b lymphocytes contribute to celiac disease pathogenesis

7
B Lymphocytes Contribute to Celiac Disease Pathogenesis Thomas Lejeune, 1,2 Celine Meyer, 3 and Valérie Abadie 3,4 1 Department of Microbiology, Infectiology, and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada; 2 Sainte-Justine Hospital Research Centre, Montreal, Quebec, Canada; 3 Department of Medicine; and 4 Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois C eliac disease (CD) is a complex intestinal disorder with autoimmune features that develops in geneti- cally susceptible individuals expressing HLA-DQ8 or HLA-DQ2 molecules. 1 The presence of antitissue trans- glutaminase 2 (TG2) and antideamidated gluten peptide antibodies represent strong disease markers. 2 Inammation and villous blunting in the small intestine result from an abnormal immune response to gluten proteins. CD is char- acterized by the loss of oral tolerance to gluten manifested by HLA-DQ2or HLA-DQ8restricted antigluten inamma- tory CD4 T cells and a massive expansion of cytotoxic CD8 þ intraepithelial lymphocytes (IELs) that mediate the killing of intestinal epithelial cells (IECs). 3 These observations have led to the general idea that CD is primarily a T-cellmedi- ated immune disorder. However, studies in human suggest that B cells could play a central role in CD pathogenesis. 4 We have recently engineered a mouse model of CD that de- velops villous atrophy (VA) in a gluten, HLA-DQ8, and TG2- dependent manner. Intestinal tissue destruction in this model is also dependent on CD4 þ and CD8 þ T cells and requires interferon-g. We now demonstrate that B cells are required for the development of VA and the associated full licensing of cytotoxic IELs in this pathophysiologically relevant mouse model of CD, providing support for the exploration of B-celldirected therapies for the treatment of CD. Methods DQ8-D d -villin-IL-15tg mice developing the main features of CD upon ingestion of gluten 3 and DQ8-D d -villin-IL-15tg crossed with muMT mice lacking mature B cells were used. B-Cell depletion was achieved by treating DQ8-D d -villin-IL-15tg mice with 250 mg antimouse CD20 (clone 5D2, isotype IgG2a; Genentech). Villous height-to-crypt depth ratio was quantita- tively determined on cross-sections of the terminal ileum, which is the primary site where VA develops in our mouse model. 3 The acquisition of cytotoxic properties by IELs was assessed by measuring the expression of the activating natural killer cell (NK) receptor NKG2D, and cytolytic granules (gran- zyme B, perforin) in IELs with the use of ow cytometry and quantitative polymerase chain reaction. 3 Detailed methods are described in the Supplementary Methods. Results To determine whether B cells were required for the development of tissue damage, we analyzed the develop- ment of VA in our DQ8-D d -villin-IL-15tg mouse model maintained on a gluten-free diet (sham), fed with gluten, or fed with gluten and treated with an anti-CD20 depleting antibody (Supplementary Figure 1 and Figure 1A and B). Despite B-cell depletion (Supplementary Figure 1BD) pre- venting the development of antigluten IgA and IgG anti- bodies upon introduction of gluten (Figure 1E and F), intestinal IgA-producing plasma cells (Supplementary Figure 1G) were still present after anti-CD20 treatment. Strikingly, the development of VA was impaired when B cells were depleted in gluten-fed mice (Figure 1A and B) demonstrating the requirement of gluten-specic humoral responses to promote a pathogenic response. Likewise, the development of VA was abrogated in DQ8-D d -villin-IL-15tg- muMT mice lacking mature B cells (Supplementary Figure 2A and B). Given that ablation of B cells signicantly reduced the development of villous atrophy and that studies in humans and in DQ8-D d -villin-IL-15tg mice 3 have demonstrated that IELs inltrating the celiac lesion are critically involved in tis- sue damage, we next assessed the impact of B-cell depletion on IELs. As shown in Figure 1C, the reduction of intestinal tissue damage in the absence of B lymphocytes was associated with a decrease in the number of IELs. Intestinal tissue destruction arises from the killing of IECs by IELs that have acquired a cytolytic phenotype. We found that the amount of cytotoxic CD8 þ IELs expressing the activating NK receptor NKG2D in the absence of inhibitory CD94/NKG2A receptors (Figure 1D) as well as the levels of the cytotoxic molecules granzyme B and perforin (Figure 1E and F and Supplementary Figure 2C and D) were decreased in mice lacking B cells, while the expression of the mouse NKG2D ligand, rae1, and the nonclassical major histocompatibility complex (MHC) class I molecule qa1 remained unchanged (Supplementary Figure 2E and 2F). Altogether, these results indicate that B cells play a role in CD pathogenesis by promoting the cytotoxic potential of IELs and the development of VA. Discussion B Cells as antigen-presenting cells have been involved in the development of organ-specic disorders. 5 In the context of CD, in vitro assays have shown that such cooperation between B cells and gluten-specic CD4 T cells can take place. 4 The strict dependence on gluten exposure for the Abbreviations used in this paper: CD, celiac disease; IELs, intraepithelial lymphocytes; IECs, intestinal epithelial cells; TG2, tissue transglutaminase 2; VA, villous atrophy. Most current article © 2021 by the AGA Institute 0016-5085/$36.00 https://doi.org/10.1053/j.gastro.2021.02.063 Gastroenterology 2021;160:26082610 BRIEF COMMUNICATIONS

Upload: others

Post on 17-Jun-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: B Lymphocytes Contribute to Celiac Disease Pathogenesis

Gastroenterology 2021;160:2608–2610

BRIEFCOM

MUNICATIONS

B Lymphocytes Contribute to Celiac Disease Pathogenesis

Thomas Lejeune,1,2 Celine Meyer,3 and Valérie Abadie3,4

1Department of Microbiology, Infectiology, and Immunology, Faculty of Medicine, University of Montreal, Montreal, Quebec,Canada; 2Sainte-Justine Hospital Research Centre, Montreal, Quebec, Canada; 3Department of Medicine; and 4Section ofGastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois

eliac disease (CD) is a complex intestinal disorder

Abbreviations used in this paper: CD, celiac disease; IELs, intraepitheliallymphocytes; IECs, intestinal epithelial cells; TG2, tissue transglutaminase2; VA, villous atrophy.

Most current article

© 2021 by the AGA Institute0016-5085/$36.00

https://doi.org/10.1053/j.gastro.2021.02.063

Cwith autoimmune features that develops in geneti-cally susceptible individuals expressing HLA-DQ8 orHLA-DQ2 molecules.1 The presence of anti–tissue trans-glutaminase 2 (TG2) and anti–deamidated gluten peptideantibodies represent strong disease markers.2 Inflammationand villous blunting in the small intestine result from anabnormal immune response to gluten proteins. CD is char-acterized by the loss of oral tolerance to gluten manifestedby HLA-DQ2– or HLA-DQ8–restricted antigluten inflamma-tory CD4 T cells and a massive expansion of cytotoxic CD8þ

intraepithelial lymphocytes (IELs) that mediate the killing ofintestinal epithelial cells (IECs).3 These observations haveled to the general idea that CD is primarily a T-cell–medi-ated immune disorder. However, studies in human suggestthat B cells could play a central role in CD pathogenesis.4 Wehave recently engineered a mouse model of CD that de-velops villous atrophy (VA) in a gluten, HLA-DQ8, and TG2-dependent manner. Intestinal tissue destruction in thismodel is also dependent on CD4þ and CD8þ T cells andrequires interferon-g. We now demonstrate that B cells arerequired for the development of VA and the associated fulllicensing of cytotoxic IELs in this pathophysiologicallyrelevant mouse model of CD, providing support for theexploration of B-cell–directed therapies for the treatmentof CD.

MethodsDQ8-Dd-villin-IL-15tg mice developing the main features of

CD upon ingestion of gluten3 and DQ8-Dd-villin-IL-15tg crossedwith muMT mice lacking mature B cells were used. B-Celldepletion was achieved by treating DQ8-Dd-villin-IL-15tg micewith 250 mg anti–mouse CD20 (clone 5D2, isotype IgG2a;Genentech). Villous height-to-crypt depth ratio was quantita-tively determined on cross-sections of the terminal ileum,which is the primary site where VA develops in our mousemodel.3 The acquisition of cytotoxic properties by IELs wasassessed by measuring the expression of the activating naturalkiller cell (NK) receptor NKG2D, and cytolytic granules (gran-zyme B, perforin) in IELs with the use of flow cytometry andquantitative polymerase chain reaction.3 Detailed methods aredescribed in the Supplementary Methods.

ResultsTo determine whether B cells were required for the

development of tissue damage, we analyzed the develop-ment of VA in our DQ8-Dd-villin-IL-15tg mouse modelmaintained on a gluten-free diet (sham), fed with gluten, orfed with gluten and treated with an anti-CD20 depleting

antibody (Supplementary Figure 1 and Figure 1A and B).Despite B-cell depletion (Supplementary Figure 1B–D) pre-venting the development of antigluten IgA and IgG anti-bodies upon introduction of gluten (Figure 1E and F),intestinal IgA-producing plasma cells (SupplementaryFigure 1G) were still present after anti-CD20 treatment.Strikingly, the development of VA was impaired when Bcells were depleted in gluten-fed mice (Figure 1A and B)demonstrating the requirement of gluten-specific humoralresponses to promote a pathogenic response. Likewise, thedevelopment of VA was abrogated in DQ8-Dd-villin-IL-15tg-muMT mice lacking mature B cells (SupplementaryFigure 2A and B).

Given that ablation of B cells significantly reduced thedevelopment of villous atrophy and that studies in humansand in DQ8-Dd-villin-IL-15tg mice3 have demonstrated thatIELs infiltrating the celiac lesion are critically involved in tis-sue damage, we next assessed the impact of B-cell depletionon IELs. As shown in Figure 1C, the reduction of intestinaltissue damage in the absence of B lymphocyteswas associatedwith a decrease in the number of IELs. Intestinal tissuedestruction arises from the killing of IECs by IELs that haveacquired a cytolytic phenotype. We found that the amount ofcytotoxic CD8þ IELs expressing the activating NK receptorNKG2D in the absence of inhibitory CD94/NKG2A receptors(Figure 1D) as well as the levels of the cytotoxic moleculesgranzymeB and perforin (Figure 1E and F and SupplementaryFigure 2C andD) were decreased inmice lacking B cells, whilethe expression of the mouse NKG2D ligand, rae1, and thenonclassical major histocompatibility complex (MHC) class Imolecule qa1 remained unchanged (Supplementary Figure 2Eand 2F). Altogether, these results indicate that B cells play arole in CD pathogenesis by promoting the cytotoxic potentialof IELs and the development of VA.

DiscussionB Cells as antigen-presenting cells have been involved in

the development of organ-specific disorders.5 In the contextof CD, in vitro assays have shown that such cooperationbetween B cells and gluten-specific CD4 T cells can takeplace.4 The strict dependence on gluten exposure for the

Page 2: B Lymphocytes Contribute to Celiac Disease Pathogenesis

Fol

d ch

ange

Prf

1 ex

pres

sion

rel

ativ

e to

sha

m-f

ed m

ice

#IE

Ls /

100

IEC

s

P = .0005

isotype anti-CD20

P = .0913

isotype anti-CD20

Fol

d ch

ange

Gzm

B e

xpre

ssio

n r

ela t

i ve

to s

ham

-fed

mic

e

CD

8+ N

KG

2D+ IE

Ls /

100

IEC

s

P = .0498

P = .0101

Agluten

+ anti-CD20gluten

+ isotypesham

B ns

C

P < .0001P = .0121

P = .0156D

ns

E F

sham

glute

n

+ iso

type

glute

n

+ an

ti-CD20

sham

glute

n

+ iso

type

glute

n

+ an

ti-CD20

sham

glute

n

+ iso

type

glute

n

+ an

ti-CD20

Vill

ous/

cryp

t rat

io

P = .0003

P = .0027

0

1

2

3

4

0

10

20

30

40

0

0.5

1

1.5

0

0.5

1

1.5

2

2.5

0

0.5

1

1.5

2

2.5

Figure 1. The development of villous atrophy and the acquisition of cytotoxic properties by intraepithelial CD8þ T cells in DQ8-Dd-villin-IL-15tg mice is impaired in the absence of B cells. DQ8-Dd-villin-IL-15tg mice were maintained on a gluten free diet(sham), or fed with gluten every other day for 30 days and treated with 250 mg anti-CD20 antibody (gluten þ anti-CD20) or itsisotype control (gluten þ isotype) every 2 weeks. (A) Hematoxylin-stained ileum sections showing villous atrophy—as evi-denced by villous height-to-crypt depth ratio �2—in the gluten-fed DQ8-Dd-villin-IL-15tg mice. Scale bar ¼ 250 mm. (B)Morphometric assessment of the villous height-to-crypt depth ratio demonstrating villous atrophy in gluten-fed mice (ratio �2)compared with sham-fed mice and gluten-fed mice treated with the anti-CD20 antibody. (C) Quantification of intraepitheliallymphocytes (IELs) among intestinal epithelial cells (IECs); mean. (D) The intestinal epithelium was isolated and analyzed byflow cytometry. IELs were identified as TCRbþ CD4� CD8þ cells, and the frequency of CD8þ NKG2Dþ TCRabþ cells wereobtained. The total amount of IELs per 100 IECs was determined on hematoxylin and eosin–stained slides. NKG2Dþ NKG2�

IELs are indicated by absolute number per 100 IECs. (E) The expression of perforin (prf1) in the epithelial compartment wasmeasured by quantitative polymerase chain reaction. Relative expression levels in gluten-fed mice were normalized to theexpression levels observed in sham-fed mice. (F) The expression of granzyme B (GzmB) was measured as in E. Data arerepresentative of 5 (B, C) or 4 (D–F) independent experiments, shown as mean ± SEM; ANOVA/Tukey multiple comparison wasperformed for B, C, and D. Unpaired Student t test was used for E and F.

June 2021 B Cells and Celiac Disease 2609

BRIEFCO

MMUN

ICAT

IONS

production of anti-TG2 antibodies suggests that an interac-tion between gluten-specific CD4þ T cells and TG2-specific Bcells having internalized TG2-gluten complexes is requiredfor their generation (as reviewed by Iversen and Sollid4). Inaddition, a transcriptional B-cell signature was associatedwith the extent of tissue damage in CD.6 Finally, plasma cellswere found to be the most abundant gluten peptide MHC-expressing cells in the lamina propria of CD patients.7

The present study demonstrates that B cells play a rolein the activation of IELs and tissue damage, both of whichare key features of active CD.1,2 Although B-cell depletiondid not restore the villous height-to-crypt depth ratio andthe levels of cytotoxic IELs to steady-state level, our resultsdemonstrate that B cells significantly contribute to both thedevelopment of VA and the acquisition of cytotoxic prop-erties by CD8þ IELs, which are known to be responsible for

enterocytes cytolysis. These results in combination with ourprevious work highlighting a role for TG2 activation andCD4þ T cells in promoting tissue damage and IEL activa-tion,3 suggest that B cells may be involved in the amplifi-cation of the CD4þ T-cell response to a magnitude sufficientto affect the activation of cytotoxic IELs and the ensuingtissue destruction. While providing evidence for a role for Bcells in the activation of IELs and tissue damage, this studydoes not rule out that antibodies also could play a role in CDpathogenesis.8 Interestingly, the critical role of antigenpresentation by B cells to CD4þ T cells for enhancingdestructive immune reactions was also suggested in thecontext of experimental autoimmune encephalomyelitis andlupus.5 Future studies will determine the role of HLA-DQ2/DQ8 expression on B cells and assess their role as antigen-presenting cells in CD pathogenesis.

Page 3: B Lymphocytes Contribute to Celiac Disease Pathogenesis

2610 Lejeune et al Gastroenterology Vol. 160, No. 7

BRIEFCOM

MUNICATIONS

Supplementary MaterialNote: To access the supplementary material accompanyingthis article, visit the online version of Gastroenterology atwww.gastrojournal.org, and at https://doi.org/10.1053/j.gastro.2021.02.063.

References

1. Abadie V, Sollid LM, Barreiro LB, et al. Integration of

genetic and immunological insights into a model of celiacdisease pathogenesis. Annu Rev Immunol 2011;29:493–525.

2. Husby S, Koletzko S, Korponay-Szabo I, et al. EuropeanSociety Paediatric Gastroenterology, Hepatology andNutrition Guidelines for Diagnosing Coeliac Disease2020. J Pediatr Gastroenterol Nutr 2020;70:141–156.

3. Abadie V, Kim SM, Lejeune T, et al. IL-15, gluten andHLA-DQ8 drive tissue destruction in coeliac disease.Nature 2020;578:600–604.

4. Iversen R, Sollid LM. Autoimmunity provoked by foreignantigens. Science 2020;368:132–133.

5. Getahun A, Cambier JC. Non–antibody-secreting func-tions of b cells and their contribution to autoimmunedisease. Annu Rev Cell Dev Biol 2019;35:337–356.

6. Garber ME, Saldanha A, Parker JS, et al. A B-cell genesignature correlates with the extent of gluten-inducedintestinal injury in celiac disease. Cell Mol GastroenterolHepatol 2017;4:1–17.

7. Hoydahl LS, Richter L, Frick R, et al. Plasma cells are themost abundant gluten peptide MHC-expressing cells ininflamed intestinal tissues from patients with celiac dis-ease. Gastroenterology 2019;156:1428–1439.e10.

8. Maglio M, Troncone R. Intestinal anti-tissue trans-glutaminase2 autoantibodies: pathogenic and clinicalimplications for celiac disease. Front Nutr 2020;7:73.

Author names in bold designate shared co-first authorship.

CorrespondenceAddress correspondence to: Valérie Abadie, PhD, Department of Medicine,University of Chicago, 900 East 57th Street, Mailbox #9, Chicago, Illinois.e-mail: [email protected].

CRediT Authorship ContributionsThomas Lejeune, PhD (Data curation: Lead; Formal analysis: Supporting;Investigation: Lead);

Celine Meyer, PhD (Data curation: Supporting; Formal analysis: Supporting);Valerie Abadie, PhD (Conceptualization: Lead; Formal analysis: Lead;

Funding acquisition: Lead; Methodology: Lead; Supervision: Lead; Writing –

original draft: Lead; Writing – review & editing: Lead)

Conflicts of InterestThe authors declare no conflicts.

FundingThis work was supported by a grant from the SickKids Foundation (NI15-040),a Canadian Institute of Health Research operating grant (376783), a Pilot andFeasibility award from the Digestive Diseases Research Core Center (P30DK42086), and a Young Investigator Award from the Celiac DiseaseFoundation to Valérie Abadie, and awards from the Wallonie-BruxellesInternational-World Excellence and the Fonds de Recherche duQuébec—Nature et Technologies to Thomas Lejeune.

Page 4: B Lymphocytes Contribute to Celiac Disease Pathogenesis

Supplementary Methods

MiceMice used in these studies were on the C57BL/6 back-

ground. Mice were maintained under specific pathogen-freeconditions at the University of Chicago and at the Sainte-Justine University Hospital Research Centre. DQ8-Dd-villin-IL-15tg mice were recently described.3 DQ8-Dd-villin-IL-15tg-muMT mice were obtained by crossing DQ8-Dd-villin-IL-15tg mice with muMT mice obtained from Jackson Lab-oratories (Bar Harbor, ME, USA). All strains were main-tained from birth on a gluten-free chow (AIN76A; Envigo).For all experiments, mice were used at 10 weeks of age. Allexperiments were performed in accordance with the Insti-tutional Biosafety Committee, the Institutional Care and UseCommittee of the University of Chicago, the CanadianCouncil on Animal Care guidelines, and the InstitutionalCommittee for Animal Care in Research of the Sainte-JustineUniversity Hospital Research Centre.

Gluten feeding and B-cell depletionTo study the response to dietary gluten, mice were

transferred from a gluten-free diet to a standard rodentchow at the beginning of each experiment and allowed toconsume the gluten-containing chow ad libitum. In addition,supplemental gluten (20 mg crude gliadin; Sigma-Aldrich)was administered via intragastric gavage every other dayfor 30 days, with the use of a 22-gauge round-tipped needle(Cadence Science). To study the response to dietary glutenin the absence of mature B cells, DQ8-Dd-villin-IL-15tg micewere injected intraperitoneally every 2 weeks, starting at 7days before the introduction of gluten, with 250 mg of aCD20-specific monoclonal antibody (anti-CD20 antibody,clone 5D2, Genentech) or its isotype control (IgG2a; Bio-XCell), as shown in Supplementary Figure 1A.

HistologyHematoxylin and eosin staining was performed on 5mM

thick sections of 10% formalin-fixed paraffin-embeddedileum. The segments of the distal ileum analyzed wereconsistently taken 0.5 cm from the cecum. Slides wereanalyzed with the use of a Leica DMi8 microscope with HCPL Fluotar L �20/0.40 and HC PL APO �40/0.75 objectivesand equipped with the image processing and analysis soft-ware LasX (Leica). All assessments were performed blindlyby 2 independent investigators. The villous height/cryptdepth ratios were obtained from morphometric measure-ments of 5 well-oriented villi. The villous height-to-cryptratio was calculated by dividing the villous height by thecorresponding crypt depth. Villous height was measuredfrom the tip to the shoulder of the villous or up to the top ofthe crypt of Lieberkuhn. The crypt depth was measured asthe distance from the top of the crypt of Lieberkuhn to thedeepest level of the crypt. If the poor orientation of a sectionprevented a correct morphometric assessment of the sam-ple, additional tissue sections were cut and analyzed. Theintraepithelial lymphocyte count was assessed by counting

the number of intraepithelial lymphocytes among at least100 enterocytes.

Epithelial compartment, lamina propria, spleen,mesenteric lymph nodes, Peyer’s patches, andblood cell isolation

Epithelial cells including intraepithelial lymphocytes(IELs) and lamina propria cells were isolated as previouslydescribed with the use of EDTA containing calcium-freemedium and collagenase VIII, respectively. For the anal-ysis of the natural killer cell (NK) receptors by means offlow cytometry, a cell purification step using a 40% Percoll(GE Healthcare) was used to enrich lymphocyte cell pop-ulations as previously described.3 Spleen, mesenteric lymphnodes, and Peyer’s patches were dissected, made into asingle-cell suspension by means of mechanical disruption,and passed through a 70-mm nylon cell strainer (Corning).Blood was collected from the submandibular vein intoheparinized tubes, and red blood cells were lysed with theuse of the lysis buffer solution from BD Biosciences.

Flow cytometryThe following conjugated antibodies were purchased

from eBioscience: TCRb APC (H57-597) CD8a APC-eFluor780 (53-6.7), CD8bPE-Cy5 (eBio H35-17.2), and CD314(NKG2D) PE (CX5). The following antibodies were pur-chased from BD Biosciences: CD4 PE-Cy7 (GK1.5), NKG2A/C/E FITC (20d5), IgA FITC (C10–3), CD16/CD32 (Fc Block)(2.4G2), CD3 V500 (UCHT1), and CD19 BUV661 (1D3).CD45 Pacific Blue (30-F11) was purchased from Biolegend.CD8a APC-eFluor 780 (53-6.70), B220 PE-Cyanine7 (RA3-6B2), and Live/Dead Fixable Violet Dead Cell Stain Kit werepurchased from Thermo Fisher Scientific. Flow cytometrywas performed with a BD LSRFortessa II cell analyzer (BDBiosciences) and data were analyzed with the use of FlowJosoftware (Treestar).

Intraepithelial lymphocytes expressing NKG2D wereidentified as TCRabþ CD8þ NKG2Dþ by means of flowcytometry. The quantification of NKG2D-expressing CD8þ

IELs per 100 intestinal epithelial cells was determined aspreviously described.3

Anti-gliadin enzyme-linked immunosorbent assaySerum was harvested 30 days after mice received the

first gliadin feeding. High-binding 96-well plates enzyme-linked immunosorbent assay (Nunc; Thermo-Scientific)were coated with 50 mL of 100 mg/ml of pepsin-trypsin–digested gliadin in 100 mmol/L Na2HPO4 overnight at 4�C.Plates were washed 3 times with phosphate-buffered salinesolution containing 0.05% Tween-20 (PBS-T) and blockedwith 200 mL of 2% bovine serum albumin in PBS-T for 2hours at room temperature. Serum was assessed in dupli-cate and at 2 dilutions, typically 1:50 and 1:200. Sera wereincubated overnight at 4�C, and plates were washed 3 timeswith PBS-T. Anti–mouse Ig–horseradish peroxidase (HRP;Southern Biotech) in blocking buffer was added to theplates and incubated for 1 hour at room temperature. The

June 2021 B Cells and Celiac Disease 2610.e1

Page 5: B Lymphocytes Contribute to Celiac Disease Pathogenesis

plates were washed 5 times with PBS-T. Fifty mL HRPsubstrate TMB (Thermo-Scientific) was added and the re-action stopped by the addition of 50 mL 2N H2SO4 (FlukaAnalytical). Absorbance was read at 450 nm on a ThermoScientific Multiskan Go microplate reader. Levels of anti-gliadin IgG and IgA were expressed in optical densityvalues.

RNA extraction, cDNA synthesis, andquantitative real-time PCR

Total RNA isolation was performed on epithelial cellswith the use of the RNeasy Mini Kit (Qiagen). RNA con-centration and quality were determined by means of ul-traviolet spectrophotometry (Epoch MicroplateSpectrophotometer; BioTek). cDNA synthesis was

performed with the use of qScript cDNA SuperMix (Quan-taBio) according to the manufacturer’s instructions.Expression analysis for murine gzm, prf1, rae1, and qa1 wasperformed with the use of TaqMan gene expression assaysand normalized to gapdh (Thermo Fisher Scientific). Rela-tive gene expression levels were determined using thedeltadelta Ct method to calculate the relative changes ingene expression relative to sham-fed mice.

Statistical analysisTests were performed as indicated in the figure legends

with the use of GraphPad Prism. Data are presented asmean ± SEM. The statistical tests used and P values areindicated in each figure or figure legend. P values <.05 wereconsidered to be statistically significant.

2610.e2 Lejeune et al Gastroenterology Vol. 160, No. 7

Page 6: B Lymphocytes Contribute to Celiac Disease Pathogenesis

0

-103

103

104

105

0

-103

103

104

105

A

ShamGluten + isotypeGluten + anti-CD20

% C

D19

+ B c

ells

Peyer’s Patches

glutenanti-CD20

glutenisotype

sham

Peyer’s Patches

Blood

MesentericLymph Nodes

Spleen

26.9 21.6 0.12

% C

D19

+ B c

ells

Blood

P = .0004ns

% C

D19

+ B c

ells

Spleen

P < .0001ns

% C

D19

+ B c

ells

Mesenteric Lymph Nodes

P = .023ns

P < .0001ns

D

G

44.1 40.6 0.46

36.3 15.6 0.22

s

29.6 34.7 2.24

% B

220- I

gA+

plas

ma

cells

E

Ant

i-glia

din

IgG

(O

.D. 45

0nm)

sham anti-CD20isotype

gluten

P = .007P = .0017

sham anti-CD20isotype

gluten

Ant

i-glia

din

IgA

(O

.D. 45

0nm)

P = .0016P = .056

F

CD19

CD

45Untreated d14 d28 d37

% C

D19

+ B c

ells

am

ong

CD

45+ c

ells

anti-CD20

ShamGluten + isotypeGluten + anti-CD20

C

*** *** ***

DQ8-Dd-villin-IL-15tg miceborn and raised on a gluten free diet

anti-CD20 i.pday -7

day 0

Analysis of celiac disease features

gluten

day 7 day 21 day 30

anti-CD20 i.p anti-CD20 i.p

B

ShamGluten + isotypeGluten + anti-CD20

0

10

20

30

40

50

0

10

20

30

40

0

20

40

60

0

10

20

30

40

0

20

40

60

0

10

20

30

40

50

0

0.5

1

1.5

0

0.1

0.2

0.3

0.4

0.5

Supplementary Figure 1. Anti-CD20 treatment eliminates B cells and anti-gluten antibodies while preserving mucosal plasmacells. (A) Experimental scheme. (B) Frequency of blood CD19þ B220þ B lymphocytes, demonstrating the efficacy of B-celldepletion. ***P < .001. (C) Representative dot-plots showing depletion efficacy in the blood, spleen, mesenteric lymph nodes,and Peyer’s patches. (D) Frequency of CD19þ B lymphocytes in the blood, spleen, mesenteric lymph nodes, and Peyer’spatches. (E, F) Serum anti-gliadin IgG (E) and IgA (F) levels were measured by means of enzyme-linked immunosorbent assay30 days after gluten feeding. (G) Percentage of B220� IgAþ plasma cells among CD45þ CD3� cells found in the lamina propriaof the different groups of mice. Results are representative of 3 (C–F) or 2 (G) independent experiments pooled together andshown as mean ± SEM. Analysis of variance/Tukey multiple comparison.

June 2021 B Cells and Celiac Disease 2610.e3

Page 7: B Lymphocytes Contribute to Celiac Disease Pathogenesis

P = .0118

B

C D

sham

glute

nsh

amglu

ten

Vill

ous/

cryp

t rat

io

P = .0331

P = .0119

ns

Fol

d ch

ange

Prf

1 ex

pres

sion

rela

tive

to s

ham

-fed

mic

e

DQ8-Dd-villin-IL-15tgDQ8-Dd-villin-IL-15tg muMT

Fol

d ch

ange

Gzm

B e

xpre

ssio

nre

l ativ

et o

sh a

m-f

ed m

iceP = .0042

0

1

2

3

4

Fol

d ch

ange

in R

ae1

expr

essi

onre

l ativ

eto

sha

m-f

ed m

ice

isotype anti-CD200

1

2

3

Fol

d ch

ange

in Q

a-1

expr

essi

onre

l at iv

et o

sha

m-f

ed m

ice

isotype anti-CD20

ns

ns

E F

A

DQ8-Dd-villin-IL-15tgDQ8-Dd-villin-IL-15tg muMT

gluten

sham

DQ8-Dd-villin-IL-15tg DQ8-Dd-villin-IL-15tg muMT

100

100

200

100

0

1

2

3

4

0

1

2

3

4

5

0

1

2

3

4

Supplementary Figure 2.Gluten-fed DQ8-Dd-villin-IL-15tg-muMT mice have decreased levels of cytotoxic molecules andintestinal tissue destruction. (A–D) DQ8-Dd-villin-IL-15tg mice and DQ8-Dd-villin-IL-15tg-muMT were maintained on a gluten-free diet (sham), or fed with gluten every other day for 30 days (gluten). (A) Hematoxylin-stained ileum sections. (B) Villousheight-to-crypt depth ratio measured from ileum sections of sham and gluten-fed DQ8-Dd-villin-IL-15tg and DQ8-Dd-villin-IL-15tg-muMT mice. Four independent experiments are presented as mean ± SEM. Analysis of variance/Tukey multiple com-parison. (C) Expression of perforin (prf1) in the epithelial compartment was measured by means of quantitative polymerasechain reaction (qPCR). Relative expression levels in gluten-fed mice were normalized to the expression levels observed insham-fed mice. (D) The expression of granzyme B (gzmb) was measured as in (C). (E, F) DQ8-Dd-villin-IL-15tg mice weremaintained on a gluten-free diet (sham), or fed with gluten every other day for 30 days and treated with 250 mg anti-CD20antibody (gluten þ anti-CD20) or its isotype control (gluten þ isotype) every 2 weeks: (E) expression of retinoic acid earlyinducible gene 1 (rae1) was measured as in (C); (F) expression of major histocompatibility class Ib molecule qa-1 wasmeasured as in (C). qPCR results are representative of 3 independent experiments, shown as mean ± SEM. Unpaired Studentt test.

2610.e4 Lejeune et al Gastroenterology Vol. 160, No. 7