, T.; Hiepe, F.; Radbruch, A.; Burmester, G.R.; Riemekasten, G. Rapid induction of clinical remission by low-dose interleukin-2 in a patient with refractory SLE. Ann. Rheum. Dis. 2015, 74, 791–792. [Google Scholar] [CrossRef]
  50. Spee-Mayer, C.; von Siegert, E.; Abdirama, D.; Rose, A.; Klaus, A.; Alexander, T.; Enghard, P.; Sawitzki, B.; Hiepe, F.; Radbruch, A.; et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann. Rheum. Dis. 2016, 75, 1407–1415. [Google Scholar] [CrossRef]
  51. Hartemann, A.; Bensimon, G.; Payan, C.A.; Jacqueminet, S.; Bourron, O.; Nicolas, N.; Fonfrede, M.; Rosenzwajg, M.; Bernard, C.; Klatzmann, D. Low-dose interleukin 2 in patients with type 1 diabetes: A phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol. 2013, 1, 295–305. [Google Scholar] [CrossRef]
  52. Raffin, C.; Vo, L.T.; Bluestone, J.A. Treg cell-based therapies: Challenges and perspectives. Nat. Rev. Immunol. 2020, 20, 158–172. [Google Scholar] [CrossRef] [PubMed]
  53. Tang, Q.; Vincenti, F. Transplant trials with Tregs: Perils and promises. J. Clin. Investig. 2017, 127, 2505–2512. [Google Scholar] [CrossRef] [PubMed]
  54. Atif, M.; Conti, F.; Gorochov, G.; Oo, Y.H.; Miyara, M. Regulatory T cells in solid organ transplantation. Clin. Transl. Immunol. 2020, 9, e01099. [Google Scholar] [CrossRef] [PubMed]
  55. Todo, S.; Yamashita, K.; Goto, R.; Zaitsu, M.; Nagatsu, A.; Oura, T.; Watanabe, M.; Aoyagi, T.; Suzuki, T.; Shimamura, T.; et al. A pilot study of operational tolerance with a regulatory T-cell-based cell therapy in living donor liver transplantation. Hepatology 2016, 64, 632–643. [Google Scholar] [CrossRef] [Green Version]
  56. Chandran, S.; Tang, Q.; Sarwal, M.; Laszik, Z.G.; Putnam, A.L.; Lee, K.; Leung, J.; Nguyen, V.; Sigdel, T.; Tavares, E.C.; et al. Polyclonal Regulatory T Cell Therapy for Control of Inflammation in Kidney Transplants. Am. J. Transplant. 2017, 17, 2945–2954. [Google Scholar] [CrossRef]
  57. Mathew, J.M.; H-Voss, J.; LeFever, A.; Konieczna, I.; Stratton, C.; He, J.; Huang, X.; Gallon, L.; Skaro, A.; Ansari, M.J.; et al. A Phase I Clinical Trial with Ex Vivo Expanded Recipient Regulatory T cells in Living Donor Kidney Transplants. Sci. Rep. 2018, 8, 7428. [Google Scholar] [CrossRef] [Green Version]
  58. Cohen, J.L.; Trenado, A.; Vasey, D.; Klatzmann, D.; Salomon, B.L. CD4+CD25+ immunoregulatory T Cells: New therapeutics for graft-versus-host disease. J. Exp. Med. 2002, 196, 401–406. [Google Scholar] [CrossRef]
  59. Jones, S.C.; Murphy, G.F.; Korngold, R. Post-hematopoietic cell transplantation control of graft-versus-host disease by donor CD425 T cells to allow an effective graft-versus-leukemia response. Biol. Blood Marrow Transplant. 2003, 9, 243–256. [Google Scholar] [CrossRef] [Green Version]
  60. Taylor, P.A.; Lees, C.J.; Blazar, B.R. The infusion of ex vivo activated and expanded CD4+CD25+ immune regulatory cells inhibits graft-versus-host disease lethality. Blood 2002, 99, 3493–3499. [Google Scholar] [CrossRef]
  61. Mancusi, A.; Piccinelli, S.; Velardi, A.; Pierini, A. CD4+FOXP3+ Regulatory T Cell Therapies in HLA Haploidentical Hematopoietic Transplantation. Front. Immunol. 2019, 10, 2901. [Google Scholar] [CrossRef] [PubMed]
  62. Boomer, J.S.; To, K.; Chang, K.C.; Takasu, O.; Osborne, D.F.; Walton, A.H.; Bricker, T.L.; Jarman, S.D.; Kreisel, D.; Krupnick, A.S.; et al. Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 2011, 306, 2594–2605. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, M.; Fang, H.; Du, M.; Li, C.; Tang, R.; Liu, H.; Gao, Z.; Ji, Z.; Ke, B.; Chen, X.-L. The Modulation of Regulatory T Cells via HMGB1/PTEN/β-Catenin Axis in LPS Induced Acute Lung Injury. Front. Immunol. 2019, 10, 1612. [Google Scholar] [CrossRef] [PubMed]
  64. Ruiz, S.; Vardon-Bounes, F.; Merlet-Dupuy, V.; Conil, J.M.; Buléon, M.; Fourcade, O.; Tack, I.; Minville, V. Sepsis modeling in mice: Ligation length is a major severity factor in cecal ligation and puncture. Intensive Care Med. Exp. 2016, 4, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Song, T.; Yin, H.; Chen, J.; Huang, L.; Jiang, J.; He, T.; Huang, H.; Hu, X. Survival advantage depends on cecal volume rather than cecal length in a mouse model of cecal ligation and puncture. J. Surg. Res. 2016, 203, 476–482. [Google Scholar] [CrossRef] [PubMed]
  66. Knethen, A.; von Schäfer, A.; Kuchler, L.; Knape, T.; Christen, U.; Hintermann, E.; Fißlthaler, B.; Schröder, K.; Brandes, R.P.; Genz, B.; et al. Tolerizing CTL by Sustained Hepatic PD-L1 Expression Provides a New Therapy Approach in Mouse Sepsis. Theranostics 2019, 9, 2003–2016. [Google Scholar] [CrossRef]
  67. Gao, M.; Ou, H.; Jiang, Y.; Wang, K.; Peng, Y.; Zhang, H.; Yang, M.; Xiao, X. Tanshinone IIA attenuates sepsis-induced immunosuppression and improves survival rate in a mice peritonitis model. Biomed. Pharmacother. 2019, 112, 108609. [Google Scholar] [CrossRef]
  68. Gao, Y.L.; Chai, Y.F.; Qi, A.L.; Yao, Y.; Liu, Y.C.; Dong, N.; Wang, L.J.; Yao, Y.M. Neuropilin-1highCD4⁺CD25⁺ Regulatory T Cells Exhibit Primary Negative Immunoregulation in Sepsis. Mediat. Inflamm. 2016, 2016, 7132158. [Google Scholar] [CrossRef] [Green Version]
  69. Jeremias, I.C.; Victorino, V.J.; Barbeiro, H.V.; Kubo, S.A.; Prado, C.M.; Lima, T.M.; Soriano, F.G. The Role of Acetylcholine in the Inflammatory Response in Animals Surviving Sepsis Induced by Cecal Ligation and Puncture. Mol. Neurobiol. 2016, 53, 6635–6643. [Google Scholar] [CrossRef]
  70. Luan, Y.; Yin, C.; Qin, Q.; Dong, N.; Zhu, X.; Sheng, Z.; Zhang, Q.; Yao, Y. Effect of Regulatory T Cells on Promoting Apoptosis of T Lymphocyte and Its Regulatory Mechanism in Sepsis. J. Interferon Cytokine Res. 2015, 35, 969–980. [Google Scholar] [CrossRef] [Green Version]
  71. Zheng, Y.; Wu, Z.; Ni, H.; Ke, L.; Tong, Z.; Li, W.; Li, N.; Li, J. Codonopsis pilosula polysaccharide attenuates cecal ligation and puncture sepsis via circuiting regulatory T cells in mice. Shock 2014, 41, 250–255. [Google Scholar] [CrossRef] [PubMed]
  72. Andrade, M.M.C.; Ariga, S.S.K.; Barbeiro, D.F.; Barbeiro, H.V.; Pimentel, R.N.; Petroni, R.C.; Soriano, F.G. Endotoxin tolerance modulates TREG and TH17 lymphocytes protecting septic mice. Oncotarget 2019, 10, 3451–3461. [Google Scholar] [CrossRef] [PubMed]
  73. Hu, Z.Q.; Yao, Y.; Chen, W.; Bian, J.; Zhao, L.; Chen, L.; Hong, G.; Lu, Z.; Zhao, G. Partial Depletion of Regulatory T Cells Enhances Host Inflammatory Response Against Acute Pseudomonas aeruginosa Infection After Sepsis. Inflammation 2018, 41, 1780–1790. [Google Scholar] [CrossRef] [PubMed]
  74. Yoon, S.J.; Kim, S.J.; Lee, S.M. Overexpression of HO-1 Contributes to Sepsis-Induced Immunosuppression by Modulating the Th1/Th2 Balance and Regulatory T-Cell Function. J. Infect. Dis. 2017, 215, 1608–1618. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, Y.; Kong, B.B.; Yang, W.P.; Zhao, X.; Zhang, R. Immunomodulatory intervention with Gamma interferon in mice with sepsis. Life Sci. 2017, 185, 85–94. [Google Scholar] [CrossRef]
  76. Restagno, D.; Venet, F.; Paquet, C.; Freyburger, L.; Allaouchiche, B.; Monneret, G.; Bonnet, J.M.; Louzier, V. Mice Survival and Plasmatic Cytokine Secretion in a “Two Hit” Model of Sepsis Depend on Intratracheal Pseudomonas Aeruginosa Bacterial Load. PLoS ONE 2016, 11, e0162109. [Google Scholar] [CrossRef]
  77. Sharma, A.; Yang, W.L.; Matsuo, S.; Wang, P. Differential alterations of tissue T-cell subsets after sepsis. Immunol. Lett. 2015, 168, 41–50. [Google Scholar] [CrossRef] [Green Version]
  78. Molinaro, R.; Pecli, C.; Guilherme, R.F.; Alves-Filho, J.C.; Cunha, F.Q.; Canetti, C.; Kunkel, S.L.; Bozza, M.T.; Benjamim, C.F. CCR4 Controls the Suppressive Effects of Regulatory T Cells on Early and Late Events during Severe Sepsis. PLoS ONE 2015, 10, e0133227. [Google Scholar] [CrossRef] [Green Version]
  79. Wang, H.W.; Yang, W.; Gao, L.; Kang, J.R.; Qin, J.J.; Liu, Y.P.; Lu, J.Y. Adoptive transfer of bone marrow-derived dendritic cells decreases inhibitory and regulatory T-cell differentiation and improves survival in murine polymicrobial sepsis. Immunology 2015, 145, 50–59. [Google Scholar] [CrossRef]
  80. Hasan, Z.; Palani, K.; Zhang, S.; Lepsenyi, M.; Hwaiz, R.; Rahman, M.; Syk, I.; Jeppsson, B.; Thorlacius, H. Rho kinase regulates induction of T-cell immune dysfunction in abdominal sepsis. Infect. Immun. 2013, 81, 2499–2506. [Google Scholar] [CrossRef] [Green Version]
  81. Zhang, S.; Luo, L.; Wang, Y.; Rahman, M.; Lepsenyi, M.; Syk, I.; Jeppsson, B.; Thorlacius, H. Simvastatin protects against T cell immune dysfunction in abdominal sepsis. Shock 2012, 38, 524–531. [Google Scholar] [CrossRef] [PubMed]
  82. Zhu, J.; Wang, J.; Sheng, Y.; Zou, Y.; Bo, L.; Wang, F.; Lou, J.; Fan, X.; Bao, R.; Wu, Y.; et al. Baicalin improves survival in a murine model of polymicrobial sepsis via suppressing inflammatory response and lymphocyte apoptosis. PLoS ONE 2012, 7, e35523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Hiraki, S.; Ono, S.; Tsujimoto, H.; Kinoshita, M.; Takahata, R.; Miyazaki, H.; Saitoh, D.; Hase, K. Neutralization of interleukin-10 or transforming growth factor-β decreases the percentages of CD4+ CD25+ Foxp3+ regulatory T cells in septic mice, thereby leading to an improved survival. Surgery 2012, 151, 313–322. [Google Scholar] [CrossRef] [PubMed]
  84. Yang, W.; Yamada, M.; Tamura, Y.; Chang, K.; Mao, J.; Zou, L.; Feng, Y.; Kida, K.; Scherrer-Crosbie, M.; Chao, W.; et al. Farnesyltransferase inhibitor FTI-277 reduces mortality of septic mice along with improved bacterial clearance. J. Pharmacol. Exp. Ther. 2011, 339, 832–841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chen, X.; Bäumel, M.; Männel, D.N.; Howard, O.M.Z.; Oppenheim, J.J. Interaction of TNF with TNF receptor type 2 promotes expansion and function of mouse CD4+CD25+ T regulatory cells. J. Immunol. 2007, 179, 154–161. [Google Scholar] [CrossRef] [Green Version]
  86. Wisnoski, N.; Chung, C.S.; Chen, Y.; Huang, X.; Ayala, A. The contribution of CD4+ CD25+ T-regulatory-cells to immune suppression in sepsis. Shock 2007, 27, 251–257. [Google Scholar] [CrossRef] [Green Version]
  87. Martin, A.N.; Alexander-Miller, M.; Yoza, B.K.; Vachharajani, V.; McCall, C.E. Sirtuin1 Targeting Reverses Innate and Adaptive Immune Tolerance in Septic Mice. J. Immunol. Res. 2018, 2018, 2402593. [Google Scholar] [CrossRef]
  88. Zhang, S.; Luo, L.; Wang, Y.; Gomez, M.F.; Thorlacius, H. Nuclear factor of activated T cells regulates neutrophil recruitment, systemic inflammation, and T-cell dysfunction in abdominal sepsis. Infect. Immun. 2014, 82, 3275–3288. [Google Scholar] [CrossRef] [Green Version]
  89. Shih, J.M.; Shih, Y.M.; Pai, M.H.; Hou, Y.C.; Yeh, C.L.; Yeh, S.L. Fish Oil-Based Fat Emulsion Reduces Acute Kidney Injury and Inflammatory Response in Antibiotic-Treated Polymicrobial Septic Mice. Nutrients 2016, 8, 165. [Google Scholar] [CrossRef] [Green Version]
  90. Kim, J.S.; Kim, S.J.; Lee, S.M. Genipin attenuates sepsis-induced immunosuppression through inhibition of T lymphocyte apoptosis. Int. Immunopharmacol. 2015, 27, 15–23. [Google Scholar] [CrossRef]
  91. Mohr, A.; Polz, J.; Martin, E.M.; Griessl, S.; Kammler, A.; Pötschke, C.; Lechner, A.; Bröker, B.M.; Mostböck, S.; Männel, D.N. Sepsis leads to a reduced antigen-specific primary antibody response. Eur. J. Immunol. 2012, 42, 341–352. [Google Scholar] [CrossRef] [PubMed]
  92. Zhou, M.; Yang, W.L.; Aziz, M.; Ma, G.; Wang, P. Therapeutic effect of human ghrelin and growth hormone: Attenuation of immunosuppression in septic aged rats. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2584–2593. [Google Scholar] [CrossRef] [PubMed]
  93. Chao, Y.H.; Wu, H.P.; Wu, K.H.; Tsai, Y.G.; Peng, C.T.; Lin, K.C.; Chao, W.R.; Lee, M.S.; Fu, Y.C. An increase in CD3+CD4+CD25+ regulatory T cells after administration of umbilical cord-derived mesenchymal stem cells during sepsis. PLoS ONE 2014, 9, e110338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yu, W.; Du, H.; Fu, Q.; Cui, N.; Du, C. The influence of Th1/Th2 and CD4+ regulatory t cells of mesenteric lymph nodes on systemic lipopolysaccharide. PJP 2014, 2, 125–129. [Google Scholar] [CrossRef]
  95. Chen, H.H.; Chang, C.L.; Lin, K.C.; Sung, P.H.; Chai, H.T.; Zhen, Y.Y.; Chen, Y.C.; Wu, Y.C.; Leu, S.; Tsai, T.H.; et al. Melatonin augments apoptotic adipose-derived mesenchymal stem cell treatment against sepsis-induced acute lung injury. Am. J. Transl. Res. 2014, 6, 439–458. [Google Scholar]
  96. Chen, H.H.; Lin, K.C.; Wallace, C.G.; Chen, Y.T.; Yang, C.C.; Leu, S.; Chen, Y.C.; Sun, C.K.; Tsai, T.H.; Chen, Y.L.; et al. Additional benefit of combined therapy with melatonin and apoptotic adipose-derived mesenchymal stem cell against sepsis-induced kidney injury. J. Pineal Res. 2014, 57, 16–32. [Google Scholar] [CrossRef]
  97. Chang, C.L.; Leu, S.; Sung, H.C.; Zhen, Y.Y.; Cho, C.L.; Chen, A.; Tsai, T.H.; Chung, S.Y.; Chai, H.T.; Sun, C.K.; et al. Impact of apoptotic adipose-derived mesenchymal stem cells on attenuating organ damage and reducing mortality in rat sepsis syndrome induced by cecal puncture and ligation. J. Transl. Med. 2012, 10, 244. [Google Scholar] [CrossRef] [Green Version]
  98. Dhamne, C.; Chung, Y.; Alousi, A.M.; Cooper, L.J.N.; Tran, D.Q. Peripheral and thymic foxp3+ regulatory T cells in search of origin, distinction, and function. Front. Immunol. 2013, 4, 253. [Google Scholar] [CrossRef] [Green Version]
  99. Deng, G.; Song, X.; Fujimoto, S.; Piccirillo, C.A.; Nagai, Y.; Greene, M.I. Foxp3 Post-translational Modifications and Treg Suppressive Activity. Front. Immunol. 2019, 10, 2486. [Google Scholar] [CrossRef] [Green Version]
  100. Brunkow, M.E.; Jeffery, E.W.; Hjerrild, K.A.; Paeper, B.; Clark, L.B.; Yasayko, S.A.; Wilkinson, J.E.; Galas, D.; Ziegler, S.F.; Ramsdell, F. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 2001, 27, 68–73. [Google Scholar] [CrossRef]
  101. Torgerson, T.R.; Ochs, H.D. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome: A model of immune dysregulation. Curr. Opin. Allergy Clin. Immunol. 2002, 2, 481–487. [Google Scholar] [CrossRef] [PubMed]
  102. Kitagawa, Y.; Ohkura, N.; Kidani, Y.; Vandenbon, A.; Hirota, K.; Kawakami, R.; Yasuda, K.; Motooka, D.; Nakamura, S.; Kondo, M.; et al. Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat. Immunol. 2017, 18, 173–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Alvarez, J.D.; Yasui, D.H.; Niida, H.; Joh, T.; Loh, D.Y.; Kohwi-Shigematsu, T. The MAR-binding protein SATB1 orchestrates temporal and spatial expression of multiple genes during T-cell development. Genes Dev. 2000, 14, 521–535. [Google Scholar] [PubMed]
  104. Chen, C.; Rowell, E.A.; Thomas, R.M.; Hancock, W.W.; Wells, A.D. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J. Biol. Chem. 2006, 281, 36828–36834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ogawa, C.; Tone, Y.; Tsuda, M.; Peter, C.; Waldmann, H.; Tone, M. TGF-β-mediated Foxp3 gene expression is cooperatively regulated by Stat5, Creb, and AP-1 through CNS2. J. Immunol. 2014, 192, 475–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Polansky, J.K.; Kretschmer, K.; Freyer, J.; Floess, S.; Garbe, A.; Baron, U.; Olek, S.; Hamann, A.; Boehmer, H.; von Huehn, J. DNA methylation controls Foxp3 gene expression. Eur. J. Immunol. 2008, 38, 1654–1663. [Google Scholar] [CrossRef]
  107. Li, X.; Liang, Y.; LeBlanc, M.; Benner, C.; Zheng, Y. Function of a Foxp3 cis-element in protecting regulatory T cell identity. Cell 2014, 158, 734–748. [Google Scholar] [CrossRef] [Green Version]
  108. Hori, S. c-Rel: A pioneer in directing regulatory T-cell lineage commitment? Eur. J. Immunol. 2010, 40, 664–667. [Google Scholar] [CrossRef]
  109. Huehn, J.; Beyer, M. Epigenetic and transcriptional control of Foxp3+ regulatory T cells. Semin. Immunol. 2015, 27, 10–18. [Google Scholar] [CrossRef]
  110. Liu, B.; Tahk, S.; Yee, K.M.; Fan, G.; Shuai, K. The ligase PIAS1 restricts natural regulatory T cell differentiation by epigenetic repression. Science 2010, 330, 521–525. [Google Scholar] [CrossRef] [Green Version]
  111. Mantel, P.-Y.; Ouaked, N.; Rückert, B.; Karagiannidis, C.; Welz, R.; Blaser, K.; Schmidt-Weber, C.B. Molecular mechanisms underlying FOXP3 induction in human T cells. J. Immunol. 2006, 176, 3593–3602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Mouly, E.; Chemin, K.; Nguyen, H.V.; Chopin, M.; Mesnard, L.; Leite-de-Moraes, M.; Burlen-defranoux, O.; Bandeira, A.; Bories, J.-C. The Ets-1 transcription factor controls the development and function of natural regulatory T cells. J. Exp. Med. 2010, 207, 2113–2125. [Google Scholar] [CrossRef] [Green Version]
  113. Polansky, J.K.; Schreiber, L.; Thelemann, C.; Ludwig, L.; Krüger, M.; Baumgrass, R.; Cording, S.; Floess, S.; Hamann, A.; Huehn, J. Methylation matters: Binding of Ets-1 to the demethylated Foxp3 gene contributes to the stabilization of Foxp3 expression in regulatory T cells. J. Mol. Med. 2010, 88, 1029–1040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Kerdiles, Y.M.; Stone, E.L.; Beisner, D.R.; Beisner, D.L.; McGargill, M.A.; Ch’en, I.L.; Stockmann, C.; Katayama, C.D.; Hedrick, S.M. Foxo transcription factors control regulatory T cell development and function. Immunity 2010, 33, 890–904. [Google Scholar] [CrossRef] [Green Version]
  115. Sekiya, T.; Kashiwagi, I.; Yoshida, R.; Fukaya, T.; Morita, R.; Kimura, A.; Ichinose, H.; Metzger, D.; Chambon, P.; Yoshimura, A. Nr4a receptors are essential for thymic regulatory T cell development and immune homeostasis. Nat. Immunol. 2013, 14, 230–237. [Google Scholar] [CrossRef] [PubMed]
  116. Takeuchi, H.; Yokota-Nakatsuma, A.; Ohoka, Y.; Kagechika, H.; Kato, C.; Song, S.-Y.; Iwata, M. Retinoid X receptor agonists modulate Foxp3⁺ regulatory T cell and Th17 cell differentiation with differential dependence on retinoic acid receptor activation. J. Immunol. 2013, 191, 3725–3733. [Google Scholar] [CrossRef] [Green Version]
  117. Xu, L.; Kitani, A.; Stuelten, C.; McGrady, G.; Fuss, I.; Strober, W. Positive and negative transcriptional regulation of the Foxp3 gene is mediated by access and binding of the Smad3 protein to enhancer I. Immunity 2010, 33, 313–325. [Google Scholar] [CrossRef] [Green Version]
  118. Bruno, L.; Mazzarella, L.; Hoogenkamp, M.; Hertweck, A.; Cobb, B.S.; Sauer, S.; Hadjur, S.; Leleu, M.; Naoe, Y.; Telfer, J.C.; et al. Runx proteins regulate Foxp3 expression. J. Exp. Med. 2009, 206, 2329–2337. [Google Scholar] [CrossRef]
  119. Zorn, E.; Nelson, E.A.; Mohseni, M.; Porcheray, F.; Kim, H.; Litsa, D.; Bellucci, R.; Raderschall, E.; Canning, C.; Soiffer, R.J.; et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006, 108, 1571–1579. [Google Scholar] [CrossRef] [Green Version]
  120. 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]
  121. Yang, X.O.; Nurieva, R.; Martinez, G.J.; Kang, H.S.; Chung, Y.; Pappu, B.P.; Shah, B.; Chang, S.H.; Schluns, K.S.; Watowich, S.S.; et al. Molecular antagonism and plasticity of regulatory and inflammatory T cell programs. Immunity 2008, 29, 44–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Song, X.; Li, B.; Xiao, Y.; Chen, C.; Wang, Q.; Liu, Y.; Berezov, A.; Xu, C.; Gao, Y.; Li, Z.; et al. Structural and biological features of FOXP3 dimerization relevant to regulatory T cell function. Cell Rep. 2012, 1, 665–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Xiao, Y.; Nagai, Y.; Deng, G.; Ohtani, T.; Zhu, Z.; Zhou, Z.; Zhang, H.; Ji, M.Q.; Lough, J.W.; Samanta, A.; et al. Dynamic interactions between TIP60 and p300 regulate FOXP3 function through a structural switch defined by a single lysine on TIP60. Cell Rep. 2014, 7, 1471–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Van Loosdregt, J.; Vercoulen, Y.; Guichelaar, T.; Gent, Y.Y.J.; Beekman, J.M.; van Beekum, O.; Brenkman, A.B.; Hijnen, D.-J.; Mutis, T.; Kalkhoven, E.; et al. Regulation of Treg functionality by acetylation-mediated Foxp3 protein stabilization. Blood 2010, 115, 965–974. [Google Scholar] [CrossRef]
  125. Kwon, H.-S.; Lim, H.W.; Wu, J.; Schnölzer, M.; Verdin, E.; Ott, M. Three novel acetylation sites in the Foxp3 transcription factor regulate the suppressive activity of regulatory T cells. J. Immunol. 2012, 188, 2712–2721. [Google Scholar] [CrossRef] [Green Version]
  126. Beier, U.H.; Wang, L.; Bhatti, T.R.; Liu, Y.; Han, R.; Ge, G.; Hancock, W.W. Sirtuin-1 targeting promotes Foxp3+ T-regulatory cell function and prolongs allograft survival. Mol. Cell. Biol. 2011, 31, 1022–1029. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, D.; Quiros, J.; Mahuron, K.; Pai, C.C.; Ranzani, V.; Young, A.; Silveria, S.; Harwin, T.; Abnousian, A.; Pagani, M.; et al. Targeting EZH2 Reprograms Intratumoral Regulatory T Cells to Enhance Cancer Immunity. Cell Rep. 2018, 23, 3262–3274. [Google Scholar] [CrossRef]
  128. DuPage, M.; Chopra, G.; Quiros, J.; Rosenthal, W.L.; Morar, M.M.; Holohan, D.; Zhang, R.; Turka, L.; Marson, A.; Bluestone, J.A. The chromatin-modifying enzyme Ezh2 is critical for the maintenance of regulatory T cell identity after activation. Immunity 2015, 42, 227–238. [Google Scholar] [CrossRef] [Green Version]
  129. Kwon, H.K.; Chen, H.M.; Mathis, D.; Benoist, C. Different molecular complexes that mediate transcriptional induction and repression by FoxP3. Nat. Immunol. 2017, 18, 1238–1248. [Google Scholar] [CrossRef] [Green Version]
  130. Li, Q.; Zou, J.; Wang, M.; Ding, X.; Chepelev, I.; Zhou, X.; Zhao, W.; Wei, G.; Cui, J.; Zhao, K.; et al. Critical role of histone demethylase Jmjd3 in the regulation of CD4+ T-cell differentiation. Nat. Commun. 2014, 5, 5780. [Google Scholar] [CrossRef] [Green Version]
  131. Mock, J.R.; Dial, C.F.; Tune, M.K.; Norton, D.L.; Martin, J.R.; Gomez, J.C.; Hagan, R.S.; Dang, H.; Doerschuk, C.M. Transcriptional analysis of Foxp3+ Tregs and functions of two identified molecules during resolution of ALI. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Tao, R.; Zoeten, E.F.; de Ozkaynak, E.; Chen, C.; Wang, L.; Porrett, P.M.; Li, B.; Turka, L.A.; Olson, E.N.; Greene, M.I.; et al. Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat. Med. 2007, 13, 1299–1307. [Google Scholar] [CrossRef] [PubMed]
  133. Samanta, A.; Li, B.; Song, X.; Bembas, K.; Zhang, G.; Katsumata, M.; Saouaf, S.J.; Wang, Q.; Hancock, W.W.; Shen, Y.; et al. TGF-beta and IL-6 signals modulate chromatin binding and promoter occupancy by acetylated FOXP3. Proc. Natl. Acad. Sci. USA 2008, 105, 14023–14027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Akimova, T.; Ge, G.; Golovina, T.; Mikheeva, T.; Wang, L.; Riley, J.L.; Hancock, W.W. Histone/protein deacetylase inhibitors increase suppressive functions of human FOXP3+ Tregs. Clin. Immunol. 2010, 136, 348–363. [Google Scholar] [CrossRef] [Green Version]
  135. Wing, K.; Onishi, Y.; Prieto-Martin, P.; Yamaguchi, T.; Miyara, M.; Fehervari, Z.; Nomura, T.; Sakaguchi, S. CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008, 322, 271–275. [Google Scholar] [CrossRef]
  136. Finn, P.W.; He, H.; Wang, Y.; Wang, Z.; Guan, G.; Listman, J.; Perkins, D.L. Synergistic induction of CTLA-4 expression by costimulation with TCR plus CD28 signals mediated by increased transcription and messenger ribonucleic acid stability. J. Immunol. 1997, 158, 4074–4081. [Google Scholar]
  137. Perkins, D.; Wang, Z.; Donovan, C.; He, H.; Mark, D.; Guan, G.; Wang, Y.; Walunas, T.; Bluestone, J.; Listman, J.; et al. Regulation of CTLA-4 expression during T cell activation. J. Immunol. 1996, 156, 4154–4159. [Google Scholar]
  138. Brunner-Weinzierl, M.C.; Rudd, C.E. CTLA-4 and PD-1 Control of T-Cell Motility and Migration: Implications for Tumor Immunotherapy. Front. Immunol. 2018, 9, 1972. [Google Scholar] [CrossRef] [Green Version]
  139. Takahashi, T.; Tagami, T.; Yamazaki, S.; Uede, T.; Shimizu, J.; Sakaguchi, N.; Mak, T.W.; Sakaguchi, S. Immunologic self-tolerance maintained by CD25+CD4+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J. Exp. Med. 2000, 192, 303–310. [Google Scholar] [CrossRef]
  140. Read, S.; Malmström, V.; Powrie, F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25+CD4+ regulatory cells that control intestinal inflammation. J. Exp. Med. 2000, 192, 295–302. [Google Scholar] [CrossRef] [Green Version]
  141. Schmidt, E.M.; Wang, C.J.; Ryan, G.A.; Clough, L.E.; Qureshi, O.S.; Goodall, M.; Abbas, A.K.; Sharpe, A.H.; Sansom, D.M.; Walker, L.S.K. Ctla-4 controls regulatory T cell peripheral homeostasis and is required for suppression of pancreatic islet autoimmunity. J. Immunol. 2009, 182, 274–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Yamaguchi, T.; Kishi, A.; Osaki, M.; Morikawa, H.; Prieto-Martin, P.; Wing, K.; Saito, T.; Sakaguchi, S. Construction of self-recognizing regulatory T cells from conventional T cells by controlling CTLA-4 and IL-2 expression. Proc. Natl. Acad. Sci. USA 2013, 110, E2116–E2125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Zhang, X.; Han, S.; Kang, Y.; Guo, M.; Hong, S.; Liu, F.; Fu, S.; Wang, L.; Wang, Q.X. SAHA, an HDAC inhibitor, synergizes with tacrolimus to prevent murine cardiac allograft rejection. Cell. Mol. Immunol. 2012, 9, 390–398. [Google Scholar] [CrossRef] [PubMed]
  144. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Koenen, H.J.P.M.; Smeets, R.L.; Vink, P.M.; van Rijssen, E.; Boots, A.M.H.; Joosten, I. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17-producing cells. Blood 2008, 112, 2340–2352. [Google Scholar] [CrossRef] [Green Version]
  146. Bieliauskas, A.V.; Pflum, M.K.H. Isoform-selective histone deacetylase inhibitors. Chem. Soc. Rev. 2008, 37, 1402–1413. [Google Scholar] [CrossRef]
  147. Reichert, N.; Choukrallah, M.-A.; Matthias, P. Multiple roles of class I HDACs in proliferation, differentiation, and development. Cell. Mol. Life Sci. 2012, 69, 2173–2187. [Google Scholar] [CrossRef] [Green Version]
  148. Asfaha, Y.; Schrenk, C.; Alves Avelar, L.A.; Hamacher, A.; Pflieger, M.; Kassack, M.U.; Kurz, T. Recent advances in class IIa histone deacetylases research. Bioorg. Med. Chem. 2019, 27, 115087. [Google Scholar] [CrossRef]
  149. Felice, C.; Lewis, A.; Armuzzi, A.; Lindsay, J.O.; Silver, A. Review article: Selective histone deacetylase isoforms as potential therapeutic targets in inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2015, 41, 26–38. [Google Scholar] [CrossRef]
  150. Warren, J.L.; MacIver, N.J. Regulation of Adaptive Immune Cells by Sirtuins. Front. Endocrinol. (Lausanne) 2019, 10, 466. [Google Scholar] [CrossRef]
  151. Yanginlar, C.; Logie, C. HDAC11 is a regulator of diverse immune functions. Biochim. Biophys. Acta Gene Regul. Mech. 2018, 1861, 54–59. [Google Scholar] [CrossRef] [PubMed]
  152. Holmes, D.; Gao, J.; Su, L. Foxp3 inhibits HDAC1 activity to modulate gene expression in human T cells. Virology 2011, 421, 12–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Murken, D.; Aufhauser, D., Jr.; Concors, S.; Wang, Z.; Ge, G. Nuclear Co-Repressor Complex Inhibition Reverses Benefit of HDAC2 Deletion in Renal Ischemia. Am. J. Transplant. 2017, 17 (Suppl. 3), 555. [Google Scholar] [CrossRef] [Green Version]
  154. Li, X.; Wang, W.; Wang, J.; Malovannaya, A.; Xi, Y.; Li, W.; Guerra, R.; Hawke, D.H.; Qin, J.; Chen, J. Proteomic analyses reveal distinct chromatin-associated and soluble transcription factor complexes. Mol. Syst. Biol. 2015, 11, 775. [Google Scholar] [CrossRef]
  155. Wang, L.; Liu, Y.; Han, R.; Beier, U.H.; Bhatti, T.R.; Akimova, T.; Greene, M.I.; Hiebert, S.W.; Hancock, W.W. FOXP3⁺ regulatory T cell development and function require histone/protein deacetylase 3. J. Clin. Investig. 2015, 125, 1111–1123. [Google Scholar] [CrossRef] [Green Version]
  156. Wang, L.; Beier, U.H.; Akimova, T.; Dahiya, S.; Han, R.; Samanta, A.; Levine, M.H.; Hancock, W.W. Histone/protein deacetylase inhibitor therapy for enhancement of Foxp3+ T-regulatory cell function posttransplantation. Am. J. Transplant. 2018, 18, 1596–1603. [Google Scholar] [CrossRef]
  157. Xiao, H.; Jiao, J.; Wang, L.; O’Brien, S.; Newick, K.; Wang, L.C.S.; Falkensammer, E.; Liu, Y.; Han, R.; Kapoor, V.; et al. HDAC5 controls the functions of Foxp3+ T-regulatory and CD8+ T cells. Int. J. Cancer 2016, 138, 2477–2486. [Google Scholar] [CrossRef] [Green Version]
  158. Huang, J.; Wang, L.; Dahiya, S.; Beier, U.H.; Han, R.; Samanta, A.; Bergman, J.; Sotomayor, E.M.; Seto, E.; Kozikowski, A.P.; et al. Histone/protein deacetylase 11 targeting promotes Foxp3+ Treg function. Sci. Rep. 2017, 7, 8626. [Google Scholar] [CrossRef] [Green Version]
  159. Kasler, H.G.; Lim, H.W.; Mottet, D.; Collins, A.M.; Lee, I.S.; Verdin, E. Nuclear export of histone deacetylase 7 during thymic selection is required for immune self-tolerance. EMBO J. 2012, 31, 4453–4465. [Google Scholar] [CrossRef] [Green Version]
  160. Beier, U.H.; Wang, L.; Han, R.; Akimova, T.; Liu, Y.; Hancock, W.W. Histone deacetylases 6 and 9 and sirtuin-1 control Foxp3+ regulatory T cell function through shared and isoform-specific mechanisms. Sci. Signal. 2012, 5, ra45. [Google Scholar] [CrossRef] [Green Version]
  161. De Zoeten, E.F.; Wang, L.; Sai, H.; Dillmann, W.H.; Hancock, W.W. Inhibition of HDAC9 increases T regulatory cell function and prevents colitis in mice. Gastroenterology 2010, 138, 583–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. De Zoeten, E.F.; Wang, L.; Butler, K.; Beier, U.H.; Akimova, T.; Sai, H.; Bradner, J.E.; Mazitschek, R.; Kozikowski, A.P.; Matthias, P.; et al. Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3+ T-regulatory cells. Mol. Cell. Biol. 2011, 31, 2066–2078. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Kalin, J.H.; Butler, K.V.; Akimova, T.; Hancock, W.W.; Kozikowski, A.P. Second-generation histone deacetylase 6 inhibitors enhance the immunosuppressive effects of Foxp3+ T-regulatory cells. J. Med. Chem. 2012, 55, 639–651. [Google Scholar] [CrossRef] [PubMed]
  164. Bodas, M.; Mazur, S.; Min, T.; Vij, N. Inhibition of histone-deacetylase activity rescues inflammatory cystic fibrosis lung disease by modulating innate and adaptive immune responses. Respir. Res. 2018, 19, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Nijhuis, L.; Peeters, J.G.C.; Vastert, S.J.; van Loosdregt, J. Restoring T Cell Tolerance, Exploring the Potential of Histone Deacetylase Inhibitors for the Treatment of Juvenile Idiopathic Arthritis. Front. Immunol. 2019, 10, 151. [Google Scholar] [CrossRef]
  166. Regna, N.L.; Vieson, M.D.; Luo, X.M.; Chafin, C.B.; Puthiyaveetil, A.G.; Hammond, S.E.; Caudell, D.L.; Jarpe, M.B.; Reilly, C.M. Specific HDAC6 inhibition by ACY-738 reduces SLE pathogenesis in NZB/W mice. Clin. Immunol. 2016, 162, 58–73. [Google Scholar] [CrossRef] [Green Version]
  167. Dahiya, S.; Wang, L.; Beier, U.H.; Han, R.; Hancock, W.W. HDAC10 Targeting Regulates Foxp3 Promoter, Enhances T-regulatory (Treg) Function and Suppresses Autoimmune Colitis. J. Immunol. 2018, 200, 54.11. [Google Scholar]
  168. Géraldy, M.; Morgen, M.; Sehr, P.; Steimbach, R.R.; Moi, D.; Ridinger, J.; Oehme, I.; Witt, O.; Malz, M.; Nogueira, M.S.; et al. Selective Inhibition of Histone Deacetylase 10: Hydrogen Bonding to the Gatekeeper Residue is Implicated. J. Med. Chem. 2019, 62, 4426–4443. [Google Scholar] [CrossRef]
  169. Akimova, T.; Xiao, H.; Liu, Y.; Bhatti, T.R.; Jiao, J.; Eruslanov, E.; Singhal, S.; Wang, L.; Han, R.; Zacharia, K.; et al. Targeting sirtuin-1 alleviates experimental autoimmune colitis by induction of Foxp3+ T-regulatory cells. Mucosal Immunol. 2014, 7, 1209–1220. [Google Scholar] [CrossRef]
  170. Levine, M.H.; Wang, Z.; Xiao, H.; Jiao, J.; Wang, L.; Bhatti, T.R.; Hancock, W.W.; Beier, U.H. Targeting Sirtuin-1 prolongs murine renal allograft survival and function. Kidney Int. 2016, 89, 1016–1026. [Google Scholar] [CrossRef] [Green Version]
  171. Shu, L.; Xu, C.Q.; Yan, Z.Y.; Yan, Y.; Jiang, S.Z.; Wang, Y.R. Post-Stroke Microglia Induce Sirtuin2 Expression to Suppress the Anti-inflammatory Function of Infiltrating Regulatory T Cells. Inflammation 2019, 42, 1968–1979. [Google Scholar] [CrossRef] [PubMed]
  172. Beier, U.H.; Angelin, A.; Akimova, T.; Wang, L.; Liu, Y.; Xiao, H.; Koike, M.A.; Hancock, S.A.; Bhatti, T.R.; Han, R.; et al. Essential role of mitochondrial energy metabolism in Foxp3⁺ T-regulatory cell function and allograft survival. FASEB J. 2015, 29, 2315–2326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Lin, W.; Chen, W.; Liu, W.; Xu, Z.; Zhang, L. Sirtuin4 suppresses the anti-neuroinflammatory activity of infiltrating regulatory T cells in the traumatically injured spinal cord. Immunology 2019, 158, 362–374. [Google Scholar] [CrossRef] [PubMed]
  174. Dequiedt, F.; van Lint, J.; Lecomte, E.; van Duppen, V.; Seufferlein, T.; Vandenheede, J.R.; Wattiez, R.; Kettmann, R. Phosphorylation of histone deacetylase 7 by protein kinase D mediates T cell receptor-induced Nur77 expression and apoptosis. J. Exp. Med. 2005, 201, 793–804. [Google Scholar] [CrossRef]
  175. Xu, K.; Yang, W.Y.; Nanayakkara, G.K.; Shao, Y.; Yang, F.; Hu, W.; Choi, E.T.; Wang, H.; Yang, X. GATA3, HDAC6, and BCL6 Regulate FOXP3+ Treg Plasticity and Determine Treg Conversion into Either Novel Antigen-Presenting Cell-Like Treg or Th1-Treg. Front. Immunol. 2018, 9, 45. [Google Scholar] [CrossRef] [Green Version]
  176. Ellmeier, W. Molecular control of CD4+ T cell lineage plasticity and integrity. Int. Immunopharmacol. 2015, 28, 813–817. [Google Scholar] [CrossRef]
  177. Zhou, L.; Zhang, M.; Wang, Y.; Dorfman, R.G.; Liu, H.; Yu, T.; Chen, X.; Tang, D.; Xu, L.; Yin, Y.; et al. Faecalibacterium prausnitzii Produces Butyrate to Maintain Th17/Treg Balance and to Ameliorate Colorectal Colitis by Inhibiting Histone Deacetylase 1. Inflamm. Bowel Dis. 2018, 24, 1926–1940. [Google Scholar] [CrossRef] [Green Version]
  178. Wang, X.; Buechler, N.L.; Woodruff, A.G.; Long, D.L.; Zabalawi, M.; Yoza, B.K.; McCall, C.E.; Vachharajani, V. Sirtuins and Immuno-Metabolism of Sepsis. Int. J. Mol. Sci. 2018, 19, 2738. [Google Scholar] [CrossRef] [Green Version]
  179. Vachharajani, V.T.; Liu, T.; Wang, X.; Hoth, J.J.; Yoza, B.K.; McCall, C.E. Sirtuins Link Inflammation and Metabolism. J. Immunol. Res. 2016, 2016, 8167273. [Google Scholar] [CrossRef] [Green Version]
  180. Chen, W.J.; Hu, X.F.; Yan, M.; Zhang, W.Y.; Mao, X.B.; Shu, Y.W. Human umbilical vein endothelial cells promote the inhibitory activation of CD4+CD25+Foxp3+ regulatory T cells via PD-L1. Atherosclerosis 2016, 244, 108–112. [Google Scholar] [CrossRef]
Figure 1. Foxp3 gene expression in Tregs. (A) The gene structure of mouse Foxp3 in naïve cluster of differentiation (CD)4+ T cells. The Foxp3 gene contains five regulatory elements. 5′ starting with one out of four conserved non-coding sequences (CNS0-3). The CNS0 site has recently been identified as a super-enhancer, bound by special AT-rich sequence-binding protein 1 (SATB1), being responsible for Treg-lineage-specific gene expression. Between CNS0 and CNS1 the Foxp3 promoter region is located. The binding of the SUMO E3 ligase protein inhibitor of activated signal transducer and activator of transcription (STAT) (PIAS)1 to the promoter region enables the tying of methyltransferases and heterochromatin protein 1 (HP1) to this site, maintaining the Foxp3 gene in a methylated and inactive, so called condensed, state. The methylation of the CNS2 region is also contributing to this heterochromatin structure. The Foxp3 gene contains 11 translated exons, encoding a protein of 431 amino acids in humans and 429 amino acids in mice. (B) The induction of Foxp3 expression in Tregs is initiated by the binding of self-antigens to the T cell receptor (TCR) in combination with a co-stimulatory signal such as CD28. Moreover, transforming growth factor (TGF)-β and IL-2 are essential for effective Foxp3 gene transcription. These three activation signals provoke the recruitment of nuclear factor of activated T cells (NFAT), activator protein 1 (AP-1), STAT5, FoxO1, Runt-related transcription factor 1 (RUNX), and nuclear receptor 4a (NR4a) to the promoter region, NFAT and SMAD2/3 to CNS1, Ets1, cAMP-responsive element binding protein (CREB), STAT5, RUNX, and Foxp3 to CNS2, and finally c-Rel to the CNS3 site. Additionally, Smad4 is required for Foxp3 expression. Moreover, retinoid x receptor/retinoid acid receptor (RAR/RXR) heterodimers enhance Foxp3 expression following retinoid acid stimulation, whereas STAT3 is important for Foxp3 downregulation. (C) Domain structure of Foxp3. (D) Foxp3 target genes.
Figure 1. Foxp3 gene expression in Tregs. (A) The gene structure of mouse Foxp3 in naïve cluster of differentiation (CD)4+ T cells. The Foxp3 gene contains five regulatory elements. 5′ starting with one out of four conserved non-coding sequences (CNS0-3). The CNS0 site has recently been identified as a super-enhancer, bound by special AT-rich sequence-binding protein 1 (SATB1), being responsible for Treg-lineage-specific gene expression. Between CNS0 and CNS1 the Foxp3 promoter region is located. The binding of the SUMO E3 ligase protein inhibitor of activated signal transducer and activator of transcription (STAT) (PIAS)1 to the promoter region enables the tying of methyltransferases and heterochromatin protein 1 (HP1) to this site, maintaining the Foxp3 gene in a methylated and inactive, so called condensed, state. The methylation of the CNS2 region is also contributing to this heterochromatin structure. The Foxp3 gene contains 11 translated exons, encoding a protein of 431 amino acids in humans and 429 amino acids in mice. (B) The induction of Foxp3 expression in Tregs is initiated by the binding of self-antigens to the T cell receptor (TCR) in combination with a co-stimulatory signal such as CD28. Moreover, transforming growth factor (TGF)-β and IL-2 are essential for effective Foxp3 gene transcription. These three activation signals provoke the recruitment of nuclear factor of activated T cells (NFAT), activator protein 1 (AP-1), STAT5, FoxO1, Runt-related transcription factor 1 (RUNX), and nuclear receptor 4a (NR4a) to the promoter region, NFAT and SMAD2/3 to CNS1, Ets1, cAMP-responsive element binding protein (CREB), STAT5, RUNX, and Foxp3 to CNS2, and finally c-Rel to the CNS3 site. Additionally, Smad4 is required for Foxp3 expression. Moreover, retinoid x receptor/retinoid acid receptor (RAR/RXR) heterodimers enhance Foxp3 expression following retinoid acid stimulation, whereas STAT3 is important for Foxp3 downregulation. (C) Domain structure of Foxp3. (D) Foxp3 target genes.
Ijms 21 02356 g001
Figure 2. HAT and HDAC binding to Foxp3. (A) Amino groups located at the ε-CH2 group of lysines can be actetylated by histone acetylases (HATs) such as p300 or TI60 leading to acetylysine, which enhances Foxp3 stability by preventing its proteasomal degradation. Reciprocally histone deacetylases (HDACs) such as Sirt1 deacetylate lysines of Foxp3, which are acetylated at the amino-group next to the ε-C atom. Deacetylated Foxp3 is prone to proteasomal degradation [124]. (B) Lysines of Foxp3, which have been identified as targets for acetylation [122,125,132,133].
Figure 2. HAT and HDAC binding to Foxp3. (A) Amino groups located at the ε-CH2 group of lysines can be actetylated by histone acetylases (HATs) such as p300 or TI60 leading to acetylysine, which enhances Foxp3 stability by preventing its proteasomal degradation. Reciprocally histone deacetylases (HDACs) such as Sirt1 deacetylate lysines of Foxp3, which are acetylated at the amino-group next to the ε-C atom. Deacetylated Foxp3 is prone to proteasomal degradation [124]. (B) Lysines of Foxp3, which have been identified as targets for acetylation [122,125,132,133].
Ijms 21 02356 g002
Table 1. Polymicrobial sepsis induced by a cecal ligation and puncture (CLP) operation in mice and rats increased regulatory T cell (Treg) count in the blood and spleen. Different mouse and rat strains have been used. The severity of the model is affected by the needle diameter, the number of punctures, and the ligation length [64,65]. (Ø, diameter; CLP, cecal ligation and puncture; f, female; G, gauge; m, male; MLN, mesenteric lymph nodes; and PC, peritoneal cavity.)
Table 1. Polymicrobial sepsis induced by a cecal ligation and puncture (CLP) operation in mice and rats increased regulatory T cell (Treg) count in the blood and spleen. Different mouse and rat strains have been used. The severity of the model is affected by the needle diameter, the number of punctures, and the ligation length [64,65]. (Ø, diameter; CLP, cecal ligation and puncture; f, female; G, gauge; m, male; MLN, mesenteric lymph nodes; and PC, peritoneal cavity.)
StrainSexWeightAgeCLPTregsRef.
[g][weeks]LigationNeedle ØPerfo-RationDura-tionOrgan
MiceBALB/cm-8immediately distal to the ileocecal valve twice ↑ (spleen)[67]
m20 ± 26–81/3, 2/3, 3/323Gsingle24 h↑ (spleen)[68]
m20–25850%21Gonce15 d↑ (spleen)[69]
m18–22-50%21Gtwice24 + 48 h↑ (spleen)[70]
m20 ± 16–8below the ileocecal valve18Gonce1/2/3/4 d↑ (blood)[71]
C57BL/6m258caecum ligated at its base18Gtwice ↓ (blood)[72]
f-6–850%27Gtwice3/7 d↑ (spleen)[73]
m25–27--23G-48 h↑ (spleen)[74]
m25–35-50%18Gtwice24 h↑ (spleen)[75]
m20–257–930% of its length21Gtwice5 d↑ (spleen)[76]
m20–258–101.5 cm from the tip22Gtwice20 h↑ (spleen)[77]
m/f 6–8below the ileocecal valve21Gnine24 h↑ (PC, MLN)[78]
m22–256–8 22Gonce3 d↑ (spleen)[79]
m20–25-75%21Gtwice24 h↑ (spleen)[80]
m20–25-75%21Gtwice24 h↑ (spleen)[81]
m22–308–10below the ileocecal valve22Gtwice24 h↑ (spleen)[82]
m258at its base21Gonce1/3 d↑ (MLN)[83]
m-71 cm from the apex18Gtwice16 h↑ (spleen)[84]
f-8–1230%27Gonce24 + 48 h↑ (spleen)[85]
m-8–12 22Gtwice24 h↑ (spleen)[86]
m-6–8 22Gtwice30 h↑ (spleen)[87]
FVB/N
9xNFAT luc
---75%21Gtwice24 h↑ (spleen)[88]
ICRm30–356–850%23Gtwice24 + 72 h↑ (blood)[89]
m27–29-at its distal site20Gtwice26 h↑ (spleen)[90]
NMRI 20–30-30%27Gonce1/2/3 d↑ (spleen)[91]
RatsFischerm-10470% of its length18Gtwice20 h↑ (spleen)[92]
Wistarm250–300 50%18Gtwice18 h↓ (blood)[93]
Wistar Hannoverm200–2508below the ileocecal valve18Gtwice24 h↑ (MLN)[94]
Sprague Dawleym350–400-distal ligation18Gtwice3 d↑ (blood)[95]
Sprague Dawleym320–350-distal ligation18Gtwice3 d↑ (spleen)[96]
Sprague Dawleym400–450-distal ligation18Gtwice72 h↑ (blood + spleen)[97]
Table 2. HDAC isoforms expressed in Tregs promoting/attenuating their function. (C, colitis; CAT, cardiac allograft transplantation; CF, cystic fibrosis; C/JIA, collagen/juvenile-induced arthritis; CLP, cecal ligation and puncture; MCAO, mouse transient middle cerebral artery occlusion.)
Table 2. HDAC isoforms expressed in Tregs promoting/attenuating their function. (C, colitis; CAT, cardiac allograft transplantation; CF, cystic fibrosis; C/JIA, collagen/juvenile-induced arthritis; CLP, cecal ligation and puncture; MCAO, mouse transient middle cerebral artery occlusion.)
ClassIsoformLocalizationEffect of HDAC TargetingSpecific HDACiHDAC-Foxp3 InteractionModelsRef.
IHDAC1nucleusnoinhibits HDAC1CAT, C[152,153]
HDAC2nucleusin progressassociates with Foxp3CAT, C[153,154]
HDAC3nucleus/cytosolnodestabilizes Foxp3CAT, C[155]
HDAC8nucleusavailable?CAT[155,156]
IIaHDAC5nucleus/cytosolno?CAT[157]
HDAC7nucleus/cytosolnoforms a transcriptional complex with Foxp3thymic positive and negative T cell selection[158,159]
HDAC9nucleus/cytosolnodestabilizes Foxp3C[132,160,161]
IIbHDAC6nucleus/cytosolavailabledestabilizes Foxp3CF, CIA, JIA,
lupus prone mice
[160,162,163,164,165,166]
HDAC10nucleus/cytosolin progressdestabilizes Foxp3, re-presses Foxp3 transcriptionCAT, C[167,168,169]
IIISIRT1nucleusavailabledestabilizes Foxp3CLP, heterotrophic cardiac and ortho-tropic renal allo-graft, C[87,126,160,169,170]
SIRT2cytosolnodestabilizes Foxp3MCAO[171]
SIRT3mitono-CAT[172]
SIRT4mitonoinhibits Foxp3 expressionmouse spinal cord compression in-jury[173]
IVHDAC11nucleusavailabledestabilizes Foxp3CAT[158]

Share and Cite

MDPI and ACS Style

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. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072356

AMA Style

von Knethen A, Heinicke U, Weigert A, Zacharowski K, Brüne B. Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells. International Journal of Molecular Sciences. 2020; 21(7):2356. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072356

Chicago/Turabian Style

von Knethen, Andreas, Ulrike Heinicke, Andreas Weigert, Kai Zacharowski, and Bernhard Brüne. 2020. "Histone Deacetylation Inhibitors as Modulators of Regulatory T Cells" International Journal of Molecular Sciences 21, no. 7: 2356. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21072356

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