Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1 - PubMed (original) (raw)

. 2011 Sep 2;146(5):772-84.

doi: 10.1016/j.cell.2011.07.033. Epub 2011 Aug 25.

Joseph Barbi, Huang-Yu Yang, Dilini Jinasena, Hong Yu, Ying Zheng, Zachary Bordman, Juan Fu, Young Kim, Hung-Rong Yen, Weibo Luo, Karen Zeller, Larissa Shimoda, Suzanne L Topalian, Gregg L Semenza, Chi V Dang, Drew M Pardoll, Fan Pan

Affiliations

Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1

Eric V Dang et al. Cell. 2011.

Abstract

T cell differentiation into distinct functional effector and inhibitory subsets is regulated, in part, by the cytokine environment present at the time of antigen recognition. Here, we show that hypoxia-inducible factor 1 (HIF-1), a key metabolic sensor, regulates the balance between regulatory T cell (T(reg)) and T(H)17 differentiation. HIF-1 enhances T(H)17 development through direct transcriptional activation of RORγt and via tertiary complex formation with RORγt and p300 recruitment to the IL-17 promoter, thereby regulating T(H)17 signature genes. Concurrently, HIF-1 attenuates T(reg) development by binding Foxp3 and targeting it for proteasomal degradation. Importantly, this regulation occurs under both normoxic and hypoxic conditions. Mice with HIF-1α-deficient T cells are resistant to induction of T(H)17-dependent experimental autoimmune encephalitis associated with diminished T(H)17 and increased T(reg) cells. These findings highlight the importance of metabolic cues in T cell fate determination and suggest that metabolic modulation could ameliorate certain T cell-based immune pathologies.

Copyright © 2011 Elsevier Inc. All rights reserved.

PubMed Disclaimer

Figures

Figure 1

Figure 1. HIF-1α mRNA is up-regulated in T cells under TH17 skewing conditions in Stat3 dependent manner

(A) Naïve (CD4+CD25-CD62Lhigh) T cells were cultured under Th-cell subset inducing conditions, and HIF-1 mRNA was detected by qRT-PCR. (B, C and D) Naïve T cells from wild type (WT, HIF-1α+/+) or CD4Cre x HIF-1flox/flox mice (ko, HIF-1α−/−) were stimulated in the presence of TGFβ and IL-6. RNA was isolated from these cells and qRT-PCR was performed at different time points during culture to measure HIF-1α (B), IL-17 (C) and RORγt (D) transcript levels. (E) WT or Stat3−/− (obtained from CD4Cre x Stat3flox/flox mice) naïve CD4+ T cells were isolated and cultured under the indicated conditions for 2 days, followed by SDS-PAGE and Western blotting using antibodies against HIF-1α (top), HIF-1β (middle) and tubulin (bottom), respectively. (F) A ChIP assay was used to examine direct Stat3 binding to the HIF-1α promoter. Panels A–D and F depict the mean + s.d. of at least three experiments while Panel E is a representative result. See also Figures S1.

Figure 2

Figure 2. HIF-1α is required for TH17 development in vitro

(A) Naïve T cells isolated from wild type (WT, HIF-1α+/+) or CD4Cre x HIF-1αflox/flox (HIF-1α−/−) mice were cultured under TH17 skewing conditions with anti-CD3/CD28 and TGFβ, IL-6 and anti-IFNγ, IL-12 and IL-4 antibodies for 6 days. Cells were then stained for IL-17 and Foxp3 (see Methods). Numbers represent the percentage of CD4+ cells positive for the indicated marker. (B) FACS-sorted naïve CD4+ T cells from WT or T-HIF-1α−/− mice were activated and cultured under TH17 skewing for 4 days. Total RNA was isolated and mRNA expression of IL-17, IL-17F and IL-23R genes was assessed by qRT-PCR. For each gene, expression level in HIF-1−/− T cells was set to 1. The mean + s.d. of at least 3 trials is shown. (C) Naïve CD4+ T cells were activated with anti-CD3/CD28 under either neutral (anti-IL-4 and anti-IFNγ Ig) or TH17-skewing conditions, and transduced with a bicistronic retrovirus expressing HIF-1α-GFP or GFP alone. Intracellular cytokines were stained and analyzed in GFP+ cells. Numbers represent the percentage of gated GFP+ cells. (D) Naïve T cells isolated from WT or T-HIF-1α−/− mice were activated as described for (A) under normoxia or hypoxia (see Methods). Panels C and D represent at least 2 independent experiments. See also Figure S2.

Figure 3

Figure 3. HIF-1α transactivates RORγt transcription and is necessary for RORγt-driven TH17 differentiation in vitro

(A) Jurkat T cells were co-transfected with a luciferase reporter under the control of a wild type RORγt promoter or one with a mutated HIF-1-binding site. 24hrs post-transfection, cells were either treated with PMA and ionomycin or left untreated prior to analysis of luciferase activity, which was normalized to that of Renilla luciferase. (B) Jurkat T cells were transfected with a RORγt-luciferase reporter plasmid (incorporating the RORγt promoter) along with plasmid encoding either wild type HIF-1 or a HIF-1 mutant (with a DNA binding domain deletion). 24hrs post-transfection, cells were stimulated and luciferase activity was assessed as described in (A). Data are shown for three independent experiments (mean and s.d. of triplicate transfections). (C) A ChIP assay was used to measure direct HIF-1 binding to the RORγt promoter directly. (D and E) Naïve WT or HIF-1−/− CD4+ T cells were activated with anti-CD3/anti-CD28 under neutral (anti-IL4 and anti-IFNγ) conditions, and transduced with either a bicistronic retrovirus expressing RORγt-GFP or the GFP containing empty vector. Intracellular IL-17 and Foxp3 were stained. The plots shown are gated on GFP+ cells. These experiments were repeated at least twice with consistent results. See also Figures S3.

Figure 4

Figure 4. RORγt, HIF-1α and p300 bind to the IL-17 promoter to regulate its gene expression

(A) A HIF-1α mutant (HIF-1-ΔDBD, with the DNA binding domain deleted) retains the capacity to activate IL-17 promoter-driven luciferase activity in the presence of RORγt. Jurkat T cells were transfected with an IL-17 promoter-driven luciferase reporter along with the indicated plasmids followed by stimulation and assessment as described for Figure 3A. Data are representative of at least 3 experiments (mean and s.d. of triplicate transfections). (B) The interaction between HIF-1 and RORγt was examined with co-immunoprecipitation. FACS-sorted CD4+ T cells were activated and cultured under TH17-skewing conditions for 5 days. The whole cell lysate were immunoprecipitated with either anti-RORγt (left panel) or anti-HIF-1α antibodies (right panel), resolved by SDS-PAGE and blotted with the indicated antibodies. (C) RORγt interacts with the N-terminus of HIF-1. His-tagged RORγt purified from E. coli was incubated with different fragments of GST-HIF-1 also purified from E. coli as indicated, followed by pull-down with GST beads, resolution by SDS-PAGE and Westen blotting with anti-RORγt (top and bottom) antibodies, or anti-GST (middle). (D and E) FACS-sorted CD4+ T cells from WT or HIF-1−/− mice were activated under TH17-skewing conditions (as described for B) prior to harvest for ChIP assay utilizing anti-RORγt, anti-HIF-1 or anti-p300 antibodies. (F) Hypoxia enhances the activation of IL-17 promoter-driven luciferase activity by HIF-1, RORγt and p300. Jurkat T cells were transfected with a IL-17 promoter-driven luciferase reporter along with the indicated plasmids under normoxia or hypoxia, followed by stimulation and assessment as described for Figure 3B. Data are representative of at least 3 independent experiments (mean and s.d. of triplicate transfections). (G) Histone hyperacetylation at the IL-17 promoter was detected by ChIP assay in WT and HIF-1α−/− T cells under TH17 skewing conditions for 5 days. See also Figures S4–S6 and Table S1.

Figure 5

Figure 5. HIF-1α mediated Foxp3 degradation through proteasomal degradation pathways

(A) Naïve T cells from either WT or T-HIF-1−/− mice were cultured and stimulated in the presence of TGFβ and IL-6 and qRT-PCR was performed to measure Foxp3 mRNA at different times. (B) HIF-1α−/− T cells displayed enhanced Foxp3 accumulation during in vitro Treg differentiation. Naïve T cells from HIF-1+/+ and T-HIF-1−/− were isolated by FACS and activated under Treg skewing (5ng/ml TGFβ, 100U IL-2). Cells were stained for Foxp3 after 72 hours. Shown are representative histograms for HIF-1+/+ (red line) and HIF-1−/− (blue line) cells from three experiments. An isotype control is shown in green. Numbers represent the mean percentage of Foxp3+ cells. (C) Foxp3 protein is lost upon culture of WT T cells with IL-6 but remains unchanged in HIF-1α−/− Foxp3+ T cells. Naive T cells were activated under Treg-skewing (TGFβ, 5ng/ml) with the indicated doses of IL-6 for 4 days. Western blotting was used to measure Foxp3 protein level. (D) Hypoxia reduced expression of Foxp3 by naïve T cells during in vitro differentiation. Naïve T cells isolated from Foxp3-GFP reporter mice (CD4+GFP-CD62Lhigh) were cultured under Treg skewing conditions (see B) in either a hypoxic chamber or under normoxia (blue and red lines, respectively). Numbers represent the mean percentage of CD4+ cells expressing GFP (Foxp3+) from three experiments. (E) HIF-1 interacts with Foxp3 in iTreg cells. FACS-sorted CD4+ T cells were activated and cultured under iTreg-skewing for 4 days. Cell lysates were immunoprecipitated with anti-HIF-1 (left panel) or anti-Foxp3 antibodies (right panel), followed by SDS-PAGE and western blotting. (F, G and H) HIF-1α mediates Foxp3 degradation. 293T cells were cotransfected with Foxp3 +/− ubiquitin and increasing amounts of WT HIF-1 (F) or mutant HIF-1 (p420A and p564A) (G) or a deleted ODD domain (CA5-HIF) (H)). CoIP and Western blots were probed as indicated. (I and J) Naïve T cells were cultured under Treg-skewing conditions in normoxia (N) or hypoxia (H) for 4 days. Cells were harvested and lysed, followed by immunoprecipitation with anti-Foxp3 or control IgG antibodies. The pulled down protein along with an input control were resolved by SDS-PAGE, followed by western blot. Depicted are typical findings from 3 independent experiments. See also Figure S7

Figure 6

Figure 6. Mice lacking HIF-1α in CD4+ T cells are deficient in IL-17 production, have increased numbers of Foxp3 Treg and are more resistant to EAE

(A) EAE was induced in HIF-1+/+ and T-HIF-1−/− mice by injection of MOG35–55 in CFA and Pertussis Toxin. Disease severity was monitored and scored daily. T-HIF-1−/− mice failed to develop the severe disease seen in HIF-1+/+ mice. Mean scores for HIF-1+/+ and T-HIF-1−/− mice over time are represented (+/− SEM; *p<0.05). Shown is a representative of 4 experiments (N=7–10 per group). (B, C) During peak disease (day12), draining lymph node, splenic, and CNS infiltrating T cells were recovered and stained for IL-17 and IFNγ. The mean percentage of CNS CD4+ cells during peak disease (days 12–14) positive for IL-17 was determined (C). (D and E) Similarly, during the recovery phase (day 21), tissue infiltrating cells were isolated and the percentage of CD4+ that wereFoxp3+ was found (D and E). Panels C and E present the mean (± SEM, *p<0.05) of at least 3 trials and dot plots are representative analyses.

Figure 7

Figure 7. A model for the multi-factorial role of HIF-1α in modulating the TH17/Treg balance

Stat3 activation by factors such as IL-6 transcriptionally activates HIF-1. HIF-1 levels are further regulated by oxygen tension and other metabolites, representing a key molecular link between metabolic cues and T cell lineage commitment. HIF-1 directly activates RORγt gene transcription and furthermore, recruits p300 to RORγt transcription complexes to the promoters of TH17 genes (ie IL-17). These activities promote Th17 differentiation. Concomitantly, HIF-1 induces Foxp3 protein degradation via targeting for ubiquitination and proteasomal degradation.

Comment in

Similar articles

Cited by

References

    1. Ahern PP, Schiering C, Buonocore S, McGeachy MJ, Cua DJ, Maloy KJ, Powrie F. Interleukin-23 drives intestinal inflammation through direct activity on T cells. Immunity. 2010;33:279–288. - PMC - PubMed
    1. Arany Z, Huang LE, Eckner R, Bhattacharya S, Jiang C, Goldberg MA, Bunn HF, Livingston DM. An essential role for p300/CBP in the cellular response to hypoxia. Proc Natl Acad Sci U S A. 1996;93:12969–12973. - PMC - PubMed
    1. Barnes MJ, Powrie F. Regulatory T cells reinforce intestinal homeostasis. Immunity. 2009;31:401–411. - PubMed
    1. Bauquet AT, Jin H, Paterson AM, Mitsdoerffer M, Ho IC, Sharpe AH, Kuchroo VK. The costimulatory molecule ICOS regulates the expression of c-Maf and IL-21 in the development of follicular T helper cells and TH-17 cells. Nat Immunol. 2009;10:167–175. - PMC - PubMed
    1. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–238. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources