21
This is an Open Access document downloaded from ORCA, Cardiff University's institutional repository: http://orca.cf.ac.uk/102239/ This is the author’s version of a work that was submitted to / accepted for publication. Citation for final published version: Cook, Laura, Munier, C. Mee Ling, Seddiki, Nabila, van Bockel, David, Ontiveros, Noé, Hardy, Melinda Y., Gillies, Jana K., Levings, Megan K., Reid, Hugh H., Petersen, Jan, Rossjohn, Jamie, Anderson, Robert P., Zaunders, John J., Tye-Din, Jason A. and Kelleher, Anthony D. 2017. Circulating gluten-specific FOXP3 + CD39 + regulatory T cells have impaired suppressive function in patients with celiac disease. Journal of Allergy and Clinical Immunology 140 (6) , pp. 1592-1603. 10.1016/j.jaci.2017.02.015 file Publishers page: http://dx.doi.org/10.1016/j.jaci.2017.02.015 <http://dx.doi.org/10.1016/j.jaci.2017.02.015> Please note: Changes made as a result of publishing processes such as copy-editing, formatting and page numbers may not be reflected in this version. For the definitive version of this publication, please refer to the published source. You are advised to consult the publisher’s version if you wish to cite this paper. This version is being made available in accordance with publisher policies. See http://orca.cf.ac.uk/policies.html for usage policies. Copyright and moral rights for publications made available in ORCA are retained by the copyright holders.

INSTRUCTIONS ON THE ANNOTATION OF Sorca.cf.ac.uk/102239/1/Circulating gluten specific forkhead box... · Noe Ontiveros, MSc,c,d Melinda Y. Hardy, ... Hugh Reid, PhD, fJan Peterson,

Embed Size (px)

Citation preview

This is an Open Access document downloaded from ORCA, Cardiff University's institutional

repository: http://orca.cf.ac.uk/102239/

This is the author’s version of a work that was submitted to / accepted for publication.

Citation for final published version:

Cook, Laura, Munier, C. Mee Ling, Seddiki, Nabila, van Bockel, David, Ontiveros, Noé, Hardy,

Melinda Y., Gillies, Jana K., Levings, Megan K., Reid, Hugh H., Petersen, Jan, Rossjohn, Jamie,

Anderson, Robert P., Zaunders, John J., Tye-Din, Jason A. and Kelleher, Anthony D. 2017.

Circulating gluten-specific FOXP3 + CD39 + regulatory T cells have impaired suppressive function

in patients with celiac disease. Journal of Allergy and Clinical Immunology 140 (6) , pp. 1592-1603.

10.1016/j.jaci.2017.02.015 file

Publishers page: http://dx.doi.org/10.1016/j.jaci.2017.02.015

<http://dx.doi.org/10.1016/j.jaci.2017.02.015>

Please note:

Changes made as a result of publishing processes such as copy-editing, formatting and page

numbers may not be reflected in this version. For the definitive version of this publication, please

refer to the published source. You are advised to consult the publisher’s version if you wish to cite

this paper.

This version is being made available in accordance with publisher policies. See

http://orca.cf.ac.uk/policies.html for usage policies. Copyright and moral rights for publications

made available in ORCA are retained by the copyright holders.

Circulating gluten-specific forkhead box protein

3–positive CD391 regulatory T cells have impaired

suppressive function in patients with celiac disease

Q12 Laura Cook, PhD,a,b* C. Mee Ling Munier, PhD,a Nabila Seddiki, PhD,a,b� David van Bockel, PhD,a

No�e Ontiveros, MSc,c,d Melinda Y. Hardy, PhD,c,d Jana K. Gillies, MSc,e Megan K. Levings, PhD,e

Hugh Reid, PhD,f Jan Peterson, PhD,f Jamie Rossjohn, PhD,f,g,h Robert P. Anderson, PhD,c,d,i

John Zaunders, PhD,a,b Jason A. Tye-Din, PhD,c,d,j and Anthony D. Kelleher, PhDa,b Sydney, Parkville, Clayton, and

Brisbane, Australia; Vancouver, British Columbia, Canada; Cardiff, United Kingdom; and Cambridge, Mass

Background: Celiac disease is a chronic immune-mediated

inflammatory disorder of the gut triggered by dietary gluten.

Although the effector T-cell response in patients with celiac

disease has been well characterized, the role of regulatory

T (Treg) cells in the loss of tolerance to gluten remains poorly

understood.

Objective: We sought to define whether patients with celiac

disease have a dysfunction or lack of gluten-specific forkhead

box protein 3 (FOXP3)1 Treg cells.

Methods: Treated patients with celiac disease underwent oral

wheat challenge to stimulate recirculation of gluten-specific T

cells. Peripheral blood was collected before and after challenge.

To comprehensively measure the gluten-specific CD41 T-cell

response, we paired traditional IFN-g ELISpot with an assay to

detect antigen-specific CD41 T cells that does not rely on

tetramers, antigen-stimulated cytokine production, or

proliferation but rather on antigen-induced coexpression of

CD25 and OX40 (CD134).

Results: Numbers of circulating gluten-specific Treg cells and

effector T cells both increased significantly after oral wheat

challenge, peaking at day 6. Surprisingly, we found that

approximately 80% of the ex vivo circulating gluten-specific

CD41 T cells were FOXP31CD391 Treg cells, which reside

within the pool of memory CD41CD251CD127lowCD45RO1

Treg cells. Although we observed normal suppressive function

in peripheral polyclonal Treg cells from patients with celiac

disease, after a short in vitro expansion, the gluten-specific

FOXP31CD391 Treg cells exhibited significantly reduced

suppressive function compared with polyclonal Treg cells.

Conclusion: This study provides the first estimation of

FOXP31CD391 Treg cell frequency within circulating gluten-

specific CD41 T cells after oral gluten challenge of patients with

celiac disease. FOXP31CD391 Treg cells comprised a major

proportion of all circulating gluten-specific CD41 T cells but

had impaired suppressive function, indicating that Treg cell

dysfunction might be a key contributor to disease pathogenesis.

(J Allergy Clin Immunol 2017;nnn:nnn-nnn.)

Key words: Regulatory T cells, CD39, forkhead box protein 3, celiac

disease, gluten, OX40

Celiac disease is a chronic inflammatory disorder with featuresof autoimmune disease that results from a loss of glutentolerance.1 It is characterized by villous atrophy and the presenceof autoantibodies to tissue transglutaminase 2 (tTG), an enzyme

From athe Immunovirology and Pathogenesis Program, Kirby Institute, UNSWAustralia,

Sydney; bSt Vincent’s Centre for Applied Medical Research, St Vincent’s Hospital,

Sydney; cthe Immunology Division, Walter and Eliza Hall Institute, Parkville; dthe

Department of Medical Biology, University of Melbourne, Parkville; ethe Department

of Surgery, University of British Columbia, Vancouver; fthe Infection and Immunity

Program and Department of Biochemistry and Molecular Biology, Biomedicine Dis-

covery Institute, Monash University, Clayton; gthe ARC Centre of Excellence in

Advanced Molecular Imaging, University of Queensland, Brisbane; hthe Institute of

Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff;iImmusanT, Cambridge; and jthe Department of Gastroenterology, Royal Melbourne

Hospital, Parkville.

*Laura Cook, PhD, is currently affiliated with the Department of Medicine, University of

British Columbia, Vancouver, British Columbia, Canada.

�C. Mee Ling Munier, PhD, is currently affiliated with INSERM U955 and Universit�e

Paris-Est Cr�eteil (UPEC)/Vaccine Research Institute, Cr�eteil, France.

Supported by the Australian Government Department of Health and Ageing; the

NHMRC through a program (510448) grant, NHMRC project grant (1085875), an

NHMRC Australia Fellowship (to J.R.), and a Practitioner Fellowship (to A.D.K.); a

Coeliac Research Fund Grant (to N.S., R.P.A., J.T.-D., and A.D.K.); an Australian

Postgraduate Award; and a UNSW Research Excellence Scholarship (to L.C.).

Disclosure of potential conflict of interest: L. Cook has received a grant from the

Australian Postgraduate Award and the University of New South Wales Research

Excellence Award. N. Seddiki has received a grant from the Coeliac Research Fund

and is named inventor on a patent for the use of CD39 and the OX40 assay to identify

antigen-specific regulatory T cells held by St Vincent’s Hospital, Sydney, Australia. J.

Rossjohn has received payment from the National Health and Medical Research

Council Australia Fellowship. R. P. Anderson has received a grant from the Coeliac

Research Fund; is Chief Scientific Officer of ImmusanT; is a coinventor of patents

pertaining to the use of gluten peptides in therapeutics, diagnostics, and nontoxic

gluten; and is a shareholder of Nexpep and ImmusanT. J. Zaunders is named inventor

on a patent for the use of CD39 and the OX40 assay to identify antigen-specific

regulatory T cells held by St Vincent’s Hospital, Sydney, Australia. J. A. Tye-Din has

received a grant from the Coeliac Research Fund; has consultant arrangements with

ImmusanT; is coinventor of patents pertaining to the use of gluten peptides in

therapeutics, diagnostics, and nontoxic gluten; and is a shareholder in Nexpep. A. D.

Kelleher has received grants from the Australian Government Department of Health

and Ageing, the National Health and Medical Research Council (510448 and

1085875), and the Coeliac Research Fund; has received a Practitioner Fellowship

from the Australian Government Department of Health and Ageing; and is named

inventor on a patent for the use of CD39 and the OX40 assay to identify antigen-

specific regulatory T cells held by St Vincent’s Hospital, Sydney, Australia. The rest of

the authors declare that they have no relevant conflicts of interest.

Received for publication March 24, 2015; revised February 3, 2017; accepted for publi-

cation February 16, 2017.

Corresponding author: Laura Cook, PhD, Levings Lab, Child and Family Research

Institute, Room A4-102, 950 West 28th Ave, Vancouver, BC V5Z 4H4, Canada.

E-mail: [email protected].

0091-6749/$36.00

� 2017 American Academy of Allergy, Asthma & Immunology

http://dx.doi.org/10.1016/j.jaci.2017.02.015

1

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

Abbreviations used

APC: Antigen-presenting cell

CTV: CellTrace Violet

FOXP3: Forkhead box protein 3

PE: Phycoerythrin

SEB: Staphylococcal enterotoxin B

Tconv: Conventional T

Treg: Regulatory T

TSDR: Regulatory T cell–specific demethylated region

tTG: Tissue transglutaminase 2

that deamidates gluten. Intestinal damage is caused by CD41

T cells, which recognize deamidated gluten peptides presentedin complex with HLA-DQ2.5, HLA-DQ2.2, and/or HLA-DQ8,2,3

and the immunodominant hierarchy of wheat gliadin T-cellepitopes in HLA-DQ2.51 patients with celiac disease has beencomprehensively mapped.4 Although HLA susceptibilityhaplotypes are expressed by 30% to 40% of the generalpopulation, celiac disease affects only approximately 1%,indicating that immune tolerance to gluten is the norm. However,the mechanisms that underpin maintenance of gluten toleranceremain poorly described.

Gluten-responsive effector CD41 T cells can be detected in theperipheral blood of patients with celiac disease on a gluten-freediet 6 to 8 days after a 3-day oral gluten challenge.5 Onactivation, these cells secrete high levels of IFN-g,6,7 support Bcell–mediated production of antibodies to tTG and modifiedgluten peptides, and enhance lysis of stressed epithelial cells byCD81 T cells.4,6 Studies of total regulatory T (Treg) cells inpatients with celiac disease have provided evidence for bothnormal suppressive function8-10 and impaired function,11,12 aswell as suggesting that effector T cells have become resistant toTreg cell suppression.10,13,14 However, although forkhead boxprotein 3 (FOXP3)1 Treg cells have an important role inmaintaining peripheral tolerance, until now, the frequency andfunction of gluten-specific FOXP31 Treg cells in patients withceliac disease have not been studied.

For the first time, this study exploited acute in vivo glutenchallenge in patients with celiac disease to interrogate botheffector and regulatory components of the recall response togluten. Specifically, we aimed to estimate the frequency of Tregcells within gluten-specific CD41T-cell recall responses; identifychanges in the frequency of peripheral gut-homing memoryCD41 T-cell populations after gluten challenge; andphenotypically and functionally characterize gluten-specificTreg cells.

METHODS

Subjects and samplesPatients with celiac disease were recruited after provision of informed

consent (Human Research Ethics Committees: Royal Melbourne Hospital ID

2003.009; Walter and Eliza Hall Institute of Medical Research ID 03/04).

Enrollment criteria were biopsy-proved disease conforming to European

Society for Paediatric Gastroenterology Hepatology and Nutrition

guidelines,15HLA-DQ21, and compliancewith a gluten-free diet for 6months

or more. Healthy donor blood was obtained from the Australian Red Cross

Blood Service and volunteers (St Vincent’s Hospital Human Research Ethics

Committee ID HREC/13/SVH/145). Peripheral blood was collected into

lithium heparin vacutainers (BD, San Jose, Calif), transported at ambient

temperature, and processed within 8 hours of collection. Mononuclear cells

were obtained by means of centrifugation over Ficoll-Paque (GE Healthcare,

Fairfield, Conn).

Serology and HLA typingSerum titers of tTG IgA and deamidated gliadin peptide IgGwere evaluated

with commercial kits (INV 708760 and 704525; INOVA Diagnostics, San

Diego, Calif) by a diagnostic laboratory (Gribbles-Healthscope, nnn,

Australia) Q1. The presence of alleles encoding HLA-DQ2.5, HLA-DQ2.2, and

HLA_DQ8 was determined by detecting 5 single nucleotide polymorphisms

(rs2187668, rs2395182, rs4713586, rs7454108, and rs7775228), as previously

described.16,17 HLA-DQB1 and HLA-DQA1 alleles were determined by

using PCR sequence–specific oligonucleotide hybridization (Victorian

Transplantation and Immunogenetics Service, Victoria, Australia).

Oral gluten challengeAll participants undertook a gluten challenge5 from days 1 to 3 by

consuming 4 slices of commercial white bread daily (approximately 10 g/d

wheat gluten) and recorded symptoms daily to day 6, grading them as mild,

moderate, or severe.18

ReagentsWe used 2 HLA-DQ2.5–restricted 15mers that encompass the

immunodominant deamidated wheat gliadin T-cell epitopes DQ2.5-glia-a1/

a2 (LQPFPQPELPYPQPQ) andDQ2.5-glia-v1/v2 (QPFPQPEQPFPWQP).4

Gluten peptide mix contained an equimolar mixture of these two 15mers. An

HLA-DQ2.5–restricted 15mer that encompasses the immunodominant barley

hordein T-cell epitope DQ2.5-hor-3 (PEQPIPEQPQPYPQQ) acted as a

specificity control.4 Peptides were synthesized to 95% purity or greater, as

confirmed by means of high-performance liquid chromatography (Pepscan,

Lelystad, The Netherlands); dissolved in dimethyl sulfoxide (Sigma-Aldrich,

St Louis, Mo); and stored at2808C until use.5,18 Chymotrypsin digestion and

deamidation of gliadin (#101778; ICN Biomedicals, Cost Mesa, Calif) were

performed, as previously described.5,18 Tetanus toxoid was from CSL (nnn,

Australia) Q2, and staphylococcal enterotoxin B (SEB) and PHA were from

Sigma-Aldrich.

IFN-g secretion assaysAntigen-stimulated IFN-g secretion from PBMCs was assessed by using

either ELISpot (Mabtech, Nacka Strand, Sweden) or ELISA (Mabtech),

assays that are equivalent in their ability to detect gluten-specific responses.18

Gluten peptide mix and deamidated gliadin were used at 100 mg/mL and

tetanus toxoid was used at 10 LfU/mL, and assays were performed in

triplicate, as previously described.5,18,19

OX40 assayThe OX40 assay was performed, as previously described,20,21 with either

fresh whole blood (diluted 1:1 with RPMI 1640 media, Invitrogen) or PBMCs

at 2 3 106 cells/mL and incubation with antigen for 44 hours at 378C (5%

CO2). Antigen concentrations were as follows: SEB, 1mg/mL; tetanus toxoid,

2 LfU/mL; deamidated gliadin, 100 mg/mL; and DQ2.5-hor-3m 50 mg/mL.

Optimal concentrations of gluten antigens were determined in a pilot study

(see Fig E1, B, in this article’s Online Repository at www.jacionline.org).

DQ2.5-glia-a1/a2 and DQ2.5-glia-v1/v2 were used separately at

50 mg/mL and in an equimolar gluten peptide mix. Assay cutoffs were as

follows: greater than 0.02% of CD41 T cells (mean1 3 SDs Q3of unstimulated

wells) and greater than 20 cells.

Population tracking within the OX40 assayPostchallenge PBMCs were fluorescence-activated cell sorting purified

into 4 CD41 T-cell populations (Fig 3, A) Q4: (1) CD45RO2 T cells and

J ALLERGY CLIN IMMUNOL

nnn 2017

2 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

121

122

123

124

125

126

127

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

(2) CD45RO1CD127highCD25low conventional T (Tconv) cells, with the

CD45RO1CD127lowCD25high Treg cells subdivided into (3) CD391 and

(4) CD392 cells. Portions of the 3 CD45RO1 populations were stained with

CellTrace Violet (CTV) to enable cell tracking. In some experiments

PBMCs were stained with DQ2.5-glia a21/a2 tetramer and sorted into

tetramer-negative and tetramer-positive CD41 T cells that were then labeled

with CTV. PBMCs were reconstituted, maintaining ex vivo cell ratios, and

supplemented with CD3-depleted autologous PBMCs (Dynabeads CD3,

Invitrogen) to one sixth of the total cell number. Each well contained a single

CTV-labeled population, and the OX40 assay was performed and analyzed, as

described above.

Flow cytometryStaining was performed, as previously described,22 with anti-CD3–

peridinin-chlorophyll-protein complex–Cy5.5 (SK7), CD4–Alexa Fluor 700

(RPA-T4), CD25–allophycocyanin (2A3), OX40 (CD134)–phycoerythrin

(PE; L106), cytotoxic T lymphocyte–associated antigen 4–PE (BNI3),

GARP-BV711 (7B11), integrin b7–PE (FIB504; BD); CD45RO-ECD

(UCHL1; Beckman Coulter, Fullerton, Calif); CD127-eFluor450

(eBioRDR5), CD39-PECy7 (A1); LAP-PECy7Q5 (FNLAP; eBioscience),

Helios–Alex Fluor 488 (22F6), and anti-FOXP3–Alexa Fluor 488 (259D;

BioLegend, San Diego, Calif). FOXP3 staining was performed with the

FOXP3 buffer kit (BD), and the IgG1k-FITC antibody (BD) was used to set

analysis gates. All mAbswere used at manufacturers’ recommended dilutions.

Cell labeling was performed with the CTV Cell Proliferation Kit (Invitrogen)

and PKH26 Red Fluorescent Cell Linker Kit for General Cell Membrane

Labelling (Sigma-Aldrich) per the manufacturers’ recommendations by using

5 mmol/L dye concentrations. HLA-DQ2 proteins were synthesized with

bound DQ2.5-glia-a1, DQ2.5-glia-a2, or the HLA class II invariant peptide

by using previously described constructs and methods.23 Tetramers were

produced by means of addition of either NeutrAvidin R-allophycocyanin or

R-PE conjugate (Invitrogen) to biotinylated protein, as previously described.24

Staining was performed with 50 mg/mL tetramer in complete media at 378C

for 1 hour. A 4-laser LSRII flow cytometer (BD) was used, and analysis

was performed with FlowJo software (v8.8.7; TreeStar, Ashland, Ore).

In vitro expansion of T-cell populationsThe protocol used to expand Treg cells, Tconv cells, and DQ2.5-glia-a1/

a2–specific CD391 T cells was adapted from Gregori et al,25 as previously

described20; T-cell cloning was performed by plating cells at 1 cell/well.

Irradiated feeder cell mix consisted of 5 3 105 cells/mL of mixed PBMCs

(equal mix of autologous PBMCs and PBMCs from 2 allogeneic healthy

donors) and 5 3 104 cells/mL of an autologous EBV-transformed B-cell

line generated and maintained, as previously described.26

Quantitative RT-PCR and T-cell receptor clonotype

analysis by 59 Rapid Amplification of cDNA EndsQuantitative RT-PCR was performed on resting T-cell populations, and

relative expression levels to b-actin were calculated, as previously

described20: FOXP3 forward, 59-TCACCTACGCCACGCTCAT-39;

FOXP3 reverse, 59-TCATTGAGTGTCCGCTGCTT-39; TGF-b forward,

59-CCCTGGACACCAACTATTGC-39; and TGF-b reverse, 59-CAGA

AGTTGGCATGGTAGCC-39. T-cell receptor clonotypes were analyzed by

using 59RapidAmplification of cDNAEnds (Clontech,MountainView, Calif)

PCR, as previously described.27 Sequences were analyzed by using the

ImmunoGenetics V-quest database.28

Suppression assaySuppression assays were performed, as previously described.20

CD41CD1271CD252 responder T cells were labeled with CTV (Invitrogen),

and suppressor cells were labeled with PKH26 (Sigma-Aldrich). Wells

contained 50,000 irradiated autologous antigen-presenting cells (APCs) and

20,000 responder T cells, with suppressor T cells added at the ratios indicated.

Assays were stimulated for 4 days with 0.25 mg/mL soluble anti-CD3

(Invitrogen). CD39 enzyme activity was blocked in some assays with

250 mmol/L ARL67156 (Sigma-Aldrich). The division index was used to

calculate the percentage of suppression.29

Treg cell–specific demethylated region analysisGenomic DNAwas isolated, and bisulfite conversion was performed with

the EZ DNAMethylation-Direct kit (Zymo Research, Irvine, Calif). PCR was

performedwith the PyroMark PCR kit (Qiagen, Hilden, Germany) andHuman

FoxP3 Methylation Assay ADS783FS2 (EpigenDx, Ashland, Mass),

which reports the methylation of 8 representative CpG sites in the regulatory

T cell–specific demethylated region (TSDR). Pyrosequencing was performed

on a PyroMark Q96 ID (Qiagen) with PyroMark Gold Q96 reagents (Qiagen)

and Streptavidin Sepharose (GE Healthcare). All kits/reagents were used,

according to the manufacturer’s instructions. Analysis was performed on

female subjects, and the levels of methylation have not been adjusted to

account for X-inactivation.

StatisticsMann-Whitney U tests or 1-way ANOVAwere used unless samples were

matched, and then Wilcoxon signed-rank tests were performed. Correlation

analyses used Spearman rho (rs). P values were considered significant at

less than .05. Prism 6.0 software (GraphPad Software, La Jolla, Calif) was

used for all statistical analyses.

RESULTS

Numbers of circulating gluten-specific

FOXP31CD391 Treg cells are significantly increased

after gluten challengeTo investigate CD41 T-cell recall responses to gluten, we

recruited a cohort of 17 treated patients with celiac disease(see Tables E1 and E2 in this article’s Online Repository atwww.jacionline.org). We used our previously developed OX40assay, which detects antigen-specific CD41 T cells throughantigen-induced coexpression of CD25 and OX40,21 to measurechanges in the frequency of circulating gluten-specific CD41

T cells in patients with celiac disease after gluten challenge(Fig 1 [F1-4/C], A). In a pilot study we found the optimal time for detectingresponses was 6 to 8 days after gluten challenge (termed days 6and 8; see Fig E1, A). At day 6, we observed significant increasesin deamidated gliadin levels (n5 15,P5.007) and gluten peptideresponses (n5 9,P5.008; Fig 1,B). The overall peak response togluten antigens occurred at day 6 (median response 0.27% ofCD41 T cells). There were no detectable responses to the barleyhordein peptide DQ2.5-hor-3 (Fig 1, A), indicating that the wheatpeptide responses are specifically induced by oral wheat glutenchallenge. Patients with detectable gluten peptide responsesalso had a significant increase in numbers of both total andgut-homing CD391FOXP31 Treg cells at day 6 (Fig 1,C), a trendnot seen in gluten peptide nonresponders (see Fig E2, A, in thisarticle’s Online Repository at www.jacionline.org). Q6There wereno significant changes observed within numbers of total orgut-homing CD41 Tconv cells (data not shown).

We assessed the presence of gluten-specific CD41 T cells in 6healthy volunteers (HLA-DQ genotypes were known for 4, andonly 1 carried HLA-DQ2.5). The median deamidatedgliadin-specific response for the non-HLA-DQ2.5 subjects was0.09% of CD41 T cells (range, 0% to 0.15%), and the responsefor all 6 subjects was 0.19% (range, 0% to 0.77%; see Fig E1,C). No deamidated gluten peptide responses were detected in

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 3

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

241

242

243

244

245

246

247

248

249

250

251

252

253

254

255

256

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

these subjects (data not shown), indicating that CD41 T-cellresponses to deamidated gluten peptides are a more specificmarker of celiac disease.

The majority of gluten peptide–specific

CD41CD251OX401 T cells express CD39We have previously shown that within antigen-responsive

CD251OX401 T cells, a subset of Treg cells can beidentified on the basis of CD39 expression, providing asensitive and specific way to measure and isolate viableantigen-specific FOXP31 Treg cells.20 We used this approach todetermine the contribution of CD391FOXP31 Treg cells togluten-specific OX40 assay responses (Fig 2[F2-4/C]

[F3-4/C], A). On average,

72% of deamidated gliadin-specific T cells were CD391

(75% coexpressed FOXP31), and 89% of gluten peptide-specific T cells were CD391, with 82% of these cells expressingFOXP3 (Fig 2, B).

We found similar CpG methylation patterns within the TSDRin DQ2.5-glia-a1/a2–specific CD391 T cells sorted directly aftera 44-hour OX40 assay compared with ex vivo Treg cellsisolated from both healthy subjects and patients with celiacdisease (Fig 2, C). These data are consistent with gluten-specific CD251OX401CD391 T cells being highly enriched forTreg cells.

We confirmed OX40 assay specificity through severalexperiments using DQ2.5-glia-a1/a2 tetramer reagents. Wecostained DQ2.5-glia-a1/a2 peptide–stimulated OX40 assayswith DQ2.5-glia-a1/a2 tetramer and observed that of allquadrants in the CD25 versus OX40 plot, the CD251OX401

quadrant had the highest proportion of tetramer-positive cells(Fig 2, D). We confirmed that, similar to OX40 responses,tetramer staining was only observed in day 6 postchallengePBMCs (not prechallenge PBMCs) and that the majority of theCD41 tetramer-positive cells were CD391 (median, 70.3%;n 5 7; Fig 2, E). Finally, we sorted CD41CD251OX401CD391

FIG 1. OX40 assay responses to gluten antigen peak at day 6 after gluten challenge. A, Representative OX40

assay responses from patients with celiac disease at day 6. B, For 15 patients with celiac disease, the

percentage of CD41 T cells responding to deamidated gliadin, gluten peptide mix, SEB, or tetanus toxoid

(Tet Tox) are shown. Dotted line, Assay cutoff. C, Flow cytometric analysis of total and integrin

b71CD391FOXP31 Treg cell (CD45RO1CD127lowCD25high) frequencies in ex vivo peripheral blood at days

0, 6, and 8 for patients with celiac disease and detectable gluten peptide responses (n 5 7). Red lines,

Patients with gluten peptide mix responses (n 5 9); black lines, patients with undetectable gluten peptide

responses (n 5 7). Statistical analyses used Wilcoxon signed-rank tests.

J ALLERGY CLIN IMMUNOL

nnn 2017

4 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

407

408

409

410

411

412

413

414

415

416

417

418

419

420

421

422

423

424

425

426

427

428

429

430

431

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

457

458

459

460

461

462

463

464

465

466

467

468

469

470

471

472

473

474

475

476

477

T cells and labeled with cell proliferation dye beforerestimulating with a panel of antigens in the presence ofautologous APCs. We observed that stimulation with cognateantigen generated substantially more proliferation (87%)than DQ2.5-glia-v1/v2 (29%), DQ2.5-hor-3 (29%), or noantigen (21.7%). DQ2.5-glia-a1/a2 restimulation of matchedCD252OX402 T cells did not stimulate cell proliferation(see Fig E3, D, in this article’s Online Repository atwww.jacionline.org).

Importantly, although CD39 expression varies betweensubjects, in each subject the proportion of CD391 cells withinCD41T cells did not vary within the timeframe of theOX40 assay

(see Fig E4, C, in this article’s Online Repository atwww.jacionline.org). The proportion of CD391 cells withinrecall responses also did not significantly vary from beforechallenge through day 8 after challenge (see Fig E2, C, OnlineRepository). Responses to the mitogen SEBwere similar betweenthe celiac cohort (n 5 15; mean response, 5.8% CD41 T-cells)and healthy subjects (n 5 15; mean age, 37; 60% female; meanresponse, 7.2% of CD41 T cells) and consisted of less than30% CD391 T cells (20.7% in healthy subjects vs 29.1% inpatients with celiac disease; see Fig E2, E). This suggests that ahigh proportion of CD391 cells within antigen-specific responsesis not an inherent feature of the OX40 assay but is instead a unique

FIG 2. The majority of circulating gluten peptide–specific CD41 T cells are CD391FOXP31. A and B, CD39

and FOXP3 expression within OX40 assay responses: representative data (Fig 2, A) and within responses

to deamidated gliadin (n 5 14) and gluten peptide mix (n 5 9; mean 6 SEM; Fig 2, B). C, Percentage

methylation at 8 CpG sites in the TSDR of FOXP3 within ex vivo Treg and Tconv cells (n 5 3 healthy female

subjects and n 5 3 female patients with celiac disease) and DQ2.5-glia-a1/a2–specific CD391 cells (n 5 3;

median 6 interquartile range). ns, Not significant. D, The frequency of DQ2.5-glia-a1/a2 tetramer-positive

cells is shown within each quadrant of the CD25/OX40 plot for DQ2.5-glia-a1/a2–stimulated OX40 assays

(n 5 3; mean 6 SEM). E, Representative DQ2.5-glia-a1/a2 tetramer and CD39 staining of unstimulated

PBMCs at days 0 and 6 after gluten challenge. For 3 patients, tetramer staining is shown at both day

0 and day 6. The proportion of tetramer-positive cells that are CD391 is shown for 7 patients with celiac

disease at day 6 after gluten challenge (mean 6 SEM). Statistical analyses in Fig 2, B and D, used

Mann-Whitney U tests, and those in Fig 2, E, used Wilcoxon signed-rank tests.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 5

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

478

479

480

481

482

483

484

485

486

487

488

489

490

491

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

585

586

587

588

589

590

591

592

593

594

595

596

597

feature of the recall response to gluten antigen in patients withceliac disease.

To confirm that gluten-specific CD251OX401CD391 T cellsoriginated from the pool of circulating Treg cells, we used ourpreviously described method for population tracking within theOX40 assay (Fig 3, A and B).20 We found that CD391 memoryTreg cells comprised an average of 88% of DQ2.5-glia-a1/a2-specific CD251OX401CD391 T cells, with CD392 Treg cellsalso the dominant population (average, 76%; n 5 4) within theCD251OX401CD392 T cells (Fig 3, E). Again, we observed

that during a 44-hour OX40 assay, CD39 expression was notaffected by cell activation (Fig 3, C). For 2 subjects, we trackedsorted, CTV-labeled CD41DQ2.5-glia-a1/a2 tetramer-positivecells within OX40 assays stimulated with DQ2.5-glia-a1/a2peptide. We observed that approximately 60% of theCD251OX401 cells were composed of CD391CTV1 cells(Fig 3, F). These data support our findings that peripheralCD41CD45RO1CD25highCD127low Treg cells constitute morethan 80% of the total DQ2.5-glia-a1/a2–specific CD251OX401

T-cell response after gluten challenge.

FIG 3. DQ2.5-glia-a1/a2–specific CD251OX401CD391 T cells originate from peripheral CD391 Treg cells.

A and B, Overview of the method and gating strategy used to conduct population tracking within an

OX40 assay performed as described in the Methods section with peripheral blood after gluten challenge.

C, Changes in CD39 expression between ex vivo isolation and after a 44-hour OX40 assay for Tconv cells,

CD392 Treg cells, and CD391 Treg cells (mean 6 SEM). D, Representative plots for unstimulated wells

and DQ2.5-glia-a1/a2–stimulated wells that contained CTV-labeled CD391 Treg cells. E, Proportion of Tconv

cells (gray), CD392 Treg cells (blue), and CD391 Treg cells (red) within the total DQ2.5-glia-a1/a2–specific

CD251OX401 T-cell response and the CD392 and CD391 fractions of this response. Data in Fig 3, C and E,

are the mean of 5 independent experiments with 4 patients with celiac disease. F, Tetramer-positive cells

were sorted from day 6 PBMCs, labeled with CTV, and either left unstimulated or stimulated for 44 hours

with DQ2.5-glia-a1/a2 peptide in the presence of autologous APCs. The proportion of CD391CTV1 cells

within the CD251OX401 quadrant is representative of 2.

J ALLERGY CLIN IMMUNOL

nnn 2017

6 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

598

599

600

601

602

603

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

707

708

709

710

711

712

713

714

715

716

717

DQ2.5-glia-a1/a2–specific CD391 T cells have

impaired suppressive functionDQ2.5-glia-a1/a2–specificCD41CD251OX401CD391Tcells

were expanded in vitro from post–gluten challenge PBMCs (sortpurities, >90%) to further investigate the phenotype and functionof gluten-specific FOXP31CD391 Treg cells. For femalepatients with celiac disease and healthy donors, we also expandednon–gluten-specific CD252OX402 T cells and, from unstimulatedPBMCs, CD25highCD127lowTreg cells and CD252CD1271Tconvcells. We generated 3 T-cell clones, 2 from patient #0174 (C1and C2) and 1 from patient #0251 (C3). Clonality wasconfirmed through sequencing the T-cell receptor b chain variableregion. Both T-cell clones from patient #0174 expressed thesame TRBV7-2 clonotype (CASSLRYTDTQYF), which mightbe a public clonotype, as previously identified in another celiaccohort.30

Suppression assays used soluble anti-CD3 stimulus for 4 daysin the presence of autologous APCs with a 1:1 ratio of suppressorto responder cells (Fig 4[F4-4/C] , A). Suppressive function of ex vivo

celiac Treg cells (CD25highCD127low; median, 73.4%; n 5 5)before oral gluten challenge was comparable with that of healthyTreg cells (median, 70.5%; n 5 6) but significantly greater thanthat of Tconv cells (CD252CD1271; median, 12%; n 5 5), asexpected (P 5 .016; Fig 4, B). Interestingly, the subset ofex vivo CD25highCD127low Treg cells that was isolated asCD391 cells before challenge had reduced suppressive function(median, 46.3%; n 5 5; P 5 .016) that was not further affectedby the CD39 inhibitor ARL67156 (median, 45.5%). Similarly,56-day expanded DQ2.5-glia-a1/a2–specific CD391 T cellshad slightly reduced suppression comparedwith ex vivoTreg cellsfrom patients with celiac disease (mean, 55.8%; range, 30.1% to74.8%), as did the CD391 T-cell clones C1 and C2 (62% and 52%suppression, respectively; Fig 4, C). Addition of the CD39inhibitor ARL67156 had a minimal effect (mean reduction insuppression, 11%; Fig 4, C).

We assessed in vitro suppressive function with suppressor/responder cell ratios of 1:1 to 1:32 for 14 days expandedQ7 :DQ2.5-glia-a1/a2–specific CD391 T cells (n5 4), a T-cell clone(C3), and, for 4 healthy subjects and patients with celiac disease,Treg cells (CD25highCD127low) and Tconv cells(CD252CD1271). The celiac Treg cells exerted suppressionacross all cell ratios comparable with that of Treg cells fromhealthy subjects, whereas the gluten-specific CD391 T-cell cloneC3 began to exhibit markedly lower suppressive function at a1:8 cell ratio (Fig 4, D). The expanded gluten-specific CD391

T cells had significantly reduced suppressive function comparedwith that of polyclonal Treg cells from both healthysubjects and patients with celiac disease across all ratios tested(Fig 4, D). These data indicate that in patients with celiac diseaseafter gluten challenge, the expanded subset of peripheralgluten-specific CD391 Treg cells, but not polyclonal Treg cells,has impaired suppressive function.

Approximately 50% of expanded gluten-specificCD251OX401CD391 T cells stained positive for DQ2.5-glia-a1/a2 tetramer compared with greater than 2%tetramer-positive cells within non–gluten-specific CD252OX402

T cells (see Fig E3, B). These cells had substantial expression ofCD39, CD25, cytotoxic T lymphocyte–associated antigen 4, andintegrin b7, but FOXP3 expression was low or absent (see TableE3 in this article’s Online Repository at www.jacionline.org).

Interestingly, gluten-specific CD391 T cells were Heliosnegative, suggesting they originate from a peripherally derivedTreg cell population (see Fig E3, C). Quantitative RT-PCRconfirmed that expanded gluten-specific CD391 T cells hadvery low levels of FOXP3 and moderate-to-high levels ofTGF-b expression that corresponded to increased surfaceexpression of GARP and LAP, which tether latent TGF-b to thecell membrane (see Fig E3, A and E). Loss of FOXP3protein expression corresponded to increased CpG methylationin the TSDR of expanded cells. CD391 T cells had anaverage 2.8-fold increase in methylation, whereas Tregcells from healthy subjects and patients with celiac diseasehad 1.5- and 1.4-fold increases in methylation, respectively (seeFig E3, F).

Antigen-stimulated expression of CD25 and OX40

detects significantly more gluten-specific T cells

than conventional IFN-g secretion assaysWe performed correlation analyses to compare the sensitivity

of the OX40 assay with that of conventional IFN-g ELISpotassays. We observed a positive correlation between the IFN-gELISpot assay at day 6 and peak OX40 assay responses to glutenpeptide stimulus (n 5 13, rs 5 0.876, P 5 .0002; Fig 5 ½F5�, A, andTable I ½T1�). For the 12 HLA-DQ2.5 patients with celiac diseaseand detectable IFN-g responses to gluten peptide antigen, 10(83%) also had detectable OX40 assay responses. A linearregression analysis of responses to gluten peptide antigendetected by using each assay generated a line of best fit with aslope (m) of 4.806. This indicates the OX40 assay detectsapproximately 5 times the number of gluten peptide–specificCD41 T cells than the IFN-g ELISpot (Fig 5, B).

A high proportion of memory Treg cells from

patients with celiac disease express CD39Flow cytometry was used to measure the frequency of

peripheral lymphocyte populations (see gating in Fig E4, B) inhealthy volunteers (n 5 13; mean age, 47; 69% female) andpatients before gluten challenge (n 5 13; mean age, 58;69% female). The proportion of CD391 cells within memoryTreg cells was significantly higher in patients (mean,74.65%; range, 59.7% to 83.7%) than healthy control subjects(mean, 48.07%; range, 11.10% to 73.30%; P < .0001; Fig 5,C). Patients with celiac disease also had significantly reducedtotal memory Treg cell numbers at day 0 (mean, 3.14% ofCD41 T cells) compared with healthy control subjects(mean, 7.97%; P < .0001; Fig 5, D), which persisted at days6 and 8 after challenge (data not shown), and significantlymore CD391 memory Treg cells within CD41 T cells at day0 (P 5 .037; Fig 5, E).

Symptom severity associated with stronger gluten

peptide recall responses in the OX40 assayPatients with celiac disease were split into 2 groups based on

their symptom severity to identify associations betweenimmunologic variables and clinical symptoms (Table I). Nosignificant differences were observed for the frequency of totalCD45RO1CD391 Treg cells at day 0 or for ELISpot responses

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 7

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

828

829

830

831

832

833

834

835

836

to gluten peptide mix (see Fig E5, A and C, in this article’s OnlineRepository at www.jacionline.org). However, patients with celiacdisease andmore severe symptoms had significantlymore CD391

cells within deamidated gliadin-specific CD251OX401 T cells(P 5 .014) and significantly larger OX40 assay responses togluten peptides (P 5 .011; see Fig E5, B and D).

DISCUSSIONThese data are the first report of the contribution of

FOXP31CD391 Treg cells to gluten-specific CD41 T-cellresponses in patients with celiac disease after in vivo glutenchallenge. Surprisingly, we observed that FOXP31CD391

Treg cells comprised more than 80% of circulating gluten

FIG 4. In vitro–expanded DQ2.5-glia-a1/a2–specific T cells retain suppressive function. A, Representative

responder cell proliferation showing division index (DI). B, Percentage of suppression of ex vivo Treg cells

from healthy subjects (n 5 6), Treg cells from patients with celiac disease (n 5 5), CD391 Treg cells from

patients with celiac disease (n 5 5), and Tconv cells from patients with celiac disease (n 5 5) at a 1:1 ratio

with responder T cells. ns, Not significant. C, Percentage suppression of 56-day expanded DQ2.5-glia-a1/a2–

specific CD391 T-cell populations (n5 2) and CD391 T-cell clones (C1 and C2). The CD39 inhibitor ARL67156

was added as indicated. D, Percentage suppression of 14-day expanded Treg cells (n 5 4), Tconv cells

(n 5 4), and DQ2.5-glia-a1/a2–specific CD391 T cells (n 5 4) from healthy subjects and patients with celiac

disease and T-cell clone C3 for 1:1 to 1:32 suppressor/responder cell ratios. Data in Fig 4, B-D, are

medians 1 interquartile ranges Q8of 1 to 3 independent experiments, and statistical analyses used

Mann-Whitney U tests.

J ALLERGY CLIN IMMUNOL

nnn 2017

8 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

896

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

914

915

916

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

942

943

944

945

946

947

948

949

950

951

952

953

954

955

956

peptide–specific CD41T cells in patients with celiac disease aftergluten challenge. We confirmed that more than 85% of the glutenpeptide–specific FOXP31CD391 T cells originated from theperipheral pool of CD391 Treg cells and that the extent of CpGmethylation in the TSDR within the FOXP3 loci of these cellsis similar to that seen in CD127lowCD25high Treg cells fromhealthy subjects.

Because the majority of FOXP31 Treg cells do not secreteIFN-g, our data indicate that IFN-g–based methods only detectapproximately 20% of the total CD41 T-cell response to glutenantigen (supported by our linear regression analysis). TheOX40 assay correlated with the IFN-g ELISpot for the detectionof gluten-specific CD41 T-cell responses. This concurs withprevious studies that found the OX40 assay has strong agreementwith IFN-g release assays for Mycobacterium tuberculosis31,32

and concordance with serology, proliferation, and cytokineresponses to HIV-1,21 hepatitis C virus,33 human papilloma-virus,34 Mycobacterium avium complex, varicella zostervirus, EBV, CMV, Candida albicans, and Streptococcus

pneumonia.21,35 For the first time, we also show that the OX40

assay corresponds with class II tetramer staining for responsesto the DQ2.5-glia-a1/a2 epitopes in patients with celiac diseaseafter gluten challenge.

These data complement our previous study showing thatCD391 Treg cells comprise a substantial proportion of CD41

T-cell recall responses to viral and bacterial antigens.20 Previousstudies of FOXP31 Treg cells within in vivo recall responses tovaricella zoster virus in human subjects36,37 and within secondaryimmune responses to influenza virus in mice38,39 identified a keyrole for pathogen-specific Treg cells in controlling the cellular im-mune response. Of particular interest is the recent discovery thatparticle-associated antigens drive a Treg cell response, whereasdistinct soluble antigens instead drive an effector T-cellresponse.40 In addition to the type of antigen, the balance betweenTreg and Tconv cells within antigen-specific responses is alsoinfluenced by chronicity of antigen exposure.41 This study is alarge contribution to the relatively underexplored area ofantigen-specific human Treg cells and shows that, for patientswith celiac disease on a gluten-free diet, the CD41 T-cellresponse to acute dietary gluten re-exposure is skewed toward

FIG 5. Analysis of assay correlation and peripheral Treg cell frequency and phenotype. A and B, Gluten

peptide mix IFN-g ELISpot responses (SFU/106 PBMCs; n 5 13) are correlated with OX40 assay responses

(n 5 13) expressed as either CD251OX401 cells as a percentage of CD41 T cells (Fig 5, A) or CD251OX401

cells/106 PBMCs (Fig 5, B). In Fig 5, A, the calculated Spearman rho (rs) and P values are shown, and in Fig 5,

B, the line of best fit (solid line) and 95% CIs (dashed line) are shown. C-E, Flow cytometric phenotyping data

from 13 patients with celiac disease were compared with those in healthy subjects (n 5 13) for the

proportion of CD41CD45RO1CD127lowCD25high Treg cells that expressed CD39 (Fig 5, C), the proportion

of CD45RO1 Treg cells within CD41 T cells (Fig 5, D), and the proportion of CD41 T cells that were

CD45RO1CD391 Treg cells (Fig 5, E). Error bars represent medians 6 interquartile ranges, and statistical

analyses used Mann-Whitney U tests.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 9

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

957

958

959

960

961

962

963

964

965

966

967

968

969

970

971

972

973

974

975

976

977

978

979

980

981

982

983

984

985

986

987

988

989

990

991

992

993

994

995

996

997

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

1019

1020

1021

1022

1023

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

1048

1049

1050

1051

1052

1053

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

Treg cells. Furthermore, we demonstrate that functionaldefects can be unmasked by studying the relevant diseaseantigen-specific population rather than polyclonal Treg cells.

On average, maximal OX40 responses to gluten antigensoccurred at day 6 after gluten challenge, a similar time courseto that previously noted for IFN-g responses.7,18 These dynamicchanges were restricted to the gluten-derived antigens becausethere was little change in responses to mitogen or controlantigens. Importantly, CD391 proportions within all OX40 assayresponses were not significantly altered after gluten challenge,indicating the high CD391 proportions observed within glutenpeptide responses occur independently of immune activation.For those patients with celiac disease who did not respond tothe gluten peptides in this study, there is no evidence of durableimmune tolerance because the gluten challenge still causedclinical symptoms for these patients. It is more likely that theimmune response is present but either less than the assay’s limitof detection or not detectable in peripheral blood. The severityof gastrointestinal symptoms also corresponded to larger immuneresponses to gluten and a higher frequency of CD391 cells withinthese responses. This is similar to a previous observation that thedensity of intestinal FOXP31Treg cells positively correlated withthe severity of histologic damage.10

Both expanded DQ2.5-glia-a1/a2–specific CD391 T-cell linesand clones and total ex vivo CD391 Treg cells from patients withceliac disease had reduced suppressive function in response to apolyclonal stimulus that was not dependent on CD39 functionbut corresponded to a loss of stable FOXP3 expression, indicatingthat gluten-specific CD391 Treg cells from patients with celiac

disease might have an inherent functional defect. These datacontrast with a previous observation that found expandedIL-10–secreting gluten-specific Treg type 1 cell clones hadnormal in vitro suppressive function.8 Our in vitro–expandedDQ2.5-glia-a1/a2–specific CD391 T cells had aCD251FOXP32TGF-b1GARP1LAP1 phenotype that mostclosely resembles that of human regulatory TH3 cells.42 Incontrast to the previously observed stable FOXP3 expressionseen in expanded CMV-P1–specific T-cell clones,20 expandedgluten-specific CD391 T-cell populations lost FOXP3 expressionin vitro within 14 days, which corresponded to increased CpGmethylation in the TSDR. This might indicate thatgluten-specific CD391 Treg cells retain a high degree ofplasticity,43 although expanded healthy Treg cells also acquiredmethylation in vitro to a lesser extent.

Compared with healthy control subjects, patients with celiacdisease before challenge had significantly more CD391 cellswithin CD45RO1 Treg cells yet lower absolute numbers ofCD45RO1 Treg cells. This is likely due to a single nucleotidepolymorphism in the CD39 gene that determines CD39expression levels in Treg cells, and futurework should investigatethe association of such CD39 single nucleotide polymorphismswith celiac disease.44 CD391 Treg cells have been shown to bepotent suppressors of IFN-g and IL-17 and to be increased inthe synovia of patients with juvenile arthritis.44 ThereforeCD391 Treg cells might be preferentially expanded, yet theirnumbers are insufficient to control inflammation after glutenexposure in patients with celiac disease. CD39 expression onTreg cells might also be useful in predicting clinical outcomes

TABLE I. Celiac disease cohort symptoms and CD41 T-cell responses to gluten antigens Q9

ID Symptoms after gluten challenge

Post–gluten challenge

gluten peptide mix response,

OX40 assayz

Post–gluten challenge

gluten peptide mix response,

IFN-g secretion

0062* Mild depressed mood and lethargy (days 4-6) Detected Detected

0077* Asymptomatic Not detected§ Detected

0080* Mild nausea (days 1-3) Not detected Not detected

0152� Severe vomiting, lethargy, and diarrhea (days 1-3); moderate nausea, bloating,

and abdominal pain (days 1-3)

Detected Detected

0159* Mild bloating, abdominal pain, and lethargy (days 2-3) Detected Detected

0174� Severe nausea, vomiting, abdominal pain, and diarrhea (days 1-3); moderate

lethargy, hot flushes, cold sweats, and flatulence (days 2-4)

Detected Detected

0196� Moderate-to-severe abdominal pain, nausea, and lethargy and mild diarrhea

(days 1-3)

Detected Detected (ELISA)

0230* Mild nausea and diarrhea (days 1-3) Detected Detected

0239* Asymptomatic Not detected Detected (at assay

limit of detection)

0250� Severe vomiting, nausea, abdominal pain, lethargy, and cold sweats

(days 1-3); mild bloating and diarrhea (days 2-4)

Detected Detected

0251� Moderate-to-severe abdominal pain, bloating, nausea, and lethargy Detected Detected (ELISA)

0505* Severe lethargy and mild diarrhea (days 1-3) Not detected Not detected

0506* Mild constipation (days 1-3) Not detected Not detected

0509* Mild bloating, abdominal pain, and diarrhea (day 1) Not detected Not detected

0510� Severe bloating, abdominal pain, and constipation (days 203) Not detected Not detected

0512* Asymptomatic Detected Detected

0072� Moderate-to-severe nausea, bloating, and vomiting (day 1);

moderate abdominal pain and lethargy (day 1)

Detected Detected

*Nonsevere group: reported mild or no symptoms.

�Severe group: reported moderate-to-severe symptoms.

�Post–gluten challenge OX40 assay responses were only listed as detected if they were greater than the baseline response.

§Cohort analyses were performed with 15 patients: patient #0077 was not included because day 8 OX40 assays used cryopreserved PBMCs, and patient #0072 was not included

because day 8 analysis was not performed.

J ALLERGY CLIN IMMUNOL

nnn 2017

10 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1077

1078

1079

1080

1081

1082

1083

1084

1085

1086

1087

1088

1089

1090

1091

1092

1093

1094

1095

1096

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114

1115

1116

1117

1118

1119

1120

1121

1122

1123

1124

1125

1126

1127

1128

1129

1130

1131

1132

1133

1134

1135

1136

1137

1138

1139

1140

1141

1142

1143

1144

1145

1146

1147

1148

1149

1150

1151

1152

1153

1154

1155

1156

1157

1158

1159

1160

1161

1162

1163

1164

1165

1166

1167

1168

1169

1170

1171

1172

1173

1174

1175

1176

1177

1178

1179

1180

1181

1182

1183

1184

1185

1186

1187

1188

1189

1190

1191

1192

1193

1194

1195

1196

because low CD39 expression has been associated with betterCD41 T-cell recovery after antiretroviral therapy in HIV1

patients20 and methotrexate resistance in patients withrheumatoid arthritis.45

Together, the data presented here indicate that in patients withceliac disease after gluten challenge, FOXP31CD391 Treg cellsdominate peripheral recall responses to gluten and can be readilyexpanded on in vivo antigen challenge yet exhibit impairedin vitro suppressive function. Therefore one interpretation ofthese data is that, in response to gluten challenge,FOXP31CD391 Treg cells are induced in vivo in an attempt torestore homeostasis. However, the generated cells have impairedsuppressive function, possibly as a result of generation underinflammatory conditions in vivo. A key area for furtherinvestigation is whether in vivo challenge conditions could bemanipulated to drive expansion of functional Treg cells.

We thank Dr Anne Pesenacker and Dr Kate MacDonald for helpful

discussions; Ms Cathy Pizzey for her assistance with patient visit scheduling

and data and sample collection; andMs Lisa Xu,MsYinXu,MsAnnett Howe,

andMsMichelle Bailey for fluorescence-activated cell sorting isolation of cell

populations. All healthy subject and patients with celiac disease are thanked

for their participation in the study.

Key messages

d In patients with celiac disease, 6 days after gluten

challenge in vivo, a surprisingly large proportion of

circulating gluten-specific CD41 T cells are FOXP31

CD391 Treg cells.

d In patients with celiac disease after gluten challenge,

gluten-specific Treg cells exhibit impaired polyclonal sup-

pressive function in vitro, suggesting that an intrinsic

dysfunction of expanded CD391 Treg cells might

contribute to the loss of tolerance to gluten.

d Detection of gluten-specific CD41 T cells based on

antigen-induced coexpression of CD25 and OX40 is

more sensitive than traditional methods relying on

antigen-induced cytokine production and, for the first

time, allows detailed characterization of antigen-specific

Treg cells in patients with this disease.

REFERENCES

1. Sollid LM. Molecular basis of celiac disease. Annu Rev Immunol 2000;18:

53-81.

2. Molberg O, McAdam SN, Korner R, Quarsten H, Kristiansen C, Madsen L, et al.

Tissue transglutaminase selectively modifies gliadin peptides that are recognized

by gut-derived T cells in celiac disease. Nat Med 1998;4:713-7.

3. Sollid LM, Qiao S-W, Anderson RP, Gianfrani C, Koning F. Nomenclature and

listing of celiac disease relevant gluten T-cell epitopes restricted by HLA-DQ

molecules. Immunogenetics 2012;64:455-60.

4. Tye-Din JA, Stewart JA, Dromey JA, Beissbarth T, van Heel DA, Tatham A, et al.

Comprehensive, quantitative mapping of T cell epitopes in gluten in celiac disease.

Sci Transl Med 2010;2:41ra51.

5. Anderson RP, Degano P, Godkin AJ, Jewell DP, Hill AV. In vivo antigen challenge

in celiac disease identifies a single transglutaminase-modified peptide as the

dominant A-gliadin T-cell epitope. Nat Med 2000;6:337-42.

6. Abadie V, Discepolo V, Jabri B. Intraepithelial lymphocytes in celiac disease

immunopathology. Semin Immunopathol 2012;34:551-66.

7. Anderson RP, van Heel DA, Tye-Din JA, Barnardo M, Salio M, Jewell DP, et al.

T cells in peripheral blood after gluten challenge in coeliac disease. Gut 2005;

54:1217-23.

8. Gianfrani C, Levings MK, Sartirana C, Mazzarella G, Barba G, Zanzi D, et al.

Gliadin-specific type 1 regulatory T cells from the intestinal mucosa of treated

celiac patients inhibit pathogenic T cells. J Immunol 2006;177:4178-86.

9. Frisullo G, Nociti V, Iorio R, Patanella AK, Marti A, Assunta B, et al. Increased

CD41CD251Foxp31 T cells in peripheral blood of celiac disease patients:

correlation with dietary treatment. Hum Immunol 2009;70:430-5.

10. Zanzi D, Stefanile R, Santagata S, Iaffaldano L, Iaquinto G, Giardullo N, et al.

IL-15 interferes with suppressive activity of intestinal regulatory T cells expanded

in celiac disease. Am J Gastroenterol 2011;106:1308-17.

11. Granzotto M, dal Bo S, Quaglia S, Tommasini A, Piscianz E, Valencic E, et al.

Regulatory T-cell function is impaired in celiac disease. Dig Dis Sci 2009;54:

1513-9.

12. Hmida NB, Ben Ahmed M, Moussa A, Rejeb MB, Said Y, Kourda N, et al. Impaired

control of effector T cells by regulatory T cells: a clue to loss of oral tolerance and

autoimmunity in celiac disease? Am J Gastroenterol 2012;107:604-11.

13. Ben Ahmed M, Belhadj Hmida N, Moes N, Buyse S, Abdeladhim M, Louzir H,

et al. IL-15 renders conventional lymphocytes resistant to suppressive functions

of regulatory T cells through activation of the phosphatidylinositol 3-kinase

pathway. J Immunol 2009;182:6763-70.

14. DePaolo RW, Abadie V, Tang F, Fehlner-Peach H, Hall JA, Wang W, et al.

Co-adjuvant effects of retinoic acid and IL-15 induce inflammatory immunity

to dietary antigens. Nature 2011;471:220-4.

15. Revised criteria for diagnosis of coeliac disease. Report of Working Group of

European Society of Paediatric Gastroenterology and Nutrition. Arch Dis Child

1990;65:909-11.

16. Koskinen L, Romanos J, Kaukinen K, Mustalahti K, Korponay-Szabo I, Barisani D,

et al. Cost-effective HLA typing with tagging SNPs predicts celiac disease risk hap-

lotypes in the Finnish, Hungarian, and Italian populations. Immunogenetics 2009;61:

247-56.

17. Monsuur AJ, de Bakker PI, Zhernakova A, Pinto D, Verduijn W, Romanos J, et al.

Effective detection of human leukocyte antigen risk alleles in celiac disease using

tag single nucleotide polymorphisms. PLoS One 2008;3:e2270.

18. Ontiveros N, Tye-Din JA, Hardy MY, Anderson RP. Ex vivo whole blood secretion

of interferon (IFN)-g and IFN-g-inducible protein-10 (IP-10) measured by ELISA

are as sensitive as IFN-g ELISpot for the detection of gluten-reactive T cells in

HLA-DQ2.51 associated celiac disease. Clin Exp Immunol 2014;175:305-15.

19. Tye-Din JA, Anderson RP, Ffrench RA, Brown GJ, Hodsman P, Siegel M, et al. The

effects of ALV003 pre-digestion of gluten on immune response and symptoms in

celiac disease in vivo. Clin Immunol 2010;134:289-95.

20. Seddiki N, Cook L, Hsu DC, Phetsouphanh C, Brown K, Xu Y, et al. Human

antigen-specific CD41CD251CD1341CD391 T cells are enriched for regulatory

T cells and comprise a substantial proportion of recall responses. Eur J Immunol

2014;44:1644-61.

21. Zaunders JJ, Munier ML, Seddiki N, Pett S, Ip S, Bailey M, et al. High levels of

human antigen-specific CD4(1) T cells in peripheral blood revealed by stimulated

coexpression of CD25 and CD134 (OX40). J Immunol 2009;183:2827-36.

22. Seddiki N, Santner-Nanan B, Martinson J, Zaunders J, Sasson S, Landay A, et al.

Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human

regulatory and activated T cells. J Exp Med 2006;203:1693-700.

23. Petersen J, Montserrat V, Mujico JR, Loh KL, Beringer DX, van Lummel M, et al.

T-cell receptor recognition of HLA-DQ2-gliadin complexes associated with celiac

disease. Nat Struct Mol Biol 2014;21:480-8.

24. Broughton SE, Petersen J, Theodossis A, Scally SW, Loh KL, Thompson A, et al.

Biased T cell receptor usage directed against human leukocyte antigen

DQ8-restricted gliadin peptides is associated with celiac disease. Immunity

2012;37:611-21.

25. Gregori S, Bacchetta R, Passerini L, Levings MK, Roncarolo MG. Isolation,

expansion, and characterization of human natural and adaptive regulatory T cells.

Methods Mol Biol 2007;380:83-105.

26. Kelleher AD, Long C, Holmes EC, Allen RL, Wilson J, Conlon C, et al.

Clustered mutations in HIV-1 gag are consistently required for escape from

HLA-B27-restricted cytotoxic T lymphocyte responses. J Exp Med 2001;193:

375-86.

27. Quigley MF, Almeida JR, Price DA, Douek DC. Unbiased molecular analysis of

T cell receptor expression using template-switch anchored RT-PCR. Curr Protoc

Cytom 2011 Chapter 10:Unt 10.33.

28. Giudicelli V, Brochet X, Lefranc MP. IMGT/V-QUEST: IMGT standardized

analysis of the immunoglobulin (IG) and T cell receptor (TR) nucleotide

sequences. Cold Spring Harb Protoc 2011;2011:695-715.

29. McMurchy AN, Levings MK. Suppression assays with human T regulatory cells: a

technical guide. Eur J Immunol 2012;42:27-34.

30. Qiao SW, Raki M, Gunnarsen KS, Loset GA, Lundin KE, Sandlie I, et al.

Posttranslational modification of gluten shapes TCR usage in celiac disease.

J Immunol 2011;187:3064-71.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 11

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1197

1198

1199

1200

1201

1202

1203

1204

1205

1206

1207

1208

1209

1210

1211

1212

1213

1214

1215

1216

1217

1218

1219

1220

1221

1222

1223

1224

1225

1226

1227

1228

1229

1230

1231

1232

1233

1234

1235

1236

1237

1238

1239

1240

1241

1242

1243

1244

1245

1246

1247

1248

1249

1250

1251

1252

1253

1254

1255

1256

1257

1258

1259

1260

1261

1262

1263

1264

1265

1266

1267

1268

1269

1270

1271

1272

1273

1274

1275

1276

1277

1278

1279

1280

1281

1282

1283

1284

1285

1286

1287

1288

1289

1290

1291

1292

1293

1294

1295

1296

1297

1298

1299

1300

1301

1302

1303

1304

1305

1306

1307

1308

1309

1310

1311

1312

1313

1314

1315

1316

31. Hsu DC, Zaunders JJ, Plit M, Leeman C, Ip S, Iampornsin T, et al. A novel assay

detecting recall response to Mycobacterium tuberculosis: comparison with existing

assays. Tuberculosis (Edinb) 2012;92:321-7.

32. Dan JM, Lindestam Arlehamn CS, Weiskopf D, da Silva Antunes R,

Havenar-Daughton C, Reiss SM, et al. A cytokine-independent approach

to identify antigen-specific human germinal center T follicular helper

cells and rare antigen-specific CD41 T cells in blood. J Immunol

2016;197:983-93.

33. Keoshkerian E, Helbig K, Beard M, Zaunders J, Seddiki N, Kelleher A, et al. A

novel assay for detection of hepatitis C virus-specific effector CD41 T cells via

co-expression of CD25 and CD134. J Immunol Methods 2012;375:148-58.

34. Tong WW, Shepherd K, Garland S, Meagher A, Templeton DJ, Fairley CK, et al.

HPV16-specific T-cell responses and spontaneous regression of anal high-grade

squamous intraepithelial lesions. J Infect Dis 2015;211:405-15.

35. Sadler R, Bateman EA, Heath V, Patel SY, Schwingshackl PP, Cullinane AC, et al.

Establishment of a healthy human range for the whole blood ‘OX40’ assay for the

detection of antigen-specific CD41 T cells by flow cytometry. Cytometry B Clin

Cytom 2014;86:350-61.

36. Vukmanovic-Stejic M, Sandhu D, Sobande TO, Agius E, Lacy KE, Riddell N, et al.

Varicella zoster-specific CD41Foxp31 T cells accumulate after cutaneous antigen

challenge in humans. J Immunol 2013;190:977-86.

37. Vukmanovic-Stejic M, Sandhu D, Seidel JA, Patel N, Sobande TO, Agius E, et al.

The characterization of varicella zoster virus-specific T cells in skin and blood

during aging. J Invest Dermatol 2015;135:1752-62.

38. Brincks EL, Roberts AD, Cookenham T, Sell S, Kohlmeier JE, Blackman MA,

et al. Antigen-specific memory regulatory CD41Foxp31 T cells control memory

responses to influenza virus infection. J Immunol 2013;190:3438-46.

39. Betts RJ, Prabhu N, Ho AW, Lew FC, Hutchinson PE, Rotzschke O, et al. Influenza

A virus infection results in a robust, antigen-responsive, and widely disseminated

Foxp31 regulatory T cell response. J Virol 2012;86:2817-25.

40. Bacher P, Heinrich F, Stervbo U, Nienen M, Vahldieck M, Iwert C, et al.

Regulatory T cell specificity directs tolerance versus allergy against aeroantigens

in humans. Cell 2016;167:1067-78.e16.

41. Su LF, Del Alcazar D, Stelekati E, Wherry EJ, Davis MM. Antigen exposure

shapes the ratio between antigen-specific Tregs and conventional T cells in human

peripheral blood. Proc Natl Acad Sci U S A 2016;113:E6192-8.

42. Weiner HL, da Cunha AP, Quintana F, Wu H. Oral tolerance. Immunol Rev 2011;

241:241-59.

43. He H, Ni B, Tian Y, Tian Z, Chen Y, Liu Z, et al. Histone methylation mediates

plasticity of human FOXP3(1) regulatory T cells by modulating signature gene

expressions. Immunology 2014;141:362-76.

44. Rissiek A, Baumann I, Cuapio A, Mautner A, Kolster M, Arck PC, et al. The

expression of CD39 on regulatory T cells is genetically driven and further

upregulated at sites of inflammation. J Autoimmun 2015;58:12-20.

45. Peres RS, Liew FY, Talbot J, Carregaro V, Oliveira RD, Almeida SL, et al. Low

expression of CD39 on regulatory T cells as a biomarker for resistance to

methotrexate therapy in rheumatoid arthritis. Proc Natl Acad Sci U S A 2015;

112:2509-14.

J ALLERGY CLIN IMMUNOL

nnn 2017

12 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1317

1318

1319

1320

1321

1322

1323

1324

1325

1326

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340

1341

1342

1343

1344

1345

1346

1347

1348

1349

1350

1351

1352

1353

1354

1355

1356

1357

1358

1359

1360

1361

1362

1363

1364

1365

1366

1367

1368

1369

1370

1371

1372

1373

1374

1375

1376

1377

1378

1379

1380

1381

1382

1383

1384

1385

1386

1387

1388

1389

1390

1391

1392

1393

1394

1395

1396

1397

1398

1399

1400

1401

1402

1403

1404

1405

1406

1407

1408

1409

1410

1411

1412

1413

1414

1415

1416

1417

1418

1419

1420

1421

1422

1423

1424

1425

1426

1427

1428

1429

1430

1431

1432

1433

1434

1435

1436

FIG E1. Optimization of OX40 assay time course and gluten antigen concentration. A, In a pilot study OX40

assays were performed with peripheral blood from 3 patients (#0080, #0152, and #0250) and 1 HLA-DQ21

subject without celiac disease (#0458). Blood was collected before gluten challenge (day 0) and at days 4,

6, 8, and 10 after challenge. OX40 assay responses (CD251OX401) are shown for gluten peptide mix,

DQ2.5-glia-a1/a2, DQ2.5-glia-v1/v2 deamidated gliadin, SEB, or tetanus toxoid (Tet Tox). B, OX40 assay

responses are shown at days 6 and 8 to 10, 50, and 100 mg/mL deamidated gliadin; 50 and 125 mg/mL

DQ2.5-glia-a1/a2; or 50 and 125 mg/mL DQ2.5-glia-v1/v2. C, OX40 assays were performed with peripheral

blood of 6 healthy subjects. The deamidated gliadin responses are shown for a single experiment with

each subject. Error bars represent means 6 SEMs.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 12.e1

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1437

1438

1439

1440

1441

1442

1443

1444

1445

1446

1447

1448

1449

1450

1451

1452

1453

1454

1455

1456

1457

1458

1459

1460

1461

1462

1463

1464

1465

1466

1467

1468

1469

1470

1471

1472

1473

1474

1475

1476

1477

1478

1479

1480

1481

1482

1483

1484

1485

1486

1487

1488

1489

1490

1491

1492

1493

1494

1495

1496

1497

1498

1499

1500

1501

1502

1503

1504

1505

1506

1507

1508

1509

1510

1511

1512

1513

1514

1515

1516

1517

1518

1519

1520

1521

1522

1523

1524

1525

1526

1527

1528

1529

1530

1531

1532

1533

1534

1535

1536

1537

1538

1539

1540

1541

1542

1543

1544

1545

1546

1547

1548

1549

1550

1551

1552

1553

1554

1555

1556

FIG E2. Changes in peripheral Treg cell frequencies and OX40 assay CD391 proportions after gluten

challenge. A and B, Frequencies of CD41 T cells that were memory CD391FOXP31 Treg cells (Fig E3, A)

and integrin b71CD391 memory Treg cells (Fig E3, B) are shown for patients who did not have detectable

gluten peptide responses (n 5 6). C, The proportion of CD39 expression within OX40 assay responses is

plotted for assays stimulated with SEB (n 5 15), tetanus toxoid (Tet Tox; n 5 15), deamidated gliadin

(n 5 10), and gluten peptide mix (n 5 7). Subjects were only included in analyses if they had detectable

responses at all 3 time points or, for gluten peptide mix, if they had detectable responses at both day 6

and day 8. Statistical analyses were performed with Wilcoxon signed-rank tests. ND, Not detected; ns,

not significant. Each subject is represented by a different symbol, and red connecting lines indicate those

subjects with gluten peptide responses. D and E, Total CD41CD251OX401 T-cell responses to SEB and

the proportion of CD391 cells within these responses are shown for healthy subjects (n 5 15; Fig E3, D)

and patients with celiac disease (n 5 15; Fig E3, E). Means 6 SEMs are shown.

J ALLERGY CLIN IMMUNOL

nnn 2017

12.e2 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1557

1558

1559

1560

1561

1562

1563

1564

1565

1566

1567

1568

1569

1570

1571

1572

1573

1574

1575

1576

1577

1578

1579

1580

1581

1582

1583

1584

1585

1586

1587

1588

1589

1590

1591

1592

1593

1594

1595

1596

1597

1598

1599

1600

1601

1602

1603

1604

1605

1606

1607

1608

1609

1610

1611

1612

1613

1614

1615

1616

1617

1618

1619

1620

1621

1622

1623

1624

1625

1626

1627

1628

1629

1630

1631

1632

1633

1634

1635

1636

1637

1638

1639

1640

1641

1642

1643

1644

1645

1646

1647

1648

1649

1650

1651

1652

1653

1654

1655

1656

1657

1658

1659

1660

1661

1662

1663

1664

1665

1666

1667

1668

1669

1670

1671

1672

1673

1674

1675

1676

FIG E3. Characterization of expanded DQ2.5-glia-a1/a2–specific CD391 T cells. A, Quantitative RT-PCR was

used to compare FOXP3 and TGF-b mRNA expression levels in 14-day expanded gluten-specific CD391 T

cells to levels in expanded CD127lowCD25high Treg cells from healthy subjects (n 5 3) and expanded Treg

and CD1271CD252 Tconv cells from patients with celiac disease (n 5 2). Relative expression is shown after

normalization to b-actin. Means 6 SEMs are shown for 2 independent experiments. B, DQ2.5

tetramer (loaded with HLA class II invariant peptide or DQ2.5-glia-a1/a2) staining of 14-day expanded

DQ2.5-glia-a1/a2–specific CD251OX401 and non–gluten-specific CD252OX402 T cells. Frequencies of

tetramer-positive cells are representative of expanded CD391 cells and clones from 2 subjects. C, CD39

and Helios staining (representative of n 5 3) of expanded CD127lowCD25high Treg cells and

gluten-specific CD391 T cells from healthy subjects and patients with celiac disease. D, DQ2.5-glia-a1/a2–

specific CD251OX401CD391 (red) and non–gluten specific CD252OX402 (black) T cells were

fluorescence-activated cell sorting isolated directly after an OX40 assay and restimulated for 5 days with

the antigens indicated. Percentage proliferation shown is representative of 3 subjects. E, Histograms

showing expression of GARP and LAP in cells expanded from patient #0251 with celiac disease and a

healthy control subject. Median fluorescence intensity (MFI) Q10is shown in the legend. F, Percentage

methylation in the TSDR of FOXP3 is shown for 14-day expanded CD127lowCD25high Treg cells from healthy

subjects (n 5 3) and patients with celiac disease (n 5 3) and gluten-specific CD391 T cells (n 5 3;

median 6 interquartile range; all donors were female). ns, Not significant.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 12.e3

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1677

1678

1679

1680

1681

1682

1683

1684

1685

1686

1687

1688

1689

1690

1691

1692

1693

1694

1695

1696

1697

1698

1699

1700

1701

1702

1703

1704

1705

1706

1707

1708

1709

1710

1711

1712

1713

1714

1715

1716

1717

1718

1719

1720

1721

1722

1723

1724

1725

1726

1727

1728

1729

1730

1731

1732

1733

1734

1735

1736

1737

1738

1739

1740

1741

1742

1743

1744

1745

1746

1747

1748

1749

1750

1751

1752

1753

1754

1755

1756

1757

1758

1759

1760

1761

1762

1763

1764

1765

1766

1767

1768

1769

1770

1771

1772

1773

1774

1775

1776

1777

1778

1779

1780

1781

1782

1783

1784

1785

1786

1787

1788

1789

1790

1791

1792

1793

1794

1795

1796

FIG E4. Flow cytometric gating strategies. A, Gating strategy for OX40 assay analysis. Lymphocytes were

gated based on their forward- and side-scatter profiles, followed by gating CD31CD41 T cells. The

crosshatch gates used to define the antigen-specific CD251OX401 T cells were set initially on unstimulated

wells and refined by using responses in SEB-stimulated positive control wells. B,Gating strategy shown for

isolation and phenotypic analysis of peripheral CD41 memory T-cell populations. After gating on

lymphocytes and CD31CD41 T cells, the CD45RO1 memory cells were divided into Treg cells

(CD127lowCD25high) and the remaining non-Treg cells, with the Treg cells separated further into CD391

and CD392 populations. Expression of phenotypic markers, such as the gut-homing marker integrin b7,

was analyzed within each gated T-cell population. C, For 12 patients with celiac disease, the percentage

of CD391 cells within CD41 T cells is shown for PBMCs sourced directly ex vivo and from OX40 assay wells

after 44 hours of stimulation with no antigen, SEB, or gluten peptide mix.

J ALLERGY CLIN IMMUNOL

nnn 2017

12.e4 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1797

1798

1799

1800

1801

1802

1803

1804

1805

1806

1807

1808

1809

1810

1811

1812

1813

1814

1815

1816

1817

1818

1819

1820

1821

1822

1823

1824

1825

1826

1827

1828

1829

1830

1831

1832

1833

1834

1835

1836

1837

1838

1839

1840

1841

1842

1843

1844

1845

1846

1847

1848

1849

1850

1851

1852

1853

1854

1855

1856

1857

1858

1859

1860

1861

1862

1863

1864

1865

1866

1867

1868

1869

1870

1871

1872

1873

1874

1875

1876

1877

1878

1879

1880

1881

1882

1883

1884

1885

1886

1887

1888

1889

1890

1891

1892

1893

1894

1895

1896

1897

1898

1899

1900

1901

1902

1903

1904

1905

1906

1907

1908

1909

1910

1911

1912

1913

1914

1915

1916

FIG E5. Symptom severity associated with greater responses to gluten peptide antigen. Patients with celiac

disease were divided into groups based on symptom severity: severe, n 5 7; nonsevere, n 5 10. Groups

were compared by identifying differences in the proportion of CD391 cells within ex vivomemory Treg cells

(A), the proportion of CD39 expression within deamidated gliadin-specific CD251OX401 T cells (B), the

magnitude of day 6 postchallenge IFN-g ELISpot response to gluten peptide mix (n 5 5 in severe group

because n 5 2 only had ELISA data; (C), and the maximal OX40 assay response to gluten peptide mix

(D). Error bars represent means 6 SEMs. Mann-Whitney U tests were performed. ns, Not significant.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 12.e5

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

1917

1918

1919

1920

1921

1922

1923

1924

1925

1926

1927

1928

1929

1930

1931

1932

1933

1934

1935

1936

1937

1938

1939

1940

1941

1942

1943

1944

1945

1946

1947

1948

1949

1950

1951

1952

1953

1954

1955

1956

1957

1958

1959

1960

1961

1962

1963

1964

1965

1966

1967

1968

1969

1970

1971

1972

1973

1974

1975

1976

1977

1978

1979

1980

1981

1982

1983

1984

1985

1986

1987

1988

1989

1990

1991

1992

1993

1994

1995

1996

1997

1998

1999

2000

2001

2002

2003

2004

2005

2006

2007

2008

2009

2010

2011

2012

2013

2014

2015

2016

2017

2018

2019

2020

2021

2022

2023

2024

2025

2026

2027

2028

2029

2030

2031

2032

2033

2034

2035

2036

TABLE E1. Celiac disease cohort characteristics

ID Age (y) Sex Time since diagnosis tTG IgA (U/mL)* DGP IgA (U/mL)* HLA-DQ Haplotype

0062 66 F 5 y, 9 mo <20 <20 HLA-DQ2.5

0077 48 M 21 y <20 <20 HLA-DQ2.5

0080� 68 F 31 y <20 <20 HLA-DQ2.5

0152� 54 M 1 y, 9 mo <20 <20 HLA-DQ2.5

0159 67 F 14 y, 9 mo <20 <20 HLA-DQ2.5 (homozygous)

0174 51 F 15 y, 7 mo <20 <20 HLA-DQ2.5

0196 60 F 9 y, 3 mo <20 <20 HLA-DQ2.5

0230 53 F 12 y, 9 mo <20 <20 HLA-DQ2.5

0239 71 F 39 y, 3 mo <20 <20 HLA-DQ2.5 (homozygous)

0250 62 M 8 y, 5 mo <20 <20 HLA-DQ2.5

0251� 53 F 8 y, 2 mo <20 <20 HLA-DQ2.5

0505 35 F 34 y, 11 mo <20 <20 HLA-DQ2.2

0506 69 M 2 y, 1 mo <20 <20 HLA-DQ2.5 (heterozygous)

0509 29 F 7 y, 11 mo <20 <20 HLA-DQ2.5 (heterozygous)

0510 38 M 4 y, 11 mo 62 (strongly positive) 27 (weakly positive) HLA-DQ2.5 and HLA-DQ2.2

0512 52 F 1 y, 4 mo <20 29 (weakly positive) HLA-DQ2.5 (heterozygous)

0072 57 F 20 y, 2 mo <20 <20 HLA-DQ2.5 (heterozygous)

Mean 55 71% F

29% M

14 y, 1 mo <20 <20

All data were collected at the time of gluten challenge. All subjects have been on a gluten-free diet since diagnosis.

DGP, Deamidated gliadin peptide; F, female; M, male.

*Normal serology is less than 20 U/mL.

�Indicates subjects analyzed in the pilot study.

J ALLERGY CLIN IMMUNOL

nnn 2017

12.e6 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

2037

2038

2039

2040

2041

2042

2043

2044

2045

2046

2047

2048

2049

2050

2051

2052

2053

2054

2055

2056

2057

2058

2059

2060

2061

2062

2063

2064

2065

2066

2067

2068

2069

2070

2071

2072

2073

2074

2075

2076

2077

2078

2079

2080

2081

2082

2083

2084

2085

2086

2087

2088

2089

2090

2091

2092

2093

2094

2095

2096

2097

2098

2099

2100

2101

2102

2103

2104

2105

2106

2107

2108

2109

2110

2111

2112

2113

2114

2115

2116

2117

2118

2119

2120

2121

2122

2123

2124

2125

2126

2127

2128

2129

2130

2131

2132

2133

2134

2135

2136

2137

2138

2139

2140

2141

2142

2143

2144

2145

2146

2147

2148

2149

2150

2151

2152

2153

2154

2155

2156

TABLE E2. HLA-DQ alleles detected in the celiac disease

cohort

ID HLA-DQA HLA-DQB

0062 DQA1*05 DQB1*02

DQB1*03

0077 DQA1*01

DQA1*05

DQB1*02

DQB1*05

0080* DQA1*05:01 DQB1*02:01

DQB1*02:02

0152* DQA1*01

DQA1*05

DQB1*02

DQB1*06

0159 DQA1*05:01 DQB1*02:01

0174 DQA1*05:01 DQB1*02:01/02

DQB1*05:02

0196 DQA1*05:01 DQB1*02:01/02

DQB1*06:02

0230 DQA1*05:01 DQB1*02:01

DQB1*05:02

0239 DQA1*05:01 DQB1*02:01

0250* DQA1*02

DQA1*05

DQB1*02

0251 DQA1*01

DQA1*05

DQB1*02:01

DQB1*05:01

0505 DQA1*02

DQA1*04/06

DQB1*02

DQB1*04

0506 N/A N/A

0509 N/A N/A

0510 DQA1*02:01

DQA1*05:01

DQB1*02:01

DQB1*02:02

0512 NA NA

0072 NA NA

HLA-DQ2.5 refers to the DQA1*0501:DQB1*0201 haplotype, and HLA-DQ2.2

refers to the DQA1*0201:DQB1*0202 haplotype.

NA, HLA-DQB1 and HLA-DQA1 alleles were not assessed, and instead only the

presence of alleles encoding HLA-DQ2.5, HLA-DQ2.2, and HLA-DQ8 were detected,

as described in section 2.4.Q11

*Indicates subjects analyzed in the pilot study.

J ALLERGY CLIN IMMUNOL

VOLUME nnn, NUMBER nn

COOK ET AL 12.e7

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

2157

2158

2159

2160

2161

2162

2163

2164

2165

2166

2167

2168

2169

2170

2171

2172

2173

2174

2175

2176

2177

2178

2179

2180

2181

2182

2183

2184

2185

2186

2187

2188

2189

2190

2191

2192

2193

2194

2195

2196

2197

2198

2199

2200

2201

2202

2203

2204

2205

2206

2207

2208

2209

2210

2211

2212

2213

2214

2215

2216

2217

2218

2219

2220

2221

2222

2223

2224

2225

2226

2227

2228

2229

2230

2231

2232

2233

2234

2235

2236

2237

2238

2239

2240

2241

2242

2243

2244

2245

2246

2247

2248

2249

2250

2251

2252

2253

2254

2255

2256

2257

2258

2259

2260

2261

2262

2263

2264

2265

2266

2267

2268

2269

2270

2271

2272

2273

2274

2275

2276

TABLE E3. Phenotype of in vitro–expanded DQ2.5-glia-a1/a2–specific CD41CD251CD1341CD391 T cells

Molecule Clone C1 (#0174) Clone C2 (#0174) Clone C3 (#0251) CD391 cells (#0174) CD391 cells (#0230) CD391 cells (#0196) CD391 cells (#0251)

CD25 111 111 111 111 111 111 111

CD39 111 111 111 111 111 111 111

FOXP3 1 2 11 1 2 1 1

CTLA-4 111 111 11 111 111 11 111

CD45RO 111 111 111 111 111 111 111

Integrin b7 11 11 11 11 111 1 1

LAP NA NA 111 NA NA 1 1

GARP NA NA 11 NA NA 11 11

Helios NA NA 2 NA NA 2 2

CTLA-4, Cytotoxic T lymphocyte–associated antigen 4; NA, not assessed; 111, expression on greater than 90% of cells; 11, expression on 50% to 90% of cells; 1, expression

on 10% to 50% of cells; 2, expression on less than 10% of cells.

J ALLERGY CLIN IMMUNOL

nnn 2017

12.e8 COOK ET AL

FLA 5.4.0 DTD � YMAI12681_proof_–15-00411 � 20 March 2017 � 12:51 pm

2277

2278

2279

2280

2281

2282

2283

2284

2285

2286

2287

2288

2289

2290

2291

2292

2293

2294

2295

2296

2297

2298

2299

2300

2301

2302

2303

2304

2305

2306

2307

2308

2309

2310

2311

2312

2313

2314

2315

2316

2317

2318

2319

2320

2321

2322

2323

2324

2325

2326

2327

2328

2329

2330

2331

2332

2333

2334

2335

2336

2337

2338

2339

2340

2341

2342

2343

2344

2345

2346

2347

2348

2349

2350

2351

2352

2353

2354

2355

2356

2357

2358

2359

2360

2361

2362

2363

2364

2365

2366

2367

2368

2369

2370

2371

2372

2373

2374

2375

2376

2377

2378

2379

2380

2381

2382

2383

2384

2385

2386

2387

2388

2389

2390

2391

2392

2393

2394

2395

2396