scholarly journals Epigenetic Programming during thymic development sets the stage for optimal function in effector T cells via DNA demethylation

2021 ◽  
Author(s):  
Athmane Teghanemt ◽  
Priyanjali Pulipati ◽  
Kenneth Day ◽  
Matthew S. Yorek ◽  
Ren Yi ◽  
...  

The repressive effect of DNA methylation at promoters is well-known. However, its role within conserved sequences in intragenic and intergenic regions is less clear. Using Cd4 as a model gene, here we show that DNA methylation regulates the function of stimulus-responsive regulatory elements in effector T cells. Two cis-elements orchestrate intra-and intergenic DNA demethylation of the Cd4 gene during thymic development, which in turn licenses a stimulusresponsive element, E4a, for its later function in effector cells. Deficiency in DNA demethylation leads to impaired E4a function, reduced H3K4me3 promoter levels and an inability to repel de novo DNA methylation during replication, ultimately leading to gene silencing. This physiological reduction in CD4 expression leads to a defect in Th1 polarization during cutaneous Leishmaniasis. Similar patterns of regulation were observed in a broad number of genes, highlighting an essential role for DNA demethylation during thymic development in modulating the function of stimulus-responsive elements.

2021 ◽  
Author(s):  
Priya Issuree ◽  
Athmane Teghanemt ◽  
Priyanjali Pulipati ◽  
Kenneth Day ◽  
Matt Yorek ◽  
...  

Abstract The potential for early thymic developmental events to program epigenetic states that influence adult T cell physiology remains an important question in health. Herein using the Cd4 locus as a paradigm for early developmental programming, we demonstrate that DNA demethylation during thymic development is critical for the licensing of a novel stimulus-responsive element that serves to maintain CD4 gene expression in effector T cells. We document the importance of maintaining high CD4 expression during parasitic infection and show that by driving transcription, this stimulus-responsive element allows for the maintenance of H3K4me3 levels during T cell replication, which is critical for repelling de novo DNA methylation at the Cd4 promoter. A failure to undergo epigenetic programming during development leads to gene silencing during effector T cell replication, thus providing evidence that early development can program stimulus-responsive elements to propagate a stable epigenetic state in effector T cells, with important biological consequences.


2021 ◽  
Vol 22 (7) ◽  
pp. 3735
Author(s):  
Guillaume Velasco ◽  
Damien Ulveling ◽  
Sophie Rondeau ◽  
Pauline Marzin ◽  
Motoko Unoki ◽  
...  

DNA methylation (DNAme) profiling is used to establish specific biomarkers to improve the diagnosis of patients with inherited neurodevelopmental disorders and to guide mutation screening. In the specific case of mendelian disorders of the epigenetic machinery, it also provides the basis to infer mechanistic aspects with regard to DNAme determinants and interplay between histone and DNAme that apply to humans. Here, we present comparative methylomes from patients with mutations in the de novo DNA methyltransferases DNMT3A and DNMT3B, in their catalytic domain or their N-terminal parts involved in reading histone methylation, or in histone H3 lysine (K) methylases NSD1 or SETD2 (H3 K36) or KMT2D/MLL2 (H3 K4). We provide disease-specific DNAme signatures and document the distinct consequences of mutations in enzymes with very similar or intertwined functions, including at repeated sequences and imprinted loci. We found that KMT2D and SETD2 germline mutations have little impact on DNAme profiles. In contrast, the overlapping DNAme alterations downstream of NSD1 or DNMT3 mutations underlines functional links, more specifically between NSD1 and DNMT3B at heterochromatin regions or DNMT3A at regulatory elements. Together, these data indicate certain discrepancy with the mechanisms described in animal models or the existence of redundant or complementary functions unforeseen in humans.


2009 ◽  
Vol 206 (11) ◽  
pp. 2329-2337 ◽  
Author(s):  
Ludovica Bruno ◽  
Luca Mazzarella ◽  
Maarten Hoogenkamp ◽  
Arnulf Hertweck ◽  
Bradley S. Cobb ◽  
...  

Runx proteins are essential for hematopoiesis and play an important role in T cell development by regulating key target genes, such as CD4 and CD8 as well as lymphokine genes, during the specialization of naive CD4 T cells into distinct T helper subsets. In regulatory T (T reg) cells, the signature transcription factor Foxp3 interacts with and modulates the function of several other DNA binding proteins, including Runx family members, at the protein level. We show that Runx proteins also regulate the initiation and the maintenance of Foxp3 gene expression in CD4 T cells. Full-length Runx promoted the de novo expression of Foxp3 during inducible T reg cell differentiation, whereas the isolated dominant-negative Runt DNA binding domain antagonized de novo Foxp3 expression. Foxp3 expression in natural T reg cells remained dependent on Runx proteins and correlated with the binding of Runx/core-binding factor β to regulatory elements within the Foxp3 locus. Our data show that Runx and Foxp3 are components of a feed-forward loop in which Runx proteins contribute to the expression of Foxp3 and cooperate with Foxp3 proteins to regulate the expression of downstream target genes.


2014 ◽  
Vol 211 (9) ◽  
pp. 1807-1819 ◽  
Author(s):  
Christian Neumann ◽  
Frederik Heinrich ◽  
Katrin Neumann ◽  
Victoria Junghans ◽  
Mir-Farzin Mashreghi ◽  
...  

Secretion of the immunosuppressive cytokine interleukin (IL) 10 by effector T cells is an essential mechanism of self-limitation during infection. However, the transcriptional regulation of IL-10 expression in proinflammatory T helper (Th) 1 cells is insufficiently understood. We report a crucial role for the transcriptional regulator Blimp-1, induced by IL-12 in a STAT4-dependent manner, in controlling IL-10 expression in Th1 cells. Blimp-1 deficiency led to excessive inflammation during Toxoplasma gondii infection with increased mortality. IL-10 production from Th1 cells was strictly dependent on Blimp-1 but was further enhanced by the synergistic function of c-Maf, a transcriptional regulator of IL-10 induced by multiple factors, such as the Notch pathway. We found Blimp-1 expression, which was also broadly induced by IL-27 in effector T cells, to be antagonized by transforming growth factor (TGF) β. While effectively blocking IL-10 production from Th1 cells, TGF-β shifted IL-10 regulation from a Blimp-1–dependent to a Blimp-1–independent pathway in IL-27–induced Tr1 (T regulatory 1) cells. Our findings further illustrate how IL-10 regulation in Th cells relies on several transcriptional programs that integrate various signals from the environment to fine-tune expression of this critical immunosuppressive cytokine.


2016 ◽  
Vol 311 (4) ◽  
pp. G713-G723 ◽  
Author(s):  
Beng San Yeoh ◽  
Piu Saha ◽  
Vishal Singh ◽  
Xia Xiao ◽  
Yun Ying ◽  
...  

Stearoyl-CoA desaturase-1 (SCD1) is a lipogenic enzyme involved in the de novo biosynthesis of oleate (C18:1, n9), a major fatty acid in the phospholipids of lipid bilayers of cell membranes. Accordingly, Scd1KO mice display substantially reduced oleate in cell membranes. An altered SCD1 level was observed during intestinal inflammation; however, its role in modulating inflammatory bowel disease remains elusive. Herein, we investigated the colitogenic capacity of Scd1KO effector T cells by employing the adoptive T-cell transfer colitis model. Splenic effector T cells (CD4+CD25−) from age- and sex-matched wild-type (WT) and Scd1KO mice were isolated by FACS and intraperitoneally administered to Rag1KO mice, which were monitored for the development of colitis. At day 60 postcell transfer, Rag1KO mice that received Scd1KO CD4+CD25− T cells displayed accelerated and exacerbated colitis than mice receiving WT CD4+CD25− T cells. Intriguingly, Scd1KO CD4+CD25− T cells display augmented inflammatory cytokine profile and cellular membrane fluidity with a concomitant increase in proinflammatory saturated fatty acids, which we postulate to potentially underlie their augmented colitogenic potential.


Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1141-1149 ◽  
Author(s):  
Ilona Kryczek ◽  
Mousumi Banerjee ◽  
Pui Cheng ◽  
Linhua Vatan ◽  
Wojciech Szeliga ◽  
...  

Abstract Th17 cells play an active role in autoimmune diseases. However, the nature of Th17 cells is poorly understood in cancer patients. We studied Th17 cells, the associated mechanisms, and clinical significance in 201 ovarian cancer patients. Tumor-infiltrating Th17 cells exhibit a polyfunctional effector T-cell phenotype, are positively associated with effector cells, and are negatively associated with tumor-infiltrating regulatory T cells. Tumor-associated macrophages promote Th17 cells through interleukin-1β (IL-1β), whereas tumor-infiltrating regulatory T cells inhibit Th17 cells through an adenosinergic pathway. Furthermore, through synergistic action between IL-17 and interferon-γ, Th17 cells stimulate CXCL9 and CXCL10 production to recruit effector T cells to the tumor microenvironment. The levels of CXCL9 and CXCL10 are associated with tumor-infiltrating effector T cells. The levels of tumor-infiltrating Th17 cells and the levels of ascites IL-17 are reduced in more advanced diseases and positively predict patient outcome. Altogether, Th17 cells may contribute to protective human tumor immunity through inducing Th1-type chemokines and recruiting effector cells to the tumor microenvironment. Inhibition of Th17 cells represents a novel immune evasion mechanism. This study thus provides scientific and clinical rationale for developing novel immune-boosting strategies based on promoting the Th17 cell population in cancer patients.


Blood ◽  
2013 ◽  
Vol 121 (22) ◽  
pp. 4595-4602 ◽  
Author(s):  
Amar Nijagal ◽  
Chris Derderian ◽  
Tom Le ◽  
Erin Jarvis ◽  
Linda Nguyen ◽  
...  

Key Points Tolerance induction after in utero hematopoietic cell transplantation involves both direct and indirect antigen presentation. Tolerance is achieved by deletion of effector T cells, which results in Treg enrichment without de novo Treg induction.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2760-2760
Author(s):  
Jennifer J. Trowbridge ◽  
Mingjie Li ◽  
Charles W.M. Roberts ◽  
Stuart H. Orkin

Abstract Abstract 2760 The significance of mutations in components of the DNA methylation machinery in blood cancer has become a topic of intense investigation. Unlike genetic modifications, the reversible nature of DNA methylation and other epigenetic changes makes them attractive therapeutic targets. Very recently, mutations in the DNA methyltransferase DNMT3A and the DNA demethylase TET2 were identified in human peripheral T cell lymphoma (PTCL) [1]. These findings provided a novel link between the development and progression of PTCL with deregulation of DNA methylation processes. Importantly, this finding also extended the few known mutations associated with both T-cell lymphoma and myeloid leukemia. Our previous work identified acute sensitivity of MLL-AF9–induced myeloid leukemia (AML) to DNA demethylation through loss or haploinsufficiency of the DNA methyltransferase Dnmt1 [2]. Here, we investigated the sensitivity of PTCL to DNA demethylation. Lymphoma was induced in mice by inactivation of Snf5, a core subunit of the SWI/SNF chromatin remodeling complex, driven by CD4Cre (CD4Cre-Snf52lox). Inactivation of Snf5 leads to rapid onset of mature CD8+ PTCL with a median survival of 10 weeks of age. Strikingly, loss of Dnmt1 in this model (CD4Cre-Snf52lox-Dnmt12lox) completely abrogated development of lymphoma. Furthermore, haploinsufficiency of Dnmt1 was sufficient to increase event-free survival to 13 weeks of age (p=0.0008). Loss or haploinsufficiency of Dnmt1 did not impact normal T cell development in the thymus with the exception of a modest reduction in CD8+ CD44hi memory T cells. Based on the selective response of PTCL to reduced levels of Dnmt1 and DNA methylation, we screened a panel of pharmacological DNA demethylating agents for efficacy in PTCL. We found three putative DNA methylation inhibitors; the nucleoside inhibitor zebularine and non-nucleoside inhibitors RG108 and procainamide, which inhibited proliferation of primary murine PTCL in vitro. These inhibitors were effective at doses that did not restrict the proliferation of normal CD8+ T cells. When these inhibitors were evaluated for efficacy in vivo, both zebularine and procainamide were found to inhibit growth of primary murine PTCL. Together, these results suggest that therapy of PTCL with DNA methylation inhibitors or other DNA demethylating agents may achieve a favorable therapeutic index. Further, these results support the concept of a shared competitive advantage of myeloid leukemia and T-cell lymphoma in carrying mutations in the DNA methylation machinery. [1] Couronne L et al., NEJM, 2012, 366:95-6; [2] Trowbridge et al., Genes Dev, 2012, 26:344-9. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Sandip Ashok Sonar ◽  
Jennifer L Uhrlaub ◽  
Christopher P Coplen ◽  
Gregory D Sempowski ◽  
Jarrod A Dudakov ◽  
...  

Secondary lymphoid organs (SLO; including the spleen and lymph nodes) are critical both for the maintenance of naive T (TN) lymphocytes and for the initiation and coordination of immune responses. How they age, including the exact timing, extent, physiological relevance, and the nature of age-related changes, remains incompletely understood. We used time-stamping to indelibly mark cohorts of newly generated naive T cells (a.k.a. recent thymic emigrants - RTE) in mice, and followed their presence, phenotype and retention in SLO. We found that SLO involute asynchronously. Skin-draining lymph nodes (LN) atrophied early (6-9 months) in life and deeper tissue-draining LN and the spleen late (18-20 months), as measured by the loss of both TN numbers and the fibroblastic reticular cell (FRC) network. Time-stamped RTE cohorts of all ages entered SLO and successfully completed post-thymic differentiation. However, in older mice, these cells were poorly retained, and those found in SLO exhibited an emigration phenotype (CCR7loS1P1hi). Transfers of adult RTE into recipients of different ages formally demonstrated that the defect segregates with the age of the SLO microenvironment and not with the age of T cells. Finally, upon intradermal immunization, RTE generated in mice as early as 6-7 months of age barely participated in de novo immune responses and failed to produce well-armed effector cells. These results highlight changes in structure and function of superficial secondary lymphoid organs in laboratory mice that are earlier than expected and are consistent with the long-appreciated and pronounced reduction of cutaneous immunity with aging.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 70-70
Author(s):  
Hidenori Kasahara ◽  
Shinichiro Okamoto ◽  
Takashi Sekiya ◽  
Akihiko Yoshimura

Abstract Antigen-specific regulatory T cells (Tregs) possess the potential to reduce excess of immune responses following allogeneic hematopoietic stem cell transplantation. Although antigen-specific in vitro- expanded Tregs (iTregs) have long been considered as a promising therapeutic agent against allo-immune reactions such as graft versus host disease (GVHD), accumulating evidences have suggested that iTregs easily lose their characteristics with quick cessation of Foxp3 expression , a lineage specifying transcription factor of Tregs, compared with in vivo -generated natural Tregs (nTregs). It has been revealed that insufficient demethylation of the CpG islands in conserved noncoding sequence 2 (CNS2) region of the Foxp3 locus mainly explains such instability of Foxp3 expression in iTregs. In order to overcome this drawback, we investigated the optimum way to generate stable iTregs for the prevention of a murine model of acute GVHD. We created a major-MHC mismatched GVHD model by transferring CD45.1 (H-2Kb) bone marrow (BM) cells together with effector T cells (CD4+25- and CD8 cells) into lethally irradiated (8Gy/body) 8-12-week old BALB/C (H-2Kd) recipients. In this model, recipient mice usually die within 7-10 days after bone marrow transplant (BMT) due to severe acute GVHD. Alloantigen-specific iTregs were generated by co-culturing naive T cells from human CD2 Foxp3 reporter mice (C57BL6/J background, H-2Kb) with BALB/C-derived antigen presenting cells in the presence of TGF-β, IL-2 and retinoic acid. Alloantigen-specific iTregs were then harvested 6-7 days after co-culture, by FACS-sorting the CD4+Foxp3+ population. Subsequently, sorted iTregs (106 cells) were transferred intravenously together with effector T cells (106 cells) and BM cells into recipients. We tested a panel of pharmacological agents and gene transduction during co-culture for their effects on iTreg stability, with FACS-based evaluation on day 7 after BMT. Among a panel of agents and gene modification tested, we found that only vitamin C-treated iTregs effectively improved Foxp3 maintenance compared with untreated iTregs (90%, vitamin C-treated iTregs vs 40%, untreated iTregs) in the GVHD model mice. As reported, vitamin C facilitated DNA demethylation of the Foxp3 CNS2 in iTregs in a Tet DNA demethylase-dependent manner. Bisulfite sequencing revealed a significant acceleration of CpG demethylation at the Foxp3 CNS2 by vitamin C, and the extent of demethylation achieved with vitamin C treatment reached to an equivalent level to those seen in nTregs. Furthermore, vitamin C mediated demethylation was extended to other Treg cell-specific regulatory lesions such as Tnfrsf18, Ikzf4, Ctla4 . On the other hand, untreated iTregs remain methylated at these loci to the same degree as naïve T cells (p<0.05) . At the same time, interestingly, production of inflammatory cytokines such as IFNγ and IL-21 that was observed in untreated iTregs which lost Foxp3 expression (ex- Foxp3 iTreg) was not observed in vitamin C-treated counterparts. Additionally, we further evaluated the benefits of vitamin C treatment, by investigating effects of another well-known demethylating agent, 5-aza-2'-deoxytidine (5-aza-dC). As a result, 5-aza-dC did not only induce DNA demethylation of Foxp3 CNS2, but also of the CNS1 enhancer region of the inflammatory cytokine IFNγ locus, even in iTreg conditioning culture. This nonspecific demethylation was not observed in vitamin C-treated iTregs (p<0.01). Finally , adoptive transfer of vitamin C-treated iTregs ameliorated GVHD in mice. Vitamin C-treated iTreg prolonged survival at BMT day 45 compared with untreated iTreg (100%, N=8, vitamin C treated iTreg group, vs 44.4%, untreated iTreg group, N=9, p<0.01). Amelioration of acute GVHD by vitamin C-treated iTreg was also confirmed by colon length on day 7 after BMT (p<0.05) and pathological findings of colon and small intestine. In summary, vitamin C stabilized Foxp3 expression with specific demethylation of Foxp3 CNS2 as well as Treg-associated genes in antigen-specific iTregs, preventing them from conversion into inflammatory ex- Foxp3 iTregs, thus ameliorating the pathogenesis of a murine model of GVHD. Our findings strongly suggest the potential of clinical application of Treg therapy combined with vitamin C treatment, for GVHD and autoimmune diseases. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document