• 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)

and Future Perspectives

7

(3)

Anti-neutrophil cytoplasmic autoantibody (ANCA)-associated vasculitides (AAV) is

a rare systemic autoimmune disease characterized by necrotizing inflammation of

small- to medium-sized blood vessels

2

. AAV comprises different clinicopathological

phenotypes including granulomatosis with polyangiitis (GPA), microscopic polyangiitis

(MPA), eosinophilic GPA and renal limited necrotizing crescentic glomerulonephritis

2

.

ANCA are considered to play an important role in the pathogenesis of AAV. These

autoantibodies (autoAbs) are primarily directed against the neutrophil- and

monocyte-derived enzymes proteinase (PR) 3 or myeloperoxidase (MPO)

3

.

Although the disease etiology is unknown, the pathogenesis of AAV is generally

considered to be multifaceted involving genetic predisposition, environmental

exposure (e.g. infections) and a complex interplay between multiple immune cells.

Particularly T- and B cells belonging to the cellular adaptive immune response play an

important role in AAV disease mechanisms. An altered distribution of CD4

+

T helper (Th)

cell subsets reflected by an expansion of a subset of circulating memory Th cells, termed

effector memory T cells (T

EM

), has been observed in AAV patients in remission

29

. Also, in

these patients a disbalance has been demonstrated in circulating Th cell responses with

a relative skewing towards Th17 cells as evidenced by increased circulating frequencies

of T

EM

17 cells

31

, increased serum interleukin (IL) 17A levels

32

, and an expanded

population of pathogenic PR3-specific Th17 cells

20,32

. Importantly, regulatory T cells

(Tregs), characterized by their immune modulating capabilities, are defective in AAV

patients

26

. Besides Th cells, B cells are well known to be involved in the pathogenesis of

GPA as ANCA-producing cells. Next to their capacity to produce (auto)Abs, B cells are

also considered to contribute to autoimmune responses by Ab-independent functions

such as antigen (Ag) presentation

43

and cytokine production

44

. Recently, an additional

subset of B cells with immunomodulatory capability, termed regulatory B cells (Bregs),

has been identified

131,132,141

. However, their role in AAV remains largely unexplored.

To suppress the effector cells of the cellular immune compartment, patients with AAV

receive standard remission induction and subsequent maintenance immunosuppressive

therapies. Two of the commonly used immunosuppressive agents in AAV used as

maintenance treatment include mycophenolate mofetil (MMF) and azathioprine (AZA).

AZA acts by inhibiting enzymes of the iosine-5’-monophosphate dehydrogenase (IMDP)

family thereby inhibiting leucocyte proliferation whereas MMF inhibits IMDP type 2

resulting in inhibition of lymphocyte proliferation

60

. Although it is widely acknowledged

these drugs block immune cell proliferation, the effects of MMF and AZA on the

distribution and function of B cell subsets is poorly understood. Of note, it is currently

unknown whether differential immunomodulatory effects of MMF and AZA are related

to the fact that MMF-treated AAV patients are more prone to future disease relapses

than AZA-treated patients

59

. Although immunosuppressive therapies have significantly

improved the prognosis of the disease, treatment is often associated with significant

(4)

7

toxicity, including an enhanced risk of infection, myelosuppression, malignancy, and

cardiovascular disease. Therefore, less toxic and more specific treatment is needed for AAV.

Previously, B cell-depleting therapy using rituximab has been shown to be as effective

as cyclophosphamide for induction of remission in patients with newly diagnosed AAV,

despite not targeting (the autoAb-producing) plasma cells

40,41

. Given that the initial

response to rituximab is seen within 72 hours of treatment, it is not likely to be due to

reduced Ab production alone

218

. It is well known that

B cell-derived cytokines affect the

differentiation of naive into effector Th cells

219

.

Thus, the beneficial effect of rituximab

may result from its indirect impact on Th cell responses. Therefore, it is important to

investigate the Ab-independent roles of B cells in AAV to reveal potential anomalies in B

cell functioning and their effect on Th cells.

Unravelling the role of cellular immunity (involving both B- and T cells) in the

pathogenesis of AAV could aid in the discovery of potential biomarkers to predict

relapses and the identification of more specific targets for therapeutic intervention. One

type of AAV, i.e. GPA, is particularly a relapsing disease in which ±60% of the patients

experience one or multiple relapses during the course of their disease

61

. Hence, it is

essential to identify GPA patients at risk for relapse (already) during disease remission

in order to prevent these relapses and there is still a pressing need for novel and more

reliable biomarkers that accurately predict disease relapses in GPA patients. Therefore,

the aims of this thesis were to:

1. Study the functional role of Bregs and effector B cells in the pathogenesis of GPA

2. Assess the effect of immunosuppressive therapy on B cell functioning in GPA

3. Examine the B cell repertoire as biomarker for future GPA disease relapses

In Chapter 3, we determined the frequency of circulating Bregs (i.e. phenotypically

characterized as CD19

+

CD24

hi

CD38

hi

cells). The frequencies of circulating Bregs were

significantly decreased in GPA patients receiving immunosuppressive therapy at the

time of measurement, whereas these frequencies were not different in untreated GPA

patients compared to healthy controls (HCs). Indeed, immunosuppressive treatment

is well known to induce lymphopenia in the circulation

220

. Hence, it is possible that

the discrepancy in the literature with respect to Breg frequencies in remission GPA

patients (discussed in Chapter 2) exists because patients were not stratified according

to immunosuppressive therapy at the time of sampling

45

. Also, in other autoimmune

diseases such as rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE)

numerical Breg deficiencies have been demonstrated, although in both studies

untreated and treated patients were included

131,135

.

Functional studies on Bregs have shown that these cells can inhibit Th cell responses

45,131

.

Together with the observations that in several autoimmune diseases frequencies and/

or suppressive function of Bregs are reduced

131,221–225

, this prompted us to study whether

(5)

reductions in Breg frequencies in GPA patients had an impact on the distribution of

circulating eff ector Th cells in Chapter 3. To this end, we assessed the frequency

of circulating Bregs in relation to the distribution of both circulating Th

EM

17 cells

(phenotypically characterized as CC-chemokine receptor (CCR) 6

+

CXC-chemokine

receptor (CXCR) 3

-

CCR4

+

CCR7

-

CD45RO

+

CD4

+

CD3

+

cells) and Th

EM

1 cells (phenotypically

characterized as CCR6

-

CXCR3

+

CCD4

-

CCR7

-

CD45RO

+

CD4

+

CD3

+

cells) in GPA patients.

As mentioned above, Breg frequencies were decreased in GPA patients treated with

immunosuppressive drugs, whereas these frequencies were not altered in untreated

GPA patients compared to HCs. The Th

EM

17 cell frequencies were increased in both

treated and untreated GPA patients compared to HCs, whereas the Th

EM

1 cell frequency

was decreased albeit in treated patients only. Importantly, we observed an inverse

correlation between Th

EM

17 cell and CD24

hi

CD38

hi

Breg frequencies in the circulation

of untreated GPA patients whereas a positive correlation was found between these

Bregs and Th

EM

1 cells. No correlations between these cell subsets were found in treated

patients. Although associative, these observations suggest that Bregs are important

regulators of Th

EM

cells in untreated GPA patients. To investigate whether Bregs were

indeed capable of suppressing Th

EM

cell responses, we sorted and co-cultured CD4

+

Th

cells with either CD24

hi

CD38

hi

Breg-depleted B cells or with total B cells of GPA patients

in remission in the presence of CpG-deoxynucleotides (CpG) and Staphylococcal

enterotoxin B. As explained in Figure 1, we showed that depletion of CD24

hi

CD38

hi

Bregs resulted in an increased IL-17

+

Th cell frequency, whereas no changes in IFNγ

+

Th cell frequencies were detected in GPA patients in remission. These fi ndings are in

line with a study in tuberculosis patients, although Bregs in this study were defi ned as

CD19

+

CD1d

+

CD5

+156

.

Th cell IL-17 IFNɣ Breg IL-17 Breg IFNɣ Th cell ThEM17 cells ThEM1 cells

Circulation

in vitro

Figure 1. Disturbed ThEM17- and ThEM1 cell frequency distribution within CD4+ Th cells and

the in vitro eff ect of CD24hiCD38hi Bregs on the Th cell response in GPA patients. Left: In the

circulation of untreated GPA patients increased ThEM17 cell frequencies are present, whereas the ThEM1 cell frequencies are decreased compared to HCs. Right: In vitro co-cultures of total B cells or CD24hiCD38hi Breg-depleted B cells with CD4+ Th cells of GPA patients showed that Bregs reduced

(6)

7

In support of our findings, studies assessing the B cell repertoire after B cell depletion

with rituximab in RA and SLE patients revealed that this treatment reduced the Th17

cell response and restored the Th cell balance

157

. In these patients, B cells with the

CD24

hi

CD38

hi

phenotype were the first B cell subset that emerged from the bone

marrow after B cell depletion with rituximab and became the dominant circulating

B cell subset

226,227

. Indeed, it has been speculated that the effectiveness of rituximab

in AAV can in part be ascribed to the expansion of Bregs and subsequent inhibition

of Th17 cell responses

157

. Alternatively, a recent study showed that in vitro CD8

+

T cell

pro-inflammatory cytokine response was decreased in AAV patients treated with

rituximab

228

. Moreover, a co-culture of B cells and naive CD8

+

T cells of AAV patients

resulted in increased pro-inflammatory cytokine production by these T cells compared

to HCs

228

. Thus, rituximab treatment might normalize the CD8

+

T cell response and during

B cell repopulation an enrichment in circulating Bregs may have a therapeutic effect by

inhibiting the pathogenic Th17 cells. However, as discussed before, the identification

and function of Bregs is, compared to Tregs, less well defined and we are only beginning

to understand the functions of these cells. Studies such as ours (Chapter 3) are required

to unravel the role of Bregs in autoimmune disease pathogenesis. Possibly, Bregs could

ultimately be used for in vitro expansion and autologous transfer to inhibit autoimmune

disease development. However, before we can even think of such therapeutic

applications, extensive functional studies are required first to really understand Breg

biology and behavior in health and disease.

Besides enhancing the suppressive function of Bregs, inhibiting activated effector

B cells might be a promising treatment option for GPA patients. Circulating B cells of

active GPA patients have been shown to be in an increased state of activation compared

to HCs as evidenced by increased CD38 expression

28

. Moreover, it has been recently

demonstrated that activated B cells from patients with autoimmune disease (RA and

Sjogren’s syndrome (SS)) show increased phosphorylation levels of signaling molecules

downstream of the B cell receptor (BCR) complex that could potentially serve as

therapeutic targets

50

. One of these important molecules in B cells is Bruton’s tyrosine

kinase (BTK), which is a pivotal signaling molecule that directly links BCR signaling to

B cell effector function. Therapeutic BTK blockade is already widely used to inhibit B

cell activation in chronic lymphocytic leukemia patients

229

and has recently been shown

to ameliorate kidney disease in a lupus nephritis mouse model

230

. Thus, BTK might be

a promising drug candidate for modulating B cell activation and effector function in

AAV. In Chapter 4, we examined the BTK protein levels and its phosphorylation status

in B cell subsets of GPA patients with active disease and in remission and HCs. We

showed that BTK protein levels were significantly increased in transitional and naïve

B cells of active GPA patients compared to remission patients and HCs (Figure 2). In

(7)

addition, increased BCR sensitivity to in vitro stimulation with anti-IgM, reflected by

increased BTK phosphorylation and signaling, and increased phospholipase C (PLC) γ2

phosphorylation, was detected in active GPA patients. Together, these results indicate

that in GPA patients newly emerging B cells are in a heightened state of activation,

which is in line with previous work in other autoimmune diseases i.e. RA and SS

50

.

We next assessed the impact of a BTK blocker on B cell cytokine production, plasma

cell formation and, IgG and ANCA production in vitro. The BTK blocker used in our

study is a small molecule that binds to the phosphorylation site of the BTK protein,

thereby preventing phosphorylation and subsequent activation of the BCR signaling

pathway

170

. Upon in vitro BTK blockade, we found that the frequencies of IFNγ

+

, IL-6

+

and IL-10

+

B cells were decreased in peripheral blood mononuclear cell (PBMC) samples

of active and remission GPA patients and HCs, whereas the TNFα

+

B cell frequency was

only decreased in remission GPA patients. Interestingly, plasma cell formation was only

inhibited by the BTK blocker in remission patients, whereas no difference was found in

active GPA patients and HCs. Importantly, plasma cell formation was increased in active

GPA patients with and without addition of the BTK blocker to the culture compared to

remission patients. Lastly, total IgG was decreased in remission patients and HCs in the

presence of BTK blockade and seemed to decrease in active patients. Interestingly,

PR3-ANCA levels tended to be lower in samples cultured in the presence of the BTK blocker

although this did not reach statistical significance, possibly due to low sample size.

Breg- and Treg-derived IL-10 is classically known as an anti-inflammatory cytokine

capable of suppressing the activation of other immune cells

231

. In Chapter 4 we found

that in vitro IL-10

+

Breg frequencies were decreased when a BTK inhibitor was added

to PBMC cultures. At first glance, such an effect seems undesirable given the

well-known anti-inflammatory properties of IL-10. However, IL-10 is also well-known to exert

non-inhibitory functions on various immune cells including B cells (reviewed in

231

). For

example, IL-10 has been shown to inhibit B cell apoptosis

232

, enhance B cell proliferation

and differentiation into plasma cells

233

, and promote immunoglobulin class switching

234

.

Therefore, in the context of B cell-mediated autoimmune diseases, IL-10 could also be

regarded as a pro-inflammatory cytokine. This contention is supported by GWAS studies

showing cytosine-adenine repeat polymorphisms in the IL-10 gene of GPA patients

235

,

multiple myeloma patients

236

and SLE patients

237

. Functionally, these polymorphisms

were associated with increased in vitro IL-10 production by lipopolysaccharide

stimulated PBMCs of multiple myeloma patients

238

. Moreover, polymorphisms in the

IL-10 gene have been reported to correlate with increased autoAb production in SLE

patients

237

. Although the functional consequences of IL-10 polymorphisms in GPA

remain to be investigated, a recent meta-analysis demonstrated that polymorphisms

in the IL-10 gene are associated with susceptibility for development of vasculitis, in

particular Behçets disease and GPA

239

. Collectively, these studies clearly indicate the

(8)

7

BTK

P

IL-10 Pro-inflammatory cytokines ANCA/IgG

No BTK Inhibition

BTK levels

Naive B cell Transitional B cell Memory B cell Plasmablast/ Plasma cell

BTK

P

IL-10 Pro-inflammatory cytokines ANCA/IgG

BTK Inhibition

Figure 2. BTK protein and phosphorylation levels in B cell subsets of active and remission GPA patients and HCs and the eff ect of BTK blockade on B cell functions. Top: BTK protein and phosphorylation levels were increased in newly emerging B cells (transitional and naïve B cells) of active GPA patients. Bottom: In vitro BTK inhibition decreased (auto)Ab secretion, as well as IL-10 and pro-infl ammatory cytokine (i.e. IFNγ and IL-6) production by B cells.

important role of IL-10 in the pathogenesis of autoimmune diseases. Moreover, we

speculate that while IL-10 may be essential for maintaining peripheral tolerance to

self-Ags and thus prevent autoimmunity, once tolerance is lost it could in fact promote

autoimmune disease by stimulating the diff erentiation of B cells into autoAb-producing

plasma cells.

(9)

Based on our in vitro findings the effectiveness of BTK blockade for active GPA patients

can be questioned as, for example, plasma cell formation in active GPA patients was

not inhibited by the BTK blocker. One possible explanation why BTK blockade did not

inhibit plasma cell formation is that B cells of active patients are already in an elevated

state of activation and do not require BCR stimulation. Additionally, experimental

evidence exists indicating that BTK inhibition not only suppresses B cell activation but

also cell migration. The latter was demonstrated in B cell lines where BTK- and

PLCγ2-deficient B cell lines were found to be unable to migrate in vitro. Moreover, treatment of

primary B cells with BTK or PLCγ2 blockers inhibited B cell migration in vitro indicating

that phosphorylation of these proteins is essential in this process

240

. Thus, blocking

BTK phosphorylation may be an attractive novel therapeutic strategy for AAV to not

only decrease effector B cell functioning but also effector B cell (e.g. memory B cell)

migration to sites of inflammation.

The current therapy options for remission maintenance for AAV include rituximab, AZA

or MMF combined with glucocorticoids. Interestingly, one open label clinical research

trial showed that MMF-treated AAV patients were more prone for disease relapses than

AZA-treated AAV patients

59

. Interestingly, the opposite is seen in SLE patients, where

MMF is more effective in the maintenance of remission

241

. The effect of MMF and

AZA on the immune system, and particularly on B cells, remains poorly understood.

We hypothesized that the increased relapse rate in MMF-treated AAV patients may be

due to a disbalance in Breg and effector B cell frequencies induced by MMF treatment.

Therefore, we investigated in Chapter 5 whether the distribution of Bregs and effector

B cells (identified based on their cytokine production) are differentially affected by

AZA and MMF in AAV patients. This study consisted of two parts. In part I, PBMCs of

untreated remission GPA patients and HCs were stimulated with CpG combined with

mycophenolic acid (MpA) or 6-mercaptopurine (6-MP) (i.e. the active compounds of

MMF and AZA, respectively) for three days. In part II, B cell functioning was determined

in GPA patients that were on maintenance therapy consisting of either AZA or MMF at

the time of blood sampling. To do so, we cultured PBMCs of these GPA patients and

stimulated the cells with CpG for three days. We showed that B cell proliferation was

decreased by MpA and 6-MP compared to CpG only, whereas no difference was found

in B cell proliferation in samples of actively treated GPA patients. Interestingly, in vitro

stimulation of PBMCs from GPA patients in the presence of MpA showed a decreased

IL-6

+

B cell frequency compared to 6-MP-treated GPA samples (Figure 3). Importantly,

the IL-10

+

B cell frequency was decreased by MpA in HC samples only whereas 6-MP did

not affect the cytokine-positive B cell frequencies at all. Although these results seem to

indicate that the cytokine profile of B cells in MpA-treated samples is shifted towards a

less pro-inflammatory state, these results need to be validated in larger patient cohorts.

(10)

7

Additionally, these immunosuppressive drugs are broad inhibitors of immune cell

activation not only aff ecting B cells but other immune cells as well.

B cell IL-10 regulatory effector TNF⍺

+ Azathioprine

/6-mercaptopurine

B cell regulatory effector IL-6

+ Mycophenolate

Mofetil

/Mycophenolic Acid

IL-6 TNF⍺ IL-10 B cell IL-10 regulatory effector TNF⍺

CpG

IL-6

Figure 3. B cell cytokine production in PBMCs of remission GPA patients and HCs in vitro upon exposure to 6-MP or MpA and in CpG-stimulated PBMCs of GPA patients receiving AZA or MMF treatment. Top: The eff ect of CpG stimulation alone on B cell cytokine production. Middle: The eff ect of in vitro AZA or 6-MP on B cell cytokine production compared to MMF/MpA treatment. Bottom: The eff ect of in vitro MMF or MpA on B cell cytokine production compared to AZA/6-MP treatment.

After GPA patients have achieved remission, the most important question for the

treating clinician is how to prevent a relapse most eff ectively with the least amount of

toxicity. To accomplish this, clinical and/or serological disease related factors need to

be identifi ed that reliably predict the risk for disease relapse in each patient. To date,

the most extensively investigated disease-related factor in GPA to predict relapse is the

(11)

serum PR3-ANCA titer and changes herein over time. However, results of these studies

are inconsistent and the value of serial PR3-ANCA measurements in clinical practice has

been a matter of debate. A persistently positive ANCA titer during disease remission has

been reported to be associated with future disease relapses

242

, whereas others found

that rather a rise in serum PR3-ANCA titer over time preceded a relapse

64

. In contrast,

others did not find an association between serum PR3-ANCA levels and ensuing

disease relapses

67

. Indeed, a meta-analysis by Tomasson et al.

68

showed that changes

in serum PR3-ANCA titer over time are only weak predictors of disease relapses and

are ineffective in predicting disease relapses for all AAV patients. However, more recent

research suggests that serial PR3-ANCA measurements may be helpful in identifying

relapses in patients presenting with renal involvement and alveolar hemorrhage

66,243

.

Additionally, PR3-ANCA levels should also be determined using novel methods (e.g. by

Phadia analyzer) to investigate whether such methods are more sensitive in detecting

rises in PR3-ANCA levels which may improve relapse prediction based on changes in

ANCA levels.

It has also been proposed that the prevalence of particular B cell subsets in the circulation

of AAV patients in remission may be an alternative disease related indicator for relapse

risk. The introduction of rituximab treatment particularly allowed for a unique way to

investigate whether specific distribution profiles of repopulated peripheral B cell subsets

associate with relapses

38

. Repopulation of circulating memory B cells at six months post

last rituximab infusion was found to be associated with increased relapse risk, whereas

patients with naïve B cell repopulation experienced less relapses

244

. Interestingly,

another study showed that increased proportions of circulating CD5

+

Bregs upon B cell

repopulation in AAV patients were related to prolonged remission whereas patients

with lower CD5

+

Breg frequencies showed a shorter time to relapse

144

. However, not all

GPA patients are treated with rituximab. Thus, in Chapter 6 we examined whether B cell

subset frequencies could predict future disease relapses in GPA patients in remission

not treated with rituximab. We showed that an increased frequency of circulating

plasmablasts was associated with decreased relapse-free survival. Circulating

plasmablast frequencies showed a trend towards a decrease in the last blood sample

collected 1-6 months prior to the relapse, which might indicate plasmablast migration

to sites of inflammation in GPA. Therefore, we stained plasmablasts in kidney biopsies

and urine of active AAV patients with renal involvement. Together, these results indeed

suggest that plasmablasts migrate from the circulation to sites of inflammation and that

monitoring circulating plasmablast frequencies might be a useful indicator for future

disease activity.

Plasmablasts are a result of the germinal center reaction and are migrating to plasma cell

niches mainly in the bone marrow to become plasma cells. In contrast to plasma cells,

plasmablasts are capable of producing only low amounts of antibodies. Plasmablasts

(12)

7

are, like plasma cells, typically present in low numbers in the circulation

245

. Importantly,

in other autoimmune diseases increased plasmablast frequencies were found to

correlate with autoAb levels and disease activity

198,199,206,207

. The data presented in

Chapter 6 suggest that in patients with upcoming relapses, B cells are already activated

and instructed to diff erentiate towards plasmablasts. The decrease in plasmablasts in

the last sample before relapse might highlight diff erentiation into plasma cells and

migration of these cells to sites of infl ammation. Although plasmablasts were related to

decreased relapse free survival when present in higher frequencies, we did not detect

a correlation between plasmablast frequencies and ANCA titers. Nonetheless, others

have shown that both memory B cells and plasmablasts are the predominant B cell

subsets of GPA patients to react with PR3

35

. Recently, PR3-ANCA-positive B cells have

been detected in infl amed tissues of GPA patients which is in line with our view that

autoreactive B cells migrate from the circulation to sites of infl ammation

246

. However, it

is currently not known whether plasmablasts at sites of infl ammation can secrete

PR3-ANCA.

+

Future disease relapse Remission

effector regulatory

Plasmablasts

Urine PeripheralBlood

Active Disease

Kidney

Figure 4. Plasmablasts as indicators for future disease relapses. During remission (top), future relapsing GPA patients demonstrated with increased plasmablast frequencies. During active disease (bottom), plasmablast frequencies were increased in the urine of GPA patients with renal involvement, whereas this frequency was lower in peripheral blood. Additionally, plasmablasts infi ltrated the infl amed kidney in these patients.

Although plasmablast frequencies might provide a novel marker to identify patients

at risk for relapse, a predictive B cell marker at an earlier stage of the disease is highly

(13)

preferred. Plasmablasts are already at a “late” differentiation stage and are in the process

of becoming Ab-producing plasma cells. Ideally, in AAV, but also in other autoimmune

diseases, the formation of pathogenic autoAb-producing cells is prevented. This would

be possible if patients at risk for relapse are identified earlier during disease remission

but this requires a marker that indicates increased activation of B cells at earlier

differentiation stages. BTK protein and its phosphorylation levels in newly emerging

B cells might provide such a B cell-specific early marker. In Chapter 4 we showed

that BTK levels were increased in transitional and naïve B cells of active patients only.

Importantly, these increased BTK levels correlated with B cell activation. Although in

our study BTK levels were not increased in remission GPA patients, it is likely that B cells

become increasingly activated in patients that are about to relapse. Currently, data on

BTK protein and phosphorylation levels in newly emerging transitional and naive B cells

of remission patients with approaching relapse is lacking. This is however of interest

because if newly emerging B cells in remission patients with future relapses indeed

show increased BTK levels, it would indicate increased B cell activation and might

identify patients at risk for relapse. The possibility of measuring B cell BTK levels as an

indicator for future relapse should be tested in a larger cohort with multiple fixed time

points to analyze changes in BTK levels over time.

Future Perspectives

The work presented in this thesis contributes to our knowledge on the role of B cells

in the pathogenesis of GPA and explored their potential as target for novel treatment

strategies and predictors of relapses. We have studied the distribution of B cell subsets

in GPA patients and found that an increased frequency of circulating plasmablasts was

associated with decreased relapse-free survival. Moreover, plasmablast frequencies

were found to be decreased in blood samples collected 1-6 months prior to relapse

of the disease and these cells could be detected in kidney biopsies and urine of GPA

patients with active renal disease. To this end, analyses of B cell subset distribution and

monitoring the frequency of plasmablasts in blood and urine could be informative as an

indicator of disease status and possibly aid in the recognition of an upcoming relapse in

AAV patients. Future studies in larger patient cohorts, including a cohort of

rituximab-treated GPA patients, are however necessary to substantiate whether plasmablast

frequencies are indeed predictive of relapses in GPA and should determine whether

these include plasmablasts that produce ANCA. Such studies will open up new avenues

for future use of these cells as markers for (upcoming) disease activity or as targets for

novel therapeutic strategies.

We also investigated the association between Bregs and the expanded Th17 cell

response in GPA patients. We observed an inverse correlation between circulating Bregs

(14)

7

and Th

EM

17 cells in GPA patients, and in vitro Breg depletion resulted in an increased Th17

cell frequency. Thus, a reduction of circulating Bregs in GPA patients may contribute

to increased numbers of Th17 cells which release IL-17, a pro-inflammatory cytokine

implicated in cell migration and granuloma formation in GPA patients. Future studies

are required to identify the signals that induce Breg expansion, as this may provide clues

to develop novel strategies to control Th17 cell responses in GPA patients and perhaps

autoimmune diseases in general. Moreover, it is essential that consensus is reached on

Breg phenotype(s), e.g. based on (novel) surface or functional markers or combinations

thereof, to truly discover the regulatory potential of these cells. Only then the potential

of therapeutic Breg transfer upon ex vivo expansion can be investigated as a possible

treatment strategy to restore immune balance.

In addition to the disturbed B cell subset distribution, we demonstrated alterations in

the BCR signaling pathway in newly emerging transitional and naive B cells of active

GPA patients. These B cells showed increased BTK levels whereas blocking BTK activity

inhibited B cell cytokine and IgG production, and plasma cell formation in vitro. Hence,

BTK might be a novel therapeutic target to dampen B cell activation in GPA patients

and future preclinical and clinical trials should establish whether BTK is a potential

novel treatment option for these patients. Our findings on intracellular BCR signaling

molecules such as BTK may also hold promise for the discovery of novel biomarkers

for (upcoming) disease activity. To investigate the potential of BTK as a biomarker, the

dynamics of BTK expression in B cells should be investigated in larger (longitudinal)

studies of future-relapsing and non-relapsing AAV patients.

Classically, the therapeutic strategy for GPA consists of remission induction and

maintenance therapy using immunosuppressive medication. However, B cell depletion

therapy by rituximab is increasingly applied for remission induction and maintenance in

GPA patients. Although rituximab is an efficacious therapeutic strategy, it is not specific

for the autoreactive B cells since it depletes all B cells including Bregs. The suggested

Breg expansion or BTK blockade might provide an additional therapeutic strategy

for AAV, and this could be more specific by inhibiting Th17 cell activation and B cell

activation, respectively.

However, the most ideal therapy for autoAb mediated autoimmune diseases would

be one that specifically depletes autoreactive B cells only. Interestingly, Ellebrecht et

al. tested such a novel therapy to deplete autoreactive B cells in pemphigus vulgaris

(PV)

247

. PV is a severe autoimmune disease that involves blistering of the skin and

oral mucosa and is characterized by autoAbs directed against keratinocyte adhesion

proteins (e.g. Dsg3). The authors engineered T cells with a chimeric autoAg receptor

(CAAR) and showed that these CAAR T cells specifically eliminated autoreactive

Dsg3-specific B cells in vitro and in a PV mouse model

247

. This elegant study provides proof of

principle that this form of cellular immunotherapy might be a future approach to treat

(15)

autoimmune diseases. The fact that this arising therapy specifically targets autoreactive

B cells makes it a very promising research field that could ultimately lead to an effective

and durable treatment of AAV and other autoimmune diseases.

(16)

7

References

1. Ntatsaki, E., Watts, R. A. & Scott, D. G. Epidemiology of ANCA-associated vasculitis. Rheum. Dis. Clin. North Am. 36, 447–461 (2010).

2. Jennette, J. C. et al. 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides.

Arthritis Rheum. 65, 1–11 (2013).

3. Harper, L., Cockwell, P., Adu, D. & Savage, C. O. S. Neutrophil priming and apoptosis in anti-neutrophil cytoplasmic autoantibody-associated vasculitis.

Kidney Int. 59, 1729–1738 (2001).

4. Jennette, J. C. Overview of the 2012 revised International Chapel Hill Consensus Conference nomenclature of vasculitides. Clin. Exp. Nephrol. 17, 603– 606 (2013).

5. Chen, M., Yu, F., Zhang, Y. & Zhao, M.-H. Antineutrophil cytoplasmic autoantibody-associated vasculitis in older patients. Medicine (Baltimore). 87, 203–209 (2008).

6. Lyons, P. A. et al. Genetically distinct subsets within ANCA-associated vasculitis. N. Engl. J. Med. 367, 214–223 (2012).

7. Xie, G. et al. Association of Granulomatosis with Polyangiitis (Wegener’s) with HLA-DPB1*04 and SEMA6A Gene Variants Evidence From Genome-Wide Analysis.

Arthritis Rheum. 65, 2457–2468 (2013).

8. Cohen, A. B. Mechanism of Action of alpha-1-antitrypsin. J. Biol. Chem. 248, 7055–7059 (1973).

9. Stegeman, C. A. et al. Association of Chronic Nasal Carriage of Staphylococcus aureus and Higher Relapse Rates in Wegener Granulomatosis. Ann. Intern.

Med. 120, 12–17 (1994).

10. Zycinska, K., Wardyn, K. A., Zielonka, T. M., Demkow, U. & Traburzynski, M. S. Chronic crusting, nasal carriage of staphylococcus aureus and relapse rate in pulmonary Wegener’s granulomatosis. J. Physiol.

Pharmacol. 59, 825–831 (2008).

11. Zycinska, K., Wardyn, K. A., Zielonka, T. M., Krupa, R. & Lukas, W. Co-Trimoxazole and Prevention of Relapses of PR3-ANCA Positive Vasculitis with Pulmonary Involvement. Eur. J. Med. Res. 14, 265–267 (2009).

12. Pendergraft, W. F. et al. Autoimmunity is triggered by cPR-3(105-201), a protein complementary to human autoantigen proteinase-3. Nat. Med. 10, 72–79 (2004). 13. Tadema, H., Kallenberg, C. G. M.,

Stegeman, C. A. & Heeringa, P. Reactivity against complementary proteinase-3 is not increased in patients with PR3-ANCA-associated vasculitis. PLoS One 6, (2011). 14. De Jong, N. W. M. et al. A structurally

dynamic N-terminal region drives function of the staphylococcal peroxidase inhibitor (SPIN). J. Biol. Chem. 293, 2260– 2271 (2018).

15. de Jong, N. W. M. et al. Immune evasion by a staphylococcal inhibitor of myeloperoxidase. Proc. Natl. Acad. Sci. 114, 9439–9444 (2017).

16. Stapels, D. A. C. et al. Staphylococcus aureus secretes a unique class of neutrophil serine protease inhibitors.

Proc. Natl. Acad. Sci. 111, 13187–13192

(2014).

17. Xu, P. C., Cui, Z., Chen, M., Hellmark, T. & Zhao, M. H. Comparison of characteristics of natural autoantibodies against myeloperoxidase and anti-myeloperoxidase autoantibodies from patients with microscopic polyangiitis.

Rheumatology 50, 1236–1243 (2011).

18. Cui, Z., Zhao, M. H., Segelmark, M. & Hellmark, T. Natural autoantibodies to myeloperoxidase, proteinase 3, and the glomerular basement membrane are present in normal individuals. Kidney Int. 78, 590–597 (2010).

19. Oliveira, D. B. G. Linked help from bacterial proteins drives autoantibody production in small vessel vasculitis. Med. Hypotheses 112, 24–26 (2018).

(17)

20. Abdulahad, W. H., Stegeman, C. A., Limburg, P. C. & Kallenberg, C. G. M. Skewed distribution of Th17 lymphocytes in patients with Wegener’s granulomatosis in remission. Arthritis

Rheum. 58, 2196–2205 (2008).

21. Land, J. et al. Prospective monitoring of in vitro produced PR3-ANCA does not improve relapse prediction in granulomatosis with polyangiitis. PLoS

One 12, e0182549 (2017).

22. Tadema, H. et al. Bacterial DNA motifs trigger ANCA production in ANCA-associated vasculitis in remission.

Rheumatology 50, 689–696 (2011).

23. Lepse, N. et al. Toll-like receptor 9 activation enhances B cell activating factor and interleukin-21 induced anti-proteinase 3 autoantibody production in vitro. Rheumatology 55, 162–172 (2016). 24. Fraser, J. D. Clarifying the mechanism of

superantigen toxicity. PLoS Biol. 9, 1–4 (2011).

25. Brouwer, E. et al. Predominance of IgG1 and IgG4 subclasses of anti-neutrophil cytoplasmic autoantibodies (ANCA) in patients with Wegener’s granulomatosis and clinically related disorders. Clin. Exp.

Immunol. 83, 379–86 (1991).

26. Abdulahad, W. H. et al. Functional Defect of Circulating Regulatory CD4+ T Cells in Patients With Wegener’s Granulomatosis in Remission. Arthritis Rheum. 56, 2080– 2091 (2007).

27. Brouwer, E., Stegeman, C. A., Huitema, M. G., Limburg, P. C. & Kallenberg, C. G. T cell reactivity to proteinase 3 and myeloperoxidase in patients with Wegener’s granulomatosis (WG). Clin Exp

Immunol 98, 448–453 (1994).

28. Popa, E. R., Stegeman, C. A., Bos, N. A., Kallenberg, C. G. M. & Cohen Tervaert, J. W. Differential B- and T-cell activation in Wegener’s granulomatosis. J. Allergy Clin.

Immunol. 103, 885–894 (1999).

29. Abdulahad, W. H., van der Geld, Y. M., Stegeman, C. A. & Kallenberg, C. G. M. Persistent expansion of CD4+ effector memory T cells in Wegener’s granulomatosis. Kidney Int. 70, 938–947 (2006).

30. Baron, B. J. L., Madri, J. A., Ruddle, N. H., Hashim, G. & Janeway, C. A. Surface Expression of alpha 4 Integrin by CD4 T cells is required for their entry into Brain parenchyma. J. Exp. Med. 177, 57–68 (1993).

31. Lintermans, L. L., Rutgers, A., Stegeman, C. A., Heeringa, P. & Abdulahad, W. H. Chemokine receptor co-expression reveals aberrantly distributed TH effector memory cells in GPA patients. Arthritis

Res. Ther. 19, 136 (2017).

32. Nogueira, E. et al. Serum IL-17 and IL-23 levels and autoantigen-specific Th17 cells are elevated in patients with ANCA-associated vasculitis. Nephrol. Dial.

Transplant. 25, 2209–2217 (2010).

33. Crotty, S. T follicular helper cell differentiation, function, and roles in disease. Immunity 16, 529–542 (2014). 34. Manz, R. A. et al. Humoral immunity

and long-lived plasma cells. Curr. Opin.

Immunol. 14, 517–521 (2002).

35. Cornec, D. et al. Identification and phenotyping of circulating autoreactive proteinase 3-specific B cells in patients with PR3-ANCA associated vasculitis and healthy controls. J. Autoimmun. 84, 122– 131 (2017).

36. Sanders, J.-S. F., Huitma, M. G., Kallenberg, C. G. M. & Stegeman, C. A. Plasma levels of soluble interleukin 2 receptor, soluble CD30, interleukin 10 and B cell activator of the tumour necrosis factor family during follow-up in vasculitis associated with proteinase 3-antineutrophil cytoplasmic antibodies: associations with di. Ann.

(18)

7

37. Hutton, H. L., Holdsworth, S. R. & Kitching, A. R. ANCA-Associated Vasculitis: Pathogenesis, Models, and Preclinical Testing. Semin. Nephrol. 37, 418–435 (2017).

38. Kallenberg, C. G. M., Stegeman, C. A., Abdulahad, W. H. & Heeringa, P. Pathogenesis of ANCA-associated vasculitis: New possibilities for intervention. Am. J. Kidney Dis. 62, 1176– 1187 (2013).

39. Söderberg, D. & Segelmark, M. Neutrophil extracellular traps in ANCA-associated vasculitis. Front. Immunol. 7, 1–9 (2016). 40. Jones, R. B. et al. Rituximab versus

cyclophosphamide in ANCA-associated renal vasculitis: 2-year results of a randomised trial. Ann. Rheum. Dis. 74, 1178–1182 (2015).

41. Stone, J. H. et al. Rituximab versus Cyclophosphamide for ANCA-Associated Vasculitis. N. Engl. J. Med. 363, 221–232 (2010).

42. Popa, C., Leandro, M. J., Cambridge, G. & Edwards, J. C. W. Repeated B lymphocyte depletion with rituximab in rheumatoid arthritis over 7 yrs. Rheumatology 46, 626–630 (2007).

43. Barnett, L. G. et al. B Cell Antigen Presentation in the Initiation of Follicular Helper T Cell and Germinal Center Differentiation. J. Immunol. 192, 3607– 3617 (2014).

44. Lino, A. C., Dörner, T., Bar-Or, A. & Fillatreau, S. Cytokine-producing B cells: A translational view on their roles in human and mouse autoimmune diseases.

Immunol. Rev. 269, 130–144 (2016).

45. Todd, S. K. et al. Regulatory B cells are numerically but not functionally deficient in anti-neutrophil cytoplasm antibody-associated vasculitis. Rheumatology

(Oxford). 53, 1693–1703 (2014).

46. Lepse, N. et al. Altered B cell balance, but unaffected B cell capacity to limit monocyte activation in anti-neutrophil cytoplasmic antibody-associated vasculitis in remission. Rheumatology 53, 1683–1692 (2014).

47. Aybar, L. T. et al. Reduced CD5+CD24hiCD38hi and interleukin-10+ regulatory B cells in active anti-neutrophil cytoplasmic autoantibody-associated vasculitis permit increased circulating autoantibodies. Clin. Exp. Immunol. 180, 178–188 (2015).

48. Bouaziz, J. D., Yanaba, K. & Tedder, T. F. Regulatory B cells as inhibitors of immune responses and inflammation. Immunol.

Rev. 224, 201–214 (2008).

49. Verstappen, G. M. et al. B cell depletion therapy normalizes circulating follicular TH cells in primary Sjögren syndrome. J.

Rheumatol. 44, 49–58 (2017).

50. Corneth, O. B. J. et al. Enhanced Bruton’s Tyrosine Kinase Activity in Peripheral Blood B Lymphocytes From Patients With Autoimmune Disease. Arthritis

Rheumatol. 69, 1313–1324 (2017).

51. Harwood, N. E. & Batista, F. D. New Insights into the Early Molecular Events Underlying B Cell Activation. Immunity 28, 609–619 (2008).

52. Baeuerle, P. A. & Henkel, T. Function and Activation of. (1994).

53. Weiner, M. et al. Outcome and treatment of elderly patients with ANCA-associated vasculitis. Clin. J. Am. Soc. Nephrol. 10, 1128–1135 (2015).

54. Guillevin, L. et al. Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. N. Engl. J. Med. 371, 1771–1780 (2014).

55. Specks, U. et al. Efficacy of remission-induction regimens for ANCA-associated vasculitis. N. Engl. J. Med. 369, 417–427 (2013).

(19)

56. Yates, M. et al. EULAR/ERA-EDTA recommendations for the management of ANCA-associated vasculitis. Ann.

Rheum. Dis. 75, 1583–1594 (2016).

57. Charles, P. et al. Comparison of individually tailored versus fixed-schedule rituximab regimen to maintain ANCA-associated vasculitis remission: results of a multicentre, randomised controlled, phase III trial (MAINRITSAN2).

Ann. Rheum. Dis. 77, 1143–1149 (2018).

58. de Joode, A. A. E., Sanders, J.-S. F., Rutgers, A. & Stegeman, C. A. Maintenance therapy in antineutrophil cytoplasmic antibody-associated vasculitis: Who needs what and for how long? Nephrol.

Dial. Transplant. 30, i150–i158 (2015).

59. Hiemstra, T. F. et al. Mycophenolate Mofetil vs Azathioprine for Remission Maintenance in Antineutrophil Cytoplasmic Antibody – Associated Vasculitis. J. Am. Med. Assoc. 304, 2381– 2388 (2011).

60. Dayton, J. S., Turka, L. A., Thompson, C. B. & Mitchell, B. S. Comparison of the effects of mizoribine with those of azathioprine, 6- mercaptopurine, and mycophenolic acid on T lymphocyte proliferation and purine ribonucleotide metabolism. Mol.

Pharmacol. 41, 671–676 (1992).

61. Luqmani, R. Maintenance of clinical remission in ANCA-associated vasculitis.

Nat. Rev. Rheumatol. 9, 127–132 (2013).

62. Walsh, M. et al. Risk factors for relapse of antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheum. 64, 542–548 (2012).

63. Pagnoux, C. et al. Predictors of treatment resistance and relapse in antineutrophil cytoplasmic antibody - Associated small-vessel vasculitis: Comparison of two independent cohorts. Arthritis Rheum. 58, 2908–2918 (2008).

64. Boomsma, M. M. et al. Prediction of relapses in Wegener’s granulomatosis by measurement of antineutrophil cytoplasmic antibody levels: a prospective study. Arthritis Rheum. 43, 2025–2033 (2000).

65. Sanders, J.-S. F., Huitma, M. G., Kallenberg, C. G. M. & Stegeman, C. A. Prediction of relapses in PR3-ANCA-associated vasculitis by assessing responses of ANCA titres to treatment. Rheumatology 45, 724–729 (2006).

66. Kemna, M. J. et al. ANCA as a Predictor of Relapse: Useful in Patients with Renal Involvement But Not in Patients with Nonrenal Disease. J. Am. Soc. Nephrol. 26, 537–542 (2015).

67. Finkielman, J. D., Merkel, P. A., Schroeder, D., Hoffman, G. S. & Spiera, R. Antiproteinase 3 Antineutrophil Cytoplasmic Antibodies and Disease. Ann. Intern. Med. 147, 611– 619 (2013).

68. Tomasson, G., Grayson, P. C., Mahr, A. D., LaValley, M. & Merkel, P. A. Value of ANCA measurements during remission to predict a relapse of ANCA-associated vasculitis--a meta-analysis. Rheumatology 51, 100–109 (2012).

69. Jennette, J. & Falk, R. Small-vessel vasculitis. N. Engl. J. Med. 337, 1512–1523 (1997).

70. van der Woude, F. J. et al. Autoantibodies against neutrophils and monocytes: tool for diagnosis and marker of disease activity in Wegener’s granulomatosis.

Lancet 1, 425–429 (1985).

71. Niles, J. L., McCluskey, R. T., Ahmad, M. F. & Arnaout, M. A. Wegener’s granulomatosis autoantigen is a novel neutrophil serine protease. Blood 74, 1888–1893 (1989). 72. Falk, R. J. & Jennette, J. C. Anti-Neutrophil

cytoplasmic autoantibodies with specificity for myeloperoxidase in patients with systemic vasculitis and idiopathic necrotizing and crescentic glomerulonephritis. N. Engl. J. Med. 318, 1651–1657 (1988).

(20)

7

73. Fauci, A. S., Haynes, B. F., Katz, P. & Wolff, S. M. Wegener’s Granulomatosis: Prospective clinical and Therapeutic experience with 85 patients for 21 years.

Ann. Intern. Med. 98, 76–85 (1983).

74. Jayne, D. The diagnosis of vasculitis. Best

Pract. Res. Clin. Rheumatol. 23, 445–453

(2009).

75. Jennette, J. C., Falk, R. J., Hu, P. & Xiao, H. Pathogenesis of Antineutrophil Cytoplasmic Autoantibody–Associated Small-Vessel Vasculitis. Annu. Rev. Pathol.

Mech. Dis. 8, 139–160 (2013).

76. Chen, M., Jayne, D. R. W. & Zhao, M.-H. Complement in ANCA-associated vasculitis: mechanisms and implications for management. Nat Rev Nephrol 13, 359–367 (2017).

77. Sakaguchi, S., Sakaguchi, N., Masanao, A., Misako, I. & Masaaki, T. Immunologic Self-Tolerance Maintained by Activated T cells Expressing IL-2 receptor alpha-chains (CD25). J. Immunol. 155, 1151–64 (1995). 78. Fillatreau, S., Sweenie, C. H., McGeachy,

M. J., Gray, D. & Anderton, S. M. B cells regulate autoimmunity by provision of IL-10. Nat. Immunol. 3, 944–50 (2002). 79. Abdulahad, W. H., Stegeman, C. A. &

Kallenberg, C. G. M. Review Article: The role of CD4+ T cells in ANCA-associated systemic vasculitis. Nephrology 14, 26–32 (2009).

80. Marinaki, S. et al. Abnormalities of CD4+ T cell subpopulations in ANCA-associated vasculitis. Clin. Exp. Immunol. 140, 181– 191 (2005).

81. Marinaki, S. et al. Persistent T-cell activation and clinical correlations in patients with ANCA-associated systemic vasculitis. Nephrol. Dial. Transplant. 21, 1825–1832 (2006).

82. Lúdvíksson, B. R. et al. Active Wegener’s granulomatosis is associated with HLA-DR+ CD4+ T cells exhibiting an unbalanced Th1-type T cell cytokine pattern: reversal with IL-10. J. Immunol. 160, 3602–3609 (1998).

83. Schönermarck, U., Csernok, E., Trabandt, A., Hansen, H. & Gross, W. L. Circulating cytokines and soluble CD23, CD26 and CD30 in ANCA-associated vasculitides.

Clin. Exp. Rheumatol. 18, 457–463 (2000).

84. Müller, A. et al. Localized Wegener’s granulomatosis: predominance of CD26 and IFN-γ expression. J. Pathol. 192, 113– 120 (2000).

85. Lamprecht, P. et al. Differences in CCR5 expression on peripheral blood CD4+CD28- T-cells and in granulomatous lesions between localized and generalized Wegener’s granulomatosis.

Clinical immunology 108, 1–7 (2003).

86. Szczeklik, W. et al. Skewing towards Treg and Th2 responses is a characteristic feature of sustained remission in ANCA-positive granulomatosis with polyangiitis.

Eur. J. Immunol. 47, 724–733 (2017).

87. Gershon, R. K. & Kondo, K. Cell Interactions in the Induction of Tolerance: The Role of Thymic Lymphocytes. Immunology 18, 723–737 (1970).

88. Gershon, R. K. & Kondo, K. Infectious immunological tolerance. Immunology 21, 903–914 (1971).

89. Baecher-Allan, C., Brown, J. A., Freeman, G. J. & Hafler, D. A. CD4+CD25high Regulatory Cells in Human Peripheral Blood. J. Immunol. 167, 1245–1253 (2001). 90. Brunkow, M. E. et al. Disruption of a new

forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 27, 68–73 (2001).

91. Chatila, T. A. et al. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. J. Clin. Invest. 106, 75–81 (2000).

92. Wildin, R. S. et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat. Genet. 27, 18–20 (2001).

(21)

93. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat.

Genet. 27, 20–21 (2001).

94. Hori, S., Nomura, T. & Sakaguchi, S. Control of Regulatory T Cell Development by the Transcription Factor Foxp3. Science (80-. ). 1057, 1057–1061 (2003).

95. Miyara, M. et al. Functional Delineation and Differentiation Dynamics of Human CD4+ T Cells Expressing the FoxP3 Transcription Factor. Immunity 30, 899– 911 (2009).

96. Mailer, R. K. W., Falk, K. & Rötzschke, O. Absence of leucine zipper in the natural FOXP3-2-7 isoform does not affect dimerization but abrogates suppressive capacity. PLoS One 4, 1–11 (2009). 97. Du, J., Huang, C., Zhou, B. & Ziegler, S.

F. Isoform-Specific Inhibition of ROR-Mediated Transcriptional Activation by Human FOXP3. J. Immunol. 180, 4785– 4792 (2009).

98. Ichiyama, K. et al. Foxp3 inhibits RORgammat-mediated IL-17A mRNA transcription through direct interaction with RORgammat. J. Biol. Chem. 283, 17003–17008 (2008).

99. Zhou, L. et al. TGF-β-induced Foxp3 inhibits TH17 cell differentiation by antagonizing RORγt function. Nature 453, 236–240 (2008).

100. Koenen, H. J. P. M. et al. Human CD25highFoxp3pos regulatory T cells differentiate into IL-17–producing cells.

Blood 112, 2340–2352 (2008).

101. Ayyoub, M. et al. Human memory FOXP3+ Tregs secrete IL-17 ex vivo and constitutively express the Th17 lineage-specific transcription factor RORgammat.

Proc. Natl. Acad. Sci. USA 106, 8635–8640

(2009).

102. Voo, K. S. et al. Identification of IL-17-producing FOXP3+ regulatory T cells in humans. Proc. Natl. Acad. Sci. U. S. A. 106, 4793–4798 (2009).

103. Vignali, D. A. A., Collison, L. W. & Workman, C. J. How regulatory T cells work. Nat. Rev.

Immunol. 8, 523–532 (2008).

104. Bodor, J. Editorial: The Molecular Mechanisms of Cyclic AMP in Regulation of Immunity and Tolerance. Front.

Immunol. 8, 1–3 (2017).

105. Burks, W. A., Laubach, S. & Jones, S. M. Oral tolerance, food allergy, and immunotherapy: Implications for future treatment. J. Allergy Clin. Immunol. 121, 1344–1350 (2008).

106. Sawant, D. V. & Vignali, D. A. A. Once a Treg, always a Treg? Immunol. Rev. 259, 173–191 (2014).

107. Klapa, S. et al. Lower numbers of FoxP3 and CCR4 co-expressing cells in an elevated subpopulation of CD4+CD25high regulatory T cells from Wegener’s granulomatosis. Clin. Exp.

Rheumatol. 28, 72–80 (2010).

108. Morgan, M. D. et al. Patients with Wegener’s granulomatosis demonstrate a relative deficiency and functional impairment of T-regulatory cells.

Immunology 130, 64–73 (2010).

109. Free, M. E. et al. Patients With Antineutrophil Cytoplasmic Antibody-Associated Vasculitis Have Defective Treg Cell Function Exacerbated by the Presence of a Suppression-Resistant Effector Cell Population. Arthritis Rheum. 65, 1922–1933 (2013).

110. Wilde, B., Hoerning, A., Kribben, A., Witzke, O. & Dolff, S. Abnormal Expression Pattern of the IL-2 Receptor beta-Chain on CD4+ T cells in ANCA-Associated Vasculitis. Dis.

Markers 2014, 1–9 (2014).

111. Zhao, Y. et al. Circulating T follicular helper cell and regulatory T cell frequencies are influenced by B cell depletion in patients with granulomatosis with polyangiitis.

(22)

7

112. Chavele, K. M. et al. Regulation of Myeloperoxidase-Specific T Cell Responses During Disease Remission in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis: The Role of Treg Cells and Tryptophan Degradation.

Arthritis Rheum. 62, 1539–1548 (2010).

113. Dominguez-Villar, M., Baecher-Allan, C. M. & Hafler, D. A. Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat. Med. 17, 673–675 (2011).

114. McClymont, S. A. et al. Plasticity of Human Regulatory T Cells in Healthy Subjects and Patients with Type 1 Diabetes. J. Immunol. 186, 3918–3926 (2011).

115. Beriou, G. et al. IL-17 producing human peripheral regulatory T cells retain suppressive function. Blood 113, 4240– 4250 (2009).

116. Komatsu, N. et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 20, 62– 68 (2014).

117. Kryczek, I. et al. IL-17+ Regulatory T Cells in the Microenvironments of Chronic Inflammation and Cancer. J. Immunol. 186, 4388–4395 (2011).

118. Abdulahad, W. H., Boots, A. M. H. & Kallenberg, C. G. M. FoxP3+ CD4+ T cells in systemic autoimmune diseases: the delicate balance between true regulatory T cells and effector Th-17 cells.

Rheumatology 50, 646–656 (2011).

119. Bettelli, E. et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238 (2006). 120. Takatori, H. et al. Helios Enhances Treg

Cell Function in Cooperation With FoxP3.

Arthritis Rheumatol. 67, 1491–1502

(2015).

121. Boyman, O. & Sprent, J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat.

Rev. Immunol. 12, 180–190 (2012).

122. Floess, S. et al. Epigenetic Control of the foxp3 Locus in Regulatory T Cells. PLoS

Biol. 5, 0169–0178 (2007).

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

124. Miyao, T. et al. Plasticity of Foxp3+ T Cells Reflects Promiscuous Foxp3 Expression in Conventional T Cells but Not Reprogramming of Regulatory T Cells.

Immunity 36, 262–275 (2012).

125. Fillatreau, S. Cytokine-producing B cells as regulators of pathogenic and protective immune responses. Ann. Rheum. Dis. 0, 1–5 (2012).

126. Mauri, C., Gray, D., Mushtaq, N. & Londei, M. Prevention of arthritis by interleukin 10-producing B cells. J. Exp. Med. 197, 489–501 (2003).

127. Katz, S. I., Parker, D. & Turk, J. L. B-cell suppression of delayed hypersensitivity reactions. Nature 251, 550–551 (1974). 128. Neta, R. & Salvin, S. B. Specific Suppression

of Delayed Hypersensitivity: The possible presence of a suppressor B cell in the regulation of Delayed hypersensitivity. J.

Immunol. 113, 1716–1725 (1974).

129. Mizoguchi, A., Mizoguchi, E., Takedatsu, H., Blumberg, R. S. & Bhan, A. K. Chronic intestinal inflammatory condition generates IL-10-producing regulatory B cell subset characterized by CD1d upregulation. Immunity 16, 219–230 (2002).

130. Yanaba, K. et al. A Regulatory B Cell Subset with a Unique CD1dhiCD5+ Phenotype Controls T Cell-Dependent Inflammatory Responses. Immunity 28, 639–650 (2008). 131. Blair, P. A. et al. CD19+CD24hiCD38hi

B Cells Exhibit Regulatory Capacity in Healthy Individuals but Are Functionally Impaired in Systemic Lupus Erythematosus Patients. Immunity 32, 129–140 (2010).

(23)

132. Iwata, Y. et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells.

Blood 117, 530–541 (2011).

133. Hagn, M. et al. Human B cells secrete granzyme B when recognizing viral antigens in the context of the acute phase cytokine IL-21. J. Immunol. 183, 1838–45 (2009).

134. Klinker, M. W. & Lundy, S. K. Multiple Mechanisms of Immune Suppression by B Lymphocytes. Mol. Med. 18, 123–137 (2012).

135. Flores-Borja, F. et al. CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting Th1 and Th17 differentiation. Sci.

Transl. Med. 5, 173ra23 (2013).

136. Shen, P. et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases.

Nature 507, 366–70 (2014).

137. Wang, R.-X. et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat. Med. 20, 633– 41 (2014).

138. Tian, J. et al. Lipopolysaccharide-Activated B Cells Down-Regulate Th1 Immunity and Prevent Autoimmune Diabetes in Nonobese Diabetic Mice. J. Immunol. 167, 1081–1089 (2001).

139. Kessel, A. et al. Human CD19+CD25high B regulatory cells suppress proliferation of CD4+ T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells.

Autoimmun. Rev. 11, 670–677 (2012).

140. Chaudhry, A. et al. Interleukin-10 Signaling in Regulatory T Cells Is Required for Suppression of Th17 Cell-Mediated Inflammation. Immunity 34, 566–578 (2011).

141. O’Dell Bunch, D. et al. Decreased CD5+ B cells in active ANCA vasculitis and relapse after rituximab. Clin. J. Am. Soc. Nephrol. 8, 382–391 (2013).

142. Wilde, B. et al. Regulatory B cells in ANCA-associated vasculitis. Ann. Rheum. Dis. 72, 1416–9 (2013).

143. Unizony, S. et al. Peripheral CD5+ B Cells in Antineutrophil Cytoplasmic Antibody-Associated Vasculitis. Arthritis Rheumatol. 67, 535–544 (2015).

144. O’Dell Bunch, D. et al. Gleaning relapse risk from B cell phenotype: decreased CD5+ B cells portend a shorter time to relapse after B cell depletion in patients with ANCA-associated vasculitis. Ann.

Rheum. Dis. 74, 1784–1787 (2015).

145. Wilde, B. et al. Granzyme B producing B-cells have immunoregulatory function and are diminished in patients with ANCA-vasculitis. Nephrol. Dial. Transplant. 31, i103 (2016).

146. Rosser, E. C. et al. Regulatory B cells are induced by gut microbiota–driven interleukin-1β and interleukin-6 production. Nat. Med. 20, 1334–9 (2014). 147. van der Geest, K. S. M. et al. Towards

precision medicine in ANCA-associated vasculitis. Rheumatology 17, 1–8 (2017). 148. Rosser, E. C. & Mauri, C. Regulatory B

Cells: Origin, Phenotype, and Function.

Immunity 42, 607–612 (2015).

149. von Borstel, A. et al. Cellular immune regulation in the pathogenesis of ANCA-associated vasculitides. Autoimmun. Rev. 17, 413–421 (2018).

150. Leavitt, R. Y. et al. The American College of Rheumatology 1990 criteria for the classification of Wegener’s granulomatosis. Arthritis Rheum. 33, 1101–1107 (1990).

151. Acosta-Rodriguez, E. V. et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat. Immunol. 8, 639–646 (2007).

152. Heufler, C. et al. Interleukin-12 is produced by dendritic cells and mediates T helper 1 development as well as interferon-gamma production by T helper 1 cells.

Referenties

GERELATEERDE DOCUMENTEN

[r]

Gezien de beperkte ruimte in de Senaatskamer is het bijwonen van de promotie alleen mogelijk op vertoon van een toegangsbewijs, welke kan worden aangevraagd bij de paranimfen.

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

Additionally, plasmablasts were investigated in matched urine and blood samples and kidney biopsies from active AAV patients with renal involvement.. Materials

Onze resultaten wezen uit dat circulerende plasmablast frequenties tijdens remissie geassocieerd waren met een ziekteverloop vrij van ziekte opvlammingen en wellicht een

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

Regulatory B cells (Bregs) of patients with granulomatosis with polyangiitis (GPA) have the capacity to suppress the T helper 17 cell response in vitro (this thesis). GPA

The question remains whether atypical naive-like effector/memory CD4 T cells should remain incorporated in the naive T cell compartment, since their classical membrane phenotype