Skip to main content

Main menu

  • 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
  • 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

CpG Methylation of the IFNG Gene as a Mechanism to Induce Immunosupression in Tumor-Infiltrating Lymphocytes

Peter C. J. Janson, Per Marits, Magnus Thörn, Rolf Ohlsson and Ola Winqvist
J Immunol August 15, 2008, 181 (4) 2878-2886; DOI: https://doi.org/10.4049/jimmunol.181.4.2878
Peter C. J. Janson
*Department of Medicine, Clinical Allergy Research Unit, Karolinska Institutet, Stockholm, Sweden;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Per Marits
*Department of Medicine, Clinical Allergy Research Unit, Karolinska Institutet, Stockholm, Sweden;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Magnus Thörn
†Department of Surgery, South Stockholm General Hospital, Stockholm, Sweden; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Rolf Ohlsson
‡Department of Development and Genetics, Evolution Biology Centre, Uppsala University, Uppsala, Sweden
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ola Winqvist
*Department of Medicine, Clinical Allergy Research Unit, Karolinska Institutet, Stockholm, Sweden;
  • 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

The execution of appropriate gene expression patterns during immune responses is of eminent importance where CpG methylation has emerged as an essential mechanism for gene silencing. We have charted the methylation status of regulatory elements in the human IFNG gene encoding the signature cytokine of the Th1 response. Surprisingly, human naive CD4+ T lymphocytes displayed hypermethylation at the IFNG promoter region, which is in sharp contrast to the completely demethylated status of this region in mice. Th1 differentiation induced demethylation of the IFNG promoter and the upstream conserved nucleotide sequence 1 enhancer region, whereas Th2-differentiated lymphocytes remained hypermethylated. Furthermore, CD19+ B lymphocytes displayed hypomethylation at the IFNG promoter region with a similar pattern to Th1 effector cells. When investigating the methylation status among tumor-infiltrating CD4+ T lymphocytes from patients with colon cancer, we found that tumor-infiltrating lymphocytes cells are inappropriately hypermethylated, and thus not confined to the Th1 lineage. In contrast, CD4+ T cells from the tumor draining lymph node were significantly more demethylated than tumor-infiltrating lymphocytes. We conclude that there are obvious interspecies differences in the methylation status of the IFNG gene in naive CD4+ T lymphocytes, where Th1 commitment in human lymphocytes involves demethylation before IFNG expression. Finally, investigations of tumor-infiltrating lymphocytes and CD4+ cells from tumor draining lymph node demonstrate methylation of regulatory regions within key effector genes as an epigenetic mechanism of tumor-induced immunosupression.

The clonal expansion and differentiation of naive Th cells into effector subpopulations is a crucial part of the adaptive immune response against invading pathogens. Upon activation, naive Th cells differentiate into Th1 or Th2 phenotypes. These subpopulations are defined on the basis of the cytokines they secrete. Th1 cells secrete IFN-γ and lymphotoxin-α and are mediators of the cellular immunity and thus provide protection against intracellular pathogens and tumors, whereas Th2 cells produce IL-4, IL-5, and IL-13 and are necessary for the protection against extracellular microbes (1, 2). The cytokine milieu surrounding a naive Th cell upon Ag exposure affects the outcome and lineage decision of the immune response. IL-12 production from dendritic cells and IFN-γ from NK cells up-regulate Th1-associated transcription factors such as STAT4 and the T Box transcription factor T-bet (3). If the naive cell is exposed to IL-4 upon stimulation, other transcription factors like STAT6, GATA binding protein-3 (GATA-3),3 and c-Maf direct the naive Th cell into the Th2 pathway (4).

The appropriate program of gene expression in differentiated cells is governed by epigenetic processes like DNA methylation, histone modification, transcription factor binding, and higher order chromatin structure. Methylation of CpG dinucleotides is a widely accepted mechanism involved in cytokine suppression during Th cell differentiation (5). The methylation status of the Ifng promoter region during Th1 and Th2 differentiation has been investigated during the past years (6, 7, 8, 9, 10, 11, 12, 13) and the mechanisms leading to IFN-γ expression and silencing is, at least in mice, well known (11, 13). Reports of the methylation status in human naive and differentiated Th cells are however scarce and conflicting (8, 9, 12). This inconsistency may be due to different protocol usage for isolation and thus different purities in isolated cells, or due to interspecies differences in epigenetic regulation. To fully clarify this issue we have sought to investigate the methylation status of the human IFNG promoter as well as a conserved upstream enhancer (14, 15) during in vitro differentiation of human naive CD4+ cells into Th1 and Th2 populations.

As epigenetic involvement in immune regulation becomes increasingly accepted, attention is also focused on epigenetic alterations in disease. A role of IFNG promoter methylation in the pathogenesis of atopic syndrome has been suggested (12), which further emphasizes the importance of epigenetic balance in the development of Th1 and Th2 phenotypes. Considering the importance of IFN-γ in the defense against tumors, we determined Th1 commitment on the basis of DNA methylation among CD4+ tumor-infiltrating lymphocytes and CD4+ cells from tumor draining lymph node.

Materials and Methods

Cell purification and culture

Naive CD4+ T lymphocytes were isolated by magnetic sorting using a human naive CD4 T cell isolation kit (Miltenyi Biotec) and an AutoMACS Separator. Naive T cells were stimulated with plate-bound anti-CD3 (OKT-3). Th1 skewing cultures contained 5 ng/ml recombinant human IL-12 (cat. no. 407711; Calbiochem) and 5 μg/ml neutralizing anti-IL-4 Ab (R&D Systems, cat. no. MAB204). Th2 skewing conditions included 5 ng/ml recombinant human IL-4 (Calbiochem, cat. no. 407635) and 5 μg/ml neutralizing anti-IFN-γ Ab (R&D Systems, cat. no. MAB285). Both Th1 and Th2 cultures contained 100 units/ml IL-2 and 1 μg/ml soluble anti-CD28 (BD Biosciences, cat. no. 555725). Cells were cultured in HyQ 1640-media (HyClone) with 10% human serum from human male AB plasma (Sigma-Aldrich, cat. no. H4522) and 1% penicillin-streptomycin solution (HyClone). Before DNA and RNA isolation, CD4+ T lymphocytes were further purified from culture either by magnetic sorting using human CD4 microbeads (Miltenyi Biotech) or by FACS in the case of naive T cell isolation. CD8+ and CD19+ lymphocytes were also purified through FACS sorting. All isolated cell populations were confirmed to have a purity of >95%. Tumors from six patients with colon cancer were cut and suspended using a loose fit glass homogenizer to obtain single cell suspensions. During surgery, tumor draining lymph nodes were identified as described previously (16). Tumor-infiltrating CD4+ T lymphocytes and CD4+ cells from tumor draining lymph node were extracted using a CD4 recognizing magnetically labeled Ab and columns according to manufacturer’s description (Miltenyi Biotec). The study was approved by the local ethical committee and informed consent was obtained by the patients.

Coculture with CCL-221

Naive CD4+ T lymphocytes were cultured in Th1 conditions as described above, washed, and put in coculture with the human colon carcinoma cell line CCL221. Th1 and CCL221 cells were grown separated by a cell culture insert with 1.0 μM in pore size (BD Biosciences, cat. no. 353102) in six-well tissue culture plates (BD Biosciences, cat. no. 353046). CCL221 cells were grown on the bottom of the wells and Th1 cells were grown in suspension in the cell culture inserts. After 5 days of coculture in HyQ 1640-media with 10% human serum and 1% penicillin-streptomycin solution, the CD4+ containing cell fraction was washed and restimulated with plate-bound anti-CD3 (OKT-3) and soluble anti-CD28 as described above. Cells were removed for RNA isolation before stimulation and after 6 h of stimulation.

5-Azacytidine treatment of tumor-infiltrating lymphocytes

Tumor-infiltrating CD4+ T cells were purified as previously described and cultured in HyQ 1640-media with 10% human serum and 1% penicillin-streptomycin solution. The cells were stimulated with anti-CD3 anti-CD28 coated beads (Invitrogen, cat. no. 111.31D) in a cell:bead ratio of 1:1. After 24 h, 5-Azacytidine (5-Aza, Sigma-Aldrich, cat. no. A-1287) was added to the culture at a concentration of 5 μM. After 48 h of culture in 5-Aza containing media the cells were washed and then cultured in 1640-media without 5-Aza for an additional 48 h before RNA isolation.

ELISA

Cells grown in Th1 and Th2 culture for 1 wk were washed and restimulated with plate-bound anti-CD3 and soluble anti-CD28 Abs. After 3 days in culture IFN-γ and IL-4, levels in supernatants were analyzed by ELISA (R&D Systems, DY285 and DY204) according to the manufacturer’s protocol.

RNA isolation and quantitative real-time PCR

RNA isolation was performed using TRIzol reagent (Invitrogen, cat. no. 15596–026). In brief, 100 ng RNA per sample was included in a reverse transcriptase reaction using an iScript cDNA Synthesis Kit according to the manufacturer’s protocol. Quantitative PCR was performed on an iCyclerIQ using 2X IQ SYBR Green Supermix. Cycle thresholds were obtained using iCycler IQ Optical System Software Version 3.1 from Bio-Rad. Expression levels were normalized to RNA polymerase II using the 2−ΔΔCt method. Primers were purchased from Cybergene AB (Table I⇓).

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

Primer sequencesa

DNA isolation and methylation analysis

Methylation analysis was conducted using bisulfite sequencing. Genomic DNA was isolated using DNeasy Blood & Tissue Kit (Qiagen) and bisulfite converted using EZ DNA Methylation Kit (Zymo Research) according to the manufacturer’s instruction. Primers shown in Table I⇑ were used in PCR reactions to amplify the IFNG promoter region and the conserved nucleotide sequence 1 (CNS1) region. PCR products were cloned into a pCR4-TOPO vector using a TOPO TA Cloning Kit (Invitrogen). DNA from individual bacterial clones was then included in a sequencing reaction with T3 and T7 primers using BigDye Terminator v1.1 Cycle Sequencing Kit (Applied Biosystems) according to manufacturer’s instruction. Samples were run on a 310 Genetic Analyzer (Applied Biosystems) and sequences were analyzed using ABI Prism Sequencing Analysis Software 3.7 (Applied Biosystems). As controls for validity of the bisulfite sequencing reaction M.Sss1 (New England Biolabs) treated DNA from a bacterial artificial chromosome (BAC RPCI11–444B24, Invitrogen) and unmodified BAC DNA were used.

CFSE labeling

Naive CD4+ T lymphocytes were resuspended in PBS containing 0.1 μM CFSE solution. Cells were incubated in CFSE solution for 5 min at 37°C. Thereafter, cells were washed in RPMI 1640 medium containing 10% FCS. Cell pellets were resuspended in medium containing FCS and incubated for 30 min. Finally, cells were washed once more before resuspension in medium for culture.

Combined bisulfite restriction enzyme analysis (COBRA)

Genomic DNA was isolated and bisulfite treated as previously described. The CNS1 region was amplified by PCR with primers directed toward the same target sequence as in the case of bisulfite sequencing except with a 6-FAM conjugated forward primer (Primer sequences shown in Table I⇑). PCR products were digested with MaeII in excess (New England Biolabs). Following a phenol chloroform extraction digested products were loaded onto a 310 Genetic Analyzer (Applied Biosystems) and fragment analysis was performed using Gene Scan v3.7 application software (Applied Biosystems).

Results

Methylation status of evolutionarily conserved regulatory elements in sorted cells

To identify evolutionarily conserved regulatory elements within the IFNG region we performed alignment using the VISTA tool between human and mouse DNA sequences. In agreement with previous reports (14, 15), we a found high sequence conservation within the promoter region and the CNS1 region (Fig. 1⇓A).

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

Human naive CD4+ T lymphocytes are hypermethylated at the IFNG promoter. A, VISTA plot showing conserved nucleotide sequences relative to the IFNG gene. Conserved nucleotide sequences are defined as stretches of nucleotides more than 100 bp long that show a conservation between the human and mouse genome of more than 70%. Blue regions are conserved nucleotide sequence elements that are located in exons, while red regions are located in noncoding DNA. B, Black bars indicate percentage methylation at each CpG position relative to the IFNG transcription start site. Shown are results from bisulfite sequencing of DNA from naive CD4+ T lymphocytes (top panel), CD8+ T lymphocytes (second panel from top), and CD19+ B lymphocytes (bottom panel) isolated by FACS sorting.

We started by investigating the methylation status of the IFNG promoter region in sorted cell populations (Fig. 1⇑B). Naive CD4+CD45RA+CD45RO− T lymphocytes, CD8+ T lymphocytes, and CD19+ B lymphocytes were isolated by FACS sorting (>95% pure) and isolated DNA was subsequently processed for bisulfite sequencing. Naive CD4+CD45RA+CD45RO− lymphocytes displayed hypermethylation at the promoter region (Fig. 1⇑B, top panel, number of clones analyzed: n = 14), an observation which is in sharp contrast to previously conducted experiments on mice (11, 13) where the Ifng promoter has been shown to be demethylated. CD8+ T lymphocytes (Fig. 1⇑B, second panel from top, n = 9) displayed a more pronounced demethylation at the promoter region compared with naive CD4+ T lymphocytes (70.4 vs 81% methylation at all positions analyzed). However human CD8+ T lymphocytes did not display the complete demethylation reported in mice (11). Surprisingly, demethylation was mostly pronounced in CD19+ B lymphocytes (Fig. 1⇑B, bottom panel, n = 10), which do not produce IFN-γ under physiological conditions (46.7% methylation at all positions analyzed). This observation is also contradictory to mouse experiments where CD19+ B lymphocytes have been described to have an overall hypermethylated promoter region (11). These results would imply different mechanisms in preventing the production of IFN-γ in human vs mouse CD19+ B cells.

Next, we analyzed the methylation status of the conserved upstream region CNS1 (Fig. 2⇓A). This region shows enhancer activity and binds important transcription factors for Th1 cell development such as T-bet and NF-AT (14, 15) and it is differentially methylated between Th1 and Th2 cells in mice (13). To our knowledge, the methylation status of this region in humans has not yet been examined. We found that the CNS1 region was hypermethylated in naive CD4+ T lymphocytes where a methylation level of 83.7% was observed at all positions analyzed (Fig. 2⇓B, top panel, n = 21). These findings are in coherence with previous studies in mice (13). Sorted CD8+ T lymphocytes cells were more demethylated with a methylation level of 68.3% at all positions analyzed (Fig. 2⇓B, middle panel, n = 9), whereas CD19+ B lymphocytes were almost completely methylated (93.7% at all positions analyzed) (Fig. 2⇓B, bottom panel, n = 9). As a control for the validity of the bisulfite sequencing method, we performed analysis on M.Sss1 treated DNA (completely CpG methylated) (Fig. 2⇓C, top panel, n = 10) from a BAC containing the IFNG locus as well as unmodified (unmethylated) BAC DNA (Fig. 2⇓C, bottom panel, n = 10), where 100 and 0% methylation, respectively, was verified.

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

Methylation status of the enhancer CNS1 in sorted cells. A, VISTA plot showing conserved nucleotide sequenced in the IFNG gene as previously described in Fig. 1⇑A. B, Black bars indicate percent methylation at each CpG position relative to the IFNG transcription start site. Shown are results from bisulfite sequencing of DNA from Naive CD4+ T lymphocytes (top panel), CD8+ T lymphocytes (second panel from top), and CD19+ B lymphocytes (bottom panel, n = 9). C, Validation of the bisulfite sequencing technique by analysis of M.Sss1 treated (top panel) and untreated (bottom panel) bacterial artificial chromosome.

We conclude that the human IFNG promoter displays a high level of methylation in naive CD4+ T lymphocytes. This is in sharp contrast to mouse, where the promoter region of naive CD4+ T lymphocytes is demethylated. In addition, the human IFNG promoter region in CD19+ B lymphocytes is demethylated whereas this region is methylated in mouse suggesting that the silencing of IFN-γ production in mice and men is controlled by different means of regulation.

Methylation pattern of regulatory elements during Th cell differentiation

Establishment of proper gene expression is dependent on chromatin remodeling proteins and transcription factors. The T-box protein T-bet has emerged as a key regulator of Th1 differentiation and Ifng expression. Retroviral expression of T-bet in developed Th2 cells not only induces Ifng expression but also suppresses the expression of Th2 cytokines (17, 18). Furthermore, T-bet induces Ifng expression directly by binding to regulatory elements in the Ifng locus, but also indirectly by interfering with the binding of GATA-3 to regulatory elements in the Th2 locus (19, 20). The importance of T-bet and GATA-3 for the establishment of cytokine expression patterns during Th cell differentiation prompted us to investigate the expression of IFNG, IL-4, T-bet and GATA-3 with quantitative real-time PCR during in vitro differentiation of naive CD4+ cells into Th1 cells (Fig. 3⇓A). As expected, IFNG expression was up-regulated while IL-4 expression remained low. T-bet expression also increased during differentiation with a decrement in GATA-3 expression as a consequence. Thus, the skewing protocol used is valid for the induction of relevant regulatory transcription factors.

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

Demethylation of regulatory elements during Th1 differentiation. A, Quantitative real-time PCR analysis of IFNG (left graph, open bars), IL-4 (left graph, filled bars), T-bet (right graph, open bars), and GATA-3 (right graph, filled bars) transcripts during Th1 differentiation. Naive CD4+ T lymphocytes were with plate bound anti-CD3 and anti-CD28 Abs and cultured in Th1 skewing conditions and RNA was isolated by TRIzol extraction before activation (top panel) at day 1 (second panel from top), day 3 (third panel from top), and day 7 (bottom panel). Graphs show analysis of triplicate samples normalized to expression in naive CD4+ T lymphocytes using the 2−ΔΔCt method and RNA polymerase II as housekeeping gene. Error bars indicate mean ± SEM. B, Production of IFN-γ (left graph) and IL-4 (right graph) in Th1 cultures (□) and Th2 cultures (▪). Data represent analysis of triplicate samples and error bars indicate mean ± SEM. C, Methylation pattern of the IFNG promoter and CNS1 region during Th1 differentiation. Naive CD4+ T lymphocytes were stimulated and cultured in Th1 skewing conditions and cultures as previously described. DNA was extracted at day 1 (second panel from the top), day 3 (third panel from top), and day 7 (bottom panel) and processed for bisulfite sequencing of the promoter (right column) and the CNS1 region (left column). Black bars indicate percent methylation at each CpG position relative to the IFNG transcription start site. Gray areas in bar graph display clones where methylation status could not be determined during sequencing reaction. D, Methylation status of the IFNG promoter and CNS1 in Th2 differentiated cells. Cells were stimulated in Th2 skewing conditions and DNA was extracted after 7 days of culture and processed for bisulfite sequencing of the promoter (right graph) and the CNS1 region (left graph).

Furthermore, the differentiation into functional Th1 and Th2 effector cells was verified by cytokine production at the protein level. Secretion of IFN-γ and IL-4 in supernatants from Th1 and Th2 cultures was analyzed by ELISA (Fig. 3⇑B). Th1 differentiation was verified by IFN-γ production and low amounts of IL-4, whereas Th2-skewed cells produced high amounts of IL-4 and low levels of IFN-γ.

We next sought to investigate methylation patterns at the promoter and CNS1 regions during in vitro differentiation into Th1 and Th2 effector cells. During in vitro Th1 differentiation, a demethylation was observed at the promoter region, which was mostly pronounced at CpG positions upstream of the transcription start site (Fig. 3⇑C, right panels, number of clones analyzed: day 1, n = 15; day 3, n = 18; day 7, n = 23) whereas the whole promoter region remained hypermethylated during Th2 differentiation (Fig. 3⇑D, right panel, n = 10). These observations agree with the previously conducted experiments by White et al. (12). The upstream enhancer CNS1 became demethylated during Th1 differentiation (Fig. 3⇑C, left panels, day 1, n = 21; day 3, n = 25; day 7, n = 19), but remained methylated in differentiating Th2 cells (Fig. 3⇑C, left panel, n = 16). The demethylation of CNS1 was mostly pronounced at the 3′ end of the enhancer. Interestingly, the demethylation of both regulatory regions was initiated at day 3 of culture (Fig. 3⇑C, third panel from top), when a major increase in IFNG expression was observed (Fig. 3⇑A, third panel from top) suggesting methylation of both regions as a mechanism of limiting IFNG expression in noncommitted Th1 effector cells. Furthermore, increased T-bet expression and decreased GATA-3 expression was observed already at day 1 of culture (Fig. 3⇑A, second panel from top) and thus proceeded the transcription of IFNG. This observation agrees with the concept of T-bet and GATA-3 as determinants of lineage decision during early Th cell differentiation.

To ensure that the demethylation observed in Th1 differentiated cells were not affected by cells that had failed to become activated during differentiation, we performed CFSE staining of naive CD4+ cells before activation and subsequently isolated cells that had undergone cell cycle divisions at day 5. DNA from these cells was isolated and subjected to bisulfite sequencing of the promoter and the CNS1 region. The demethylation in dividing cells, 5 days poststimulation, showed a similar pattern to the 1 wk differentiated Th1 cells at both regions analyzed (Fig. 4⇓, B and C). Thus, the differentiation into Th1 effector cells parallels demethylation of the IFNG promoter and CNS1 regions, suggesting demethylation as an important mechanism for the control of IFN-γ production.

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

Methylation status of conserved regulatory elements in dividing cells. Naive CD4+ T lymphocytes were labeled with CFSE before activation and cultured in Th1 skewing conditions. A, At day 5 of culture, cells that had decreased CFSE emission as a consequence of cell cycle divisions were isolated by FACS sorting DNA from dividing cells were isolated and processed for bisulfite sequencing of the CNS1 region (number of clones analyzed: n = 15) (B) and the IFNG promoter region (n = 15) (C).

Methylation status of CD4+ tumor-infiltrating lymphocytes (TILs) and CD4+ cells from tumor-draining lymph node

Since bisulfite sequencing in itself is both time-consuming and limited by the number of clones analyzed in its ability to reflect the true methylation status of a cell population, we developed a COBRA based method for screening the methylation status of the IFNG gene. The −4229 position in the enhancer CNS1 region was chosen as a reporter position due to its selective demethylation in Th1 cells. Shown in Fig. 5⇓A is a schematic drawing of the COBRA-based method. After bisulfite treatment and PCR amplification, MaeII will selectively digest fragments at the −4229 position which were originally methylated at the cytosine residue and thus protected from deamination to uracil during bisulfite treatment. Fig. 5⇓B shows examples of electropherograms from Th1 and Th2 samples. Th1-skewed lymphocytes displayed a dominant peak of undigested PCR product due to their demethylated status of position −4229 (Fig. 5⇓B, top panel). In contrast when Th2 lymphocytes were subjected to the COBRA method, the major peak was recognized as a short digested PCR fragment demonstrating the presence of a methylated CpG at position −4229 (Fig. 5⇓B, bottom panel). Thus, the COBRA method using MaeII digestion of bisulfite-treated PCR product of position −4229 in the CNS1 region of the IFNG gene is useful for identification of Th1 committed lymphocytes.

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

Hypermethylation of the IFNG gene in tumor-infiltrating lymphocytes. A, Schematic drawing of the restriction enzyme based analysis of the −4229 reporter position. MaeII will selectively digest PCR products that were methylated at the −4229 CpG before bisulfite treatment and PCR amplification. Digestion is followed by Gene Scan analysis. B, Gene Scan electropherograms of MaeII digested PCR product from Th1 DNA (top panel) and Th2 DNA (bottom panel). Methylation level at the −4229 position is calculated as the relative amount of digested product based upon peak height. C, Graph showing methylation at the −4229 reporter position in Th1 cells, Th2 cells, CD4+ TIL, and CD4+ cells from sentinel lymph node (SLN). p values show level of significance from t test performed on the different populations. D, Quantitative real-time PCR analysis of IFNG and T-bet transcript from CD4+ tumor-infiltrating lymphocytes cultured in media with 5-Azacytidine (□) or without 5-Azacytidine (▪). Shown are results from experiments performed on TILs from two colon cancer patients. Ct values were normalized to expression in TILs grown without 5-Aza using the 2−ΔΔCt method and RNA polymerase II as housekeeping gene. E, Quantitative real-time PCR analysis of IFNG (left graph) and T-bet (right graph) transcript from Th1 differentiated cells grown in Transwell culture with the colon cancer cell line CCL221 (□) or without CCL221 (▴). RNA was isolated before and after stimulation with plate bound anti-CD3 (OKT-3) and soluble anti-CD28. Graphs show analysis of triplicate measures normalized to expression in the resting population using the 2−ΔΔCt method and RNA polymerase II as housekeeping gene. Error bars indicate mean ± SEM.

Altered cytokine expression in tumor-infiltrating lymphocytes suggests that the gene expression program of these cells has been shifted to the benefit of tumor survival. We therefore sought to investigate the methylation status of the IFNG gene in tumor-infiltrating CD4+ T lymphocytes isolated from colon cancer tumors. We measured the methylation level at the −4229 CNS1 reporter position using the established restriction enzyme-based method in differentiated Th1 and Th2 lymphocytes from four healthy donors, CD4+ tumor-infiltrating lymphocytes from six patients with colon cancer (Fig. 5⇑C), and CD4+ cells from tumor draining lymph nodes from three patients. The total number of draining lymph nodes analyzed was four since two independently draining lymph nodes were identified in one of the patients. As expected Th1-skewed cells displayed a significantly (p < 0.0001) lower degree of methylation (28.66 ± 0.9651 n = 4) at the −4229 reporter position compared with Th2 cells (71.40 ± 1.156 n = 4). Interestingly, sorted CD4+ tumor-infiltrating lymphocytes from six patients with colon cancer displayed hypermethylation at the −4229 CNS1 reporter position indicating a silenced IFNG gene (62.10 ± 1.520 n = 6) (Fig. 5⇑C). The level of methylation of CD4+ tumor-infiltrating lymphocytes were similar to those seen among Th2 skewed lymphocytes suggesting inappropriate silencing of the IFNG gene among TILs. The methylation level of CD4+ cells from tumor draining lymph nodes was significantly (p = 0.0005) less methylated (45.47 ± 2.934 n = 4) than CD4+ cells infiltrating the tumor. Table II⇓ shows detailed description of demethylation levels at the reporter position and detailed results from statistical analysis can be found in Table III⇓.

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

Level of methylation at position −4229 by restriction enzyme based analysisa

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

Statistical analysis of difference in methylation at the −4229 positiona

5-Azacytidine treatment induces IFN-γ expression in TILs

We have previously shown that tumor-infiltrating lymphocytes are poor producers of IFN-γ when stimulated with autologous tumor extract or ConA (16). The observation that TILs are hypermethylated at the IFN-γ (Fig. 5⇑C) region prompted us to investigate if culturing these cells in the presence of demethylating reagents would induce/restore the expression of IFN-γ. CD4+ tumor-infiltrating lymphocytes from two colon cancer patients cultured in the presence of 5-Azacytidine displayed 4- and 35-fold induced expression of IFN-γ (Fig. 5⇑D). The expression of T-bet was induced ∼3-fold in cells grown with 5-Aza. These results show that the observed hypermethylation among CD4+ TILs is the reason for the limited capacity to produce IFN-γ in these cells.

Suppressed IFN-γ production among Th1 cells in coculture with CCL221

Tumor reactive lymphocytes encounter their Ags in the tumor draining lymph node (16). The limited IFN-γ production (16) and hypermethylation (Fig. 5⇑C) in CD4+ TILs compared with CD4+ cells from tumor draining lymph nodes suggest tumor microenvironment induced cell fate reprogramming of tumor reactive T cells on the basis of DNA methylation. To investigate whether the presence of colon cancer cells would have any effects on differentiated Th1 cells in vitro we established a transwell culturing system. Differentiated Th1 cells grown in the presence of the colon cancer cell line CCL221 showed no response in IFN-γ production upon restimulation (Fig. 5⇑E). In contrast, cells grown in the absence of CCL221 were able to mount a vigorous IFN-γ response when stimulated, an observation that favors the assumption of an immunosuppressive environment surrounding the tumor.

Discussion

In this report, we show that IFNG regulatory elements of naive CD4+ Th lymphocytes are hypermethylated and thus not poised for rapid onset of Th1 responses. Furthermore, we demonstrate hypermethylation of the IFNG gene among tumor-infiltrating lymphocytes as a mechanism of tumor induced immunosuppression. Reports on methylation status of the IFNG gene during Th cell differentiation have been contradictory (7, 8, 9, 10, 11, 13). In this study, we conclude that there are obvious differences in the epigenetic regulation of IFNG transcription between mice and human T lymphocytes. Whereas recent reports conducted on mice have shown that naive Th cells and their progenitors are hypomethylated at the Ifng promoter and that de novo methylation during Th2 differentiation is an important mechanism for silencing of the Ifng gene in these cells (11, 13), we show that human naive CD4+ lymphocytes are hypermethylated at the promoter region (Fig. 1⇑B) and become demethylated during Th1 differentiation (Fig. 3⇑C). An evolutionarily conserved CpG dinucleotide positioned −53 base pairs relative to the transcription start site in the mouse Ifng gene has emerged as an important regulator of Ifng silencing in differentiating Th2 cells. This CpG undergoes rapid de novo methylation during early Th2 differentiation and subsequently inhibits activating transcription factor 2/c-Jun and CREB transcription factor binding to the promoter and thus leads to silencing of Ifng transcription (11). This site is evolutionarily conserved and our data show that this site undergoes demethylation during Th1 differentiation. Binding of this complex could thus serve as a mechanism to allow transcription of IFNG in human Th1 cells. However, the different methylation status of the IFNG promoter in naive CD4+ T lymphocytes would imply alternative mechanisms for limiting IFNG expression in these cells.

In addition, we demonstrate that human CD19+ B lymphocytes show a similar methylation profile to Th1 effector cells, which is in sharp contrast to mice studies (11). This implies that the IFNG locus of human B lymphocytes regulated by other means, e.g., the lack of essential transcription factors or positioning to heterochromatin. Th and B lymphocytes share common progenitors suggesting a de novo methylation of the human IFNG gene during differentiation from progenitor to naive CD4+ T lymphocyte. A de novo methylation during differentiation into naive CD4+ T lymphocytes could serve as a mean of limiting expression of IFNG in these cells and thus serve as a safety mechanism to avoid autoimmunity.

In mice, the Ifng and Th2 loci are juxtaposed in naive CD4+ T lymphocytes, suggesting a poised state of the Ifng locus allowing rapid onset of IFN-γ production during early Th1 differentiation (21). The mouse Ifng promoter region is already hypomethylated in naive CD4+ T lymphocytes which further supports this hypothesis (11, 13). Whereas the mouse Ifng promoter is demethylated in naive CD4+ T lymphocytes, the CNS1 enhancer region is hypermethylated suggesting recruitment during later stages of Th1 differentiation to further amplify Ifng expression and support the manifestation of a Th1 identity (13). Our data on the human CNS1 enhancer region supports this theory (Fig. 3⇑C). However the obvious interspecies difference in methylation status in the promoter region of naive CD4+ T lymphocytes implies that mouse data on the epigenetic regulation of Th1 and Th2 differentiation is not applicable to the human setting, and calls for separate investigations concerning Th cell differentiation. This is further emphasized by the fact that the human and mouse IFNG loci are separated evolutionarily by structural remodelling (13).

In order for a tumor to persist, it needs to overcome the intrinsic tumor immune defense. The immune defense against tumors involves both the innate and the adaptive immune system. Tumor-infiltrating lymphocytes as part of the adaptive immune system have been identified in several different tumor types, including colon cancer. A positive correlation between survival of patients with colon cancer and presence of tumor-infiltrating effector memory T lymphocytes have been observed (22). Moreover, mRNA expression of Th1 associated markers such as T-bet, IRF-1, and IFNG in tumor tissue is also related to prolonged survival, indicating that a Th1 anti-tumoral response is beneficial (23). Our results demonstrate that tumor-infiltrating CD4+ lymphocytes are inappropriately hypermethylated (Fig. 5⇑C) and not committed to the Th1 phenotype. Interestingly, when we investigate the functionality of TILs from patients with colon cancer they are frequently anergic and unable to produce IFN-γ upon activation with autologous tumor extract (16). Furthermore we show in this study that the limited IFN-γ expression in TILs is methylation dependent, since treatment with 5-Azacytidine is able to restore IFN-γ production in these cells (Fig. 5⇑D). In constrast to TILs, a vigorous response with proliferation and production of IFN-γ is recognized in the tumor draining lymph node, the sentinel node (16). The sentinel node is the first location of tumor drainage, recognition and clonal T cell expansion of appropriate Th1 effector cells, followed by entrance to the blood via the thoracic duct. T effector memory cells then enter tumors as TIL cells of the same clonality (24). In this report, we show that CD4+ cells from tumor draining lymph nodes are significantly more demethylated than CD4+ TILs (Fig. 5⇑C). The majority of tumor-infiltrating T lymphocytes belong to the memory T cell pool (23). It is likely that these cells, that initially were expanded and Th1 activated in the sentinel node, have undergone cell fate reprogramming of the IFNG gene as a consequence of the immunosuppressive environment of the tumor, as supported by the abolished IFN-γ secretion by Th1 cells upon coculture with colon cancer cells (Fig. 5⇑E).

One way by which the cytokine expression profile can be altered to benefit tumor survival is through the up-regulation of immunosuppressive regulatory T cells (Tregs). Increased numbers of Tregs have been observed in peripheral blood and among tumor-infiltrating lymphocytes (25, 26). These induced Tregs have been shown to impair effector cell functions in terms of proliferation, altered cytokine expression and/or cytotoxic capacities (25, 27). In addition, depletion of Tregs promote anti-tumoral responses and is associated with tumor regression in mouse models (reviewed in Ref. 28). TGF-β has been suggested as an important participant in the immunosuppressive events caused by induction of Tregs (27, 29, 30). TGF-β is known to inhibit IFN-γ secretion through down-regulation of T-bet and STAT4, transcription factors which are crucial for Th1 development and appropriate chromatin remodeling of the IFNG locus (31). In this report, we show that tumor-infiltrating lymphocytes are not epigenetically committed to the Th1 phenotype (Fig. 5⇑C). The coculture experiments performed in this report (Fig. 5⇑E) supports the assumption that soluble, noncontact dependent factors are exerting these immunosuppressive effects. It is tempting to speculate that the de novo methylation of the IFNG gene is mediated through TGF-β dependent pathways down regulating essential transcription factors of the Th1 response.

In conclusion we find major interspecies differences in the methylation status of the IFNG gene in naive CD4+ T lymphocytes. Th1 commitment in human lymphocytes involves demethylation, demonstrating that human naive T cells are not readily available for Th1 responses without prior epigenetical modifications. Finally, epigenetical silencing of key responder genes among tumor-infiltrating T effector cells serve as a mechanism of tumor-induced immunosupression.

Disclosures

The authors have no financial conflict of interest.

Footnotes

  • The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

  • ↵1 This work was supported by the Swedish cancer society, the Cancer and Allergy society, the Hedlunds foundation, the Lundberg foundation, Gustav V foundation, the Selander foundation, and the Åke Wiberg foundation.

  • ↵2 Address correspondence and reprint requests to Dr. Ola Winqvist, Department of Medicine, Clinical Allergy Research Unit L2:04, Karolinska University Hospital, S-171 76 Stockholm, Sweden. E-mail address: ola.winqvist{at}karolinska.se

  • ↵3 Abbreviations used in this paper: GATA-3, GATA binding protein-3; 5-Aza, 5-Azacytidine; CNS1, conserved nucleotide sequence 1; BAC, bacterial artificial chromosome; COBRA, combined bisulfite restriction enzyme analysis; TIL, tumor-infiltrating lymphocyte; Treg, T regulatory cell.

  • Received October 18, 2007.
  • Accepted June 6, 2008.
  • Copyright © 2008 by The American Association of Immunologists

References

  1. ↵
    Abbas, A. K., K. M. Murphy, A. Sher. 1996. Functional diversity of helper T lymphocytes. Nature 383: 787-793.
    OpenUrlCrossRefPubMed
  2. ↵
    Murphy, K. M., S. L. Reiner. 2002. The lineage decisions of helper T cells. Nat. Rev. Immunol. 2: 933-944.
    OpenUrlCrossRefPubMed
  3. ↵
    Hsieh, C. S., S. E. Macatonia, C. S. Tripp, S. F. Wolf, A. O'Garra, K. M. Murphy. 1993. Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 260: 547-549.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Szabo, S. J., B. M. Sullivan, S. L. Peng, L. H. Glimcher. 2003. Molecular mechanisms regulating Th1 immune responses. Annu. Rev. Immunol. 21: 713-758.
    OpenUrlCrossRefPubMed
  5. ↵
    Wilson, C. B., K. W. Makar, M. Shnyreva, D. R. Fitzpatrick. 2005. DNA methylation and the expanding epigenetics of T cell lineage commitment. Semin. Immunol. 17: 105-119.
    OpenUrlCrossRefPubMed
  6. ↵
    Young, H. A., P. Ghosh, J. Ye, J. Lederer, A. Lichtman, J. R. Gerard, L. Penix, C. B. Wilson, A. J. Melvin, M. E. McGurn, et al 1994. Differentiation of the T helper phenotypes by analysis of the methylation state of the IFN-γ gene. J. Immunol. 153: 3603-3610.
    OpenUrlAbstract
  7. ↵
    Melvin, A. J., M. E. McGurn, S. J. Bort, C. Gibson, D. B. Lewis. 1995. Hypomethylation of the interferon-γ gene correlates with its expression by primary T-lineage cells. Eur. J. Immunol. 25: 426-430.
    OpenUrlCrossRefPubMed
  8. ↵
    White, G. P., P. M. Watt, B. J. Holt, P. G. Holt. 2002. Differential patterns of methylation of the IFN-γ promoter at CpG and non-CpG sites underlie differences in IFN-γ gene expression between human neonatal and adult CD45RO- T cells. J. Immunol. 168: 2820-2827.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Yano, S., P. Ghosh, H. Kusaba, M. Buchholz, D. L. Longo. 2003. Effect of promoter methylation on the regulation of IFN-γ gene during in vitro differentiation of human peripheral blood T cells into a Th2 population. J. Immunol. 171: 2510-2516.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Winders, B. R., R. H. Schwartz, D. Bruniquel. 2004. A distinct region of the murine IFN-γ promoter is hypomethylated from early T cell development through mature naive and Th1 cell differentiation, but is hypermethylated in Th2 cells. J. Immunol. 173: 7377-7384.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Jones, B., J. Chen. 2006. Inhibition of IFN-γ transcription by site-specific methylation during T helper cell development. EMBO J. 25: 2443-2452.
    OpenUrlCrossRefPubMed
  12. ↵
    White, G. P., E. M. Hollams, S. T. Yerkovich, A. Bosco, B. J. Holt, M. R. Bassami, M. Kusel, P. D. Sly, P. G. Holt. 2006. CpG methylation patterns in the IFNγ promoter in naive T cells: variations during Th1 and Th2 differentiation and between atopics and non-atopics. Pediatr. Allergy Immunol. 17: 557-564.
    OpenUrlCrossRefPubMed
  13. ↵
    Schoenborn, J. R., M. O. Dorschner, M. Sekimata, D. M. Santer, M. Shnyreva, D. R. Fitzpatrick, J. A. Stamatoyonnapoulos, C. B. Wilson. 2007. Comprehensive epigenetic profiling identifies multiple distal regulatory elements directing transcription of the gene encoding interferon-γ. Nat. Immunol. 8: 732-742.
    OpenUrlCrossRefPubMed
  14. ↵
    Lee, D. U., O. Avni, L. Chen, A. Rao. 2004. A distal enhancer in the interferon-γ (IFN-γ) locus revealed by genome sequence comparison. J. Biol. Chem. 279: 4802-4810.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    Shnyreva, M., W. M. Weaver, M. Blanchette, S. L. Taylor, M. Tompa, D. R. Fitzpatrick, C. B. Wilson. 2004. Evolutionarily conserved sequence elements that positively regulate IFN-γ expression in T cells. Proc. Natl. Acad. Sci. USA 101: 12622-12627.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Marits, P., M. Karlsson, K. Dahl, P. Larsson, A. Wanders, M. Thorn, O. Winqvist. 2006. Sentinel node lymphocytes: tumour reactive lymphocytes identified intraoperatively for the use in immunotherapy of colon cancer. Br. J. Cancer 94: 1478-1484.
    OpenUrlCrossRefPubMed
  17. ↵
    Szabo, S. J., S. T. Kim, G. L. Costa, X. Zhang, C. G. Fathman, L. H. Glimcher. 2000. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100: 655-669.
    OpenUrlCrossRefPubMed
  18. ↵
    Szabo, S. J., B. M. Sullivan, C. Stemmann, A. R. Satoskar, B. P. Sleckman, L. H. Glimcher. 2002. Distinct effects of T-bet in TH1 lineage commitment and IFN-γ production in CD4 and CD8 T cells. Science 295: 338-342.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Hwang, E. S., S. J. Szabo, P. L. Schwartzberg, L. H. Glimcher. 2005. T helper cell fate specified by kinase-mediated interaction of T-bet with GATA-3. Science 307: 430-433.
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Usui, T., J. C. Preiss, Y. Kanno, Z. J. Yao, J. H. Bream, J. J. O'Shea, W. Strober. 2006. T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription. J. Exp. Med. 203: 755-766.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Spilianakis, C. G., M. D. Lalioti, T. Town, G. R. Lee, R. A. Flavell. 2005. Interchromosomal associations between alternatively expressed loci. Nature 435: 637-645.
    OpenUrlCrossRefPubMed
  22. ↵
    Naito, Y., K. Saito, K. Shiiba, A. Ohuchi, K. Saigenji, H. Nagura, H. Ohtani. 1998. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 58: 3491-3494.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Pages, F., A. Berger, M. Camus, F. Sanchez-Cabo, A. Costes, R. Molidor, B. Mlecnik, A. Kirilovsky, M. Nilsson, D. Damotte, et al 2005. Effector memory T cells, early metastasis, and survival in colorectal cancer. N. Engl. J. Med. 353: 2654-2666.
    OpenUrlCrossRefPubMed
  24. ↵
    Baner, J., P. Marits, M. Nilsson, O. Winqvist, U. Landegren. 2005. Analysis of T-cell receptor V β gene repertoires after immune stimulation and in malignancy by use of padlock probes and microarrays. Clin. Chem. 51: 768-775.
    OpenUrlAbstract/FREE Full Text
  25. ↵
    Ling, K. L., S. E. Pratap, G. J. Bates, B. Singh, N. J. Mortensen, B. D. George, B. F. Warren, J. Piris, G. Roncador, S. B. Fox, et al 2007. Increased frequency of regulatory T cells in peripheral blood and tumour infiltrating lymphocytes in colorectal cancer patients. Cancer Immun. 7: 7
    OpenUrlPubMed
  26. ↵
    Clarke, S. L., G. J. Betts, A. Plant, K. L. Wright, T. M. El-Shanawany, R. Harrop, J. Torkington, B. I. Rees, G. T. Williams, A. M. Gallimore, A. J. Godkin. 2006. CD4+CD25+FOXP3+ regulatory T cells suppress anti-tumor immune responses in patients with colorectal cancer. PLoS ONE 1: e129
    OpenUrlCrossRef
  27. ↵
    Somasundaram, R., L. Jacob, R. Swoboda, L. Caputo, H. Song, S. Basak, D. Monos, D. Peritt, F. Marincola, D. Cai, et al 2002. Inhibition of cytolytic T lymphocyte proliferation by autologous CD4+/CD25+ regulatory T cells in a colorectal carcinoma patient is mediated by transforming growth factor-β. Cancer Res. 62: 5267-5272.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Zou, W.. 2006. Regulatory T cells, tumour immunity, and immunotherapy. Nat. Rev. Immunol. 6: 295-307.
    OpenUrlCrossRefPubMed
  29. ↵
    Seo, N., S. Hayakawa, M. Takigawa, Y. Tokura. 2001. Interleukin-10 expressed at early tumour sites induces subsequent generation of CD4+ T-regulatory cells and systemic collapse of antitumour immunity. Immunology 103: 449-457.
    OpenUrlCrossRefPubMed
  30. ↵
    Larmonier, N., M. Marron, Y. Zeng, J. Cantrell, A. Romanoski, M. Sepassi, S. Thompson, X. Chen, S. Andreansky, E. Katsanis. 2007. Tumor-derived CD4+CD25+ regulatory T cell suppression of dendritic cell function involves TGF-β and IL-10. Cancer Immunol. Immunother. 56: 48-59.
    OpenUrlCrossRefPubMed
  31. ↵
    Lin, J. T., S. L. Martin, L. Xia, J. D. Gorham. 2005. TGF-β 1 uses distinct mechanisms to inhibit IFN-γ expression in CD4+ T cells at priming and at recall: differential involvement of Stat4 and T-bet. J. Immunol. 174: 5950-5958.
    OpenUrlAbstract/FREE Full Text
View Abstract
PreviousNext
Back to top

In this issue

The Journal of Immunology: 181 (4)
The Journal of Immunology
Vol. 181, Issue 4
15 Aug 2008
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Advertising (PDF)
  • Back Matter (PDF)
  • Editorial Board (PDF)
  • Front Matter (PDF)
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.
CpG Methylation of the IFNG Gene as a Mechanism to Induce Immunosupression in Tumor-Infiltrating Lymphocytes
(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
CpG Methylation of the IFNG Gene as a Mechanism to Induce Immunosupression in Tumor-Infiltrating Lymphocytes
Peter C. J. Janson, Per Marits, Magnus Thörn, Rolf Ohlsson, Ola Winqvist
The Journal of Immunology August 15, 2008, 181 (4) 2878-2886; DOI: 10.4049/jimmunol.181.4.2878

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
CpG Methylation of the IFNG Gene as a Mechanism to Induce Immunosupression in Tumor-Infiltrating Lymphocytes
Peter C. J. Janson, Per Marits, Magnus Thörn, Rolf Ohlsson, Ola Winqvist
The Journal of Immunology August 15, 2008, 181 (4) 2878-2886; DOI: 10.4049/jimmunol.181.4.2878
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
    • Disclosures
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Differential Susceptibility to Staphylococcal Superantigen (SsAg)-Induced Apoptosis of CD4+ T Cells from Atopic Dermatitis Patients and Healthy Subjects: The Inhibitory Effect of IL-4 on SsAg-Induced Apoptosis
  • HIV-1 Vaccination Administered Intramuscularly Can Induce Both Systemic and Mucosal T Cell Immunity in HIV-1-Uninfected Individuals
  • Osteopontin (Eta-1) and Fibroblast Growth Factor-2 Cross-Talk in Angiogenesis
Show more Clinical 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