Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • Log in

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • Log in
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

c-Rel Is Required for Chromatin Remodeling Across the IL-2 Gene Promoter

Sudha Rao, Steve Gerondakis, Donna Woltring and M. Frances Shannon
J Immunol April 1, 2003, 170 (7) 3724-3731; DOI: https://doi.org/10.4049/jimmunol.170.7.3724
Sudha Rao
*Division of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Steve Gerondakis
†Walter and Eliza Hall Institute, Parkville, Australia
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Donna Woltring
*Division of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
M. Frances Shannon
*Division of Molecular Bioscience, John Curtin School of Medical Research, Australian National University, Canberra, Australia; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

IL-2 gene transcription occurs in an activation-dependent manner in T cells responding to TCR and CD28 activation. One of the critical events leading to increased IL-2 transcription is an alteration in chromatin structure across the 300-bp promoter region of the gene. We initially showed that IL-2 gene transcription in CD4+ primary T cells is dependent on the NF-κB family member, c-Rel, but not RelA. We found that c-Rel is essential for global changes in chromatin structure across the 300-bp IL-2 promoter in response to CD3/CD28 in primary CD4+ T cells, but not in response to pharmacological signals, paralleling the requirement for c-Rel in IL-2 mRNA and protein accumulation. Interestingly, measurement of activation-induced localized accessibility changes using restriction enzyme digestion revealed that accessibility close to the c-Rel binding site in the CD28RR region of the promoter is specifically dependent on c-Rel. In contrast, restriction enzyme sites located at a distance from the CD28RR behave independently of c-Rel. These results suggest a nonredundant role for c-Rel in generating a correctly remodeled chromatin state across the IL-2 promoter and imply that the strength of the signal determines the requirement for c-Rel.

The NF-κB family of transcription factors plays a central role in immune and inflammatory responses, serving to rapidly activate a wide variety of genes involved in host defense against pathogens (1, 2, 3, 4). The NF-κB family is composed of p50 (NF-κB1), p52 (NF-κB2), RelA (p65), RelB, and c-Rel, which share a common region known as the Rel homology domain and function as homo- or heterodimers binding decanucleotide elements in the promoters and enhancers of genes, including those encoding cytokines (3, 4, 5). Deletion of the genes for individual family members in mice has helped elucidate specific physiological roles for each protein; however, the molecular basis of these differences is not well understood (6, 7, 8).

The secretion of IL-2 by activated T cells is an important step in an immune response, driving autocrine T cell proliferation and regulating T cell homeostasis (9). The region of the IL-2 gene that responds to T cell activation has been mapped to a 300-bp region immediately upstream of the transcription start site (10, 11) and contains binding sites for numerous transcription factors (12). The IL-2 promoter contains two NF-κB binding sites. One is in the CD28 response region (CD28RR)2 that is referred to as the CD28 response element (CD28RE). This region is required for response to T cell costimulation through the CD28 receptor (13, 14). The CD28RR (−164 to −146 bp) also contains an AP-1 binding region that functions in a cooperative manner with the CD28RE (14). The NF-κB family members that bind to the CD28RE change in a time-dependent manner following T cell activation, with RelA homodimers being a major component at early time points, and c-Rel-containing complexes dominating at later time points, reflecting the need for new c-Rel synthesis (15). Analysis of c-rel−/− mice has demonstrated that c-Rel is essential for the induced expression of several cytokines in T cells (6, 16). A 50-fold decrease is observed in IL-2 protein production compared with that in wild-type (wt) cells when cells are stimulated via TCR/CD28 pathways, but there is no decrease in response to pharmacologic stimuli (6, 16).

The genomic DNA in the nucleus is organized into a highly complex structure known as chromatin, which is an important component in the control of gene transcription. One of the first events required for inducible gene transcription is the remodeling of chromatin across promoter and enhancer regions of the gene. Modification of chromatin structure is accomplished by the directed actions of two distinct groups of enzymes, namely ATP-dependent chromatin remodelers and proteins that can covalently modify N-terminal histone tails (reviewed in Refs. 17 and 18). Chromatin remodeling associated with gene activation can be measured by altered accessibility across specific regions using agents such as micrococcal nuclease (MNase) or restriction enzymes (RE) (19, 20, 21). Whereas RE accessibility can provide information on localized changes, MNase measures alterations at a more global level (22, 23). Recently, several laboratories have developed a novel approach to study chromatin accessibility events in vivo that uses real-time PCR (21, 24). This approach has several advantages over the more conventional use of Southern hybridization: the accuracy of quantification is improved; the resolution of the assay is improved by designing primers to amplify small amplicons; and a relatively small quantity of genomic DNA is required, with the assay being in the linear range with 1.5 ng of input DNA (21, 24). These features have previously allowed us to successfully monitor chromatin accessibility events in primary T cells following cellular activation (21).

There have been numerous studies showing that DNase I-hypersensitive sites appear across the IL-2 proximal promoter following T cell activation (19, 25, 26). Other studies have shown that a specific RE site located close to the CD28RR becomes accessible for digestion following activation of EL-4 T cells (19). These experiments suggest that the structure of chromatin across the proximal IL-2 promoter is remodeled following T cell activation. We have recently shown that the 300-bp region of the IL-2 proximal promoter becomes selectively remodeled, as measured by MNase and RE accessibility, upon T cell activation not only in EL-4 T cells, but also in primary murine CD4+ T cells (21). This remodeling occurs late (1.5 h) relative to the translocation of many transcription factors to the nucleus, which generally occurs within 10–30 min of activation (21). This delay is not due to the need for prior histone acetylation, nor does it appear to be dependent on the histone kinase Msk1 (21). The roles of specific transcription factors in these events have not, however, been investigated. Here we show that c-Rel is essential for the correct pattern of chromatin remodeling across the IL-2 promoter following primary T cell activation with anti-CD3 and anti-CD28 Abs. This requirement is stimulus specific, however, and can be overcome with the pharmacological agents, PMA and calcium ionophore (I). In contrast to c-Rel, RelA is not essential for IL-2 gene transcription, pointing to a distinct role for these NF-κB proteins in immune cell activation.

Materials and Methods

Cell culture

EL-4.E1.F4 (EL-4) thymoma cells were maintained in RPMI 1640 medium supplemented with 10% FCS, 2 mM l-glutamine, 50 μM 2-ME, and antibiotics. Cells in suspension were stimulated for an optimal period of 4 h at 1.25 × 106 cells/ml with PMA (Roche, Indianapolis, IN), calcium ionophore A23187 (I; Sigma-Aldrich, St. Louis, MO) and an activating CD28 Ab (BD PharMingen, San Diego, CA) at final concentrations of 10 ng/ml, 1 μM, and 5 μg/ml, respectively.

Primary T cell preparation

All mice were maintained in a pathogen-free environment in a barrier facility. Spleens were isolated from C57BL/6 mice (4–5 wk old), c-rel−/− mice (4–5 wk old), and relA−/− mice (obtained by fetal liver reconstitution of irradiated hosts as described previously (27)), and CD4+ T cells were purified using MACS CD4+ (LT34) beads according to the manufacturer’s guidelines (Miltenyi Biotec, Auburn, CA). The cells were subsequently stained and analyzed by flow cytometry, with T cell populations shown to be ∼90% pure using Abs against CD4+ T cells, CD8+ T cells, B cells, and macrophages. Stimulations were performed as described above for EL-4 T cells. Anti-CD3 stimulation was performed by coating plates with 10 μg/ml anti-CD3 Ab (BD PharMingen) at 37°C for 3 h or overnight at 4°C. The plates were subsequently washed (four or five times) with PBS before the addition of cells at a density of 1.25 × 106/ml.

RNA isolation and real-time PCR analysis

Total RNA was extracted from stimulated and unstimulated T cells (5 × 106 cells) using the modified RNAzol B methodology described previously (28). RNA (1 μg) was subsequently treated with DNase I (1 U/μg RNA) reverse transcribed using 100 U of Superscript II reverse transcriptase (Life Technologies, Gaithersburg, MD) as detailed in the manufacturer’s guidelines. SYBR Green real-time PCR reactions were performed on an ABI PRISM 7700 sequence detector (PerkinElmer/PE Applied Biosytems, Foster City, CA) as described previously (21) with 10 ng of cDNA in a total volume of 25 μl, according to the manufacturer’s guidelines (PerkinElmer/PE Applied Biosytems; protocol 04304965). An aliquot of each sample was also analyzed by quantitative PCR for GAPDH to normalize for inefficiencies in cDNA synthesis and RNA input amounts. The primer sets used are detailed in Table II⇓. To correlate the threshold (Ct) values from the amplification plots to copy number, a standard curve was generated using the appropriate plasmid.

Chromatin accessibility by real-time PCR (CHART-PCR) assay

Accessibility to digestion with REs and MNase was analyzed using the CHART-PCR assay described previously (21). Briefly, stimulated and unstimulated T cells (5 × 106 cells/sample) were pelleted by centrifugation at 500 × g, washed in ice-cold PBS, resuspended in 1 ml of ice-cold Nonidet P-40 lysis buffer (10 mM Tris (pH 7.4), 10 mM NaCl, 3 mM MgCl2, 0.5% Nonidet P-40, 0.15 mM spermine, and 0.5 mM spermidine), and incubated on ice for 5 min. The suspension was centrifuged at 3000 rpm for 5 min to pellet the nuclei. The nuclei were subsequently washed in the respective digestion buffer (without CaCl2) and then resuspended at 5 × 106 nuclei/100 μl. MNase and RE accessibility assays were performed essentially as previously described (20). One hundred-microliter suspensions of nuclei were then treated with 100 U of HinfI or MseI (Roche) at 37°C for 45 min or with 50 U of MNase (Roche) for 5 min at 20°C. A control without the accessibility agent was used to monitor endonuclease activity. The genomic DNA was subsequently isolated using a QIAamp blood kit (Qiagen, Chatsworth, CA). Fifty micrograms of genomic DNA was used to perform SYBR Green real-time PCR on the ABI PRISM 7700 sequence detector (PerkinElmer/PE Applied Biosystems) in a total volume of 25 μl using the SYBR Green PCR master mix (PerkinElmer/PE Applied Biosystems) according to the manufacturer’s guidelines. The primer sets used are detailed in Table II⇓. To convert the Ct values from the CHART-PCR amplification plots to percent accessibility, a standard curve was generated with genomic DNA. PCR products were visualized on a 4% Nuseive 3/1 agarose gel (PerkinElmer/PE Applied Biosytems) to ensure that a single product was generated in the PCR. All samples were subjected to real-time PCR analysis using Primer Set-F to ensure equal loading of DNA in all PCR reactions.

Western blotting analysis

Nuclear extracts were prepared by a modification of the method described by Schreiber et al. (29). Unstimulated and stimulated EL4 T cells (3 × 107/sample) were centrifuged for 5 min at 500 × g, washed with ice-cold PBS, then resuspended in 1 ml of ice-cold buffer A (10 mM Tris (pH 7.4), 10 mM NaCl, 3 mM MgCl2, 0.1 mM EDTA, 0.5% Nonidet P-40, and protease inhibitors) per 1 × 107 cells, and incubated on ice for 5 min. Nuclei were recovered by centrifugation at 900 × g for 5 min, washed in buffer A without Nonidet P-40, then resuspended in 75 μl of buffer C (400 mM NaCl, 7.5 mM MgCl2, 0.2 mM EDTA, 0.1 mM EGTA, 1 mM DTT, and protease inhibitors) per 3 × 107 cells, and incubated in ice with shaking for 15 min. Nuclear debris was removed by centrifugation at 4°C for 15 min. Protein concentrations were determined by the Bradford assay (Bio-Rad, Hercules, CA). Proteins (10 μg) were resolved by SDS-PAGE, transferred to nitrocellulose, and subjected to Western blotting analysis using anti-c-Rel, anti-RelA, and anti-Sp1 (all from Santa Cruz Biotechnology, Santa Cruz, CA) Abs. Proteins were detected using Super Signal chemiluminescent substrate (Pierce, Rockford, IL), visualized using a luminescent analyzer (Las-1000 Plus; Fuji, Tokyo, Japan), and quantified using the Fuji Image Gauge software.

Results

IL-2 transcription in T cells is c-Rel, but not RelA, dependent

The CD28RE within the proximal promoter of the IL-2 gene binds NF-κB family members c-Rel and RelA in extracts from activated T cells (26), and IL-2 protein levels are dramatically reduced in CD3/CD28-activated T cells from c-rel−/− mice (6, 16). To compare the importance of c-Rel and RelA in IL-2 gene transcription, we performed quantitative PCR analysis of IL-2 mRNA levels in CD4+ T cells, treated with CD3/CD28 for 4 h, from wt, c-rel−/−, or relA−/− (obtained by fetal liver reconstitution of irradiated hosts) mice. IL-2 mRNA levels were reduced by 18-fold in c-rel−/− T cells, but were unaffected in relA−/− T cells (Table I⇓). The same results were obtained at several different times of stimulation (data not shown). In contrast to the effect observed with CD3/CD28 stimulation, the absence of c-Rel had no effect on the response of the IL-2 gene to PMA, I, and anti-CD28 (PMA/I/CD28) treatment (Table I⇓).

View this table:
  • View inline
  • View popup
Table I.

Comparison of IL-2 mRNA in wt vs c-rel−/− or RelA−/− CD4+ T cells

To further support the finding that the requirement for c-Rel is stimulus dependent, we examined the effect of the c-Rel inhibitor, pentoxyfilline, on IL-2 transcription in CD4+ T cells isolated from wt mice stimulated with PMA/I/CD28 or CD3/CD28. Pentoxyfilline has previously been shown to specifically inhibit the translocation of c-Rel, but not the other NF-κB family members or NFAT, to the nucleus (30). Consistent with data from c-rel−/− mice, pentoxyfilline inhibited CD3/CD28-mediated, but not PMA/I/CD28-mediated, IL-2 transcription (Table I⇑). Thus, c-Rel appears to be essential for IL-2 transcription in response to CD3/CD28, but dispensable in the presence of pharmacological activation, while RelA is not essential for IL-2 gene transcription.

Kinetics of c-Rel accumulation in EL-4 T cells

The initiation of gene transcription consists of several events, including alteration of chromatin structure, assembly of transcription activation complexes, and recruitment of basal transcriptional machinery (18, 31, 32), and c-Rel could be involved in one or several of these events. We wanted to determine whether c-Rel contributed to chromatin remodeling events on the IL-2 promoter or was part of the activation complex that assembled on the remodeled promoter. We first examined the time course of c-Rel mRNA and protein accumulation in EL-4 T cells to ascertain whether c-Rel was present in cells, and more specifically in the nucleus, at the time of chromatin remodeling on the IL-2 promoter. In contrast to RelA or NF-κB1/p50, little c-Rel pre-exists in the resting T cell cytoplasm, and new synthesis is required for significant nuclear accumulation (33). Significant levels of c-Rel mRNA were detected as early as 15 min following PMA/I activation of EL-4 T cells, with maximum levels observed at 2 h (Fig. 1⇓a). Thereafter, levels dropped slightly, but were then maintained at relatively high levels up to 16 h postactivation (Fig. 1⇓a). c-Rel protein began accumulating in the nucleus at 30 min following stimulation; higher levels were observed from 1 h poststimulation and were sustained until at least 4.5 h following activation (Fig. 1⇓b). These results show that c-Rel accumulates in the nucleus before the time that IL-2 promoter chromatin remodeling is initiated (1.5 h; Fig. 1⇓a) (21) in EL4 T cells, which, in turn, precedes the accumulation of IL-2 mRNA (Fig. 1⇓a). Similar kinetics of c-Rel accumulation, IL-2 mRNA production, and promoter remodeling were observed in primary CD4+ T cells (data not shown).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

The accumulation of nuclear c-Rel correlates with changes in IL-2 chromatin accessibility. a, Comparison of IL-2 chromatin remodeling kinetics, as measured by MNase digestion, with c-Rel and IL-2 mRNA kinetics measured by SYBR Green real-time PCR. IL-2 and c-Rel mRNA levels are plotted as fold change relative to the unstimulated samples. Remodeling is plotted as a percentage of the unstimulated, MNase-digested genomic DNA sample. Data shown are the mean and SE of three replicate experiments. b, Nuclear extracts were prepared from EL-4 T cells, either unstimulated (NS) or stimulated with PMA/I for 0.5–4.5 h. Ten micrograms of nuclear extract from each time point was resolved on SDS-PAGE and subjected to Western blotting analysis using anti-c-Rel (upper panel) and anti-Sp1 (lower panel) Abs.

c-Rel accumulates in the nucleus much later than RelA or many other transcription factors thought to be involved in IL-2 gene transcription (15, 33) and appears to closely precede promoter chromatin remodeling. The time frame of c-Rel accumulation and chromatin remodeling suggests that new protein synthesis may be required for these events. c-Rel, but not RelA, accumulation in the nuclei of EL-4 T cells was indeed dependent upon new protein synthesis (Fig. 2⇓a), in agreement with previous data (34). A small amount of c-Rel was detected in cycloheximide (CHX)-treated nuclear extracts at all time points examined and may represent a small pool of pre-existing c-Rel in the EL-4 cells (Fig. 2⇓a). In contrast to the inhibition of c-Rel by pretreatment with CHX, nuclear translocation and maintenance of c-Rel were not affected by addition of CHX 2 h after stimulation with PMA/I (Fig. 2⇓b). Constitutively expressed Sp1 was used to monitor equal protein loading in all lanes.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Chromatin remodeling across the IL-2 gene promoter is dependent on protein synthesis. a, Nuclear extracts were prepared from EL-4 T cells stimulated with PMA/I for the indicated times with or without pretreatment with CHX for 30 min. Ten micrograms of nuclear extract from each time point was resolved on SDS-PAGE and subjected to Western blotting analysis using anti-c-Rel (upper panel), anti-Sp1 (middle panel), and anti-RelA (lower panel) Abs. b, Nuclear extracts were prepared from EL-4 T cells unstimulated (lane 1), stimulated with PMA/I alone for 1.5 h (lane 2) or 4 h (lane 3), and unstimulated or stimulated with PMA/I for 2 h before the addition of CHX for an additional 2 h (lanes 4 and 5). Ten micrograms of nuclear extract from each time point were resolved on SDS-PAGE and subjected to Western blotting analysis using anti-c-Rel (upper panel) and anti-Sp1 (lower panel) Abs. The immunoreactive bands were visualized using a Fuji-Film FLA-1000 phosphorimager. c, Nuclei for MNase digestion or RNA for IL-2 expression analysis were prepared from EL-4 T cells pretreated with CHX for 30 min and left unstimulated (NS), stimulated with PMA/I for 4 h, pretreated with CHX for 30 min before stimulation with PMA/I (+CHX), and stimulated with PMA/I for 2 h before the addition of CHX (+CHX2). The intact nuclei were incubated with or without 50 U of MNase for 5 min at room temperature. Genomic DNA (50 ng) for each treatment was subjected to real-time PCR analysis using primer sets B–D. The Ct values generated were converted to DNA concentration and percent accessibility using a standard curve. The mean and SE of three replicate experiments are shown. IL-2 mRNA levels were measured by SYBR Green real-time PCR analysis. The fold change in IL-2 mRNA levels for each of the treatments is shown relative to the control value.

Significant amounts of c-Rel, therefore, accumulate in the nucleus following 1 h of activation, and this accumulation is driven by de novo synthesis. c-Rel appears to be relatively stable in the nucleus even in the presence of protein synthesis inhibition.

IL-2 promoter remodeling is dependent on new protein synthesis

We next asked whether chromatin remodeling across the IL-2 promoter was dependent on new protein synthesis. To investigate changes in chromatin structure associated with IL-2 gene transcription, MNase accessibility across the promoter was measured using a quantitative CHART-PCR (21), where the amount of PCR product is inversely proportional to the extent of MNase digestion occurring across the amplified region (21). The requirement for de novo protein synthesis for chromatin remodeling of the IL-2 promoter was determined by CHART-PCR using three primer sets spanning the 300-bp IL-2 promoter (Table II⇓ and Fig. 4⇓a). As we have previously shown (21), the IL-2 promoter is completely inaccessible to MNase digestion in resting EL-4 T cells, and stimulation with PMA/I resulted in 80–85% accessibility across the 300-bp promoter region (Fig. 2⇑c). Pre-treatment of EL-4 T cells with CHX 30 min before PMA/I activation inhibited accessibility across the entire IL-2 promoter (Fig. 2⇑c) and was paralleled by an inhibition of IL-2 mRNA accumulation as measured by quantitative PCR (Fig. 2⇑c).

View this table:
  • View inline
  • View popup
Table II.

Primer sequences for SYBR Green real-time PCR

To determine whether protein synthesis was required for maintenance of the remodeled state, EL-4 T cells were treated with CHX following 2-h stimulation with PMA/I (at a time when chromatin remodeling has already occurred), and remodeling was assayed 2 h later. The IL-2 proximal promoter was still accessible to MNase under these conditions (Fig. 2⇑c), and in parallel, IL-2 transcript levels were not inhibited (Fig. 2⇑c). These results show that new protein synthesis is required for the efficient initiation, but not maintenance, of chromatin remodeling across the IL-2 promoter, implicating newly synthesized proteins, such as c-Rel, in this process.

Characterization of IL-2 promoter chromatin remodeling in c-rel−/− CD4+ T cells

To explore the possibility that c-Rel is required for chromatin remodeling across the IL-2 promoter, chromatin accessibility was examined in CD4+ T cells isolated from wt and c-rel−/− mice. As we have previously shown (21), activation of primary CD4+ T cells with CD3/CD28 leads to an increase in MNase accessibility across the IL-2 promoter (Fig. 3⇓a). In contrast, there was little or no increase in MNase accessibility in CD3/CD28-activated CD4+ T cells from c-rel−/− mice, with levels of accessibility being identical with those of unstimulated cells (Fig. 3⇓a). This lack of chromatin accessibility is in agreement with the decrease in IL-2 mRNA levels seen in c-rel−/− T cells (Table I⇑). However, when c-rel−/− T cells were treated with PMA/I/CD28, almost normal levels of MNase accessibility were observed in parallel with normal levels of IL-2 mRNA accumulation (Fig. 3⇓a and Table I⇑).

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Chromatin accessibility across the IL-2 promoter is inhibited in rel−/− primary T cells activated with CD3/CD28. a, Nuclei for MNase analysis were prepared from either wt (W) or c-rel−/− (R) CD4+ T cells that were left unstimulated or stimulated with PMA/I/CD28 or CD3/CD28 for 4 h. The intact nuclei were incubated with or without 50 U of MNase for 5 min at room temperature. Genomic DNA (50 ng) for each treatment was subjected to real-time PCR analysis using primer sets B–D. The Ct values generated were converted to DNA concentration using a standard curve, and MNase accessibility was expressed as a percentage of the unstimulated, digested genomic DNA sample. b, CD4+ T cells were either untreated or pretreated with pentoxyfilline for 30 min and then left unstimulated or stimulated with CD3/CD28 for 4 h. Intact nuclei were isolated and incubated with 50 U of MNase for 5 min at room temperature, and the amount of MNase accessibility was measured and calculated as described in a.

Since the c-Rel inhibitor, pentoxyfilline, blocks CD3/CD28, but not PMA/I/CD28, activation of IL-2 in primary T cells (Table I⇑), we asked whether pentoxyfilline also inhibited chromatin remodeling across the IL-2 promoter in response to CD3/CD28 stimulation. Pentoxyfilline pretreatment partially inhibited MNase accessibility across the promoter in CD3/CD28-activated cells (Fig. 3⇑b), with the largest effect seen across the region encompassing the binding site for c-Rel at the CD28RE (Fig. 3⇑b).

These results show that c-Rel is required for correct chromatin remodeling across the IL-2 promoter in response to CD3/CD28 activation, but this requirement can be overcome by pharmacological agents. In addition, our results illustrate that MNase accessibility closely parallels IL-2 mRNA accumulation.

RE accessibility is not uniform across the IL-2 promoter following CD3/CD28 activation

To gain insight into localized changes in chromatin structure across the IL-2 promoter, we examined RE accessibility, which can be influenced by localized chromatin structure, DNA conformation, and/or transcription factor binding (22, 23). Three MseI sites and two HinfI sites, chosen to measure accessibility at a range of sites across the promoter (Fig. 4⇓a), were examined using CHART-PCR (21). The majority of the RE sites became accessible to RE digestion in wt CD4+ T cells treated with CD3/CD28 or PMA/I/CD28 (Fig. 4⇓b). The one exception to this general accessibility was the HinfI site at −100, which became accessible in response to PMA/I/CD28, but not in response to CD3/CD28 (Fig. 4⇓b). These results indicate that while there is a general change in promoter architecture, as measured by MNase accessibility, individual promoter regions have distinct modes of accessibility.

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

RE accessibility across the IL-2 promoter in primary T cells. a, Schematic representation of the murine IL-2 proximal promoter from −300 bp to the transcriptional start site. The locations of the primer sets B–D used in the CHART-PCR assay are indicated in a, and the details of the primer sets are shown in Table II⇑. The positions of the MseI (MseI-B, MseI-C, MseI-D) and HinfI (HinfI-A, HinfI-B) RE sites are also shown. Functional control elements are also indicated (▩). The approximate position of the nucleosome across the CD28RE is indicated above the promoter. b, Nuclei for RE accessibility analysis were prepared from CD4+ T cells unstimulated or stimulated with PMA/I/CD28 and CD3/CD28. Nuclei were incubated with 100 U of MseI or HinfI at 37°C for 45 min. Genomic DNA (50 ng) from each sample was subjected to real-time PCR analysis using the appropriate primer set detailed in a. The Ct values were converted to DNA concentration using a standard curve, and accessibility was expressed as a percentage of the unstimulated, uncut DNA sample. The results represent the mean ± SE of three experiments from separate groups of mice.

The absence of c-Rel specifically affects RE accessibility at the CD28RE

To examine in more detail the requirement for c-Rel in remodeling the region around its binding site at the CD28RE, the accessibility of RE sites across this region were examined in c-rel−/− T cells. The accessibility observed at the HinfI-B site (−150) in wt cells in response to CD3/CD28 was inhibited in c-rel−/− cells (Fig. 5⇓a). In contrast, the HinfI-A site (−100) that was not accessible in CD3/CD28-activated wt cells became accessible in the activated c-rel−/− cells (Fig. 5⇓a). The same pattern of RE accessibility was observed in PMA/I/CD28-activated c-rel−/− cells, with a decrease in accessibility at sites located across the CD28RE (HinfI-B, Fig. 5⇓b; DraI, data not shown) and an increase at the −100 HinfI site (Fig. 5⇓b).

FIGURE 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 5.

The CD28RE region is less accessible to RE digestion in CD4+ T cells from c-rel−/− mice. Nuclei for RE analysis were prepared from wt or c-rel−/− murine CD4+ T cells unstimulated or stimulated with CD3/CD28 (a) and PMA/I/CD28 (b). Nuclei were incubated with 100 U of MseI or HinfI at 37°C for 45 min. Genomic DNA (50 ng) from each sample was subjected to real-time PCR analysis using the appropriate primer (Fig. 4⇑a). The Ct values were converted to DNA concentration using a standard curve, and accessibility was expressed as a percentage of the unstimulated, uncut DNA sample. The results represent the mean ± SE of three experiments from separate groups of mice.

To examine accessibility at RE sites located further from the c-Rel binding site, MseI sites at −40 and −250 were compared in wt and rel−/− T cells. In contrast to the decrease in accessibility observed at RE sites close to the CD28RE, activation-induced accessibility at these MseI sites was not decreased in CD3/CD28-activated c-rel−/− cells (Fig. 5⇑a). Similarly, access to these RE sites was not affected by the absence of c-Rel following PMA/I/CD28 activation (Fig. 5⇑b).

These data indicate that while c-Rel is required for correct RE accessibility at sites close to the c-Rel binding site at the CD28RE, sites located at a distance behave in a distinct manner.

Discussion

We have shown here that c-Rel, but not RelA, is essential for correct chromatin disruption and IL-2 gene transcription in CD4+ primary T cells. The observation that RelA was not essential for IL-2 gene transcription was surprising given previous results showing that RelA accumulates in T cell nuclei following activation, binds to numerous NF-κB sites including the CD28REs of genes such as IL-2, and can trans-activate these promoter regions in transient transfection assays (12). These data clearly show that the NF-κB/Rel proteins play nonredundant roles in gene transcription in T cells.

We have presented several lines of evidence to shown here that c-Rel is required for a correctly altered chromatin state across the IL-2 proximal promoter in CD3/CD28-activated primary T cells. Firstly, MNase accessibility across the entire promoter is prevented in the absence of c-Rel. Secondly, RE accessibility close to the c-Rel binding site at the CD28RR is also dependent on the presence of c-Rel. Clearly, other NF-κB proteins are not sufficient to generate an accessible chromatin configuration across the promoter, since the expression and activity of the other NF-κB proteins are normal in c-rel−/− T cells (6, 16). In addition, we have shown here that RelA is dispensable for IL-2 transcription. Thirdly, the kinetics of c-Rel accumulation in the nucleus as well as its response to cycloheximide treatment closely correlate with chromatin remodeling kinetics on the IL-2 promoter. Thus, c-Rel may play a specific role in recruiting chromatin-modifying complexes to the promoter or may play a role in stabilizing a remodeled state.

It has recently been shown that c-Rel is not required for chromatin remodeling across the IL-12 p40 promoter in LPS-activated macrophages (35), although it is required for efficient gene transcription (35). It is not clear why these results differ from those presented here for IL-2, although c-Rel may have a cell- or gene-specific role in chromatin remodeling. Since we have shown that access to RE sites only in the region of the c-Rel binding site requires c-Rel, it should be noted that RE accessibility at a site distant from the c-Rel binding site in the IL-12 p40 promoter was used to show that c-Rel was not required (35).

It is intriguing that c-Rel is required for chromatin disruption and gene transcription in response to CD3/CD28, but not in response to PMA/I/CD28. Interestingly, our results are consistent with two separate studies showing that a mutation within the CD28RE resulted in the loss of responsiveness of the IL-2 promoter to Ag receptor stimulation (36), but had no effect on PMA/I inducibility (13). PMA/I/CD28 is clearly a stronger stimulus, leading to 100-fold more IL-2 mRNA that CD3/CD28. A strong signal such as PMA/I may generate threshold levels of other transcription factors, such as NFAT (36) and NF-MATp35 (37), which have previously been shown to interact with the IL-2 promoter, including the CD28RE, perhaps reducing the need for c-Rel. These results support a threshold model for chromatin remodeling as well as for gene transcription where the density of a specific set of factors, and not an individual factor per se, is the key to driving promoter function. The fact that CHX inhibits chromatin remodeling in response to P/I in EL-4 cells implies that in addition to c-Rel, CHX is inhibiting other factors involved in chromatin remodeling. The identity of such factors and whether they cooperate with c-Rel to allow chromatin remodeling across the IL-2 promoter will be important to determine.

It is unclear why pentoxyfilline treatment is less potent at inhibiting chromatin accessibility across the entire promoter than deletion of c-Rel from the genome, although significant inhibition was seen across the CD28RR region following pentoxyfilline treatment. Pentoxyfilline has previously been shown to inhibit c-Rel translocation into the nucleus (30), and we have shown here that it completely inhibits IL-2 mRNA accumulation in response to CD3/CD28 activation. It is possible that c-Rel has direct as well as indirect effects on chromatin remodeling and that the latter are only seen when T cells develop in the absence of c-Rel. It is clear that the effects seen in either c-rel−/− cells or pentoxyfilline-treated cells are sufficient to inhibit gene transcription in response to CD3/CD28. In addition, pentoxyfilline may inhibit other aspects of IL-2 gene transcription to give complete abrogation of mRNA production.

RE accessibility has been equated with MNase or DNase I accessibility as a means of measuring chromatin remodeling (19, 20, 35), but it is now becoming clear that these agents may be measuring different events. The data presented here imply that RE accessibility is measuring a specific local event that is affected by binding of transcription factors as well as chromatin remodeling events. This is particularly evident for the two HinfI sites at −100 and −150, which are most likely located within the same positioned nucleosome (38), but behave independently. We have previously shown that the human IL-2 promoter can assemble a highly positioned nucleosome in vitro and that a similarly positioned nucleosome is present in vivo (38). We have recently shown that the mouse promoter assembles a nucleosome in a similar position in EL-4 T cells spanning these two HinfI sites (S. Rao et al., unpublished observations). The independent behavior of RE site accessibility is consistent with several recently published findings showing that RE accessibility measures disruption of local chromatin structure as a consequence of transcription factor binding (22) and/or changes in the conformation of nucleosomes (39) without the loss or movement of any underlying nucleosomes.

Importantly, access to RE sites close to the c-Rel binding site at the CD28RE is dependent on the presence of c-Rel, while sites that are more distant are independent of c-Rel. The binding of other transcription factors, such as NFAT or AP-1, accessing their binding sites in the presence of chromatin, could be creating RE accessibility to these more distant sites. In contrast, MNase may be measuring dramatic unfolding of a higher order structure or complete disruption of nucleosomes across the promoter, events that are dependent on c-Rel. It is important to note that MNase accessibility correlates with IL-2 mRNA accumulation, whereas RE accessibility at specific sites is not indicative of the level of gene transcription. However, we need to be aware that current approaches to measure changes in chromatin structure, using nucleases and REs, do not provide detailed information on the nature of the chromatin alterations upon cell activation. It is important to realize that accessibility changes can be attributed to many mechanisms, including nucleosome sliding, displacement/replacement of activators and/or repressors, and conformational changes/alterations in the fluidity of nucleosome structure as a consequence of histone modifications such as acetylation or methylation. Thus, it will be important to determine more precisely the nature of the events measured by the different accessibility agents to gain a better understanding of the molecular processes involved in chromatin remodeling.

It is interesting that the HinfI site at −100 is inaccessible in CD3/CD28-activated T cells, while all other sites tested become accessible. This site is close to a negative regulatory region, the negative regulatory element-A (NRE) site that binds a zinc finger, E box-binding repressor, ZEB (40, 41), which is a potent repressor of IL-2 transcription. Yasui et al. (41) have shown that PMA/I stimulation reduces NRE-1 binding activity in IL-2-producing T cells by 40%. Thus, the accessibility observed at this site upon PMA/I/CD28 stimulation in CD4+ T cells could be a consequence of reduced NRE-1 binding in stimulated cells. The lack of accessibility at this site in CD3/CD28-activated cells suggests that a weaker stimulus may not provide an appropriate signal to remove a negative regulatory complex from this site. This may account in part for the 100-fold lower level of IL-2 mRNA seen in CD3/CD28 compared with PMA/I/CD28-activated T cells. Activation of c-rel−/− T cells resulted in increased accessibility at the NRE-A binding site (HinfI-A) compared with that seen in wt T cells. This suggests that the absence of c-Rel relieves repression at the NRE-1 site on the IL-2 promoter, perhaps through a direct effect on the synthesis or activity of components of the repressor complex or, alternatively, through an effect on chromatin structure from the loss of c-Rel binding at the CD28RE.

It is clear from the work presented here that for CD3/CD28 activation, c-Rel is required to generate an accessible chromatin configuration across the entire promoter, as measured by MNase accessibility, or at the CD28RR, as measured by RE accessibility. Since the strength of the stimulus appears to determine the need for c-Rel, these findings have implications in understanding how T cells respond to different Ag doses or Ags of different affinities. In addition, c-Rel and RelA do not have overlapping roles in IL-2 gene transcription, and it will be interesting to determine whether they control distinct sets of genes during T cell activation.

Acknowledgments

We are grateful to Adele Holloway and other members of the Shannon laboratory for helpful discussions, and to David Tremethick for discussions and comments on the manuscript.

Footnotes

  • ↵1 Address correspondence and reprint requests to Dr. M. Frances Shannon, John Curtin School of Medical Research, P.O. Box 334, Canberra, Australian Capital Territory 2601, Australia. E-mail address: frances.shannon{at}anu.edu.au

  • 2 Abbreviations used in this paper: CD28RR, CD28 response region; CD28RE, CD28 response element; CHART-PCR, chromatin accessibility by real-time PCR; CHX, cycloheximide; Ct, threshold; I, Ca2+ ionophore A23187; MNase, micrococcal nuclease; RE, restriction enzymes; wt, wild type; NRE, negative regulatory element.

  • Received December 11, 2002.
  • Accepted January 17, 2003.
  • Copyright © 2003 by The American Association of Immunologists

References

  1. ↵
    Baldwin, A. S., Jr.. 1996. The NF-κB and IκB proteins: new discoveries and insights. Annu. Rev. Immunol. 14: 649
    OpenUrlCrossRefPubMed
  2. ↵
    Baeuerle, P. A., T. Henkel. 1994. Function and activation of NF-κB in the immune system. Annu. Rev. Immunol. 12: 141
    OpenUrlCrossRefPubMed
  3. ↵
    Verma, I. M., J. K. Stevenson, E. M. Schwarz, D. Van Antwerp, S. Miyamoto. 1995. Rel/NF-κB/IκB family: intimate tales of association and dissociation. Genes Dev. 9: 2723
    OpenUrlFREE Full Text
  4. ↵
    Serfling, E., A. Avots, M. Neumann. 1995. The architecture of the interleukin-2 promoter: a reflection of T lymphocyte activation. Biochim. Biophys. Acta 1263: 181
    OpenUrlCrossRefPubMed
  5. ↵
    Bull, P., T. Hunter, I. M. Verma. 1989. Transcriptional induction of the murine c-rel gene with serum and phorbol-12-myristate-13-acetate in fibroblasts. Mol. Cell. Biol. 9: 5239
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Kontgen, F., R. J. Grumont, A. Strasser, D. Metcalf, R. Li, D. Tarlinton, S. Gerondakis. 1995. Mice lacking the c-rel proto-oncogene exhibit defects in lymphocyte proliferation, humoral immunity, and interleukin-2 expression. Genes Dev. 9: 1965
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Sha, W. C., H. C. Liou, E. I. Tuomanen, D. Baltimore. 1995. Targeted disruption of the p50 subunit of NF-κB leads to multifocal defects in immune responses. Cell 80: 321
    OpenUrlCrossRefPubMed
  8. ↵
    Weih, F., D. Carrasco, S. K. Durham, D. S. Barton, C. A. Rizzo, R. P. Ryseck, S. A. Lira, R. Bravo. 1995. Multiorgan inflammation and hematopoietic abnormalities in mice with a targeted disruption of RelB, a member of the NF-κB/Rel family. Cell 80: 331
    OpenUrlCrossRefPubMed
  9. ↵
    Reiner, S. L., R. A. Seder. 1995. T helper cell differentiation in immune response. Curr. Opin. Immunol. 7: 360
    OpenUrlCrossRefPubMed
  10. ↵
    Ullman, K. S., J. P. Northrop, C. L. Verweij, G. R. Crabtree. 1990. Transmission of signals from the T lymphocyte antigen receptor to the genes responsible for cell proliferation and immune function: the missing link. Annu. Rev. Immunol. 8: 421
    OpenUrlCrossRefPubMed
  11. ↵
    Crabtree, G. R., N. A. Clipstone. 1994. Signal transmission between the plasma membrane and nucleus of T lymphocytes. Annu. Rev. Biochem. 63: 1045
    OpenUrlCrossRefPubMed
  12. ↵
    Jain, J., C. Loh, A. Rao. 1995. Transcriptional regulation of the IL-2 gene. Curr. Opin. Immunol. 7: 333
    OpenUrlCrossRefPubMed
  13. ↵
    Fraser, J. D., B. A. Irving, G. R. Crabtree, A. Weiss. 1991. Regulation of interleukin-2 gene enhancer activity by the T cell accessory molecule CD28. Science 251: 313
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Shapiro, V. S., K. E. Truitt, J. B. Imboden, A. Weiss. 1997. CD28 mediates transcriptional upregulation of the interleukin-2 (IL-2) promoter through a composite element containing the CD28RE and NF-IL-2B AP-1 sites. Mol. Cell. Biol. 17: 4051
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Himes, S. R., L. S. Coles, R. Reeves, M. F. Shannon. 1996. High mobility group protein I(Y) is required for function and for c-Rel binding to CD28 response elements within the GM-CSF and IL-2 promoters. Immunity 5: 479
    OpenUrlCrossRefPubMed
  16. ↵
    Liou, H. C., Z. Jin, J. Tumang, S. Andjelic, K. A. Smith, M. L. Liou. 1999. c-Rel is crucial for lymphocyte proliferation but dispensable for T cell effector function. Int. Immunol. 11: 361
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Hassan, A. H., K. E. Neely, M. Vignali, J. C. Reese, J. L. Workman. 2001. Promoter targeting of chromatin-modifying complexes. Front. Biosci. 6: 1054
    OpenUrlCrossRef
  18. ↵
    Workman, J. L., R. E. Kingston. 1998. Alteration of nucleosome structure as a mechanism of transcriptional regulation. Annu. Rev. Biochem. 67: 545
    OpenUrlCrossRefPubMed
  19. ↵
    Ward, S. B., G. Hernandez-Hoyos, F. Chen, M. Waterman, R. Reeves, E. V. Rothenberg. 1998. Chromatin remodeling of the interleukin-2 gene: distinct alterations in the proximal versus distal enhancer regions. Nucleic Acids Res. 26: 2923
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Weinmann, A. S., S. E. Plevy, S. T. Smale. 1999. Rapid and selective remodeling of a positioned nucleosome during the induction of IL-12 p40 transcription. Immunity 11: 665
    OpenUrlCrossRefPubMed
  21. ↵
    Rao, S., E. Procko, M. F. Shannon. 2001. Chromatin remodeling, measured by a novel real-time polymerase chain reaction assay, across the proximal promoter region of the IL-2 gene. J. Immunol. 167: 4494
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Cirillo, L. A., F. R. Lin, I. Cuesta, D. Friedman, M. Jarnik, K. S. Zaret. 2002. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell. 9: 209
    OpenUrlCrossRefPubMed
  23. ↵
    Soutoglou, E., I. Talianidis. 2002. Coordination of PIC assembly and chromatin remodeling during differentiation-induced gene activation. Science 295: 1847
    OpenUrlAbstract/FREE Full Text
  24. ↵
    McGuire, K. L., M. Iacobelli. 1997. Involvement of Rel, Fos, and Jun proteins in binding activity to the IL-2 promoter CD28 response element/AP-1 sequence in human T cells. J. Immunol. 159: 1319
    OpenUrlAbstract
  25. ↵
    Siebenlist, U., D. B. Durand, P. Bressler, N. J. Holbrook, C. A. Norris, M. Kamoun, J. A. Kant, G. R. Crabtree. 1986. Promoter region of interleukin-2 gene undergoes chromatin structure changes and confers inducibility on chloramphenicol acetyltransferase gene during activation of T cells. Mol. Cell. Biol. 6: 3042
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Ghosh, P., T. H. Tan, N. R. Rice, A. Sica, H. A. Young. 1993. The interleukin 2 CD28-responsive complex contains at least three members of the NFκB family: c-Rel, p50, and p65. Proc. Natl. Acad. Sci. USA 90: 1696
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Grossmann, M., L. A. O’Reilly, R. Gugasyan, A. Strasser, J. M. Adams, S. Gerondakis. 2000. The anti-apoptotic activities of Rel and RelA required during B-cell maturation involve the regulation of Bcl-2 expression. EMBO J. 19: 6351
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Gruffat, D., C. Piot, D. Durand, D. Bauchart. 1996. Comparison of four methods for isolating large mRNA: apolipoprotein B mRNA in bovine and rat livers. Anal. Biochem. 242: 77
    OpenUrlCrossRefPubMed
  29. ↵
    Schreiber, E., P. Matthias, M. M. Muller, W. Schaffner. 1989. Rapid detection of octamer binding proteins with ‘mini-extracts,’ prepared from a small number of cells. Nucleic Acids Res. 17: 6419
    OpenUrlFREE Full Text
  30. ↵
    Wang, W., W. F. Tam, C. C. Hughes, S. Rath, R. Sen. 1997. c-Rel is a target of pentoxyfilline-mediated inhibition of T lymphocyte activation. Immunity 6: 165
    OpenUrlCrossRefPubMed
  31. ↵
    Aalfs, J. D., R. E. Kingston. 2000. What does ‘chromatin remodeling’ mean?. Trends Biochem Sci 25: 548
    OpenUrlCrossRefPubMed
  32. ↵
    Agalioti, T., S. Lomvardas, B. Parekh, J. Yie, T. Maniatis, D. Thanos. 2000. Ordered recruitment of chromatin modifying and general transcription factors to the IFN-β promoter. Cell 103: 667
    OpenUrlCrossRefPubMed
  33. ↵
    Bryan, R. G., Y. Li, J. H. Lai, M. Van, N. R. Rice, R. R. Rich, T. H. Tan. 1994. Effect of CD28 signal transduction on c-Rel in human peripheral blood T cells. Mol. Cell. Biol. 14: 7933
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Venkataraman, L., S. J. Burakoff, R. Sen. 1995. FK506 inhibits antigen receptor-mediated induction of c-rel in B and T lymphoid cells. J. Exp. Med. 181: 1091
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Weinmann, A. S., D. M. Mitchell, S. Sanjabi, M. N. Bradley, A. Hoffmann, H. C. Liou, S. T. Smale. 2001. Nucleosome remodeling at the IL-12 p40 promoter is a TLR-dependent, Rel-independent event. Nat. Immunol. 2: 51
    OpenUrlCrossRefPubMed
  36. ↵
    Rooney, J. W., Y. L. Sun, L. H. Glimcher, T. Hoey. 1995. Novel NFAT sites that mediate activation of the interleukin-2 promoter in response to T-cell receptor stimulation. Mol. Cell. Biol. 15: 6299
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Civil, A., A. Bakker, I. Rensink, S. Doerre, L. A. Aarden, C. L. Verweij. 1996. Nuclear appearance of a factor that binds the CD28 response element within the interleukin-2 enhancer correlates with interleukin-2 production. J. Biol. Chem. 271: 8321
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Attema, J. L., R. Reeves, V. Murray, I. Levichkin, M. D. Temple, D. J. Tremethick, M. F. Shannon. 2002. The human IL-2 gene promoter can assemble a positioned nucleosome that becomes remodeled upon T cell activation. J. Immunol. 169: 2466
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Soutoglou, E. T. I.. 2002. Coordination of PIC assembly and chromatin remodeling during differentiation-induced gene activation. Science 295: 1847
    OpenUrlAbstract/FREE Full Text
  40. ↵
    Brabletz, T., A. Jung, F. Hlubek, C. Lohberg, J. Meiler, U. Suchy, T. Kirchner. 1999. Negative regulation of CD4 expression in T cells by the transcriptional repressor ZEB. Int. Immunol. 11: 1701
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Yasui, D. H., T. Genetta, T. Kadesch, T. M. Williams, S. L. Swain, L. V. Tsui, B. T. Huber. 1998. Transcriptional repression of the IL-2 gene in Th cells by ZEB. J. Immunol. 160: 4433
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 170 (7)
The Journal of Immunology
Vol. 170, Issue 7
1 Apr 2003
  • Table of Contents
  • About the Cover
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
c-Rel Is Required for Chromatin Remodeling Across the IL-2 Gene Promoter
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
c-Rel Is Required for Chromatin Remodeling Across the IL-2 Gene Promoter
Sudha Rao, Steve Gerondakis, Donna Woltring, M. Frances Shannon
The Journal of Immunology April 1, 2003, 170 (7) 3724-3731; DOI: 10.4049/jimmunol.170.7.3724

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
c-Rel Is Required for Chromatin Remodeling Across the IL-2 Gene Promoter
Sudha Rao, Steve Gerondakis, Donna Woltring, M. Frances Shannon
The Journal of Immunology April 1, 2003, 170 (7) 3724-3731; DOI: 10.4049/jimmunol.170.7.3724
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Structure and Functional Characterization of a Humanized Anti-CCL20 Antibody following Exposure to Serum Reveals the Formation of Immune Complex That Leads to Toxicity
  • Loss of the Transfer RNA Wobble Uridine–Modifying Enzyme Elp3 Delays T Cell Cycle Entry and Impairs T Follicular Helper Cell Responses through Deregulation of Atf4
  • The Role of the HLA Class I α2 Helix in Determining Ligand Hierarchy for the Killer Cell Ig-like Receptor 3DL1
Show more MOLECULAR AND STRUCTURAL IMMUNOLOGY

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606