9
of March 13, 2013. This information is current as Expression Regulatory T Cells upon Loss of FOXP3 Dominant Th2 Differentiation of Human Matthias Edinger Reinhard Andreesen, Petra Hoffmann, Michael Rehli and Leo Hansmann, Christian Schmidl, Janina Kett, Lena Steger, http://www.jimmunol.org/content/188/3/1275 doi: 10.4049/jimmunol.1102288 December 2011; 2012; 188:1275-1282; Prepublished online 30 J Immunol Material Supplementary 8.DC1.html http://www.jimmunol.org/content/suppl/2011/12/30/jimmunol.110228 References http://www.jimmunol.org/content/188/3/1275.full#ref-list-1 , 20 of which you can access for free at: cites 60 articles This article Subscriptions http://jimmunol.org/subscriptions is online at: The Journal of Immunology Information about subscribing to Permissions http://www.aai.org/ji/copyright.html Submit copyright permission requests at: Email Alerts http://jimmunol.org/cgi/alerts/etoc Receive free email-alerts when new articles cite this article. Sign up at: Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists, Inc. All rights reserved. Copyright © 2012 by The American Association of 9650 Rockville Pike, Bethesda, MD 20814-3994. The American Association of Immunologists, Inc., is published twice each month by The Journal of Immunology at University of Regensburg on March 13, 2013 http://jimmunol.org/ Downloaded from

Dominant Th2 Differentiation of Human Regulatory T Cells

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Dominant Th2 Differentiation of Human Regulatory T Cells

of March 13, 2013.This information is current as

ExpressionRegulatory T Cells upon Loss of FOXP3 Dominant Th2 Differentiation of Human

Matthias EdingerReinhard Andreesen, Petra Hoffmann, Michael Rehli and Leo Hansmann, Christian Schmidl, Janina Kett, Lena Steger,

http://www.jimmunol.org/content/188/3/1275doi: 10.4049/jimmunol.1102288December 2011;

2012; 188:1275-1282; Prepublished online 30J Immunol 

MaterialSupplementary

8.DC1.htmlhttp://www.jimmunol.org/content/suppl/2011/12/30/jimmunol.110228

Referenceshttp://www.jimmunol.org/content/188/3/1275.full#ref-list-1

, 20 of which you can access for free at: cites 60 articlesThis article

Subscriptionshttp://jimmunol.org/subscriptions

is online at: The Journal of ImmunologyInformation about subscribing to

Permissionshttp://www.aai.org/ji/copyright.htmlSubmit copyright permission requests at:

Email Alertshttp://jimmunol.org/cgi/alerts/etocReceive free email-alerts when new articles cite this article. Sign up at:

Print ISSN: 0022-1767 Online ISSN: 1550-6606. Immunologists, Inc. All rights reserved.Copyright © 2012 by The American Association of9650 Rockville Pike, Bethesda, MD 20814-3994.The American Association of Immunologists, Inc.,

is published twice each month byThe Journal of Immunology

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 2: Dominant Th2 Differentiation of Human Regulatory T Cells

The Journal of Immunology

Dominant Th2 Differentiation of Human Regulatory T Cellsupon Loss of FOXP3 Expression

Leo Hansmann, Christian Schmidl, Janina Kett, Lena Steger, Reinhard Andreesen,

Petra Hoffmann, Michael Rehli, and Matthias Edinger

CD4+CD25+FOXP3+ regulatory T cells (Treg) are pivotal for peripheral self-tolerance. They prevent immune responses to auto-

and alloantigens and are thus under close scrutiny as cellular therapeutics for autoimmune diseases and the prevention or

treatment of alloresponses after organ or stem cell transplantation. We previously showed that human Treg with a memory cell

phenotype, but not those with a naive phenotype, rapidly downregulate expression of the lineage-defining transcription factor

FOXP3 upon in vitro expansion. We now compared the transcriptomes of stable FOXP3+ Treg and converted FOXP32 ex-Treg

by applying a newly developed intranuclear staining protocol that permits the isolation of intact mRNA from fixed, permeabilized,

and FACS-purified cell populations. Whole-genome microarray analysis revealed strong and selective upregulation of Th2 sig-

nature genes, including GATA-3, IL-4, IL-5, and IL-13, upon downregulation of FOXP3. Th2 differentiation of converted FOXP32

ex-Treg occurred even under nonpolarizing conditions and could not be prevented by IL-4 signaling blockade. Thus, our studies

identify Th2 differentiation as the default developmental program of human Treg after downregulation of FOXP3. The Journal

of Immunology, 2012, 188: 1275–1282.

Natural regulatory T cells (Treg) are indispensable for themaintenance of dominant self-tolerance and can suppressthe activation, proliferation, and effector function of a

wide range of immune cells, including CD4+ and CD8+ T cells,NK, NK T cells, B cells, and APC. They are thymus-derived andcharacterized by the expression of CD4, CD25, and the tran-scription factor FOXP3 (1). FOXP3 is pivotal for the developmentand function of Treg, and loss-of-function mutations of FOXP3cause lethal autoimmune syndromes in mice and man (2, 3).During Treg development and in mature peripheral Treg, FOXP3represses many inflammation-associated genes but also positivelyinduces a gene expression profile that supports Treg function (4,5). This Treg profile seems to depend on the strength and stabilityof FOXP3 expression, as Treg function is partially lost in genet-ically modified mice that express only low Foxp3 levels (6). Thestability of FOXP3 expression is in parts regulated by epigeneticmechanisms, as shown by the differential DNA methylation pat-

tern of the FOXP3 locus and differential histone modifications inTreg and conventional CD4+ T cells (7–11). Recent studies em-ploying conditional Foxp3 knockout mice revealed that periph-erally induced suppressor cell populations do not compensate forthe lack of Treg (12, 13). Thus, thymus-derived Treg are crucialfor the preservation of peripheral tolerance, and their adoptivetransfer is a promising strategy for the treatment of inflammatorybowel disease (14), autoimmunity (9, 15), and the prevention ofalloresponses after solid organ (16) or stem cell transplantation(17–19). For such applications, we previously described culturemethods that permit the 100–1000-fold expansion of human Tregin vitro within 2 to 3 wk (20, 21). Yet, we and others (22) re-ported that Treg selected on the basis of a CD4+CD25high

CD127low/neg phenotype were heterogeneous with respect tothe frequency of FOXP3+ cells after in vitro expansion, andwe confirmed the downregulation of FOXP3 in Treg clones. Theloss of FOXP3 was almost exclusively confined to CD45RA2

memory-type Treg, whereas CD45RA+ naive Treg homogene-ously maintained FOXP3 expression even after 3 wk in culture(9, 20). Based on these findings, we suggested selectingCD45RA+ Treg for the generation of Treg products for clinicaltrials, whereas the fate of CD45RA2 Treg after in vitro stimu-lation required further clarification. For this purpose, we nowdeveloped new methods that permit the isolation of intact mRNAfrom fixed, permeabilized, and FOXP3-stained, FACS-sortedcells to compare the differential gene expression profiles inconverted versus stable Treg using whole-genome microarrays.We found that Treg rapidly and strongly upregulate Th2 genesupon loss of FOXP3 expression. These findings were confirmedon a protein level as converted Treg secrete high amounts ofIL-4, IL-5, and IL-13, but hardly any Th1 or Th17 cytokines.Thus, using new technologies that permit the examination of hu-man Treg with the same accuracy as in murine Foxp3-reportermodels, we now demonstrate the dominant conversion of humanTreg into Th2 cells upon in vitro stimulation. These findings arehighly relevant for researchers planning adoptive cell therapieswith in vitro-expanded Treg.

Department of Hematology and Oncology, University Hospital Regensburg, 93053Regensburg, Germany

Received for publication August 8, 2011. Accepted for publication November 30,2011.

This work was supported by research funds from the German Research Society (KFO146 to M.E., M.R., and P.H.), the Bavarian Immunotherapy Network, the WilhelmSander Foundation (to M.E.), and the Jose Carreras Foundation (flow cytometryfacility).

The microarray data presented in this article have been submitted to the NationalCenter for Biotechnology Information/Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE26190) under accession number GSE26190.

Address correspondence and reprint requests to Prof. Matthias Edinger and Prof.Michael Rehli, Department of Hematology and Oncology, University HospitalRegensburg, Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany. E-mailaddresses: [email protected] (M.E.) and [email protected] (M.R.)

The online version of this article contains supplemental material.

Abbreviations used in this article: DEPC, diethyl pyrocarbonate; PFA, paraformal-dehyde; qRT-PCR, quantitative RT-PCR; Tconv, conventional T cell; Treg, regulatoryT cell.

Copyright� 2012 by The American Association of Immunologists, Inc. 0022-1767/12/$16.00

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1102288

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 3: Dominant Th2 Differentiation of Human Regulatory T Cells

Materials and MethodsIsolation and cultivation of human Treg

PBMC were isolated from leukapheresis products of healthy volunteers(approved by the ethics committee and after their informed consent) bydensity gradient centrifugation. CD4+CD25highCD45RA+ naive and CD4+

CD25highCD45RA2 memory Treg were purified by FACS (BD FACSAria;BD Biosciences, Heidelberg, Germany) from MACS (Miltenyi Biotec,Bergisch Gladbach, Germany) pre-enriched CD25+ cells. Treg were cul-tured for 11 d on huFcgR+ murine feeder cells (L929 cells) with anti-CD3and anti-CD28 Abs in the presence of IL-2 (300 U/ml) and rested for anadditional 3 to 4 d in medium with IL-2, as described before (20, 23). Forinhibition of IL-4 signaling, anti–IL-4 (clone MP4-25D2, 1 mg/ml; eBio-science, San Diego, CA), anti–IL-4R (clone 25463, 500 ng/ml; R&DSystems, Minneapolis, MN), or both Abs were added during expansion andresting.

Intranuclear FOXP3 staining for subsequent RNA extraction(ethanol/tryptone method)

Surface staining of CD4 (FITC conjugate; BD Biosciences) was carried outin PBS/2% FCS. For FOXP3 staining and subsequent RNA extraction, PBSwith 2% tryptone (Roth, Karlsruhe, Germany) and 0.1% diethyl pyrocar-bonate (DEPC; Roth) was used. The buffer was autoclaved, cooled to 4˚C,and used for all washing and incubation steps throughout the procedureunless otherwise stated. Following surface staining, cells were washedonce, resuspended on a vortex in ice-cold 70% ethanol (up to 73 107 cellsin 2 ml), and fixed for 15 min at 220˚C, then washed twice and resus-pended in 1 ml tryptone/DEPC buffer containing 20 ml rat serum (eBio-science) and 20 ml recombinant RNasin RNase inhibitor (20–40 U/ml;Promega, Madison, WI). After 5 min incubation at 4˚C, 50 ml anti-humanFOXP3 (allophycocyanin conjugate, clone PCH101; eBioscience) wasadded, and cells were incubated for another 25 min at 4˚C in the dark.Cells were washed, resuspended at 1 3 107 cells/ml, and FACS-sorted intotryptone/DEPC buffer. RNA was extracted immediately and its integritychecked with the RNA 6000 Nano Kit on an Agilent Bioanlayzer (AgilentTechnologies, Boblingen, Germany).

Transcription factor and intracellular cytokine staining

Cells were stimulated for 5 h with PMA (20 ng/ml)/ionomycin (1mM) in thepresence of GolgiStop (BD Biosciences) and stained using the FOXP3staining buffer set (eBioscience) and the following anti-human Abs:FOXP3 (PE-, eFluor 450-, or allophycocyanin-conjugated; clone PCH101),IL-4 (allophycocyanin- or AF488-conjugated; clone 8D4-8), IL-5 (PE-conjugated; clone TRFK5), IL-13 (FITC-conjugated; clone PVM13-1),IL-17A (FITC- or allophycocyanin-conjugated; clone eBio64DEC17) (allfrom eBioscience), IFN-g (FITC-, PE-, or allophycocyanin-conjugated;clone B27; BD Biosciences), IL-10 (AF488- or allophycocyanin-con-jugated, clone JES3-9D7; eBioscience; or clone JES3-19F1; BD Bio-sciences), anti-mouse/human GATA-3 (PE-conjugated; clone TWAJ), andanti-mouse/human T-bet (AF647-conjugated; clone eBio4B10), both fromeBioscience. Data were acquired on a BD LSR II or FACSCalibur (BDBiosciences) and analyzed with FlowJo software (Tree Star, Ashland, OR).

RNA extraction

RNA was isolated from FACS-sorted cells using the RNeasy Micro Kit(Qiagen, Hilden, Germany) or TRIzol Reagent (Invitrogen, Darmstadt,Germany). When RNA was extracted from ,1 3 106 cells, glycogen(Roche, Mannheim, Germany) was added during the TRIzol procedure.

Microarray analysis of gene expression

RNA preparations from FOXP3-sorted CD4+CD25highCD45RA2 Tregfrom five donors were analyzed using Whole Human Genome OligoMicroarrays (Agilent Technologies). Labeling and hybridization wereperformed using the Agilent Gene Expression system according to themanufacturer’s instructions (Agilent Technologies). In brief, 50–200 nghigh-quality RNAwere amplified and cyanine 3-CTP labeled with the OneColor Low Input Quick Amp Labeling Kit (Agilent Technologies). Sixhundred nanograms labeled cRNA was fragmented and hybridized on theWhole Human Genome Expression Array G4851A (8 3 60K; AgilentTechnologies). Images were scanned using a DNA microarray scanner,processed using Feature Extraction Software, and further analyzed usingGeneSpring GX (all from Agilent Technologies). Fluorescence signalswere normalized to the 75th percentile and baseline transformed to themedian of all samples. Features were discarded that did not have a mini-mum raw expression value of 40 in at least 3 out of 10 samples. Different

microarray probes covering the same gene were combined using the gene-level technology of the GeneSpring GX software (Agilent Technologies).Expression data for the donor-matched comparisons of variance (Supple-mental Fig. 2) were percentile normalized as above, but not baselinetransformed. Microarray data are available from the National Center forBiotechnology Information/Gene Expression Omnibus repository (acces-sion number GSE26190; http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE26190).

Quantitative RT-PCR

RNA was transcribed into cDNA with Reverse Transcriptase (Promega)and analyzed on an Eppendorf Realplex4 S Cycler (Eppendorf Interna-tional). mRNA expression levels were normalized to b2-microglobulin or18S RNA. Primer sequences are listed in Supplemental Table I.

Western blot analysis

For preparation of whole-cell extracts, 1–5 3 106 cells were washed withPBS, pelleted, dissolved in 100 ml 23 SDS sample buffer with completeprotease and phosphatase inhibitors (Roche) per 5 3 106 cells, heated at95˚C for 10 min, and vortexed for 1 min. For Western blots, equivalents of0.5–0.75 3 106 cells were separated by SDS-PAGE and blotted on poly-vinylidene difluoride membranes, followed by blocking (1 h at roomtemperature with TBS/5% milk powder/0.1% Tween 20) and probing withpolyclonal rabbit anti-human STAT6 (#9362; Cell Signaling Technology,Frankfurt, Germany) or anti-human phospho-STAT6 Abs (Tyr641, #9361;Cell Signaling Technology) at 4˚C overnight. Blots were washed and in-cubated with alkaline phosphatase-conjugated goat anti-rabbit Ab (Dako-Cytomation, Hamburg, Germany). Bands were visualized with ECL solu-tion on Hyperfilm, scanned using a Molecular Dynamics personal densi-tometer SI, and quantified with ImageQuant 5.2 (all from GE Healthcare).

Cytokine detection in culture supernatants

Supernatants were collected on day 11 of Treg cultures. Concentrations ofIL-4, IL-5, IL-13, IL-10, IL-17, and IFN-g were determined using CytokineBead Array Flex-Sets (BD Biosciences) according to the manufacturer’sinstructions.

Statistical analysis

Statistical significance was determined by paired or unpaired two-tailedStudent t test, as indicated in the figure legends. A p value #0.05 wasconsidered significant.

ResultsIsolation of intact RNA from FACS-sorted cells afterintranuclear FOXP3 staining

There is increasing evidence for plasticity within the Treg lineage,which includes the loss of FOXP3 and the conversion of Treg intopotentially proinflammatory Th cell subsets (24–27). The study ofgene expression patterns in FOXP3-losing Treg could extend ourunderstanding of this plasticity and reveal the underlying molec-ular mechanisms. However, paraformaldehyde (PFA) contained incommercially available FOXP3 staining kits induces conforma-tional changes and covalent as well as noncovalent cross-links innucleic acids and proteins and thus impedes the subsequent ex-traction of intact RNA. We therefore established an alternativeFOXP3 staining method using 70% ethanol for fixation and per-meabilization that does not induce such cross-links and thusallows the extraction of intact high-quality RNA. Staining forFOXP3 and subsequent flow cytometric sorting was carried outin tryptone- and RNase inhibitor-containing PBS. Both additiveswere essential for maintaining RNA integrity during the stainingand sorting procedure. The frequencies of FOXP3+ cells amongin vitro-expanded CD45RA2 Treg detected with this protocolwere comparable to those obtained with a commercial FOXP3staining kit (Fig. 1A; divergence ranged from 210.6 to 5.7%; n =4). FACS-separated populations routinely showed .95% purityupon reanalysis (96.9 6 1.2% for FOXP3+ and 98.1 6 0.9% forFOXP32 cells, respectively, n = 5; see also Supplemental Fig. 1).RNA extracted from sorted cells displayed little or no signs of

1276 DOMINANT Th2 DIFFERENTIATION OF HUMAN Treg

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 4: Dominant Th2 Differentiation of Human Regulatory T Cells

degradation (Fig. 1B), and mean RNA integrity numbers (28) were7.9 (range: 7.3–9.0; n = 14) in fixed samples and 9.4 (range: 9.2–9.5; n = 5) in unfixed controls. RNAyield and quality depended onthe time span between fixation and cell lysis for RNA extraction;best results were obtained by keeping preparation times short andsamples cold and RNase-free. Thus, this PFA-free FOXP3 stainingprotocol (referred to as the ethanol/tryptone method) allowed thereliable extraction of largely intact RNA from intranuclearlystained and FACS-sorted cells.

In vitro-expanded CD45RA2 Treg show a Th2 gene expressionsignature upon loss of FOXP3 expression

We previously showed that FOXP3 expression is primarily lost inCD45RA2 memory Treg after in vitro stimulation (20, 22). Toanalyze the fate of FOXP3-losing memory Treg on the level ofgene expression, FACS-sorted CD4+CD25highCD45RA2 Tregwere in vitro expanded and subsequently sorted into FOXP3+ andFOXP32 populations applying the ethanol/tryptone method. RNAwas extracted and analyzed using whole-genome expression mi-croarrays. Combined results of five independent experiments arepresented as heat maps for selected gene classes in Fig. 2A. De-tailed mRNA expression levels of individual cell cultures can befound in Supplemental Fig. 2.As expected, we observed significantly higher mRNA levels

of Treg signature genes (e.g., FOXP3, CTLA4, and IL2RA) inthe FOXP3+ as compared with the corresponding FOXP32 pop-ulation. Yet, surprisingly, a dramatic increase of Th2 cytokinemRNA (IL4, IL5, and IL13) was detected in FOXP32 cells, but nosignificant difference between the two subpopulations with respectto other Th cell lineage- and inflammation-associated cytokinegenes (IFNG, IL6, IL9, IL10, IL12, IL17, IL21, IL23, and IL24).Likewise, key transcription factors for Th2 differentiation, namelyGATA3 and GFI1, showed a significantly higher expression inFOXP32 cells, whereas the genes RORC and STAT1, responsiblefor Th17 and Th1 lineage commitment, respectively, were sup-

pressed. In addition, FOXP32 cells highly expressed GPR44, alsoknown as CRTH2, which encodes the Th2 cell-specific G protein-coupled receptor CD294 (29) (Fig. 2A). Thus, the FOXP32 pop-ulation showed a gene signature dominated by Th2 lineage-associated genes including cytokines, transcription factors, sig-naling molecules, and cell-surface receptors.

FIGURE 1. Isolation of intact RNA from human T cells after intra-

nuclear staining for FOXP3. A, In vitro-expanded CD45RA2 Treg were

stained for FOXP3 using either a commercially available FOXP3 staining

kit (left panel) or the ethanol-tryptone method (right panel). Plots are

representative of n = 4 independent experiments. Numbers indicate per-

centages of cells. B, Electropherograms of RNA extracted from expanded

human CD45RA2 Treg. Cells were either stained for FOXP3 using the

ethanol/tryptone method and separated by FACS (left panel, FOXP32

population; representative of n = 14 independent RNA preparations) or

remained unfixed before RNA extraction (right panel, representative of n =

5 independent preparations). RIN, RNA integrity number.

FIGURE 2. Transcriptome analysis of human FOXP3-sorted CD45RA2

Treg. A, Relative gene expression levels of selected gene classes of

FOXP3-sorted CD45RA2 Treg of five independent donors (microarray

data normalized to the median of all samples) are presented in heat maps

by a pseudocolor scale as indicated. Data were filtered as described in the

Materials and Methods section. Genes that showed significant expression

differences between FOXP3+ and FOXP32 subpopulations (paired Student

t test, asymptotic p value computation, cutoff p # 0.05) with a fold change

of $2 are marked with an asterisk. For detailed gene expression levels

of selected genes in individual Treg cultures, see Supplemental Fig. 2. B,

Microarray results were validated by qRT-PCR for the indicated genes in

FOXP3+ (blue bars) and FOXP32 cells (green bars) derived from

CD45RA2 Treg cultures. FOXP3+ cells sorted from expanded CD45RA+

Treg (red bars) and unsorted CD45RA2 Treg (gray bars) served as con-

trols. Values represent means + SD (n $ 3). Significant differences in gene

expression between FOXP3+ and FOXP32 cells derived from the

CD45RA2 Treg population are indicated above bars (*p # 0.05, **p #

0.01, ***p # 0.001, paired two-tailed Student t test). ND, not detected.

The Journal of Immunology 1277

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 5: Dominant Th2 Differentiation of Human Regulatory T Cells

Because STAT6 is critically involved in IL-4–induced Th2 de-velopment (30), we also asked whether the expression of typicalSTAT6 target genes (including LTB, SOCS1, and IRF8) would bealtered during Treg conversion. Interestingly, their expression lev-els were not significantly different between FOXP3+ and FOXP32

populations, nor were the levels of MAF encoding a trans-acti-vator of the IL-4 gene (31).Microarray data were validated at the single gene level using

quantitative RT-PCR (qRT-PCR) (Fig. 2B). As controls, RNA iso-lated from in vitro-expanded, FOXP3-stained, and sorted CD45RA+

Treg as well as from expanded bulk CD45RA2 Treg without priorfixation were analyzed (Fig. 2B). As expected, FOXP3+ cells fromcultures of CD45RA2 and CD45RA+ Treg showed comparablyhigh FOXP3 and low GATA3 mRNA expression. When totalCD45RA2 unfixed cells were compared with ethanol/tryptone-fixed subpopulations, they mainly showed intermediate mRNAexpression, confirming the reliability of the RNA preparation pro-cedure (Fig. 2B). Taken together, the gene expression profile ofFOXP32 cells developing from initially FOXP3+CD45RA2 Tregupon in vitro stimulation indicated Th2 differentiation.

FOXP32 cells from CD45RA2 Treg cultures predominantlyproduce Th2 cytokines

To correlate the mRNA expression data with cytokine produc-tion, freshly isolated as well as in vitro-expanded CD45RA2 andCD45RA+ Treg were stained for FOXP3 and Th cell lineage-defining cytokines (Fig. 3). In freshly isolated cells, the expres-sion frequency for one or more Th cell lineage signature cyto-kines (IL-4, IL-10, IL-13, IFN-g, and IL-17) never exceeded 5%.In contrast, after in vitro expansion, ∼30% of the cells withinCD45RA2 Treg cultures started to produce one or more of theTh2 cytokines IL-4, IL-5, and IL-13 (Fig. 3A). Although we alsodetected IFN-g+ and/or IL-17+ cells in these cultures [as previ-ously described by us and others (22, 25, 32, 33)], their fre-quencies were significantly lower than those of the Th2 cytokineproducers (Fig. 3A). Th2 cytokine production was largely confinedto the FOXP32 subpopulation, whereas IFN-g, IL-10, and IL-17producers were almost equally distributed between FOXP3+ andFOXP32 cells (Fig. 3B). Confirming our previous findings,CD45RA+ Treg remained uniformly FOXP3+ and comprisedhardly any cytokine-producing cells before and after in vitro ex-pansion (20, 22 and data not shown).

Th2 cytokine production in expanded CD45RA2 Treg isassociated with high GATA3 expression

GATA3, the lineage-defining transcription factor for Th2 cells,was highly expressed in CD45RA2 Treg that had downregulatedFOXP3 upon in vitro stimulation (Fig. 2). To analyze whetherGATA3 expression was restricted to Th2 cytokine producers,in vitro expanded CD45RA2 Treg were simultaneously stained forcytokine production and the presence of GATA-3. GATA-3 ex-pression was significantly higher in IL-4–producing cells whencompared with IL-17 and IFN-g producers (Fig. 4).

Inhibition of IL-4 signaling does not block Th2 differentiationin CD45RA2 Treg

Th2 differentiation from naive conventional T cells (Tconv) ismainly driven by IL-4 (30). Because CD45RA2 Treg secreted IL-4 and other Th2 cytokines under the nonpolarizing culture con-ditions applied in this study, we next asked to which extent en-dogenous IL-4 supported the conversion of initially FOXP3+ Treginto cells of the Th2 lineage. To block IL-4 signaling, CD45RA2

Treg were cultured in the presence of anti–IL-4 Abs, anti–IL-4RAbs, or both during the entire expansion period. The treatment

effectively prevented phosphorylation of the transcription factorSTAT6, a crucial step in IL-4 signaling and Th2 induction (Fig.5A, 5B). However, despite profound inhibition of STAT6 acti-vation, the frequencies of cytokine-secreting cells in CD45RA2

Treg cultures were only marginally altered, with a slight but notstatistically significant reduction of Th2 cytokine producers anda minor, statistically insignificant increase in IFN-g– or IL-17–

FIGURE 3. CD45RA2 Treg predominantly produce Th2 cytokines upon

loss of FOXP3 expression. CD45RA2 Treg, either ex vivo (d 0) or af-

ter in vitro expansion (d 15), were stimulated for 5 h with PMA/iono-

mycin and stained for FOXP3 and various cytokines. A, Percentages of

CD45RA2 Treg producing the indicated cytokines. IL-5 was not deter-

mined ex vivo. Horizontal bars represent means of n = 3–17 independent

analyses with cells from different donors. ***p # 0.001 as compared with

the number of IL-4 producers; two-tailed unpaired Student t test. B, Pro-

portion of FOXP3-expressing cells among indicated cytokine producers

after in vitro expansion. Horizontal bars represent means of n = 9–17 in-

dependent analyses. *p # 0.05, ***p # 0.001 as compared with the

number of FOXP3+ cells among IL-4 producers; two-tailed unpaired

Student t test. C, Representative example of IL-4– and IL-13–producing

cells among CD45RA2 Treg ex vivo (d 0) and after in vitro expansion

(d 15).

1278 DOMINANT Th2 DIFFERENTIATION OF HUMAN Treg

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 6: Dominant Th2 Differentiation of Human Regulatory T Cells

secreting cells (Fig. 5C, 5D). In line with these findings, cytokineconcentrations in supernatants of blocked and unblocked culturesshowed no significant differences, except for IL-4, because IL-4neutralization by anti–IL-4 Abs expectedly reduced IL-4 levels,whereas blockade of IL-4R increased IL-4 concentrations, provingthat IL-4 is consumed in such cultures (Fig. 5E). Overall, thesedata indicate that Th2 differentiation of CD45RA2 Treg does notrequire endogenous IL-4 but seems to represent the default de-velopmental pathway upon loss of FOXP3.

DiscussionThere is increasing evidence that Treg lineage commitment maynot be irreversible, as downregulation of FOXP3 and consecutiveexpression of Th1 and Th2 cytokines by Treg has been observed byseveral groups (25, 27, 32, 34–36). We showed that, in particular,human CD45RA2 memory-type Treg lose FOXP3 expressionupon in vitro stimulation, whereas CD45RA+ naive Treg maintainhigh FOXP3 levels even after extended culture periods. Impor-tantly, both FACS-purified starting populations contained equiva-lent frequencies of FOXP3+ cells before in vitro stimulation(.93%), and both showed equally homogeneous demethylationat the Treg-specific demethylated region, a sensitive epigeneticmark for the identification of Treg and currently the most re-liable marker for the exclusion of contaminations by induced ornon-Treg (7, 10, 37). Furthermore, we ultimately proved loss ofFOXP3 expression by the serial examination of human Tregclones in our previous studies (22). Because Treg instability maybe detrimental for future adoptive cell therapies [or those alreadyunderway at some centers (17, 38)], we now examined the fate ofTreg that lose FOXP3 upon in vitro stimulation. For this purpose,we developed a cell fixation and permeabilization protocol thatallowed the isolation of intact mRNA from FOXP3-stained andFACS-sorted cells. In contrast to previously described methods(39), the isolated RNA was not degraded but of high quality andsuited for sensitive downstream assays, such as qRT-PCR andmicroarray-based whole-genome expression analyses. The use ofethanol for fixation prevented the cross-linking of nucleic acidsand proteins (40) that is caused by PFA contained in commercialstaining kits. Furthermore, our method improves previously de-scribed RNA isolation protocols developed by Esser and col-leagues (41) because it permits the simultaneous detection offluorescently labeled surface markers. Because this technology isapplicable to a wide variety of transcription factors and cell types,it may advance many areas in cellular biology.Classical differentiation of naive CD4+ T cells into Th1 or Th2

cells is mainly induced by the local cytokine milieu as well as thetype of Ag and APC. IFN-g and IL-12 polarize toward Th1 dif-ferentiation via the transcription factors STAT4, STAT1, and T-bet(42, 43). Th2 cells are induced by IL-4 that induces GATA-3,usually in a STAT6-dependent fashion, and the hallmark of Th2cells is their secretion of IL-4, IL-5, and IL-13 (44–47). T-bet and

GATA-3 reciprocally repress each other (48–50), and FOXP3 inTreg is supposed to prohibit expression of both of these lineage-defining transcription factors (51). Nevertheless, coexpression ofFoxp3 and T-bet has been observed in activated murine Treg undertype 1 inflammatory conditions, which permits their migration toinflammatory sites, but does not induce Th1 cytokine secretion(52). Our comparative analysis of human CD45RA2 memoryTreg-derived FOXP3+ and FOXP32 subpopulations now revealedthat typical marker genes of Treg, such as CTLA4, LAG3, IL2RA,LGALS3, and LRRC32, are downregulated upon loss of FOXP3expression, whereas IL7R and CD40LG, usually not expressed byTreg, are upregulated. More strikingly, the cells losing FOXP3upon in vitro stimulation under nonpolarizing conditions con-verted into Th2-like cells as they started to overexpress a numberof Th2-specific genes, such as the signature cytokines IL4, IL5,and IL13, the transcription factors GATA3 and GFI1, and thesurface receptor GPR44. In contrast, expression of Th1 or Th17signature genes, such as TBX21 and RORC, was even lower inFOXP32 as compared with FOXP3+ cells isolated from thesame cultures (Fig. 2). The conversion into Th2 cells upon lossof FOXP3 expression dramatically exceeded the previously de-scribed differentiation into Th1 or Th17 cells (22, 25, 32, 33).Both the high frequency of Th2 cytokine producers amongFOXP32CD45RA2 Treg and the substantial concentrations ofTh2 cytokines in CD45RA2 Treg culture supernatants corrobo-rated these results. In murine studies, induction of cytokine se-cretion in former Treg has also been observed, either upon Foxp3deletion (53) or upon expression of a nonfunctional Foxp3 protein(54). Whereas Rudensky and coworkers (53) found only Th1cytokines in C57BL/6 animals, Chatila’s group (54) also observeda dramatic increase in IL-4 secretion after Foxp3 downregulationin BALB/c mice. Thus, a genetic predisposition may influence theconversion of Treg. Interestingly, however, artificial attenuation ofFoxp3 expression has been described to induce predominantlya Th2 phenotype in Foxp3low-expressing cells even in C57BL/6mice (6), and this conversion seems to be an intrinsic develop-mental program that occurs independently of IL4/STAT6 sig-naling, yet still requires GATA-3 expression (55). In Th2 dif-ferentiation of naive Tconv, IL-4 causes STAT6 phosphorylationby JAK1 and JAK3 kinases, leading to STAT6 dimerization andnuclear translocation that in turn supports IL4 and IL4R as well asGATA3 transcription (30). Blocking IL-4/STAT6 signaling in hu-man Treg by the addition of anti–IL-4 Abs, anti–IL-4R Abs, orboth during culture resulted in a dramatic reduction of STAT6phosphorylation, as expected. Yet, the frequencies of IL-4–, IL-5–,and IL-13–producing cells and the amount of secreted Th2 cyto-kines were only marginally diminished, suggesting that auto- orparacrine IL-4/STAT6 signaling is not the main cause of Th2conversion. To which extent IL-4–independent signaling cascadesare involved in Th2 conversion of human Treg requires furtherclarification. For murine T cells, it has previously been shown that

FIGURE 4. Th2 cytokine production in in vitro-expanded CD45RA2 Treg is associated with high GATA-3 expression. In vitro-expanded CD45RA2

Treg were stimulated for 5 h with PMA/ionomycin, then stained for GATA-3 and indicated cytokines. The histogram shows GATA-3 expression of IL-4+,

IL-17+, or IFN-g+ cells, respectively. Mean fluorescence intensity for GATA-3 was significantly different between IL-4+ and IFN-g+ cells (p = 0.046; n = 6;

two-tailed unpaired Student t test) as well as IL-4+ and IL-17+ cells (p = 0.009; n = 5; two-tailed unpaired Student t test).

The Journal of Immunology 1279

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 7: Dominant Th2 Differentiation of Human Regulatory T Cells

IL-2 combined with CD28 costimulation can induce Th2 differ-entiation in an IL-4/STAT6-independent but STAT5-dependentmanner (56, 57). Because our stimulation conditions provide allrequired components (CD3, CD28, and high-dose IL-2 stimula-tion), this pathway may be involved in Treg conversion. However,we reason instead that Th2 conversion of CD45RA2 Treg repre-sents a developmental program that is imprinted on the cells byrepeated contact to (self-) Ag in vivo, as CD45RA+ naive Tregs donot convert although exposed to the same in vitro conditions. Insupport of this hypothesis, an increased GATA-3 expression in

CD45RA2 Treg (as compared with CD45RA+ Treg and Tconv)was detectable already before in vitro expansion (SupplementalFig. 3). In mice, Th2 conversion of wild-type Tregs in vivo hasbeen described (55) as well as conversion into IL-4–secretingfollicular B Th cells (58), and it remains to be determined whetherthis is also a frequent event in humans, in whom diminishedFOXP3 expression of Treg has been observed in patients withatopic diseases (59).In summary, by performing, to our knowledge, the first com-

parative transcriptome analyses of human Treg and their converted

FIGURE 5. Inhibition of IL-4 signaling does not

block Th2 differentiation of CD45RA2 Treg during

in vitro expansion. CD45RA2 Treg were expanded

in the presence or absence of anti-IL-4, anti–IL-4R,

or both Abs. A, Phosphorylated STAT6 (pSTAT6) in

whole-cell extracts was analyzed by Western blot.

Total STAT6 was analyzed as a loading control. Shown

is one representative immunostaining (n = 3). B,

Western blot band intensities were quantified with

a densitometer. Phospho-STAT6 band intensities of

unblocked cultures were set to 100%. Values represent

means + SD (n = 3; two-tailed unpaired Student t test

performed with absolute values). C and D, Cells were

stimulated for 5 h with PMA/ionomycin, then stained

for FOXP3 and indicated cytokines. Dot plots in C

show one representative experiment. Bars in D repre-

sent mean percentages + SD of cytokine producers

among expanded CD45RA2 Treg. For better compar-

ison of n = 5–9 individual cultures, the numbers of

cytokine-producing cells in unblocked cultures were

scaled to 100%. Unblocked cultures contained on av-

erage 29.1, 15.5, and 26.4% IL-4, IL-5, and IL-13

producers, respectively, and 9.3, 9.9, and 7.1% IL-10,

IFN-g, and IL-17 producers, respectively. E, Super-

natants from treated or untreated CD45RA2 Treg

cultures were analyzed for cytokine content on day 11

of in vitro expansion. Bars represent means + SD of

n = 5–9 independent cultures. *p # 0.05, **p # 0.01,

***p # 0.001 as compared with unblocked cultures;

two-tailed unpaired Student t test.

1280 DOMINANT Th2 DIFFERENTIATION OF HUMAN Treg

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 8: Dominant Th2 Differentiation of Human Regulatory T Cells

progeny identified and separated on the basis of their FOXP3expression level, we show that differentiation toward a Th2 phe-notype represents the dominant pathway of human Treg upon lossof FOXP3 expression. These findings further elucidate the de-velopmental plasticity of Treg in humans (60) and are thus of highrelevance for current as well as future adoptive Treg therapies.

AcknowledgmentsWe thank R. Eder, J. Stahl, and I. Ritter for excellent cell sorting and tech-

nical support and N. Ahrens, Institute of Transfusion Medicine, University

Hospital Regensburg, Regensburg, Germany, for provision of leukapheresis

products.

DisclosuresThe authors have no financial conflicts of interest.

References1. Sakaguchi, S., T. Yamaguchi, T. Nomura, and M. Ono. 2008. Regulatory T cells

and immune tolerance. Cell 133: 775–787.2. Brunkow, M. E., E. W. Jeffery, K. A. Hjerrild, B. Paeper, L. B. Clark, S.-A. Yasayko,

J. E. Wilkinson, D. Galas, S. F. Ziegler, and F. Ramsdell. 2001. Disruption of anew forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferativedisorder of the scurfy mouse. Nat. Genet. 27: 68–73.

3. Wildin, R. S., F. Ramsdell, J. Peake, F. Faravelli, J.-L. Casanova, N. Buist,E. Levy-Lahad, M. Mazzella, O. Goulet, L. Perroni, et al. 2001. X-linked neo-natal diabetes mellitus, enteropathy and endocrinopathy syndrome is the humanequivalent of mouse scurfy. Nat. Genet. 27: 18–20.

4. Hill, J. A., M. Feuerer, K. Tash, S. Haxhinasto, J. Perez, R. Melamed, D. Mathis,and C. Benoist. 2007. Foxp3 transcription-factor-dependent and -independentregulation of the regulatoryT cell transcriptional signature. Immunity 27: 786–800.

5. Zheng, Y., S. Z. Josefowicz, A. Kas, T.-T. Chu, M. A. Gavin, and A. Y. Rudensky.2007. Genome-wide analysis of Foxp3 target genes in developing and matureregulatory T cells. Nature 445: 936–940.

6. Wan, Y. Y., and R. A. Flavell. 2007. Regulatory T-cell functions are subvertedand converted owing to attenuated Foxp3 expression. Nature 445: 766–770.

7. Baron, U., S. Floess, G. Wieczorek, K. Baumann, A. Grutzkau, J. Dong,A. Thiel, T. J. Boeld, P. Hoffmann, M. Edinger, et al. 2007. DNA demethylationin the human FOXP3 locus discriminates regulatory T cells from activatedFOXP3(+) conventional T cells. Eur. J. Immunol. 37: 2378–2389.

8. Kim, H. P., and W. J. Leonard. 2007. CREB/ATF-dependent T cell receptor-induced FoxP3 gene expression: a role for DNA methylation. J. Exp. Med. 204:1543–1551.

9. Miyara, M., Y. Yoshioka, A. Kitoh, T. Shima, K. Wing, A. Niwa, C. Parizot,C. Taflin, T. Heike, D. Valeyre, et al. 2009. Functional delineation and differ-entiation dynamics of human CD4+ T cells expressing the FoxP3 transcriptionfactor. Immunity 30: 899–911.

10. Schmidl, C., M. Klug, T. J. Boeld, R. Andreesen, P. Hoffmann, M. Edinger, andM. Rehli. 2009. Lineage-specific DNA methylation in T cells correlates withhistone methylation and enhancer activity. Genome Res. 19: 1165–1174.

11. Wei, G., L. Wei, J. Zhu, C. Zang, J. Hu-Li, Z. Yao, K. Cui, Y. Kanno, T. Y. Roh,W. T. Watford, et al. 2009. Global mapping of H3K4me3 and H3K27me3 revealsspecificity and plasticity in lineage fate determination of differentiating CD4+T cells. Immunity 30: 155–167.

12. Kim, J., K. Lahl, S. Hori, C. Loddenkemper, A. Chaudhry, P. deRoos,A. Rudensky, and T. Sparwasser. 2009. Cutting edge: depletion of Foxp3+ cellsleads to induction of autoimmunity by specific ablation of regulatory T cells ingenetically targeted mice. J. Immunol. 183: 7631–7634.

13. Kim, J. M., J. P. Rasmussen, and A. Y. Rudensky. 2007. Regulatory T cellsprevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Im-munol. 8: 191–197.

14. Mottet, C., H. H. Uhlig, and F. Powrie. 2003. Cutting edge: cure of colitis byCD4+CD25+ regulatory T cells. J. Immunol. 170: 3939–3943.

15. Brusko, T. M., A. L. Putnam, and J. A. Bluestone. 2008. Human regulatoryT cells: role in autoimmune disease and therapeutic opportunities. Immunol. Rev.223: 371–390.

16. Nadig, S.N., J.Wieckiewicz,D.C.Wu,G.Warnecke,W.Zhang, S.Luo,A.Schiopu,D. P. Taggart, and K. J.Wood. 2010. In vivo prevention of transplant arteriosclerosisby ex vivo-expanded human regulatory T cells. Nat. Med. 16: 809–813.

17. Di Ianni, M., F. Falzetti, A. Carotti, A. Terenzi, F. Castellino, E. Bonifacio,B. Del Papa, T. Zei, R. I. Ostini, D. Cecchini, et al. 2011. Tregs prevent GVHDand promote immune reconstitution in HLA-haploidentical transplantation.Blood 117: 3921–3928.

18. Edinger, M., P. Hoffmann, J. Ermann, K. Drago, C. G. Fathman, S. Strober, andR. S. Negrin. 2003. CD4+CD25+ regulatory T cells preserve graft-versus-tumoractivity while inhibiting graft-versus-host disease after bone marrow transplan-tation. Nat. Med. 9: 1144–1150.

19. Hoffmann, P., J. Ermann, M. Edinger, C. G. Fathman, and S. Strober. 2002.Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J. Exp. Med.196: 389–399.

20. Hoffmann, P., R. Eder, T. J. Boeld, K. Doser, B. Piseshka, R. Andreesen, andM. Edinger. 2006. Only the CD45RA+ subpopulation of CD4+CD25high T cellsgives rise to homogeneous regulatory T-cell lines upon in vitro expansion. Blood108: 4260–4267.

21. Hoffmann, P., R. Eder, L. A. Kunz-Schughart, R. Andreesen, and M. Edinger.2004. Large-scale in vitro expansion of polyclonal human CD4(+)CD25highregulatory T cells. Blood 104: 895–903.

22. Hoffmann, P., T. J. Boeld, R. Eder, J. Huehn, S. Floess, G. Wieczorek, S. Olek,W. Dietmaier, R. Andreesen, and M. Edinger. 2009. Loss of FOXP3 expressionin natural human CD4+CD25+ regulatory T cells upon repetitive in vitro stim-ulation. Eur. J. Immunol. 39: 1088–1097.

23. Hoffmann, P., R. Eder, and M. Edinger. 2011. Polyclonal expansion of humanCD4(+)CD25(+) regulatory T cells. Methods Mol. Biol. 677: 15–30.

24. Komatsu, N., M. E. Mariotti-Ferrandiz, Y. Wang, B. Malissen, H. Waldmann,and S. Hori. 2009. Heterogeneity of natural Foxp3+ T cells: a committed reg-ulatory T-cell lineage and an uncommitted minor population retaining plasticity.Proc. Natl. Acad. Sci. USA 106: 1903–1908.

25. Schmidl, C., L. Hansmann, R. Andreesen, M. Edinger, P. Hoffmann, andM. Rehli. 2011. Epigenetic reprogramming of the RORC locus during in vitroexpansion is a distinctive feature of human memory but not naıve Treg. Eur. J.Immunol. 41: 1491–1498.

26. Xu, L., A. Kitani, I. Fuss, and W. Strober. 2007. Cutting edge: regulatory T cellsinduce CD4+CD25-Foxp3- T cells or are self-induced to become Th17 cells inthe absence of exogenous TGF-beta. J. Immunol. 178: 6725–6729.

27. Zhou, X., S. L. Bailey-Bucktrout, L. T. Jeker, C. Penaranda, M. Martınez-Llordella, M. Ashby, M. Nakayama, W. Rosenthal, and J. A. Bluestone. 2009.Instability of the transcription factor Foxp3 leads to the generation of pathogenicmemory T cells in vivo. Nat. Immunol. 10: 1000–1007.

28. Schroeder, A., O. Mueller, S. Stocker, R. Salowsky, M. Leiber, M. Gassmann,S.Lightfoot,W.Menzel,M.Granzow, andT.Ragg. 2006. TheRIN: anRNAintegritynumber for assigning integrity values to RNA measurements. BMC Mol. Biol. 7: 3.

29. Cosmi, L., F. Annunziato, M. I. G. Galli, R. M. E. Maggi, K. Nagata, andS. Romagnani. 2000. CRTH2 is the most reliable marker for the detection ofcirculating human type 2 Th and type 2 T cytotoxic cells in health and disease.Eur. J. Immunol. 30: 2972–2979.

30. Kaplan, M. H., U. Schindler, S. T. Smiley, and M. J. Grusby. 1996. Stat6 isrequired for mediating responses to IL-4 and for development of Th2 cells.Immunity 4: 313–319.

31. Lin, B.-S., P.-Y. Tsai, W.-Y. Hsieh, H.-W. Tsao, M.-W. Liu, R. Grenningloh,L.-F. Wang, I. C. Ho, and S.-C. Miaw. 2010. SUMOylation attenuates c-Maf-dependent IL-4 expression. Eur. J. Immunol. 40: 1174–1184.

32. Ayyoub, M., F. Deknuydt, I. Raimbaud, C. Dousset, L. Leveque, G. Bioley, andD. Valmori. 2009. Human memory FOXP3+ Tregs secrete IL-17 ex vivo andconstitutively express the T(H)17 lineage-specific transcription factor ROR-gamma t. Proc. Natl. Acad. Sci. USA 106: 8635–8640.

33. McClymont, S. A., A. L. Putnam, M. R. Lee, J. H. Esensten, W. Liu,M. A. Hulme, U. Hoffmuller, U. Baron, S. Olek, J. A. Bluestone, andT. M. Brusko. 2011. Plasticity of human regulatory T cells in healthy subjectsand patients with type 1 diabetes. J. Immunol. 186: 3918–3926.

34. Beriou, G., C. M. Costantino, C. W. Ashley, L. Yang, V. K. Kuchroo, C. Baecher-Allan, and D. A. Hafler. 2009. IL-17-producing human peripheral regulatoryT cells retain suppressive function. Blood 113: 4240–4249.

35. Zhou, L., M. M. Chong, and D. R. Littman. 2009. Plasticity of CD4+ T celllineage differentiation. Immunity 30: 646–655.

36. Zhu, J., and W. E. Paul. 2010. Heterogeneity and plasticity of T helper cells. CellRes. 20: 4–12.

37. Hansmann, L., C. Schmidl, T. J. Boeld, R. Andreesen, P. Hoffmann, M. Rehli, andM. Edinger. 2010. Isolation of intact genomic DNA from FOXP3-sorted humanregulatory T cells for epigenetic analyses. Eur. J. Immunol. 40: 1510–1512.

38. Brunstein, C. G., J. S. Miller, Q. Cao, D. H. McKenna, K. L. Hippen, J. Curtsinger,T. Defor, B. L. Levine, C. H. June, P. Rubinstein, et al. 2011. Infusion of ex vivoexpanded T regulatory cells in adults transplanted with umbilical cord blood:safety profile and detection kinetics. Blood 117: 1061–1070.

39. Boniface, K., W. M. Blumenschein, K. Brovont-Porth, M. J. McGeachy,B. Basham, B. Desai, R. Pierce, T. K. McClanahan, S. Sadekova, and R. de WaalMalefyt. 2010. Human Th17 cells comprise heterogeneous subsets includingIFN-gamma-producing cells with distinct properties from the Th1 lineage. J.Immunol. 185: 679–687.

40. Fowler, C. B., T. J. O’Leary, and J. T. Mason. 2008. Modeling formalin fixationand histological processing with ribonuclease A: effects of ethanol dehydrationon reversal of formaldehyde cross-links. Lab. Invest. 88: 785–791.

41. Esser, C., C. Gottlinger, J. Kremer, C. Hundeiker, andA. Radbruch. 1995. Isolationof full-size mRNA from ethanol-fixed cells after cellular immunofluorescencestaining and fluorescence-activated cell sorting (FACS). Cytometry 21: 382–386.

42. Lighvani, A. A., D. M. Frucht, D. Jankovic, H. Yamane, J. Aliberti, B. D. Hissong,B. V. Nguyen, M. Gadina, A. Sher, W. E. Paul, and J. J. O’Shea. 2001. T-bet israpidly induced by interferon-gamma in lymphoid and myeloid cells. Proc. Natl.Acad. Sci. USA 98: 15137–15142.

43. Thieu, V. T., Q. Yu, H. C. Chang, N. Yeh, E. T. Nguyen, S. Sehra, andM. H. Kaplan. 2008. Signal transducer and activator of transcription 4 is requiredfor the transcription factor T-bet to promote T helper 1 cell-fate determination.Immunity 29: 679–690.

44. Shimoda, K., J. van Deursen, M. Y. Sangster, S. R. Sarawar, R. T. Carson,R. A. Tripp, C. Chu, F. W. Quelle, T. Nosaka, D. A. A. Vignali, et al. 1996. Lackof IL-4-induced Th2 response and IgE class switching in mice with disruptedStat6 gene. Nature 380: 630–633.

The Journal of Immunology 1281

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from

Page 9: Dominant Th2 Differentiation of Human Regulatory T Cells

45. Takeda, K., T. Tanaka, W. Shi, M. Matsumoto, M. Minami, S.-i. Kashiwamura,K. Nakanishi, N. Yoshida, T. Kishimoto, and S. Akira. 1996. Essential role ofStat6 in IL-4 signalling. Nature 380: 627–630.

46. Zheng, W., and R. A. Flavell. 1997. The transcription factor GATA-3 is neces-sary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell 89:587–596.

47. Zhu, J., B. Min, J. Hu-Li, C. J. Watson, A. Grinberg, Q. Wang, N. Killeen,J. F. Urban, Jr., L. Guo, andW. E. Paul. 2004. Conditional deletion of Gata3 showsits essential function in T(H)1-T(H)2 responses. Nat. Immunol. 5: 1157–1165.

48. Szabo, S. J., A. S. Dighe, U. Gubler, and K. M. Murphy. 1997. Regulation of theinterleukin (IL)-12R beta 2 subunit expression in developing T helper 1 (Th1)and Th2 cells. J. Exp. Med. 185: 817–824.

49. Kurata, H., H. J. Lee, A. O’Garra, and N. Arai. 1999. Ectopic expression ofactivated Stat6 induces the expression of Th2-specific cytokines and transcrip-tion factors in developing Th1 cells. Immunity 11: 677–688.

50. Djuretic, I. M., D. Levanon, V. Negreanu, Y. Groner, A. Rao, and K. M. Ansel.2007. Transcription factors T-bet and Runx3 cooperate to activate Ifng and si-lence Il4 in T helper type 1 cells. Nat. Immunol. 8: 145–153.

51. Zheng, Y., and A. Y. Rudensky. 2007. Foxp3 in control of the regulatory T celllineage. Nat. Immunol. 8: 457–462.

52. Koch, M. A., G. Tucker-Heard, N. R. Perdue, J. R. Killebrew, K. B. Urdahl, andD. J. Campbell. 2009. The transcription factor T-bet controls regulatory T cellhomeostasis and function during type 1 inflammation.Nat. Immunol. 10: 595–602.

53. Williams, L. M., and A. Y. Rudensky. 2007. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires con-tinued expression of Foxp3. Nat. Immunol. 8: 277–284.

54. Lin, W., D. Haribhai, L. M. Relland, N. Truong, M. R. Carlson, C. B. Williams,and T. A. Chatila. 2007. Regulatory T cell development in the absence offunctional Foxp3. Nat. Immunol. 8: 359–368.

55. Wang, Y., A. Souabni, R. A. Flavell, and Y. Y. Wan. 2010. An intrinsic mech-anism predisposes Foxp3-expressing regulatory T cells to Th2 conversionin vivo. J. Immunol. 185: 5983–5992.

56. Cote-Sierra, J., G. Foucras, L. Guo, L. Chiodetti, H. A. Young, J. Hu-Li, J. Zhu,and W. E. Paul. 2004. Interleukin 2 plays a central role in Th2 differentiation.Proc. Natl. Acad. Sci. USA 101: 3880–3885.

57. Zhu, J., J. Cote-Sierra, L. Guo, and W. E. Paul. 2003. Stat5 activation playsa critical role in Th2 differentiation. Immunity 19: 739–748.

58. Tsuji, M., N. Komatsu, S. Kawamoto, K. Suzuki, O. Kanagawa, T. Honjo,S. Hori, and S. Fagarasan. 2009. Preferential generation of follicular Bhelper T cells from Foxp3+ T cells in gut Peyer’s patches. Science 323:1488–1492.

59. Provoost, S., T. Maes, Y. M. van Durme, P. Gevaert, C. Bachert, C. B. Schmidt-Weber, G. G. Brusselle, G. F. Joos, and K. G. Tournoy. 2009. Decreased FOXP3protein expression in patients with asthma. Allergy 64: 1539–1546.

60. Zhou, X., S. Bailey-Bucktrout, L. T. Jeker, and J. A. Bluestone. 2009. Plasticityof CD4(+) FoxP3(+) T cells. Curr. Opin. Immunol. 21: 281–285.

1282 DOMINANT Th2 DIFFERENTIATION OF HUMAN Treg

at University of R

egensburg on March 13, 2013

http://jimm

unol.org/D

ownloaded from