scholarly journals Regulatory T Cells-Related Genes Are under DNA Methylation Influence

2021 ◽  
Vol 22 (13) ◽  
pp. 7144
Author(s):  
Magdalena Piotrowska ◽  
Mateusz Gliwiński ◽  
Piotr Trzonkowski ◽  
Dorota Iwaszkiewicz-Grzes

Regulatory T cells (Tregs) exert a highly suppressive function in the immune system. Disturbances in their function predispose an individual to autoimmune dysregulation, with a predominance of the pro-inflammatory environment. Besides Foxp3, which is a master regulator of these cells, other genes (e.g., Il2ra, Ctla4, Tnfrsf18, Ikzf2, and Ikzf4) are also involved in Tregs development and function. Multidimensional Tregs suppression is determined by factors that are believed to be crucial in the action of Tregs-related genes. Among them, epigenetic changes, such as DNA methylation, tend to be widely studied over the past few years. DNA methylation acts as a repressive mark, leading to diminished gene expression. Given the role of increased CpG methylation upon Tregs imprinting and functional stability, alterations in the methylation pattern can cause an imbalance in the immune response. Due to the fact that epigenetic changes can be reversible, so-called epigenetic modifiers are broadly used in order to improve Tregs performance. In this review, we place emphasis on the role of DNA methylation of the genes that are key regulators of Tregs function. We also discuss disease settings that have an impact on the methylation status of Tregs and systematize the usefulness of epigenetic drugs as factors able to influence Tregs functions.

Arthritis ◽  
2012 ◽  
Vol 2012 ◽  
pp. 1-16 ◽  
Author(s):  
Maryam Gol-Ara ◽  
Farhad Jadidi-Niaragh ◽  
Reza Sadria ◽  
Gholamreza Azizi ◽  
Abbas Mirshafiey

Rheumatoid arthritis (RA) is a common autoimmune disease and a systemic inflammatory disease which is characterized by chronic joint inflammation and variable degrees of bone and cartilage erosion and hyperplasia of synovial tissues. Considering the role of autoreactive T cells (particularly Th1 and Th17 cells) in pathophysiology of RA, it might be assumed that the regulatory T cells (Tregs) will be able to control the initiation and progression of disease. The frequency, function, and properties of various subsets of Tregs including natural Tregs (nTregs), IL-10-producing type 1 Tregs (Tr1 cells), TGF-β-producing Th3 cells, CD8+ Tregs, and NKT regulatory cells have been investigated in various studies associated with RA and collagen-induced arthritis (CIA) as experimental model of this disease. In this paper, we intend to submit the comprehensive information about the immunobiology of various subsets of Tregs and their roles and function in immunopathophysiology of RA and its animal model, CIA.


2004 ◽  
Vol 16 (11) ◽  
pp. 1643-1656 ◽  
Author(s):  
Haruhiko Yagi ◽  
Takashi Nomura ◽  
Kyoko Nakamura ◽  
Sayuri Yamazaki ◽  
Toshio Kitawaki ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4835-4835
Author(s):  
Rodrigo Haddad ◽  
Josiane Lilian dos Santos Schiavinato ◽  
Felipe Saldanha Saldanha-Araújo ◽  
Priscila S Scheucher ◽  
Amélia G Araújo ◽  
...  

Abstract Abstract 4835 The development and functionality of CD4+CD25hi regulatory T cells (Tregs) depends on stable FoxP3 expression, a central regulator of Treg differentiation. It is believed that this is accomplished by regulatory regions in the promoter and 3 evolutionarily conserved noncoding sequences, termed CNS1, CNS2 (or TSDR) and CNS3. The activation of TCR (with anti-CD2/3/28) in CD4+CD25− naïve T cells from PBMCs, in the presence of IL-2, TGF-β and atRA, induces the generation of Foxp3+ induced regulatory T cells (iTreg). While demethylation of 5mC residues in the CNS2 is associated with stable FoxP3 expression in nTregs, the epigenetic events involved in the regulation of FoxP3 in iTregs remains unexplored. Recently, the oxidation of 5-mC, originating hidroxymethylated 5-hmC residues, have been described as a key mechanism of active demethylation, with roles in biological processes, such as regulation of pluripotency and differentiation of hematopoietic stem cells. In contrast to PBMCs, in umbilical cord blood (UCB) T cells are mainly naïve making UCB an attractive source for the development of protocols for generation of iTregs. Here, we evaluated the iTregs generation from UCB naïve T cells. In addition, we compared the expression of FoxP3 on iTregs and on naturally occurring Tregs (nTregs) obtained from PBMCs. Also, we evaluated the methylation pattern of promoter and CNS2 and CNS3 in nTregs, fleshly isolated naïve T cells, activated naïve T cells (Teff), and iTregs. Finally, we evaluated the ability of iTregs, to suppress the proliferation of activated T cells, as compared to nTregs. For this, CD4+CD25-CD45RA+ naïve T cells were immunomagnetically isolated from UCB and activated with anti-human CD2/CD3/CD28 beads (1:2 beads:cell ratio) in the presence of IL-2 (50 U/ml) with (iTregs) or without (Teff) TGF-β (5 ng/ml) and atRA (100 nM) for 5 days. In parallel, PBMCs from 5 individuals were obtained for nTregs phenotypic characterization. CD4+ gated cells from iTregs and from PBMC were analyzed by flow cytometry for FoxP3 expression in the CD25+, CD25hi and CD25−population. nTregs (CD4+CD25+CD127−) were immunomagnetically isolated from PBMCs and CD4+CD25hi and CD4+CD25− populations were FACS-sorted from iTreg to observe the potential in regulate the proliferation of CD3+ T cells (CFSE staining). Finally, methylation pattern analysis of FoxP3 locus, including CNS2 and CNS3, were performed in naïve T cells, nTregs, iTreg and Teff. The mean percentage of FoxP3+ cells in CD4+CD25hi from iTreg was 98.5%, as compared to 82.4% in PBMCs. Interestingly, the percentage of FoxP3+ cells in total CD4+CD25+ was higher in cells from iTreg (97,3%) than on PBMCs (26,8%). Moreover, while the percentage of FoxP3+ cells in the CD4+CD25− population, was very low in PBMCs (2,8%), up to 55% of the cells derived from iTreg were FoxP3+. The immunossupression assay showed that, compared to activated CD3+ T cells cultured alone, nTregs (CD4+CD25+CD127−) decreased the proliferation of CD3+ T cells in 55%, while iTregs (CD4+CD25hi) decreased the proliferation in 46%. Interestingly, the CD4+CD25− population from iTreg (55% of FoxP3+ cells) also decreased the proliferation of CD3+ T cells, but to a lower extent (21%). Additionally, while naïve T cells and Teff presented low level of 5hmC in both segments evaluated of CNS2 (∼1%); upon in vitro induction, iTregs presented 5hmC levels comparable to that of nTregs (5–11% and 5% respectively), in line to FoxP3 expression. Furthermore, CNS3, which was found to be partially demethylated in naïve T cells and nTregs (45 and 50% respectively), presented even higher levels of demethylation upon activation in iTregs and nTregs (77 and 82% respectively). In summary, we show that functional Foxp3+CD4+CD25hi T cells can be generated in vitro from UCB naïve T cells. Additionally, our results indicate that active demethylation of CNS2 occurs in a TGF-β and atRA-dependent manner during iTregs generation. Moreover, the partial demethylation of CNS3 observed in naïve T cells and nTregs, and the increased demethylation promoted by activation (in Teff and iTreg), is consistent with the role of CNS3 as a pioneer element that initiates FoxP3 transcription. Our results contribute to the understanding of the epigenetic mechanisms underlying the differentiation of Tregs and may help in the development of protocols for the generation of functional iTregs for future therapeutic applications. Support: FAPESP, CNPq. Disclosures: No relevant conflicts of interest to declare.


Author(s):  
Kristy Ou ◽  
Dania Hamo ◽  
Anne Schulze ◽  
Andy Roemhild ◽  
Daniel Kaiser ◽  
...  

Adoptive transfer of regulatory T cells (Treg) is a promising new therapeutic option to treat detrimental inflammatory conditions after transplantation and during autoimmune disease. To reach sufficient cell yield for treatment, ex vivo isolated autologous or allogenic Tregs need to be expanded extensively in vitro during manufacturing of the Treg product. However, repetitive cycles of restimulation and prolonged culture have been shown to impact T cell phenotypes, functionality and fitness. It is therefore critical to scrutinize the molecular changes which occur during T cell product generation, and reexamine current manufacturing practices. We performed genome-wide DNA methylation profiling of cells throughout the manufacturing process of a polyclonal Treg product that has proven safety and hints of therapeutic efficacy in kidney transplant patients. We found progressive DNA methylation changes over the duration of culture, which were donor-independent and reproducible between manufacturing runs. Differentially methylated regions (DMRs) in the final products were significantly enriched at promoters and enhancers of genes implicated in T cell activation. Additionally, significant hypomethylation did also occur in promoters of genes implicated in functional exhaustion in conventional T cells, some of which, however, have been reported to strengthen immunosuppressive effector function in Tregs. At the same time, a set of reported Treg-specific demethylated regions increased methylation levels with culture, indicating a possible destabilization of Treg identity during manufacturing, which was independent of the purity of the starting material. Together, our results indicate that the repetitive TCR-mediated stimulation lead to epigenetic changes that might impact functionality of Treg products in multiple ways, by possibly shifting to an effector Treg phenotype with enhanced functional activity or by risking destabilization of Treg identity and impaired TCR activation. Our analyses also illustrate the value of epigenetic profiling for the evaluation of T cell product manufacturing pipelines, which might open new avenues for the improvement of current adoptive Treg therapies with relevance for conventional effector T cell products.


2015 ◽  
Vol 2015 ◽  
pp. 1-8 ◽  
Author(s):  
Érica S. S. de Araújo ◽  
Dimitrius T. Pramio ◽  
André Y. Kashiwabara ◽  
Paula C. Pennacchi ◽  
Silvya S. Maria-Engler ◽  
...  

In melanoma development, oncogenic process is mediated by genetic and epigenetic mutations, and few studies have so far explored the role of DNA methylation either as predisposition factor or biomarker. We tested patient samples for germlineCDKN2Amethylation status and found no evidence of inactivation by promoter hypermethylation. We have also investigated the association of clinical characteristics of samples with the DNA methylation pattern of twelve genes relevant for melanomagenesis. Five genes (BAP1, MGMT, MITF, PALB2, andPOT1) presented statistical association between blood DNA methylation levels and eitherCDKN2A-mutation status, number of lesions, or Breslow thickness. In tumors, five genes (KIT, MGMT, MITF, TERT, andTNF) exhibited methylation levels significantly different between tumor groups including acral compared to nonacral melanomas and matched primary lesions and metastases. Our data pinpoint that the methylation level of eight melanoma-associated genes could potentially represent markers for this disease both in peripheral blood and in tumor samples.


Sign in / Sign up

Export Citation Format

Share Document