• No results found

University of Groningen B cells in ANCA-associated vasculitides von Borstel, Anouk

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen B cells in ANCA-associated vasculitides von Borstel, Anouk"

Copied!
29
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

B cells in ANCA-associated vasculitides

von Borstel, Anouk

DOI:

10.33612/diss.93537940

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

von Borstel, A. (2019). B cells in ANCA-associated vasculitides: from pathogenic players to biomarkers. University of Groningen. https://doi.org/10.33612/diss.93537940

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Tyrosine Kinase Activity in

Transitional and Naive B cells

of Patients with Granulomatosis

with Polyangiitis

4

aDepartment of Internal Medicine, Division of Nephrology,

University Medical Center Groningen, University of

Groningen, Groningen, the Netherlands bDepartment of

Rheumatology and Clinical Immunology, University Medical Center Groningen, University of Groningen, Groningen,

the Netherlands cDepartment of Pathology and Medical

Biology, University Medical Center Groningen, University

of Groningen, Groningen, the Netherlands dDepartment of

Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands

*Authors contributed equally Published:

Rheumatology 2019, kez 205

Anouk von Borstela, Wayel H. Abdulahadb,c,

Jan Stephan Sandersa, Jasper Ripd, Stefan F.H. Neysd,

Rudi W. Hendriksd, Coen A. Stegemana, Peter Heeringac,

Abraham Rutgersb* and Odilia B.J. Cornethd*

(3)

Abstract

Objectives To determine Bruton’s tyrosine kinase (BTK) protein and phosphorylation

levels in B cell subsets of granulomatosis with polyangiitis (GPA) patients and to investigate the effect of BTK blockade on in vitro B cell cytokine production, subset distribution and (auto)antibody production.

Methods BTK protein and phosphorylation levels were determined by flow cytometry

in peripheral blood B cells of 29 untreated GPA patients [9 active and 20 remission GPA patients (10 ANCA- and 10 ANCA+)], 9 age- and sex-matched healthy controls (HCs) and 9 untreated active RA patients. The effect of BTK blockade on in vitro B cell cytokine production, subset distribution and (auto)antibody production was determined in the same donors in peripheral blood mononuclear cell cultures.

Results BTK protein levels were significantly increased in transitional and naïve B cells

of active GPA and RA patients compared with remission GPA patients and HCs. Both B cell subsets of active patients were more sensitive to B cell receptor stimulation, as BTK and phospholipase Cγ2 phosphorylation were increased in these patients. In vitro BTK blockade had profound effects on B cell cytokine production, plasma cell formation and (auto)antibody production in both GPA patients and HCs. Interestingly, the effect of BTK blockade was less pronounced in active GPA patients, possibly due to increased activation of B cells.

Conclusions We show that BTK protein and phosphorylation levels are most profoundly

increased in newly emerging B cells of active GPA patients compared with remission patients. BTK blockade greatly inhibits in vitro B cell effector functions in GPA patients and HCs. These promising data identify BTK as an interesting novel therapeutic target in the treatment of GPA.

Key Messages

• Bruton’s tyrosine kinase levels are increased in transitional and naïve B cells of active granulomatosis with polyangiitis patients

• Bruton’s tyrosine kinase blockade inhibits in vitro B cell effector functions in granulomatosis with polyangiitis patients and healthy controls

• Bruton’s tyrosine kinase might be an interesting novel therapeutic target in the treatment of granulomatosis with polyangiitis

(4)

4

Introduction

Granulomatosis with polyangiitis (GPA) is an autoimmune disease that affects small-

to medium-sized blood vessels69 and is characterized by the presence of ANCA,

predominantly directed against PR3. Although progress has been made in the understanding of the disease mechanisms, GPA and its treatment are still associated

with high disease burden and mortality58,69. Even with appropriate treatment, ~50% of

patients experience a disease relapse in <4 years, often resulting in irreversible loss of

organ function and necessitating toxic immunosuppressive therapy160. As precursors of

autoantibody-producing cells, B cells are crucially involved in the GPA pathogenesis. In

addition, B cells can also present antigen161 and produce pro- and anti-inflammatory

cytokines that have been linked to the GPA pathogenesis40,41,149. GPA patients display

shifts in circulating B cell subsets during active disease and remission46. This is

characterized by increased naïve and decreased memory B cell frequencies compared

with healthy controls (HCs)46. Additionally, increased circulating plasmablast frequencies

during remission were associated with decreased relapse-free survival162. Together,

this evidence suggests that B cells not only function as precursors of autoantibody-producing cells but also as important effector cells in GPA pathogenesis. Therefore, modulation of abnormal B cell function might be beneficial in GPA.

It has been demonstrated that aberrancies in Bruton’s tyrosine kinase (BTK) levels may contribute to abnormalities in B cell activity or subset distribution. BTK is a critical mediator of B cell receptor (BCR) signaling and has an important role in B cell

growth and differentiation163. Upon antigen binding to the BCR, phosphorylated BTK

(pBTK) initiates a downstream signaling cascade that eventually leads to activation of extracellular signal-related kinase (ERK), protein kinase B (also known as AKT) and the transcription factor nuclear factor (NF)-κB, promoting B cell survival, proliferation and

differentiation163.

Mounting evidence indicates that BTK is an important factor in autoimmune disease pathogenesis, as BTK overexpression in murine B cells is sufficient to induce a

spontaneous autoimmune phenotype164, and BTK inhibition is an effective treatment in

many murine autoimmune models165. Aberrant BTK activity was also demonstrated in

human autoimmune diseases such as primary SS and RA50,166. In untreated SS patients,

BTK levels were increased in peripheral B cell subsets, including naïve B cells, compared

with HCs50. These levels correlated with BTK phosphorylation, serum autoantibodies,

circulating T follicular helper (Tfh) cells and infiltrating T cell numbers in salivary glands. Similarly, in ACPA+ RA patients, BTK protein levels were increased compared with ACPA- RA patients and HCs, and correlated with inducible T cell co-stimulator (ICOS) expression

on Tfh cells50. As B cells and ANCA play an important role in the GPA pathogenesis, it is

(5)

In this study, we aimed to investigate BTK protein levels and phosphorylation in B cell subsets of active and remission GPA patients. Additionally, phosphorylation levels of other proteins up- and downstream of BTK were studied. Also, the effect of BTK blockade on in vitro B cell cytokine production, plasma cell formation and (auto) antibody production was investigated.

Materials and Methods

Study Population

We included nine active and 20 remission GPA patients (10 ANCA- GPA-patients; Table 1). Also, nine age-matched HCs (44.4% male, median age 56.8 years; range 22-74 years) and nine untreated ACPA+ RA patients, fulfilling ACR/EULAR 2010 classification

criteria for RA167, from the Rotterdam Early Arthritis Cohort168 were included as active

disease controls. GPA was diagnosed according to Chapel Hill Consensus Conference

definitions and ACR classification criteria2,150. Active and remission GPA was diagnosed

by clinical and laboratory evaluation. Active GPA had to result in start/increase of immunosuppressive medication. Remission was defined as absence of any clinical/ laboratory signs of active disease and a BVAS of zero. None of the patients received immunosuppressive treatment. ANCA titres of ≤1:20 were considered negative. Two patients received rituximab >3 years before sampling. This study was approved by the medical ethics committee of the University Medical Center Groningen (METc no. 2012/151), informed consent was obtained from all patients and the study complies with the Declaration of Helsinki.

Table 1. Patient characteristics.

Active

GPA-patients Remission GPA patients Active ACPA+ RA patients HCs

Subjects, n (% male) 9 (55.6) 20 (40) 9 (11.1) 9 (44.4)

Age years, mean (range) 57.2 (21.5-78.3) 59.5 (26-77.8) 56.3 (35.1-75.8) 56.8 (22-74)

Autoantibody titer, median

(range) 1:160 (1:40-1:640) 1:80 (0-1:640) (U/ml) 96 (14-1802)

-Creat µmol/mL, median

(range) 82 (62-151) 93.5 (57-409) NA

-CRP mg/mL, median

(range) 43.5 (9-268) 3.4 (0.5-38) 5 (1-17)

-BVAS, median (range) 15 (8-21) 0 DAS28: 4.3 (2.0-6.8)

(6)

4

Peripheral Blood Mononuclear Cell Culture Assays

Peripheral blood mononuclear cells (PBMCs) were isolated, frozen and thawed as

described before169. For all cultures, 1*106 PBMCs/ml were cultured with or without 1000

nM BTK blocker (BMS-986142, kindly provided by Bristol-Myers Squibb, New York, NY,

USA170). The BTK blocker was pre-incubated for 1 h.

To investigate the effect of BTK inhibition on B cell cytokine production, PBMCs were

cultured for 3 days with 1000 nM anti-IgM Fab2 fragments (Jackson ImmunoResearch,

West Grove, PA, USA) and restimulated the last 4.5 h with 50 ng/ml phorbol myristate acetate and 2 mM calcium ionophore in the presence of 10 mg/mL brefeldin A (Sigma-Aldrich, St. Louis, MO, USA).

To assess the effect of BTK inhibition on plasma cell formation, PBMCs were cultured for

7 days with 1000 nM anti-IgM Fab2 fragments (Jackson ImmunoResearch) and 100 ng/

mL B cell activating factor (BAFF; PeproTech Inc., Rocky Hill, NJ, USA).

To determine the effect of the BTK blocker on (auto)antibody production, PBMCs were

cultured for 12 days with 1000 nM anti-IgM Fab2 fragments (Jackson ImmunoResearch),

100 ng/mL BAFF (PeproTech Inc.) and 100 ng/mL IL-21 (ImmunoTools, Friesoythe,

Germany)23.

BTK Protein levels and T cell Subsets by Flow Cytometry

BTK protein levels in B- and T cell subset frequencies were assessed ex vivo by flow

cytometry as described previously50. For antibodies used, see Supplementary Table

1. For gating strategy, see Supplementary Figures 1 and 2. The BTK protein staining was validated using isotype controls and FMOs (fluorescence minus one). The levels of background staining in B and T cells were similar. To account for background in the BTK staining, ratios were calculated by dividing the mean fluorescence intensity (MFI) of BTK protein in B cell subsets by the MFI in T cells within each sample, as T cells do not express BTK (Supplementary Figure 1). Data were analyzed using FlowJo software (Treestar, Ashland, OR, USA).

Phosphorylation of Signaling Molecules by Phosphoflow

To determine ex vivo phosphorylation of intracellular signaling molecules, 4*105

PBMCs/sample were kept at 4⁰C and directly fixed with FoxP3-staining kit Fix/Perm solution (eBioscience, San Diego, CA, USA). Samples were stained for 30 min at 4⁰C with a staining cocktail to identify B cell subsets, followed by the antibody targeting the phosphorylated signaling molecule (30 min; room temperature), diluted in FoxP3-staining kit Perm/Wash buffer. To determine BCR sensitivity, samples were cultured in Rosswell Park Memorial Institute 1640 medium with 2% fetal calf serum at 37⁰C for 5 minutes in the presence/absence of 20 µg/mL anti-IgM (SouthernBiotech, Birmingham,

(7)

AL, USA) prior to fixation (Supplementary Table 1 for antibodies). Data was analyzed by FlowJo software.

Intracellular Cytokines by Flow Cytometry

After restimulation, cultured PBMCs were washed and stained as described previously169

(Supplementary Table 1 for antibodies). Data were analyzed using Kaluza v1.7 (Beckman Coulter, Brea, CA, USA). FACS plots from unstimulated samples were used to define gates (gating example in Supplementary Figure 5). One HC with <20% viable PBMCs was excluded.

In vitro Plasma Cell Formation by Flow Cytometry

After 7-day culture, PBMCs were washed twice and stained for 15 min with antibodies

(Supplementary Table 1). Data were analyzed in Kaluza v1.7. CD27hiCD38hi plasma cell

frequencies were determined among CD19+CD22+ B cells (Supplementary Figure 6).

In vitro Total IgG and PR3-ANCA

Total IgG levels (ng/ml) and PR3-ANCA IgG levels (response units) were determined by ELISA and Phadia ImmunoCAP 250 analyzer, respectively, in supernatants of anti-IgM

Fab2/BAFF/IL-21 stimulated PBMCs after a 12-day culture, as described previously23.

Statistics

Data were analyzed using GraphPad Prism software (GraphPad Prism Inc., La Jolla, CA, USA). Statistical differences between groups were determined by one-way ANOVA with a Tukey’s multiple comparison test or Dunn’s test. Paired data were analyzed using the Wilcoxon signed-rank test. Correlations were analyzed by Spearman correlation test. P-values <0.05 were considered statistically significant.

Results

Increased BTK Protein Levels in Transitional and Naïve B cells from Active GPA Patients

The frequencies of various B cell subsets were quantified in PBMCs from GPA patients and HCs by flow cytometry (Supplementary Figure 3). Likely due to low sample numbers, no significant differences were found in B cell subset distribution between HCs and the patient groups, although plasmablasts were slightly increased in active GPA patients.

(8)

4

Next, we determined BTK protein levels in different B cell subsets. Compared with HCs,

BTK protein levels were increased in total B cells of active GPA patients, which were similar to levels in the active ACPA+ RA patient control group. BTK protein levels were not different in remission GPA patients, and no differences were found between ANCA+ and ANCA- remission patients (Figure 1A and data not shown). Interestingly, comparable

differences were observed in IgD+CD27- naïve B cells and CD38+CD27- transitional B cells

from active compared with remission GPA patients (Figures 1B and C). These differences were not present in switched or non-switched memory B cells or plasmablasts (Figures 1D-F), indicating that the difference between active and remission patients involves newly emerging B cells rather than antigen-experienced B cells.

Enhanced Response to BCR Stimulation in Newly Emerging B cells of Active GPA Patients

To link BTK protein levels to its activity, we measured pBTK at Y223165, as well as

phosphorylation of several up- and downstream signaling molecules in a smaller

group of patients. As previously shown for RA patients50, BTK protein levels in total B

cells correlated with ex vivo pBTK in HCs and active GPA patients (Figure 2A). pBTK was not different between HCs and GPA patients ex vivo (Figure 2B). Phosphorylation of the direct and specific BTK downstream target phospholipase C (PLC) γ2 (Y759), which

links BCR stimulation to Ca2+ signaling, appeared increased both in transitional and

naïve B cells from active and remission GPA patients, but the increase observed ex vivo only reached significance for the difference between HC and remission patients in the transitional B cell subpopulation (Figures 2C and D). In contrast, no differences were found in the phosphorylation of the BCR-associated upstream molecule MB1 (CD79a), or the downstream molecules ERK (MAPK pathway) and S6 (AKT pathway) in directly fixed transitional or naïve B cells between active patients and HCs, nor in phosphorylation in memory B cells (ex vivo, data not shown).

To further determine BCR sensitivity of different B cell subsets, PBMCs from patients

and HCs were stimulated in vitro with anti-IgM Fab2 fragments. All groups showed

upregulation of pBTK in transitional and naïve B cells upon stimulation (Figures 3A and B). However, B cell sensitivity, measured by increased pBTK upon anti-IgM stimulation, was significantly increased in these subsets from active compared with remission GPA patients and HCs (Figure 3C), while sensitivity of memory B cell subsets was not significantly different between the groups. Similar results were obtained for pPLCγ2 expression upon anti-IgM stimulation (Figures 3D-F). Although the differences between the groups did not reach significance for pPLCγ2, the expressions of pPLCγ2 and pBTK Y223 upon stimulation were significantly correlated (ρ=0.802; p=0.007; Supplementary Figure 4).

(9)

Von Borstel et. al. Figure 1

A T ce ll B ce ll HC B ce ll act BTK protein B C D E F to ta l B H C ac tiv e re m is si on R A 0 4 8 ** * ** * * BTK M FI ( rat io) * na iv e B H C ac tiv e re m is si on R A 0 4 8 io) rat FI ( M BTK ** ** * ** * * tra ns iti on al B H C ac tiv e re m is si on R A 0 4 8 ** * * ** ** ** BTK M FI ( rat io) Ig D + no n-sw itc he d H C ac tiv e re m is si on R A 0 4 8 io) rat FI ( M BTK ** * * * sw itc he d H C ac tiv e re m is si on R A 0 4 8 io) rat FI ( M BTK * * pl as m ab la st H C ac tiv e re m is si on R A 0 4 8 io) rat FI ( M BTK 0 1 0 2 1 0 3 1 0 4 1 0 5 Figur e 1. B TK pr ot ein le vels ar e incr eased in B c ells fr om ac tiv e GP A pa tien ts c ompar ed with HCs and r emission pa tien ts . A. B TK pr ot ein lev els in t otal per ipher al B c ells fr om HCs , ac tiv e GP A, r emission GP A and R A pa tien ts , analyz ed b y in tr ac ellular flo w c yt ometr y. D epic ted r atio is B TK MFI in B cells/B TK MFI in T c ells . H ist og ram sho w s an o ver la y of a r epr esen ta tiv e HC and ac tiv e GP A pa tien t. B TK pr ot ein lev els in B. naïv e B c ells , C . tr ansitional B c ells , D. IgD + non-swit ched memor y B c ells , E. swit ched memor y B c ells and F. plasmablasts . *p<0.05; **p<0.01; ***p<0.001. ac t: ac tiv e; B TK : Brut on ’s tyr osine k inase; GP A: g ranuloma

tosis with poly

ang

iitis; HCs: health

y c

on

tr

ols; MFI: mean fluor

esc

enc

e in

tensit

(10)

4

B TK -p B TK 2 3 4 5 0 40 00 80 00 B TK pr ot ei n (M FI ra tio ) pBT KY 223 (M FI) naive B H C ac tiv e re m is si on 0 40 00 80 00 pBT KY 223 (M FI) naive B H C ac tiv e re m is si on 0 20 0 40 0 60 0 pPL Cγ2 (M FI) transitional B H C ac tiv e re m is si on 0 50 00 10 00 0 15 00 0 pBT KY 223 (M FI) transitional B H C ac tiv e re m is si on 0 50 0 10 00 15 00 ** pPL Cγ2 (M FI) B TK -p P LC γ 2 3 4 5 0 20 0 40 0 60 0 B TK pr ot ei n (M FI ra tio ) pPL Cγ2 (M FI) 2 A B C D

Von Borstel et. al. Figure 2

pBTK Y223 pPLC γ2 HC active 0. 037 0. 762 ρ= p= 0. 083 0. 900 ρ= p= 0. 327 0. 405 ρ = p= 0. 658 0. 257 ρ= p= HC active Figur e 2. I ncr eased BCR ac tivit y in tr

ansitional and naïv

e B c ells of GP A pa tien ts ex viv o. A. C or rela tions bet w een B TK pr ot ein and ex viv o pB TK in total B cells of HCs (open cir cles) and ac tiv e GP A pa tien ts (closed cir cles). H ist og ram ov er la y sho w s ex viv o pB TK expr ession in total B cells . B. Ex viv o pB TK expr ession in tr ansitional and naïv e B c ells of HCs , ac tiv e and r emission GP A pa tien ts . C. C or rela tions bet w een BTK pr ot ein and pPL Cγ2 in t otal B c

ells of HCs (open cir

cles) and ac tiv e GP A pa tien ts (closed cir cles). H ist og ram o ver la y sho w s pPL Cγ2 expr ession in t otal B c ells . D. Ex viv o pPL Cγ2 expr ession in tr

ansitional and naïv

e B c ells of HCs , ac tiv e and r emission GP A pa tien ts . **p<0.01. B: B c ells; BCR: B c ell r ec ept or ; B TK : Brut on ’s tyr osine k inase; pB TK : phosphor yla ted B TK ; GP A: g ranuloma

tosis with poly

ang

iitis; HCs: health

y c

on

tr

ols; MFI: mean fl

uor esc enc e in tensit y; pPL Cγ2: phosphor yla ted phospholipase Cγ2.

(11)

tra ns iti on al B 0 50 00 10 00 0 15 00 0 HC ac tiv e re m is si on ** ** * ** * pBT KY 223 (M FI) na iv e B 0 40 00 80 00 HC ac tiv e re m is si on ** ** * ** * pBT KY 223 (M FI) pBTK Y223 A B C 0 1 0 2 1 0 3 1 0 4 1 0 5 0 1 0 2 1 0 3 1 0 4 1 0 5

unstim HC aIgM HC aIgM act pBTK Y223 unstim HC aIgM HC aIgM act

Von Borstel et. al. Figure 3

tra ns itio na l H C ac tiv e re m is si on 0 2 4 6 8 * ** * pBT K( rat io stim /un stim ) na ive H C ac tiv e re m is si on 0 1 2 3 4 5 * Ig D + no n-sw itc he d H C ac tiv e re m is si on 0 2 4 6 sw itc he d H C ac tiv e re m is si on 0 1 2 3 D E 0 20 00 40 00 pPL Cγ2 (M FI) 0 20 00 40 00 60 00 pPL Cγ2 (M FI) na iv e B tra ns iti on al B ** ** ** **

unstim HC aIgM HC aIgM act unstim HC aIgM HC aIgM act

pPLC γ2 pPLC γ2 F na ive B H C ac tiv e re m is si on 0 5 10 15 tra ns itio na lB H C ac tiv e re m is si on 0 5 10

15) stim stim/un atio γ2(r LC pP

Ig D + no n-sw itc he d H C ac tiv e re m is si on 0 5 10 15 sw itc he d H C ac tiv e re m is si on 0 2 4 HC ac tiv e re m is si on HC ac tiv e re m is si on 1 0 3 -1 0 3 0 1 0 4 1 0 5 1 0 3 -1 0 3 0 1 0 4 1 0 5 Figur e 3. I ncr eased in vitro BCR r esp onsiv eness in tr

ansitional and naïv

e B c ells of ac tiv e GP A pa tien ts . pB TK expr ession on A. tr ansitional B c ells and B. naïv e B c ells in vitr o, unstimula ted (lef

t) and upon stimula

tion with an ti-IgM F ab2 -fr ag men ts f or 5 min (aIgM; r igh t). H ist og ram sho w s o ver la ys of pB TK MFI of a r epr esen ta tiv e HC and ac tiv e GP A pa tien t. C. R atio of pB TK MFI on diff er en t B c ell subsets of an ti-IgM stimula ted sample/unstimula ted sample . pPL Cγ2 expr ession on D. tr ansitional B c ells and E. naïv e B c ells in vitr o, unstimula ted (lef

t) and upon stimula

tion with an ti-IgM F ab2 -fr ag men ts f or 5 min (r igh t). H ist og ram sho w s ov er la ys of pPL Cγ2 MFI of a repr esen ta tiv e HC and ac tiv e GP A pa tien t. F. R atio of pPL Cγ2 MFI on diff er en t B cell subsets of an ti-IgM stimula ted sample/unstimula ted sample . *p<0.05; **p<0.01; ***p<0.001. ac t: ac tiv e; aIgM: an ti-IgM; B: B c ells; BCR: B c ell r ec ept or ; GP A: g ranuloma

tosis with poly

ang iitis; HC: health y c on tr

ol; MFI: mean fl

uor esc enc e in tensit y; pB TK : phosphor yla ted Brut on ’s t yr osine k inase; pPL Cγ2: phosphor yla

ted phospholipase Cγ2; stim:

stimula

ted; unstim: unstimula

ted

(12)

4

These data indicate that increased activity of BTK in newly emerging B cells is correlated

with increased BTK protein levels in active GPA patients, and that this may also affect its downstream target pPLCγ2. These data suggest that sensitivity of the BCR signaling pathway may be increased in transitional and naïve B cells of active GPA patients.

BTK Protein Levels Correlate with B cell Activity and Decreased Circulating

Pro-inflammatory CD4+ T cells in GPA Patients

The increased BCR signaling in B cells of active GPA patients suggested increased B cell activation. Surface expression of co-stimulation molecule CD86 on total B cells was moderately, but not significantly, increased in active GPA patients (Figure 4A). BTK protein levels in GPA patients, but not in HCs, correlated with B cell CD86 expression, indicating that high BTK levels associate with increased B cell activation in GPA patients (Figure 4B).

We previously showed that BTK protein levels were associated with increased pro-inflammatory CD4+ T cell numbers in autoimmune patients50. Therefore, the proportions

and activation status of different T cell subsets in PBMCs from GPA patients and HCs were quantified. Memory CD4+ T cell proportions were increased in remission but

not in active GPA patients compared with HCs (Supplementary Figure 5A). Within the memory CD4+ T cell pool, pro-inflammatory T cell subsets implicated in autoimmunity

were generally decreased in active GPA patients (Supplementary Figure 5B), whereas anti-inflammatory subsets and non-autoimmune associated Th2 cells were increased (Supplementary Figure 5C). Interestingly, BTK protein levels were inversely correlated with pro-inflammatory Th1 cells , and a trend was seen for Th17.1, in GPA patients (Figure 4C). Conversely, BTK levels were positively correlated with the frequencies of Th2 cells, and a trend was seen for regulatory Tfr-like cells (Figure 4D). ICOS expression on Tfh-like cells in GPA patients was not different from HCs (Supplementary Figure 5D). These data show that BTK protein levels in B cells correlate with B cell activity and decreased circulating pro-inflammatory CD4+ T cells in GPA patients.

BTK Inhibition Decreases in vitro IFNγ+, IL-10+ and IL-6+ B cell Frequencies,

Plasma cell Formation and (auto)Antibody Production

To assess the functional importance of BTK in B cells of GPA patients, PBMCs were stimulated with anti-IgM in the absence or presence of a BTK inhibitor, and in vitro B cell cytokine production was determined. Upon culture without BTK inhibitor, no significant differences in cytokine-positive B cell frequencies were found between groups. In vitro BTK inhibition significantly decreased IFNγ+, IL-10+ and IL-6+ B cell frequencies in all

groups (gating strategy is shown in Supplementary Figure 6). However, the TNFα+ B

(13)

B TK - Th 1 2 3 4 5 0 20 40 60 BT K pr ot ei n (M FI ra tio ) Th1 (% of C D4+ m em ory T) 0. 00 1 -0 .5 98 p= ρ= B TK - Th 17 .1 2 3 4 5 0 5 10 15 BT K pr ot ei n (M FI ra tio ) Th1 7.1 (% of C D4+ m em ory T) 0. 08 5 -0 .3 38 p= ρ= B TK - Th 17 2 3 4 5 0 5 10 15 BT K pr ot ei n (M FI ra tio ) Th1 7 (% of C D4+ m em ory T) 0. 24 3 -0 .2 33 p= ρ= B TK - Tf h-lik e 2 3 4 5 0 12 24 BT K pr ot ei n (M FI ra tio ) Tfh -lik e (% of C D4+ m em ory T) 0. 82 2 -0 .0 46 p= ρ= B TK - Th 2 2 3 4 5 0 20 40 BT K pr ot ei n (M FI ra tio ) Th2 (% of C D4+ m em ory T) 0. 00 03 0. 64 3 p= ρ= B TK - Tf r-l ik e 2 3 4 5 0 4 8 BT K pr ot ei n (M FI ra tio ) Tfr-lik e (% of C D4+ m em ory T) 0. 07 0 0. 35 4 p= ρ= B TK - Tr eg 2 3 4 5 0 12 24 BT K pr ot ei n (M FI ra tio ) Tre g (% of C D4+ m em ory T) 0. 49 3 0. 13 8 p= ρ= to ta l B H C ac tiv e re m is si on 0 50 0 10 00 15 00 CD 86 (M FI) 0 1 0 2 1 0 3 1 0 4 1 0 5

B cell HC B cell act

CD86 A B C D H C 2 3 4 5 0 40 0 80 0 12 00 0.843 -0.083 ρ= p= BT K pr ot ei n (M FI ra tio ) CD 86 (M FI) ac tiv e 2 3 4 5 0 40 0 80 0 12 00 0. 017 0. 783 ρ= p= BT K pr ot ei n (M FI ra tio ) re m is si on 2 3 4 5 0 40 0 80 0 12 00 0. 043 ρ= p= 0. 548 BT K pr ot ei n (M FI ra tio ) Figur e 4. B TK pr ot ein le vels of B c ells of GP A pa tien ts c orr ela te with B c ell ac tiv

ation and decr

eased pr

o-inflamma

tor

y

T c

ell subsets in the

cir cula tion. A. CD86 expr ession on t otal per ipher al B c ells of HCs , ac tiv e and r emission GP A pa tien ts . H ist og ram sho w s an o ver la y of a r epr esen ta tiv e HC and ac tiv e GP A pa tien t. C or rela tion of B. B TK pr ot ein with CD86 expr ession on total B cells of pa tien ts and HCs , C. pr opor tions of cir cula ting pr o-inflamma tor y or D. regula tor y CD4 + T c ell subsets in ac tiv e GP A pa tien ts (cir cles) and r emission GP A pa tien ts (squar es). ac t: ac tiv e; B: B c ells; BCR: B c ell rec ept or ; B TK : Brut on ’s t yr osine k inase; GP A: g ranuloma

tosis with poly

ang

iitis; HC: health

y c

on

tr

ol; MFI: mean fluor

esc enc e in tensit y; T: T c ells; Tfh-like: T f ollicular helper -like c ell; Tfr -like: T f ollicular helper r egula tor y c ell .

(14)

4

BTK inhibition between groups showed decreased IFNγ+ B cell frequencies in remission

compared with active GPA patients (Figure 5A).

The sensitivity of IL-10+ B cells to BTK inhibition was decreased in active GPA patients

compared with HCs, as the ratio of anti-IgM/anti-IgM + BTK inhibitor was decreased in active patients (Supplementary Figure 8A). Together, cytokine-positive B cell frequencies upon BTK inhibition are decreased, and the effect is possibly lower in active GPA patients.

To determine whether in vitro plasma cell formation is affected by BTK inhibition, PBMCs were stimulated with anti-IgM and BAFF in the absence or presence of a BTK inhibitor. Upon culture without BTK inhibitor, no differences were found in memory B cell formation between groups; however, plasma cell formation was increased in active compared with remission GPA patients (for gating strategy, see Supplementary Figure 7). Upon in vitro BTK inhibition, plasma cell formation was decreased in remission GPA patients, but could not be decreased in active GPA patients and HCs (Figure 5B). Also, memory B cell formation was decreased upon BTK inhibition in remission GPA patients and HCs, but not in active GPA patients. Interestingly, plasma cell frequencies were increased upon BTK inhibition in active compared with remission GPA patients (Figure 5B). Importantly, transitional B cell frequencies were not different between groups upon BTK inhibition (Supplementary Figure 8B). Naïve B cell frequencies in the absence and presence of BTK inhibition were increased in remission compared with active GPA patients and HCs (Supplementary Figure 8B). This indicates that the efficacy of BTK inhibition is lower in active GPA patients.

To determine the effect of BTK inhibition on (auto)antibody secretion, PBMCs were cultured with anti-IgM, BAFF and IL-21 in the absence or presence of the BTK inhibitor for 12 days, and PR3-ANCA and total IgG concentrations were determined in supernatants. In cultures of eight GPA patients, including 5 active patients, PR3-ANCA were secreted and BTK inhibition appeared to decrease the PR3-ANCA levels (Figure 5C). However, no significant differences were found, possibly due to large variation between samples. Total in vitro IgG production was decreased in HCs and remission patients upon BTK inhibition, whereas a trend towards a decrease was seen in active GPA patients (Figure 5C).

Together, these data show that BTK inhibition may decrease in vitro B cell differentiation towards memory B cells and antibody-producing plasma cells in HCs and remission GPA patients, but seemed less effective in active GPA patients. Furthermore, BTK inhibition diminished in vitro (auto)antibody production in remission and active GPA patients.

(15)

A

B

C

Active GPA ANCA- GPA ANCA+ GPA 0 3 6 9 IFNγ+ B cells (%) HC Active Remission *** * * * % of C D 19 +C D 22 + c el ls 0 10 20 30 40 50 TNFα + B cells (%) HC Active Remission *** anti-IgManti-IgM 0.0 2.5 5.0 7.5 10.0 12.5 21.0 22.0 IL-10 B cells (%) HC Active Remission **** * ** 0 10 20 30 IL-6+ B cells (%) HC Active Remission **** ** ** anti-IgM anti-IgM 0 10 20 30 40 50 Memory B cells (%) HC Active Remission ** * 0 5 10 15 20 25 40 60 Plasma cells (%) HC Active Remission anti-IgM + BAFF anti-IgM + BAFF *** **** ** 0 1000 2000 3500 4500 Total IgG HC Active Remission

anti-IgM + BAFF + IL-21 + BTK Blocker ** **** Ig G (n g/ m

L) anti-IgM + BAFF + IL-21

0.2 0.3 0.4 0.5 0.6 10 20 30 3*SD HCs PR3-ANCA RU /m L % of C D 19 +C D 22 + c el ls % of C D 19 +C D 22 + c el ls % of C D 19 +C D 22 + c el ls % of C D 19 +C D 22 + c el ls % of C D 19 +C D 22 + c el ls anti-IgM + BTK Blocker BAFF IL-21 anti-IgM BAFF IL-21 + BTK Blocker + BTK Blocker + BTK Blocker +

Figure 5. BTK blockade inhibits in vitro B cell cytokines production, plasma cell formation and

(auto)antibody production. A. The frequencies of IFNγ+ (top left), TNFα+ (top right), IL-10+ (bottom

left) and IL-6+ (bottom right) B cells are given in samples stimulated with anti-IgM only (open circle)

and samples stimulated with anti-IgM + BTK blocker (open squares) for HCs, active and remission GPA patients. B. For HCs, active and remission GPA patients, the frequencies of memory B cells (left) and

plasma cells (right) within CD19+CD22+ B cells are depicted. Open circles represent samples stimulated

with anti-IgM/BAFF, open squares represent samples stimulated with anti-IgM/BAFF in the presence of the BTK blocker. C. PR3-ANCA levels (left) for anti-IgM/BAFF/IL-21 and anti-IgM/BAFF/IL-21 + BTK blocker stimulated samples for active (open circles), ANCA- (open squares), and ANCA+ (open diamonds) remission GPA patients, and total IgG concentrations (right) in anti-IgM/BAFF/IL-21 (open circles) and anti-IgM/BAFF/IL-21 + BTK blocker (open squares) stimulated samples for HCs, active and remission GPA patients. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. BAFF: B cell activating factor; BTK: Bruton’s tyrosine kinase; GPA: granulomatosis with polyangiitis; HCs: healthy controls; RU: response units.

(16)

4

Discussion

GPA is characterized by a high morbidity and is fatal if left untreated. Although disease remission can be established by immunosuppressive therapy, these treatments are not

directed specifically at pathogenic processes and can lead to severe side-effects171. New

therapies that specifically target pathogenic pathways and cells involved in GPA are therefore needed. Here we assessed BTK expression and the effect of BTK blockade on B cell function as a novel treatment target in GPA.

Although GPA patients commonly receive medication at the time of diagnosis, we were able to include nine untreated active GPA patients. Despite this low number, we found clear differences in the BTK protein levels compared with remission GPA patients and HCs. These protein levels correlated with BCR sensitivity and circulating pro-inflammatory T cells. Furthermore, clear effects of in vitro BTK inhibition on B cell effector functions were observed in remission GPA patients and HCs, but were less pronounced in active GPA patients, showing that with the current sample size, active and remission patients can be distinguished.

BTK levels were increased in some memory B cell subsets in remission patients, but - most importantly - we observed a striking difference between active disease and remission in newly formed naïve and transitional B cells. Hereby only active GPA patients show increased BTK protein levels compared with HCs. Transitional and naïve B cells from these patients show increased BCR signaling ex vivo, as pPLCγ2 levels were increased, as well as increased BCR sensitivity in vitro, inferred from the enhanced pBTK levels upon BCR stimulation. These observations parallel our previous findings of

increased BTK protein levels in naïve B cells of untreated active RA and SS patients50. In

these patients, BTK levels correlated with autoantibodies. We did not find a correlation with ANCA titers, possibly due to low sample numbers. Increased BTK protein levels in transitional and naïve B cells do correlate with a more activated B cell phenotype in GPA

patients, and a decrease in pro-inflammatory CD4+ T cell proportions in the circulation.

We therefore propose that pathogenic B-T cell interaction occurs already at an early stage of B cell differentiation, and that newly emerging B cells may be actively involved in autoimmune disease pathology.

We previously demonstrated correlations between BTK levels and T cell activity in RA and SS patients and showed that inhibition of B-T cell or dendritic cell-T cell co-stimulation by

abatacept was sufficient to reduce BTK protein in naïve B cells to HC levels50. In SS, B cell

BTK protein levels correlated with infiltrating T cell numbers in parotid glands. Although target organs in GPA patients were not studied here, it is conceivable that numbers of activated T cells involved in disease pathogenesis are higher in target organs of active GPA patients than in remission patients. Normalized BTK expression in remission GPA patients and abatacept-treated SS patients both indicate that BTK is not intrinsically dysregulated in autoimmune patients, but that a pro-inflammatory micro-environment

(17)

including activated T cells is responsible for increased BTK expression and BCR activity. B cells play an essential role in GPA pathogenesis as ANCA-producing cells. As expected, BTK blockade inhibited in vitro plasma cell formation, PR3-ANCA and total IgG production upon BCR stimulation. GPA patients show increased B cell activation and activated effector B cells may be an important source of pro-inflammatory cytokines, although

the contribution of these cytokines to GPA pathogenesis is unclear169. Importantly, in

vitro BTK blockade resulted in inhibition of B cell cytokine production (IFNγ, IL-10, IL-6) in active and remission GPA patients and HCs. Thus, BTK inhibition affects several B cell effector functions in GPA B cells.

We also show a clear effect of in vitro BTK blockade on plasma cell formation in remission patients, accompanied by a decrease in memory B cells, whereas no such effect was seen in active GPA patients. Altogether, in our in vitro cultures of B cells from active GPA patients, the proportions of differentiated plasma cells were increased compared with remission patients and HCs. Although in vitro effects on BCR signaling in Ramos

cells were shown with lower doses of the BTK inhibitor170, it is possible that the dose

used in our study was insufficient to inhibit in vivo activated B cells of active patients, or that activated B cells in these patients no longer depend on BTK-mediated survival signals. In vitro BTK inhibition significantly reduced IgG production in remission GPA patients and HCs, but not in active GPA patients. This suggests that B cells in patients with active disease are more activated and may require a higher dose of BTK inhibition to significantly reduce IgG secretion. In the five samples from active patients that produced PR3-ANCA in vitro, we found reduced levels upon BTK inhibition, indicating that BTK kinase activity may play an important role in autoantibody production. Taken together, BTK protein levels were increased in most B cell subsets of active GPA patients, including newly emerging transitional and naïve B cells, but only in a limited number of memory B cell subsets in remission GPA patients. These levels are correlated with enhanced BCR activity and sensitivity, and an increased expression of the co-stimulatory molecule CD86 on B cells. The correlation of B cell BTK levels with decreased numbers of pro-inflammatory Th1 and Th17.1 cells in the circulation would be consistent with migration of these pro-inflammatory T cells to target organs during active disease. This might be supported by our previous finding in SS patients that BTK protein levels in

circulating B cells correlated with T cell infiltration in the salivary glands50.

Furthermore, we found that BTK inhibition in vitro can inhibit B cell cytokine production, plasma cell formation and total IgG (auto)antibody secretion. Although B cells from active GPA patients respond less to BTK inhibition in vitro than healthy B cells, further studies in larger patient cohorts may identify BTK as an interesting novel therapeutic target in GPA patients.

(18)

4

Acknowledgments

We thank Theo Bijma and Minke Huitema for the experimental help. J.S.S. is supported by personal grants from the Dutch Kidney Foundation (grant number 13OKJ39) and the Dutch Organization for Scientific Research (Clinical Fellow grant number 907-14-542). W.H.A. and P.H. are supported by a grant from the European Union’s Horizon-2020 research and innovation program project RELENT (grant number 668036), and O.B.J.C. is supported by a grant from the Dutch Arthritis Foundation (Reumafonds, grant number 13-2-301).

Funding

This work was supported by Brisol-Myers Squibb, the Dutch Organization for Scientific Research (grant number 907-14-542), the Jan-Kornelis de Cock foundation and the Dutch Arthritis Foundation (Reumafonds, grant number 13-2-301).

Disclosure Statement

(19)

Supplementary Files

Supplementary Table 1. Monoclonal antibodies used for flow cytometry.

BTK protein Staining

Target Fluorochrome Clone Company

CD3 AF700 UCHT1 eBioscience

CD19 PerCP Cy5.5 SJ25C1 BD Biosciences

CD27 BV421 M-T271 BD Biosciences

CD38 APC HIT2 BD Biosciences

CD86 BV650 FUN-1 BD Biosciences

IgD PE Cy7 IA6-2 BD Biosciences

IgM Biotin G20-127 BD Biosciences

BTK PE 53/BTK BD Biosciences

Aqua live/dead BV506 - eBioscience

Streptavidin APCefl780 - eBioscience

T cell subset Staining

Target Fluorochrome Clone Company

CD3 APCefl780 UCHT1 eBioscience

CD4 AF700 OKT4 eBioscience

CD45RA BV650 HI100 BD Biosciences

CCR4 FITC 205410 R&D

CCR6 APC 11A9 BD Biosciences

CXCR3 BV711 1C6/CXCR3 BD Biosciences

CXCR5 PerCP Cy5.5 RF8B2 BD Biosciences

CTLA4 BV421 BNI3 BD Biosciences

FoxP3 PE 236A/E7 eBioscience

ICOS PE Cy7 ISA-3 eBioscience

PD1 BV786 EH12.2 BD Biosciences

(20)

4

Phosphoflow Staining

Target Fluorochrome Clone Company

CD3 AF700 UCHT1 eBioscience

CD19 FITC SJ25C1 BD Biosciences

CD27 BV421 M-T271 BD Biosciences

CD38 BV786 HIT2 BioLegend

IgD BV605 IA6-2 BD Biosciences

IgM Biotin G20-127 BD Biosciences

Aqua live/dead BV506 - eBioscience

Streptavidin APCefl780 - eBioscience

pBTK Y223 PE N35-86 BD Biosciences

pPLCγ2 Y759 APC K86-1161 BD Biosciences

Intracellular cytokine Staining

Target Fluorochrome Clone Company

CD3 BV786 SK7 BD Biosciences

CD8 PerCP Cy5.5 SK1 BioLegend

CD19 BUV737 SJ25C1 BD Biosciences

CD22 APC HIB22 BioLegend

IFNγ BUV395 B27 BD Biosciences

IL-6 PE Cy7 MQ2-13A5 eBioscience

IL-10 PE JES3-907 BioLegend

IL-21 BV421 3A3-N2.1 BD Biosciences

TNFα AF488 Mab11 BioLegend

NIR live/dead APC-Cy7 - BioLegend

Plasma cell Staining

Target Fluorochrome Clone Company

CD19 Efl450 HIB19 eBioscience

CD22 APC HIB22 BioLegend

CD27 APCefl780 O323 eBioscience

(21)

0 10 3 10 4 10 5 0 10 3 10 4 10 5 B cell T cell 0 10 2 10 3 10 4 10 5 0 10 2 10 3 10 4 10 5 naive

IgD+ non-sw IgD- memory

DN ef f 0 10 2 10 3 10 4 10 5 0 10 2 10 3 10 4 10 5

IgD- non-sw switched

0 10 2 10 3 10 4 10 5 0 10 2 10 3 10 4 10 5 transitional PB CD19 CD3 CD38 CD27 IgD CD27 IgM CD27 Supplemen tar y F igur e 1. G ating str at egy f or B -c

ell subsets and signal of B

TK pr ot ein staining . G ating str at egy as sho wn w as used in B TK pr ot

ein and phosphofl

o w stainings . T he sig nal of the B TK pr ot

ein staining is sho

wn and c ompar ed t o an isot ype c on tr ol in B - and T-cells . B TK , Brut on ’s tyr osine k inase , DN eff ; double nega tiv e eff ec tor , non-sw ; non-swit ched , PB; plasmablasts .

(22)

4

0 102 103 104 105 0 102 103 104 105 0 50K 100K 150K 200K 250K 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 CD3 CD4 CD45RA FCS CXCR5 PD1 FoxP3 CTLA4 CD4+ T naive memory follicular follicularnon Tfr Tfh 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 0 102 103 104 105 FoxP3 CD4 CCR6 CD4 CXCR3 CCR4 CXCR3 CCR4 Treg Treg non CCR6+ CCR6-Th17.1 Th17 Th1 Th2

Supplementary Figure 2. Gating strategy for T cell subsets, analyzed by flow cytometry.

T; T cells, Tfh; T follicular helper cells, Tfr; T follicular helper regulatory cells, Th; T helper, Treg; regulatory T cells.

(23)

to ta l B H C ac tiv e re m is si on R A 0 5 10 15 20 25 tes ocy mph f ly % o na iv e B H C ac tiv e re m is si on R A 0 20 40 60 80 10 0 % o f to tal B ce lls * Ig D + no n-sw itc he d H C ac tiv e re m is si on R A 0 10 20 lls ce B tal f to % o tra ns iti on al B H C ac tiv e re m is si on R A 0 5 10 15 ls cel B tal f to % o sw itc he d H C ac tiv e re m is si on R A 0 20 40 lls ce B tal f to % o pl as m ab la st H C ac tiv e re m is si on R A 0. 0 1. 0 2. 0 % o f to tal B cel ls Supplemen tar y F igur e 3. P rop or tions of cir cula ting B c ell subsets in HCs , ac tiv e and r emission GP A pa tien ts and R A pa tien ts , measur ed b y flo w c yt ometr y. *p<0.05. B; B c ells , HC; health y c on tr ols , R A; r heuma toid ar thr itis .

(24)

4

Supplementary Figure 4. Correlation between pBTK and pPLCγ2 expression upon BCR stimulation. Open circles are HCs, closed circles are active patients and closed squares are

remission patients. aIgM; anti-IgM, pBTK; phosphorylated Bruton’s tyrosine kinase, pPLCγ2; phosphorylated phospholipase C gamma 2, unstim; unstimulated.

(25)

A

B

C

m em or y C D 4+ T H C ac tiv e re m is si on 0 50 10 0 % o f ly mph ocy tes ** Th 1 H C ac tiv e re m is si on 0 40 80ls cel T D4+ C ory em f m % o * ** Th 2 H C ac tiv e re m is si on 0 10 20 30 40 50ls cel T D4+ C ory em f m % o Th 17 H C ac tiv e re m is si on 0 5 10 15ls cel T D4+ C ory em f m % o * Tr eg H C ac tiv e re m is si on 0 5 10 15 20 25ls cel T D4+ C ory em f m % o * * Tf h-lik e H C ac tiv e re m is si on 0 5 10 15 20 25ls cel T D4+ C ory em f m % o Tf r-l ik e H C ac tiv e re m is si on 0 4 8ls cel T D4+ C ory em f m % o Supplemen tar y F igur e 5. C ir cula ting T c ell subsets in HCs , ac tiv e and r emission GP A pa tien ts . A-C. P ropor tions of cir cula ting T c ell subsets in HCs , ac tiv e and r emission GP A pa tien ts , measur ed b y flo w c yt ometr y. D. IC OS expr ession on Tfh-like c ells , measur ed b y flo w c yt ometr y. HC; health y con tr ols , T ; T c ells , Tfh; T f ollicular helper c ell , Tfr ; T f ollicular helper r egula tor y c ell , T h; T helper , T reg; r egula tor y T c ells .

(26)

4

IL-6 IFNγ TNFα IL-10 CD3 CD8 CD19 CD22 FSC-A SSC-A FSC-A FSC-H Live/Dead Count Unstimulated anti-IgM anti-IgM + BTK Blocker

Supplementary Figure 6. Gating strategy to determine cytokine-positive B-cells. To

determine the frequencies of IFNγ+, IL-6+, IL-10+ and TNFα+ B cells, first a gate was set on

lymphocytes using the SSC-A/FSC-A plot. After excluding doublets in the FSC-H/FSC-A plot and

excluding dead cells, we gated on CD3-CD8- cells. Within these cells, CD22+CD19+ B cells were

gated. Within the CD22+CD19+ B cells, we set the gates for IFNγ+, IL-6+, IL-10+ and TNFα+ B cells on

the unstimulated sample and the same gates were applied to the anti-IgM only and anti-IgM + BTK blocked samples. IFNγ; interferon gamma, IL; interleukin, TNFα; tumor necrosis factor alpha.

(27)

Plasma cells Memory Naive Transi-tional CD27 CD38 CD19 CD22 FSC-A SSC-A CD19 CD27 CD19 CD38

Supplementary Figure 7. Gating strategy to determine B cell subsets in in vitro studies.

Using the FSC-A/SSC-A plot, we first determined CD38hi- and CD27+-expressing cells by plotting

CD19 against CD38 and CD27, respectively (top). Then, within the lymphocytes, CD22+CD19+ B

cells were gated in the CD19/CD22 plot and in the CD38/CD27 plot (bottom), we set the exact

same gates as determined in the top two figures to assess frequencies of CD38hiCD27- transitional

(28)

4

0 10 20 30 40 50 60 70 80 % withi n C D 19 +CD2 2 + B -cell s Transitional B-cells (%) HCs Active Remisson 0 5 10 15 20 25 30 % withi n CD 19 +CD2 2 + cell s

Ratio IL-10+ B-cells (%)

HCs Active Remission *

A

B

0.0 2.5 5.0 7.5 10.0 12.5 15.0 % withi n C D 19 +CD2 2 + cell s

Ratio IL-6+ B-cells (%)

HCs Active Remission 0 10 20 30 40 50 60 70 80 90 % withi n CD 19 +CD2 2 + B -cell s Naive B-cells (%) HCs Active Remission anti-IgM anti-IgM + BTK Blocker ** # *** ***

Supplementary Figure 8. Anti-IgM:anti-IgM+BTK blocker ratio for IL-10+ and IL-6+ B-cells

and transitional and naïve B-cell proportions. A. The ratio of the anti-IgM stimulated sample

and anti-IgM with BTK blocker samples were calculated and are given for IL-10+ and IL-6+ B cells for

HCs, active and remission GPA patients. B. Transitional and naïve B cell distribution after 7-day cell culture in the presence of anti-IgM with (open squares)/without (open circles) BTK inhibition for HCs, active and remission GPA patients. BTK; Bruton’s tyrosine kinase, GPA; granulomatosis with polyangiitis, HCs; healthy controls, IL; interleukin.

(29)

Referenties

GERELATEERDE DOCUMENTEN

Granulomatosis with Polyangiitis Patients, whereas Only Mycophenolic Acid Inhibits B cell IL-6 Production.

In this thesis, several aspects concerning the contribution of B cells to the AAV pathogenesis were investigated including functional implications (e.g. eff ector and

In this study it was shown that the frequency of activated B cells, identified as B cells with high expression levels of CD38, was significantly increased during active

Circulating CD24 hi CD38 hi Bregs Suppress Th17 cell Responses in vitro To elucidate the impact of Bregs on Th17 cell expansion, we sorted and co-cultured CD4 + Th cells

To determine the ex vivo effect of the active compounds of MMF and AZA, MPA and 6-MP, respectively, on B cell proliferation and cytokine production, peripheral blood mononuclear

In reply to the first sub-question (To what extent does Golden Dawn use national identity as a means of resonance with the Greek people?), the Golden Dawn deviates from

Dit artikel beschrijft de eerste resultaten van het onderzoek door IMARES naar de teelt van deze soort in Nederlandse recirculatiesystemen.. nieuwe soorten in

By gaining in-depth insights into behavioural choices within this daily activity, the results of this research can contribute to the relationship between parents' travel