• No results found

Marginal zone B cells are naturally reactive to collagen type II and are involved in the initiation of the immune response in collagen-induced arthritis

N/A
N/A
Protected

Academic year: 2022

Share "Marginal zone B cells are naturally reactive to collagen type II and are involved in the initiation of the immune response in collagen-induced arthritis"

Copied!
20
0
0

Loading.... (view fulltext now)

Full text

(1)

Uppsala University

This is a submitted version of a paper published in Cellular & Molecular Immunology.

Citation for the published paper:

Carnrot, C., Prokopec, K., Råsbo, K., Karlsson, M., Kleinau, S. (2011)

"Marginal zone B cells are naturally reactive to collagen type II and are involved in the initiation of the immune response in collagen-induced arthritis"

Cellular & Molecular Immunology, 8(4): 296-304 URL: http://dx.doi.org/10.1038/cmi.2011.2

Access to the published version may require subscription.

Permanent link to this version:

http://urn.kb.se/resolve?urn=urn:nbn:se:uu:diva-109185

http://.diva-portal.org

(2)

Marginal zone B cells are naturally reactive to collagen type II and initiate the immune response in collagen-induced arthritis

K. E. Prokopec

1*

, C. Carnrot

1*

, K. Råsbo

1

, M. C. I. Karlsson

2

and S. Kleinau

1

Manuscript in submission

* K.E.P and C.C. contributed equally to this work

1Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden, 2Department of Medicine, Unit of Clinical Allergy Research, The Karolinska Institute, Solna, Sweden.

Supported by the Swedish Research Council, The King Gustav V´s 80 years Foundation.

Abstract

B cells are important in collagen-induced arthritis (CIA) and antibodies against type II collagen (CII) are essential for disease development. How and where the autoimmune B cell response is activated in CIA is not fully known. Here we describe where autoreactive B cells are initiated following immunization with CII. Unimmunized mice and mice immunized with ovalbumin (OVA) were used as controls.

Surprisingly, we discovered that naïve DBA/1 mice display IgM, but also IgG, positive B cells reactive to CII prior immunization. The self-reactive B cells were observed in the spleen and identified as marginal zone (MZ) B cells. After CII-immunization, CII-specific B cells expanded rapidly in the spleen, while the concurrent response in the lymph nodes was sparse. In contrast, OVA- immunization resulted in an early OVA-specific B cell response in the lymph nodes rather than in the spleen. The early IgG anti-CII response following CII-immunization was derived from MZ B cells.

After immunization the MZ B cells migrated into the follicles, possibly depositing immune complexes onto follicular dendritic cells and activating T cells and follicular B cells. Thus, around disease onset increased numbers of IgG anti-CII producing follicular B cells was seen in the spleen and lymph nodes.

These data demonstrate that CII-reactive MZ B cells are present before and expanded after CII-

immunization. This autoreactive B cell response, present prior to concurrent T cell activation, may

contribute to the breakage of T cell tolerance and consequently to the activation of follicular B cells

and the generation of pathogenic IgG anti-CII antibodies.

(3)

Introduction

Autoantibody production is a characteristic of autoimmune diseases such as rheumatoid arthritis (RA). In mouse models of autoimmune arthritis it is well established that antibodies (Ab) play an essential role in disease development. Thus, in collagen- induced arthritis (CIA) high titres of IgG against collagen type II (CII) are essential for disease onset and transfer of the Ab to naïve mice induces arthritis [1, 2]. Furthermore, mice deficient in activating Fc gamma receptors (FcγR) are protected from CIA, while absence of the inhibitory FcγRIIb, which normally regulates Ab production, results in increased anti-CII Ab titres and augmented CIA [3].

Protection of CIA in B cell-deficient mice has further demonstrated a pathogenic role of B cells in arthritis [3-5]. This has also in recent years been verified in RA by the newly introduced B cell depletion therapy, where a positive clinical response occurs in correlation with a significant drop in auto-Ab [6].

I mportantly, low levels of auto-Ab can be detected in the serum many years before clinical symptoms of RA [7], suggesting that an early break in B cell tolerance is underlying the disease.

In the B cell compartment tolerance to self- antigen (Ag) is mediated by either clonal deletion and Ag receptor editing or functional silencing through anergy. However, these procedures are naturally not complete and peripheral B cells with low self-reactivity do occur. A subtype of B cells that has been shown to be enriched for self-reactivity is the innate-like marginal zone (MZ) B cells. MZ B cells are long lived, non-circulating B cells that reside strategically on the border of the white and red pulp in the spleen, as a first line of defence against blood–borne pathogens [8].

The MZ B cells can respond to low quantities of thymus-independent and thymus-dependent Ag and can initiate a rapid Ab response [8-12].

Further, they can transport Ag to the follicles where they can efficiently activate naïve T cells and quickly differentiate into plasma cells

[10, 13]. The MZ B cell compartment together with B1 B cells contain a large number of self- reactive clones and are associated with the production of natural Ab, which arise spontaneously at low titres without previous exogenous immune exposure [14]. Natural Ab are generally of the IgM isotype and are polyreactive, with low affinity towards foreign Ag but also against self-proteins [15, 16].

Interestingly, an expansion of the MZ B cell compartment has been associated with autoimmunity, as pathogenesis in non-obese diabetic (NOD) mice and in murine lupus is correlated with intrafollicular location of MZ B cells [17-20]. However, the role of MZ B cells in the autoimmune B cell response in CIA is still to be explored. We therefore characterized the early autoreactive B cell response in CIA by analyzing different lymphoid compartments of mice immunized with CII. For comparison we immunized a group of mice with a foreign Ag (ovalbumin; OVA) that does not induce autoimmunity. Unimmunized mice were used as controls. Surprisingly, we discovered a pre- existing anti-CII B cell response in the spleen of naïve DBA/1 mice. The self-reactive B cells produced mainly IgM, but also IgG anti-CII Ab, and belonged to the MZ B cell compartment. CII-immunization rapidly increased the number of CII-reactive MZ B cells and generated an early B cell response mainly in the spleen. In contrast, OVA immunization generated Ab forming cells (AFC) mainly in the lymph nodes rather than in the spleen. These results suggest that self- responsive MZ B cells in the spleen are involved in the initiation of the autoreactive CII response in CIA.

Materials and Methods Mice

NOD, C57BL/6 and arthritis-susceptible

DBA/1 mice (Bomholtgaard Ltd., Ry,

Denmark) of both sexes were bred and

maintained at the animal unit at the Uppsala

Biomedical Centre, Uppsala University,

(4)

Sweden. Balb/c mice (Bomholtgaard Ltd., Ry, Denmark) of both sexes were bred and maintained at the animal unit at the National Veterinary Institute, Uppsala, Sweden. The animals were fed rodent chow and water ad libitum and were negative for routine screened pathogens. The mice were six to ten weeks old at the initiation of the experiments. All experiments were approved by the local ethics committee (Uppsala tingsrätt, Sweden).

Collagen preparation

Native bovine CII (BCII) was prepared from nasal cartilage by pepsin digestion and subsequent purification, as described previously [21]. BCII was solubilised in 0.01 M acetic acid overnight at + 4ºC with constant mixing to 2 mg/ml.

CII and OVA immunizations and assessment of arthritis

For CIA studies BCII was emulsified with an equal volume (1:1) of complete Freund’s adjuvant (CFA) (Difco, Detroit, MI, USA) to a final concentration of 1 mg/ml. The mice were injected intradermally at the base of the tail with 50 µl of the emulsion, giving a dose of 50 µg of CII per mouse. Mice constituting a control group were immunized in the same way but with OVA (Sigma). The CII- immunized mice were inspected 2-3 times a week for arthritis development, starting three weeks after immunization, and clinical severity of arthritis was graded as described previously [22].

ELISA

The mice were bled by the tail vein at several time points after immunization and Ag specific IgM and IgG in individual serum samples were measured by ELISA, as described previously [22]. Briefly, sera from CII- or OVA- immunized mice were added to 96-well plates (MaxiSorp, NuncBrand Thermo Fischer Scientific, Roskilde, Denmark), which were coated with BCII and OVA, respectively. After incubation, alkaline phosphatase-conjugated sheep anti-mouse IgM or IgG Ab (Sigma) were

added for total Ig measurement and biotin- conjugated goat anti-mouse IgG1, IgG2a, IgG2b and IgG3 Ab (Southern Biotech, Birmingham, Alabama, AL, USA) for subclass measurement. For detection of IgG subclasses the plates were further incubated with alkaline phosphatase-conjugated ExtrAvidin (Sigma).

The plates were washed after each incubation and finally ρ-nitrophenyl phosphate substrate (Sigma, Steinheim, Germany), diluted in diethanolamine buffer (1 mg/ml), was thereafter applied and the absorbance was measured using a spectrophotometer (VersaMax, Molecular devices, Sunnyvale, USA) at 405 nm.

Cell preparation

Naïve or immunized mice were killed at different time points after immunization and the spleen, bone marrow and pooled axillary and inguinal lymph nodes were collected.

Individual single cell suspensions were prepared from each lymphoid tissue. Bone marrow cells were flushed with cold PBS from femurs and kept on ice in DMEM culture medium (Sigma) with 100 U of penicillin (Sigma), 100 µg/ml of streptomycin (Sigma), 1

% L-Glutamine (Sigma) and 10 % FCS (Sigma) referred to as complete DMEM medium. The lymph nodes and spleen were carefully pressed through a stainless steel mesh into a single cell suspension. Splenic erythrocytes were lysed by ACK lysis buffer (0.15 M NH

4

Cl, 0.1 mM Na

2

EDTA, 1.0 M KHCO

3

) and further washed in PBS. The single cell suspensions of the spleen and the lymph nodes were finally diluted in complete DMEM medium and kept on ice. Cell counts of individual samples were made with trypan blue (Gibco).

Cell isolation and sorting

Mature B cells were sorted from splenocytes

by magnetic activated cell sorting (MACS)

followed by FACS sorting, or by FACS sorting

only. MACS was performed according to the

manufacturer’s protocol (Miltenyi Biotec,

Bergisch Gladbach, Germany). Briefly, the

(5)

splenocytes were labelled with biotin- conjugated anti-mouse CD43 (eBioscience, San Diego, CA, USA) at 4°C for 20 min. After washing the splenocytes were incubated with streptavidin microbeads (Miltenyi Biotec) at 4°C for 30 min. After additional washing, the splenocytes were resuspended in MACS buffer and put on a LS column (Miltenyi Biotec

)

in a magnetic field. The CD43 positive cells, including T cells and B1 B cells, were retained in the column and the negative cell fraction, containing B cells, was collected. The B cells were thereafter labelled with PE-conjugated anti-mouse CD1d (clone 1B1, BD Pharmingen) and FITC-conjugated anti-mouse CD23 (clone B3B4, BioLegend, San Diego, CA, USA) at 4°C for 30 min. After washing, the splenic B cells were separated into MZ and follicular (FO) B cells using a FACS Vantage DiVa and FACS Diva software 5.1 (BD Biosciences). MZ B cells were defined as CD23

low

and CD1d

high

and FO B cells as CD23

high

and CD1d

low

(figure S1).

Cell sorting using FACS only were performed accordingly: Splenocytes were stained with PE-labelled anti-B220 (clone RA3-6B2, BD Pharmingen), biotin-labelled anti-CD1d (clone 1B1, BD Pharmingen) and FITC-conjugated anti-CD23 (clone B3B4, BD Pharmingen) at 4°C for 30 min. After incubation the cells were washed twice and incubated for an additional 30 min with streptavidin-conjugated APC on ice (BD Pharmingen). The B220

+

splenocytes were gated and further sorted into MZ B cells, defined as CD23

low

and CD1d

high

and FOB cells, defined as CD23

high

and CD1d

low

, using a FACS Vantage DiVa and FACS Diva software 5.1 (BD Biosciences).

Splenocytes stained with FITC- or PE- conjugated isotype-matched control Ab (BD Pharmingen) were used as negative controls.

To determine possible contamination of transitional B cells in the different B cell populations (MZ and FO B cells) a FACS staining was performed. Accordingly, splenocytes, before and after anti-CD43 MACS separation, were stained with biotinylated anti-CD1d, FITC conjugated anti-

CD21, PE conjugated anti-CD23 (all from BD Pharmingen), APC conjugated anti-IgM (BioLegend) and anti-B220 conjugated with pacific blue (BioLegend) for 30 min on ice.

After washing an additional staining with streptavidin PerCP Cy 5.5 (BioLegend) was performed for 30 min on ice. The cells were analyzed using FACSAria and the FACSDiva software (BD) and transitional T1 cells were defined as CD23

negative

, IgM

high

and CD21

low

and T2 cells as CD23

positive

IgM

high

and CD21

intermediate

[23] (figure S1). In addition, a separate staining to elucidate possible expression of CD5 on separated splenocytes was performed. The cells were stained with FITC conjugated anti-IgD (BD Pharmingen), APC conjugated anti-IgM (BioLegend), pacific blue conjugated anti-B220 (BioLegend) and PE conjugated anti-CD5 (BD Pharmingen) for 30 minutes on ice. The cells were analyzed using FACSAria and the FACSDiva software (BD).

FcγRIIb expression

In order to study FcγRIIb expression on MZ and FO B cells, splenocytes were stained with PE-labelled anti-CD1d (clone 1B1, BD Pharmingen), FITC-conjugated anti-CD23 (clone B3B4, BD Pharmingen) and biotin labelled anti-FcγRIIb haplotype Ly-17.2 (clone K9.361) (kindly provided by Dr. Ulrich Hammerling) at 4°C for 30 min. After incubation the cells were washed twice and incubated for an additional 30 min with streptavidin-conjugated APC on ice (BD Pharmingen). The splenic B cells were thereafter separated into FO B cells and MZ B cells as described above and further analyzed for their FcγRIIb expression using FACS LSR SORP and the FACS Diva software 5.1 (BD Biosciences).

Sphingosine-1-phosphate receptor 1 expression

To investigate the sphingosine-1-phosphate

receptor 1 (S1P

1

R) mRNA expression on MZ

and FO B cells, splenocytes were isolated from

unimmunized mice and mice sacrificed 2 days

(6)

after CII- immunization. The MZ and FO B cells were separated using MACS and FACS (as described above) and RNA was extracted from the cells using Trizole (Invitrogen, Stockholm, Sweden), following the Roche Tripure Isolation Reagent protocol. After treating the RNA with DNas TURBO DNas treatment and removal reagents (Applied Biosystem, Foster City, CA, USA), complementary DNA was generated using Verso cDNA kit, Oligo dT (Thermo, Epsom, Surrey, UK) and PCR (42˚C 30 min, 95˚C 2 min) (Biometra Thermocycler, Goettingen, Germany). A quantitative PCR was performed using BioRad iCycler and the iCycler iQ software (optical system software version 3.1)(BioRad, Hercules, CA, USA) with the following S1P

1

R probes in combination with SYBR green (Applied Biosystems); forward primer, GTG TAG ACC CAG AGT CCT GCG and reverse primer, AGC TTT TCC TTG GCT GGA GAG. For the house keeping gene HPRT the forward primer, AGG TTG CAA GCT TGC TGGT and the reverse primer, TGA AGT ACT CAT TAT AGT CAA GGG CA were used.

ELISPOT

Ninety-six-well plates (MaxiSorp, NuncBrand Thermo Fischer Scientific, Roskilde, Denmark) were coated over night at 4°C with 50 µl of 0.2 mg/ml BCII or OVA, and bovine serum albumin (BSA) fraction V (Sigma) as an irrelevant control protein. The plates were washed with PBS and 10

6

splenocytes, lymph node or bone marrow cells or 5 - 10 x 10

5

sorted MZ or FO B cells diluted in 200 µl complete DMEM medium were added to each well. A minimum of eight wells per lymphoid tissue were analyzed, while MZ and FO B cells were analyzed in 1 - 5 wells, depending on the number of cells gained after cell sorting. The cells were cultured for 20 - 22 h at 37°C in a 5

% CO

2

atmosphere. After washing alkaline phosphatase-conjugated sheep anti-mouse IgM or IgG (Sigma) was added to the plates and incubated for 2 h in room temperature or over night at 4°C. After washing with 0.2 M Tris

buffer at pH 9.1, 50 µl of BCIP/NBT liquid substrate system (Sigma) was added to each well. The development of AFC seen as spots were monitored for up to 1 h and the reaction was stopped by washing the plates with distillate water. The spots were counted under an inverted microscope (Leitz Diavert, Germany). A mean value of spots in the wells were calculated and adjusted when necessary to number of spots per one million cells. The IgG subclass ELISPOT was performed as described above except that biotin-conjugated goat anti-mouse IgG1, IgG2a, IgG2b and IgG3 Ab (Southern Biotech, Birmingham, Alabama, AL, USA) were added to the plates and incubated over night at 4˚C. The plates were washed and alkaline phosphatase-conjugated ExtrAvidin (Sigma) was added and incubated for 2 h at RT. Spots were developed with BCIP/NBT liquid substrate system as described above.

Immunofluorescence

Spleens were removed from unimmunized DBA/1 mice and from CII-immunized DBA/1 mice day 7 and 28 after immunization. The spleens were snap frozen in Killik cryostat embedding medium (Bio-optica, Milan, Italy) and were kept at -70˚C until sectioned.

Cryostat sections were cut at 8-10 µm and were fixed in acetone and blocked with 1:20 diluted goat-sera for 15 minutes prior staining.

After additional washes the sections were

incubated with MOMA-1 conjugated to FITC

(anti-CD169) (AbD serotec, Düsseldorf,

Germany), anti-CD3 conjugated to PE or anti-

B220 conjugated to PE in combination with

biotinylated anti-CD9 (BD Pharmingen) for

1h. The sections were washed and either

incubated with streptavidin-conjugated APC

for 30 min or mounted in Fluoromount-G

(Southern Biotech, Birmingham, Alabama,

AL, USA). APC stained sections were further

washed and mounted. In addition, consecutive

sections were stained with isotype control Ab

(BD Pharmingen). The sections were analyzed

using a confocal microscope (LSM 510 Meta,

Carl Zeiss, Jena, Germany).

(7)

Statistics

The Student’s t-test was used for analyzing the sorted MZ and FO B cells, FcγRIIb expression, the number of AFC in different mouse strains and between CII- and OVA-immunized mice.

Figure 1. The autoimmune B cell response is initiated in the spleen of CII-immunized mice

The number of CII- and OVA-specific IgM and IgG AFC were analyzed in mice immunized with CII (n = 6-

21) or OVA (n = 6-14) by ELISPOT. Data is presented as mean AFC per million cells ± SEM at different time

points after immunization in different organs (A-F). Statistical differences CII vs. OVA: in spleen, IgM AFC

week 0, 1 and 4; p < 0.01 - 0.05 and IgG AFC week 0 and 1; p < 0.001 - 0.05. In bone marrow, IgG AFC week

9; p < 0.01 G. The combined IgM and IgG anti-CII AFC in the spleen and lymph nodes of CII-immunized mice

at early time points. H. The combined IgM and IgG anti-OVA AFC in the spleen and lymph nodes of OVA-

immunized mice at early time points. I. Mean OD (± SEM) values of IgM (diluted 1:80) and IgG (diluted 1:50)

anti-CII in sera from CII-immunized DBA/1 mice (n = 5-10). J. Mean OD (± SEM) values of IgM (diluted

1:50) and IgG (diluted 1:1250) anti-OVA in sera from OVA-immunized DBA/1 mice (n = 5-12). Where not

visible, error bars are contained within the symbol.

(8)

Results

The spleen is responsible for the initial anti- CII Ab response in CIA

To explore the sites where self-reactive B cells are initiated in CIA, lymphoid organs of CII- immunized DBA/1 mice were analyzed by ELISPOT at different time points after immunization and the number of CII-specific AFC was investigated. DBA/1 mice immunized with OVA were used as controls to study a non-autoimmune response, as OVA is a foreign Ag that does not mimic an auto-Ag nor induce arthritis. Interestingly, when analyzing AFC in unimmunized DBA/1 mice (week 0) we found that naïve splenic B cells produced Ab against CII (figure 1a, d). The majority of the CII-reactive B cells produced IgM, but IgG positive cells could also be observed. Although the number of CII-specific AFC in the naïve spleen was low it was significant, as no such reactivity was observed in the lymph nodes, the bone marrow or towards OVA (CII vs. OVA p < 0.001 for IgM AFC and p < 0.001-0.01 IgG AFC) (figure 1a- f). Neither could any AFC be observed in the naïve spleen when BSA, representing another bovine Ag, was used as coating agent (data not shown).

After CII-immunization the number of IgM anti-CII AFC in the spleen increased rapidly, peaked at two weeks and subsequently declined at four weeks after immunization (figure 1a). The AFC pattern in the lymph nodes was similar, but the number of IgM anti- CII AFC was fewer (figure 1b). The bone marrow demonstrated in general very few IgM anti-CII AFC (figure 1c).

Interestingly, and in contrast to the CII- immunized mice, OVA-immunized mice did not develop an early and strong IgM Ab response in the spleen (CII vs. OVA p < 0.01 week 1 and 2) (figure 1a). Instead, these animals displayed a somewhat greater IgM anti-OVA response in the lymph nodes one week after immunization, which increased at two weeks and subsequently declined thereafter (figure 1b). Very few IgM anti-OVA

AFC were seen in the bone marrow of OVA- immunized mice (figure 1c).

The number of splenic IgG anti-CII AFC in CII-immunized mice was fewer compared with the splenic IgM anti-CII AFC (figure 1a, d).

Nevertheless, an early production of IgG anti- CII was observed that remained relatively constant throughout the experiment (figure 1d).

The IgG anti-CII response in the lymph nodes was somewhat delayed compared to the spleen, but a significant number of IgG anti-CII AFC was demonstrated four weeks after CII- immunization (figure 1e), a time point when the mice start to develop CIA (figure S2a). At nine weeks after CII-immunization, and at a more late stage of the disease (figure S2b), the number of IgG anti-CII AFC in the lymph nodes had declined, whereas the IgG anti-CII AFC in the bone marrow instead had increased substantially (figure 1e, f).

In OVA-immunized mice the IgG anti-OVA response in the spleen was most evident four weeks after immunization (figure 1d). A similar pattern was observed in the bone marrow (figure 1f). However, very few OVA- specific IgG AFC were seen in the lymph nodes during the experiment (figure 1e). The number of OVA-specific IgG AFC cells declined even further at nine weeks after OVA- immunization, and this was also observed in the bone marrow, which displayed less IgG AFC than CII-immunized mice (CII vs. OVA p

< 0.01)(figure 1 d-f).

When compiling the ELISPOT data from spleen and lymph nodes for the first two weeks after immunization, it was evident that the majority of the humoral response to CII principally started in the spleen (figure 1g).

Even prior CII-immunization, noticeable IgM

and IgG responses to CII were present in the

spleen (week 0). In contrast, IgM and IgG

responses to OVA started essentially in the

lymph nodes rather than in the spleen of OVA-

immunized mice (figure 1h). Additionally, the

total number of IgM AFC during the first two

weeks after immunization appeared higher in

CII-immunized animals, while the total IgG

(9)

Table I. Splenic B cell response to CII in different naive mouse strains

Strain

a

Anti-CII AFC

b

IgM IgG

DBA/1 1.6 ± 0.3* 1.2 ± 0.4

NOD 1.1 ± 0.3 1.5 ± 1.3

Balb/c 0.9 ± 0.3 1.2 ± 0.8

C57BL/6 0.3 ± 0.2 0.3 ± 0.1

a3 - 6 mice were analyzed in each strain. *p ≤ 0.05 between DBA/1 and C57BL/6 mice.

bMean anti-CII AFC per million plated splenocytes ± SEM

AFC were comparable in CII- and OVA- immunized mice (figure 1g, h).

We next set out to investigate how the B cell specific response in the different lymphoid organs corresponded with the total Ab concentration in the mice. Thus, Ag specific IgM and IgG Ab were analyzed in the sera at different time points after immunization by ELISA (figure 1i, j). The CII-immunized mice developed IgM anti-CII Ab at one week after immunization, which increased steadily up to four weeks and thereafter stayed at a relatively constant level to the termination of the experiment (Figure 1i). IgG anti-CII Ab were not observed until two weeks after immunization, but increased progressively thereafter at four and nine weeks after immunization. The Ab pattern in OVA- immunized mice was quite different from the CII-immunized animals and the Ab levels were much higher. Thus, high IgM anti-OVA Ab levels were observed at one week after immunization that rapidly dropped at two and four weeks after immunization (figure 1j). The IgG levels in OVA-immunized animals were very high and peaked already at 4 weeks after immunization. Consequently, serum from OVA-immunized mice needed to be diluted 25 times more in the IgG ELISA than the sera from CII-immunized mice.

These findings indicate that the concentration of Ab in serum may not necessarily reflect the number of Ag-specific AFC in lymphoid organs. However, it should also be taken into account that anti-CII Ab may disseminate out to joints, where they bind joint cartilage, rather than staying in the circulation.

Autoimmune prone mice display self-reactivity to CII in the spleen

The demonstrated natural self-reactivity to CII in DBA/1 mice led us to investigate whether other mouse strains also display similar self- reactivity. Thus, splenic cell suspensions were prepared from naïve DBA/1, NOD, Balb/c and C57BL/6 mice and the presence of anti-CII AFC using ELISPOT was investigated.

Interestingly, all strains except C57BL/6 mice displayed notable IgM and IgG AFC to CII (table I), while AFC to a control bovine protein, BSA, were absent (data not shown).

DBA/1 mice had significantly more IgM anti- CII AFC than C57BL/6 mice (p ≤ 0.05) and the number of IgG anti-CII AFC were higher compared to C57BL/6 mice. No IgG and IgM towards CII could be demonstrated in the sera of the different naïve mouse strains (data not shown).

MZ B cells harbour the CII reactivity and expand after immunization

Since AFC to CII were first detected in the spleen we were interested to see if the Ab production could be related to a certain splenic B cell population. Thus, splenocytes from naïve and CII-immunized DBA/1 mice were separated into FO and MZ B cells by MACS and flow cytometry using CD23 and CD1d markers (figure 2a). Possible contamination of transitional B cells (T1 and T2) in the MZ B cell population was negligible as well as CD5 positive B cells as demonstrated by FACS (figure S1 and data not shown, respectively).

The percentages of FO and MZ B cells

remained constant during the experiment (data

(10)

not shown). When the populations were analyzed for IgG anti-CII reactivity, using the ELISPOT assay, it was shown that the CII- reactive B cells of the naïve mice predominantly belonged to the MZ B cell population. One week after CII-immunization the number of self-reactive MZ B cells increased and was significantly greater than the number of CII-reactive FO B cells (p < 0.05) (figure 2b). The number of MZ B cells subsequently decreased thereafter, and at four weeks after immunization the FO B cells produced the majority of the IgG anti-CII Ab (figure 2b). The increase of FO B cells producing IgG anti-CII Ab seen at week 4 correlated with CIA onset (figure 2b and S2a).

Furthermore, we investigated the FcγRIIb expression (mean fluorescence intensity; MFI) on the MZ and FO B cells following CII- immunization, since FcγRIIb is involved in the regulation of Ab production in B cells. The MFI value of FcγRIIb was significantly higher on the MZ B cells compared to the FO B cells at two and four weeks after CII-immunization (figure 2c). In addition, the FcγRIIb expression on MZ B cells at four weeks was significantly increased compared to two weeks after immunization, possibly indicating a higher level of regulation of the MZ B cells.

Enhanced migration of MZ B cell to the follicle following CII-immunization

The enhanced number of CII-reactive MZ B cells one week after immunization, led us to investigate whether this MZ B cell activation was associated with enhanced migration of the MZ B cells into the follicles. Thus, MZ B cell shuttling is related to Ag transportation into the follicles and this migration is linked to the degree of S1P

1

R expression on the cells [24, 25]. Accordingly, MZ and FO B cells were isolated from splenocytes from unimmunized mice and mice that had been immunized with CII two days previously. The mRNA levels of S1P

1

R was determined by quantitative PCR

Figure 2. Marginal zone B cells trigger the early anti-CII response A. Splenocytes were separated

into marginal zone B cells (MZB) (CD23

low

and CD1d

high

) and follicular B cells (FOB) (CD23

high

and CD1d

low

) by FACS B. The mean number of IgG anti-CII AFC ± SEM for each cell population was analyzed at different time points after CII- immunization (n = 7-12). C. The expression (mean fluorescence intensity; MFI) of the inhibitory FcγRIIb was analyzed on MZ and FO B cells at different time points in CII-immunized animals (n

= 3 - 6). * p < 0.05, ** p < 0.01, *** p < 0.001

(11)

and demonstrated that the MZ B cells from immunized mice had somewhat higher S1P

1

R mRNA levels than unimmunized animals, however, it did not reach significance (figure 3a).In contrast, similar S1P

1

R mRNA expression was observed on splenic FO B cells from unimmunized and CII-immunized animals (data not shown). These results indicate that CII-immunization may affect S1P

1

R on MZ B cells, likely stimulating the migration of the cells.

Furthermore, we analysed the localization of MZ B cells around the day of onset in the spleen of CII-immunized mice in comparison with unimmunized mice (figure 3b). Spleen sections were stained with MOMA Ab (green) to detect metallophilic macrophages, located on the border between the marginal and follicular zone [26], together with CD3 (yellow) for T cell identification, or B220 (red) and CD9 (blue) for B cell and MZ B cell detection. When comparing sections from unimmunized mice with that of CII-immunized mice it was revealed that the T cell zone in the follicle was smaller in the immunized animals (figure 3b; first row) and that the B cell area inside the follicle was larger (figure 3b; second row). However, the number of B cells located outside the follicle in the MZ was reduced in the immunized animals. By triple staining of MOMA, B220 and CD9 we could localize and identify MZ B cells as they are B220

pos

and CD9

pos

and appeared in pink. The MZ B cells were mainly located in the MZ outside the metallophilic macrophages (green) (figure 3b;

third row), where unimmunized mice displayed a dense area of MZ B cells, while CII- immunized mice had a more dispersed MZ B cell area. In addition, when using a greater magnification it was revealed that MZ B cells also were present inside the follicle, especially in the CII-immunized animals, as these mice appeared to have higher number of MZ B cells inside the follicle compared to the unimmunized mice (figure 3b, bottom row).

However, this finding did not reach statistical significance, possibly due to the large variation

in CIA onset among individual animals (3 - 6 weeks after CII-immunization) (figure S2).

The IgG subclass profiles differ between CII and OVA-immunized animals

It is known that some IgG subclasses are more pathogenic than others and IgG2a in particular have been associated with CIA [27-29].

However, IgG1 and IgG2b Ab can also induce arthritis [2]. Thus, we were interested to investigate which IgG subclasses were generated in mice immunized with CII compared to OVA. Firstly, IgG subclass specific AFC were analyzed by ELISPOT in the spleen, beginning one week after immunization and in the lymph nodes, beginning two weeks after immunization, as the total IgG AFC in the lymph nodes mainly were observed after two weeks. Interestingly, IgG2b anti-CII AFC were the dominating subclass-specific AFC in the spleen of CII- immunized mice (figure 4a), whereas OVA- immunization generated initially IgG3 anti- OVA AFC in the spleen that was replaced by a major enhancement of IgG1 anti-OVA AFC.

The lymph nodes of CII-immunized mice showed comparable percentages of IgG1, IgG2a and IgG2b AFC with a little increase of IgG1 and IgG2b AFC at 2 weeks (Figure 4b).

In OVA-immunized mice IgG1 AFC were the dominating AFC in the lymph nodes (figure 4b).

Secondly, the IgG subclass specific response in

sera of the CII and OVA-immunized mice was

further evaluated. CII-immunization generated

similar levels of IgG1, IgG2a and IgG2b anti-

CII Ab at two weeks after immunization that

increased substantially at four weeks (figure

4c). IgG1 anti-OVA Ab dominated the serum

response in OVA-immunized animals. Thus,

the titre of IgG1 was more than 3 times higher

than that of IgG2a and IgG2b at four weeks

after immunization, although the serum was

diluted 50 times more (figure 4d). IgG3 Ab

were just about detectable in both CII and

OVA-immunized mice (figure 4c, d). These

results show that the pattern of Ag-specific IgG

subclasses in serum seems to parallel the

(12)

Figure 3. Intrafollicular location of marginal zone B cells in CII-immunized mice

A. The relative amount of

S1P

1

R mRNA in sorted MZ B cells from unimmunized (n = 4) and CII-immunized mice (n = 5) two days after immunization.

B. Spleen sections from

unimmunized mice (n = 5) and CII-immunized mice (n

= 4) four weeks after immunization were stained with fluorescently labelled anti-CD3 (yellow), MOMA-1 (green), B220 (red) and CD9 (blue) to visualize B cell follicles and marginal zone B cells (10x magnification). The triple staining of B220, MOMA-1 and CD9 (third row) was further magnified (20x) (bottom row). B220

+

CD9

+

MZ B cells (in pink) are circled.

Representative sections

from one unimmunized and

one CII-immunized mouse

are shown.

(13)

Figure 4. Immunization with CII results in IgG1, IgG2a and IgG2b responses

A-B.

The number of CII- and OVA-specific IgG1, IgG2a, IgG2b and IgG3 AFC were analyzed in mice

immunized with CII or OVA by ELISPOT. Percentage of each subclass in the spleen (A) and in the lymph

nodes (B) of CII- and OVA-immunized mice at different time points is illustrated, with the mean AFC per

million cells shown in each section. C-D. The corresponding mean OD values (± SEM) of Ag-specific IgG

subclasses in the sera of CII-immunized mice (diluted 1:500) (n = 5-10) (C) and in the OVA-immunized mice

(IgG1 diluted 1:12500, IgG2a/2b/3 diluted 1:250) (n = 5-12) (D). Where not visible, error bars are contained

within the symbol.

(14)

pattern of subclass specific AFC in the lymph nodes of CII and OVA-immunized mice, with IgG1, IgG2a and IgG2b dominating CII- immunized mice and IgG1 OVA-immunized mice.

Discussion

CII has an evolutionary conserved structure and immune dominant sequences of bovine and murine CII differ only with single amino acid residue [30]. Thus, immunization with BCII will result in an immune response that is cross reactive to murine CII, which in the CIA model will elicit arthritis. The key finding in this paper is the observation that naïve DBA/1 mice exhibit clones of B cells with reactivity to CII without any previous exposure to the Ag.

Given the evolutionary conserved structure of CII among species, this finding reflects reactivity to self-Ag. One could argue that the response might be unspecific, but the anti-CII response was not seen in the lymph nodes and neither a different bovine protein (BSA) or OVA generated such response in the naïve mice. The self-reactive B cells produced mainly IgM anti-CII, but few B cells showed also IgG anti-CII reactivity. Notably, B cells recognizing CII was not unique to the DBA/1 strain, as two other strains of mice also exhibited self-reactivity. Spleens from naïve NOD and Balb/c mice displayed B cell clones reactive to CII (although somewhat fewer than DBA/1), while C57BL/6 mice had almost no response against CII. This indicates that genetic background influences the natural B cell reactivity to CII. In agreement, natural Ab is independent of internal or external stimuli, whereas genetic differences seem to influence their titres [31, 32].

B cells with a self-reactive BCR have been reported to accumulate in the splenic MZ [33, 34], a site where MZ B cells are positioned.

The MZ B cells are enriched with autoreactive and polyreactive specificities and show for example spontaneous IgM production to

double stranded DNA, a self-Ag in murine lupus [14, 35, 36]. Indeed, when we separated the splenic B cells from naïve DBA/1 mice into FO B cells and MZ B cells we found that the CII reactive B cells belonged to the MZ B cell population. After CII immunization we could observe a rapid and significant expansion of the CII-specific MZ B cells.

Thus, the activation of MZ B cells is probably facilitated by the actual structure of CII, functioning as a thymus-independent type 2 Ag, due to highly repetitive epitopes that may induce maximal cross linking of the BCR [37].

Further, MZ B cells are known to have high basal expression of CD80 and CD86, keeping them in a reactivated state and thereby making them easy to trigger, especially by thymus- independent Ag [8]. Once they have been activated they will produce large quantities of IgM [11]. Indeed, a rapid expansion of IgM anti-CII AFC was particularly observed in the spleen following CII-immunization. Splenic IgG anti-CII AFC were also expanded, but to a lesser degree, and here we noticed that not only MZ B cells but also FO B cells contributed to this response, mainly at 4 weeks after immunization. With regard to the B cell response in the lymph nodes this was somewhat delayed compared to the spleen, but demonstrated the highest numbers of IgG anti- CII AFC. Notably, this was evident at 4 weeks after immunization, a time point when the mice begin to develop CIA. Similarly, the bone marrow exhibited mainly IgG anti-CII AFC at late time points after CII-immunization, likely as a result of immigrating plasma cells.

The B cell response to a self-Ag differed substantially compared to a foreign Ag i.e.

OVA in this case. Thus, CII-immunization generated mainly an early B cell response in the spleen rather in the lymph nodes, while the opposite was observed in OVA-immunized mice, although identical immunization protocols were used. In addition, OVA- immunization generated generally fewer AFC.

This contrasts the observation of higher Ab

levels in sera of mice immunized with OVA

(15)

than with CII. Thus, the anti-OVA response in serum was faster and more potent than the anti- CII response, which was order of magnitude lower. Overall, this suggests that CII- immunization generates more Ag-specific B cells with low Ab production, while OVA- immunization generates fewer Ag-specific B cells, but with high Ab production. One possible explanation to this can be that secretion of self-reactive Ab is much more controlled. Thus, self-reactive B cells seem to be strongly regulated, as indicated by the almost 40% higher expression of the inhibitory FcγRIIb on MZ B cells compared to FO B cells following CII-immunization. High FcγRIIb expression on B cells profoundly reduces IgG responses and FcγRIIb-deficient DBA/1 mice develop more severe arthritis and higher titres of IgG anti-CII Ab than wild type DBA/1 mice [3, 38]. Additionally, MZ B cells display generally a higher degree of complement receptor (CR) 1 and 2 than FO B cells. CR are important for the maintenance of B cell tolerance. In fact, we have previously shown that the lack of CR1 and CR2 increase the incidence of CIA and Prodeus et al showed that CR1- and CR2-deficient lpr/lpr lupus mice exhibit impaired B cell tolerance and Ab against self-Ag [39, 40].

Although CIA is regarded as an IgG-mediated disease, previous studies have shown that IgM also play an important role [41]. Thus, mice deficient in IgM that lack both membrane and secreted IgM develop significantly reduced CIA and interestingly also decreased IgG2a anti-CII Ab, which are considered pathogenic in CIA. This may suggest that the high number of IgM producing AFC and the prolonged serum IgM response observed in CII- immunized mice (but not in OVA-immunized mice) may influence the generation of pathogenic IgG Ab. The IgM anti-CII response is probably induced by MZ B cell plasma blasts in the spleen, known to generate an early wave of IgM towards thymus-independent blood-borne Ag [11]. Furthermore, weak Ag receptor-derived signals (possibly induced here

by low amount of blood-borne CII) favour MZ B-cell generation, whereas relatively strong signals favour the development of mature FO B cells [42].

MZ B cells are potent activators of naïve T cells, which may help in the activation of anergic self-reactive FO B cells [13]. Thus, it has been shown that anergic B cells can be drawn into proliferation if T helper-related signals are present [43, 44]. It is therefore interesting to note that numerous of MZ B cells have left the MZ after CII-immunization and migrated into the follicles, to the border of the T cell zone. The MZ B cells may shuttle CII into the follicle where they either deposit the Ag on follicular dendritic cells [25, 45] and/or present CII to T cells [10, 13]. Indeed, the finding of stimulated S1P

1

R gene expression in the MZ B cells following CII immunization, suggested an increased shuttling. Thus, previous studies have shown that S1P

1

R directs cells to the MZ [24].

Ag-specific T cells activated by MZ B cells have been shown to produce Th1 cytokines generating high IFN-γ levels [13].

Consequently, when MZ B cell-activated T

cells, together with CII-loaded follicular

dendritic cells, stimulate FO B cells the

surrounding cytokine milieu favours a Th1

polarization. Indeed, the general view of CIA

and RA is that they are associated with a strong

Th1 response [27-29]. In agreement, the

majority of the CII-reactive AFC in the spleen

produced IgG2b and IgG2a, while IgG1 was

sparse. Interestingly, the IgG2b response was

evident early and was dominating the response

in the spleen. We have previously shown that

transfer of IgG2b anti-CII monoclonal Ab to

naïve mice trigger arthritis, demonstrating that

IgG2b anti-CII Ab can be pathogenic [2]. In

contrast to the spleen, the AFC in the lymph

node of CII-immunized mice were

predominantly of IgG1, IgG2a and IgG2b type,

with emphasis on IgG1 at 4 weeks. Despite

that IgG1 is regarded as a Th2 associated Ab,

transfer of IgG1 anti-CII to naïve mice induce

arthritis and previous findings in the

(16)

autoimmune K/BxN Ab-mediated arthritis model show that the dominant arthritis inducing subclass is IgG1, indicating the pathogenic potential of IgG1 in arthritis [2, 46]. The quality of the anti-OVA response was on the other hand dominated by IgG1, both in the spleen and in the lymph nodes.

Interestingly, the subclass specific B cell response in the lymph nodes of immunized mice reflected the subclass pattern in the sera.

Thus, IgG1, IgG2a, IgG2b responses were observed both in the lymph nodes and in serum of CII-immunized mice, and OVA-immunized animals displayed IgG1 responses in lymph nodes and in serum.

In conclusion, we have demonstrated that naïve mice display B cell reactivity to CII in the spleen. The response is derived from self- reactive MZ B cells that upon CII- immunization are spontaneously activated.

Based on the findings described, we propose that the MZ B cells direct T-cell responses against CII and the activation of FO B cells, which may give rise to pathogenic IgG anti-CII

Ab. Furthermore, our results show that an autoimmune response is, in contrast to a normal immune response, initiated by B cells in the spleen rather in the lymph nodes.

Acknowledgments

For technical assistance, we would like to thank Dr Jan Grawé at the Cell Analysis Core Facility (Rudbeck Laboratory, Uppsala University) for cell sorting, Julie Moran, (Department of Cell and Molecular Immunology, Uppsala University) for ELISPOT,. Dr Yunying Chen, Fredrik Wermeling and Sara Lind (Karolinska Institute, Solna) for FACS and quantitative PCR and finally Prof. Birgitta Heyman, Dr Magnus Åbrink (Department of Medical Biochemistry and Microbiology, Uppsala University) and Prof. Stellan Sandler (Department of Medical Cell Biology, Uppsala University) for providing the Balb/c, C57BL/6 and NOD mice.

.

(17)

Supplemented data

Figure S1. Discrimination of transitional T1 and T2 B cells before and after anti-CD43 MACS separation

Splenocytes were stained with antibodies against B220, CD1d, CD21, CD23, and IgM before and after anti- CD43 MACS separation. The cells were thereafter separated into CD23 negative and CD23 positive lymphocytes according to size (forward scatter) and CD23 staining in FACS. The CD23 negative and CD23 positive cells were further analyzed in a dot plot using the anti-IgM and anti-CD21 markers to identify MZ B cells (CD23

negative

, IgM

high

, CD21

high

), transitional T1 (CD23

negative

, IgM

high

, CD21

low

) and transitional T2 (CD23

positive

, IgM

high

CD21

high

) B cells. MZ B cells, T1, T2 B cells and FO B cells were thereafter identified among B220 positive B cells in a dot plot, showing CD1d and CD23 expression (A, B). Before anti-CD43 MACS separation (A) T1 B cells (CD1d

intermediate

, CD23

low

)(circled in red) were situated down from the MZ B cell population (CD1d

high

, CD23

low

), while T2 B cells (CD1d

intermediate

, CD23

high

) were located within the FO B cell population (CD1d

intermediate

, CD23

high

). After anti-CD43 MACS separation (B) the T1 B cells were almost completely lost, while the T2 B cells were still present in the FO B cell population.

Figure S2. Incidence and severity of CIA

DBA/1 mice of both sexes were immunized with 50 µg of CII in CFA. The arthritis development is presented

as incidence (A) i.e. the number of diseased mice out of total number of immunized mice and the mean arthritic

score (B) i.e. the severity of arthritis in diseased mice only. The graph represents pooled data from two

experiments (n = 11 - 17).

(18)

Reference:

1. Wooley, P.H., Luthra, H.S., Stuart, J.M., David, C.S. 1981. Type II collagen- induced arthritis in mice. I. Major histocompatibility complex (I region) linkage and antibody correlates. J Exp

Med, 154: 688-700.

2. Nandakumar, K.S., Andren, M., Martinsson, P., Bajtner, E., Hellstrom, S., Holmdahl, R., Kleinau, S. 2003. Induction of arthritis by single monoclonal IgG anti- collagen type II antibodies and enhancement of arthritis in mice lacking inhibitory FcgammaRIIB. Eur J Immunol,

33: 2269-77.

3. Kleinau, S., Martinsson, P., Heyman, B.

2000. Induction and suppression of collagen-induced arthritis is dependent on distinct fcgamma receptors. J Exp Med,

191: 1611-6.

4. Svensson, L., Jirholt, J., Holmdahl, R., Jansson, L. 1998. B cell-deficient mice do not develop type II collagen-induced arthritis (CIA). Clin Exp Immunol, 111:

521-6.

5. Diaz de Stahl, T., Andren, M., Martinsson, P., Verbeek, J.S., Kleinau, S. 2002.

Expression of FcgammaRIII is required for development of collagen-induced arthritis.

Eur J Immunol, 32: 2915-22.

6. Cambridge, G., Leandro, M.J., Edwards, J.C., Ehrenstein, M.R., Salden, M., Bodman-Smith, M., Webster, A.D. 2003.

Serologic changes following B lymphocyte depletion therapy for rheumatoid arthritis.

Arthritis Rheum, 48: 2146-54.

7. Rantapaa-Dahlqvist, S., de Jong, B.A., Berglin, E., Hallmans, G., Wadell, G., Stenlund, H., Sundin, U., van Venrooij, W.J. 2003. Antibodies against cyclic citrullinated peptide and IgA rheumatoid factor predict the development of rheumatoid arthritis. Arthritis Rheum, 48:

2741-9.

8. Martin, F., Kearney, J.F. 2002. Marginal- zone B cells. Nat Rev Immunol, 2: 323-35.

9. Oliver, A.M., Martin, F., Gartland, G.L., Carter, R.H., Kearney, J.F. 1997. Marginal zone B cells exhibit unique activation, proliferative and immunoglobulin secretory responses. Eur J Immunol, 27: 2366-74.

10. Oliver, A.M., Martin, F., Kearney, J.F.

1999. IgMhighCD21high lymphocytes

enriched in the splenic marginal zone generate effector cells more rapidly than the bulk of follicular B cells. J Immunol,

162: 7198-207.

11. Martin, F., Oliver, A.M., Kearney, J.F.

2001. Marginal zone and B1 B cells unite in the early response against T-independent blood-borne particulate antigens. Immunity,

14: 617-29.

12. Cesta, M.F. 2006. Normal structure, function, and histology of the spleen.

Toxicol Pathol, 34: 455-65.

13. Attanavanich, K., Kearney, J.F. 2004.

Marginal zone, but not follicular B cells, are potent activators of naive CD4 T cells.

J Immunol, 172: 803-11.

14. Goodyear, C.S., Silverman, G.J. 2005. B cell superantigens: a microbe's answer to innate-like B cells and natural antibodies.

Springer Semin Immunopathol, 26: 463-84.

15. Manson, J.J., Mauri, C., Ehrenstein, M.R.

2005. Natural serum IgM maintains immunological homeostasis and prevents autoimmunity.

Springer Semin Immunopathol, 26: 425-32.

16. Holers, V.M., Kulik, L. 2007. Complement receptor 2, natural antibodies and innate immunity: Inter-relationships in B cell selection and activation. Mol Immunol, 44:

64-72.

17. Rolf, J., Motta, V., Duarte, N., Lundholm, M., Berntman, E., Bergman, M.L., Sorokin, L., Cardell, S.L., Holmberg, D.

2005. The enlarged population of marginal zone/CD1d(high) B lymphocytes in nonobese diabetic mice maps to diabetes susceptibility region Idd11. J Immunol,

174: 4821-7.

18. Grimaldi, C.M., Michael, D.J., Diamond, B. 2001. Cutting edge: expansion and activation of a population of autoreactive marginal zone B cells in a model of estrogen-induced lupus. J Immunol, 167:

1886-90.

19. Wither, J.E., Loh, C., Lajoie, G., Heinrichs, S., Cai, Y.C., Bonventi, G., MacLeod, R.

2005. Colocalization of expansion of the splenic marginal zone population with abnormal B cell activation and autoantibody production in B6 mice with an introgressed New Zealand Black chromosome 13 interval. J Immunol, 175:

4309-19.

(19)

20. Duan, B., Niu, H., Xu, Z., Sharpe, A.H., Croker, B.P., Sobel, E.S., Morel, L. 2008.

Intrafollicular location of marginal zone/CD1d(hi) B cells is associated with autoimmune pathology in a mouse model of lupus. Lab Invest, 88: 1008-20.

21. Miller, E.J. 1972. Structural studies on cartilage collagen employing limited cleavage and solubilization with pepsin.

Biochemistry, 11: 4903-9.

22. Magnusson, S.E., Andren, M., Nilsson, K.E., Sondermann, P., Jacob, U., Kleinau, S. 2008. Amelioration of collagen-induced arthritis by human recombinant soluble FcgammaRIIb. Clin Immunol, 127: 225-33.

23. Carsetti, R. 2004. Characterization of B- cell maturation in the peripheral immune system. Methods Mol Biol, 271: 25-35.

24. Cinamon, G., Matloubian, M., Lesneski, M.J., Xu, Y., Low, C., Lu, T., Proia, R.L., Cyster, J.G. 2004. Sphingosine 1- phosphate receptor 1 promotes B cell localization in the splenic marginal zone.

Nat Immunol, 5: 713-20.

25. Cinamon, G., Zachariah, M.A., Lam, O.M., Foss, F.W., Jr., Cyster, J.G. 2008.

Follicular shuttling of marginal zone B cells facilitates antigen transport. Nat

Immunol, 9: 54-62.

26. Kraal, G., Janse, M. 1986. Marginal metallophilic cells of the mouse spleen identified by a monoclonal antibody.

Immunology, 58: 665-9.

27. Anthony, D.D., Haqqi, T.M. 1999.

Collagen-induced arthritis in mice: an animal model to study the pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol,

17: 240-4.

28. Mauri, C., Williams, R.O., Walmsley, M., Feldmann, M. 1996. Relationship between Th1/Th2 cytokine patterns and the arthritogenic response in collagen-induced arthritis. Eur J Immunol, 26: 1511-8.

29. Okamoto, Y., Gotoh, Y., Tokui, H., Mizuno, A., Kobayashi, Y., Nishida, M.

2000. Characterization of the cytokine network at a single cell level in mice with collagen-induced arthritis using a dual color ELISPOT assay. J Interferon

Cytokine Res, 20: 55-61.

30. Sakurai, Y., Tang, B., Rosloniec, E.F., Stuart, J.M., Kang, A.H., Myers, L.K.

2006. Molecular characterization of an

arthritogenic collagen peptide interacting with I-Ar. Immunology, 117: 136-42.

31. Bos, N.A., Kimura, H., Meeuwsen, C.G., De Visser, H., Hazenberg, M.P., Wostmann, B.S., Pleasants, J.R., Benner, R., Marcus, D.M. 1989. Serum immunoglobulin levels and naturally occurring antibodies against carbohydrate antigens in germ-free BALB/c mice fed chemically defined ultrafiltered diet. Eur J

Immunol, 19: 2335-9.

32. Gobet, R., Cerny, A., Ruedi, E., Hengartner, H., Zinkernagel, R.M. 1988.

The role of antibodies in natural and acquired resistance of mice to vesicular stomatitis virus. Exp Cell Biol, 56: 175-80.

33. Tighe, H., Heaphy, P., Baird, S., Weigle, W.O., Carson, D.A. 1995. Human immunoglobulin (IgG) induced deletion of IgM rheumatoid factor B cells in transgenic mice. J Exp Med, 181: 599-606.

34. Li, Y., Li, H., Weigert, M. 2002.

Autoreactive B cells in the marginal zone that express dual receptors. J Exp Med,

195: 181-8.

35. Zeng, D., Lee, M.K., Tung, J., Brendolan, A., Strober, S. 2000. Cutting edge: a role for CD1 in the pathogenesis of lupus in NZB/NZW mice. J Immunol, 164: 5000-4.

36. Takahashi, T., Strober, S. 2008. Natural killer T cells and innate immune B cells from lupus-prone NZB/W mice interact to generate IgM and IgG autoantibodies. Eur

J Immunol, 38: 156-65.

37. Prockop, D.J., Kivirikko, K.I. 1995.

Collagens: molecular biology, diseases, and potentials for therapy. Annu Rev

Biochem, 64: 403-34.

38. Brownlie, R.J., Lawlor, K.E., Niederer, H.A., Cutler, A.J., Xiang, Z., Clatworthy, M.R., Floto, R.A., Greaves, D.R., Lyons, P.A., Smith, K.G. 2008. Distinct cell- specific control of autoimmunity and infection by FcgammaRIIb. J Exp Med,

205: 883-95.

39. Nilsson, K.E., Andren, M., de Stahl, T.D., Kleinau, S. 2009. Enhanced susceptibility to low-dose collagen-induced arthritis in CR1/2-deficient female mice--possible role of estrogen on CR1 expression. FASEB J.

40. Prodeus, A.P., Goerg, S., Shen, L.M.,

Pozdnyakova, O.O., Chu, L., Alicot, E.M.,

Goodnow, C.C., Carroll, M.C. 1998. A

(20)

critical role for complement in maintenance of self-tolerance. Immunity, 9:

721-31.

41. Zheng, B., Zhang, X., Guo, L., Han, S.

2007. IgM plays an important role in induction of collagen-induced arthritis.

Clin Exp Immunol, 149: 579-85.

42. Pillai, S., Cariappa, A., Moran, S.T. 2004.

Positive selection and lineage commitment during peripheral B-lymphocyte development. Immunol Rev, 197: 206-18.

43. Noorchashm, H., Bui, A., Li, H.L., Eaton, A., Mandik-Nayak, L., Sokol, C., Potts, K.M., Pure, E., Erikson, J. 1999.

Characterization of anergic anti-DNA B cells: B cell anergy is a T cell-independent and potentially reversible process. Int

Immunol, 11: 765-76.

44. Seo, S.J., Fields, M.L., Buckler, J.L., Reed, A.J., Mandik-Nayak, L., Nish, S.A., Noelle, R.J., Turka, L.A., Finkelman, F.D., Caton, A.J. et al. 2002. The impact of T helper and T regulatory cells on the regulation of anti-double-stranded DNA B cells. Immunity, 16: 535-46.

45. Ferguson, A.R., Youd, M.E., Corley, R.B.

2004. Marginal zone B cells transport and deposit IgM-containing immune complexes onto follicular dendritic cells. Int Immunol,

16: 1411-22.

46. Maccioni, M., Zeder-Lutz, G., Huang, H., Ebel, C., Gerber, P., Hergueux, J., Marchal, P., Duchatelle, V., Degott, C., van Regenmortel, M. et al. 2002. Arthritogenic monoclonal antibodies from K/BxN mice.

J Exp Med, 195: 1071-7.

References

Related documents

Den förbättrade tillgängligheten berör framför allt boende i områden med en mycket hög eller hög tillgänglighet till tätorter, men även antalet personer med längre än

På många små orter i gles- och landsbygder, där varken några nya apotek eller försälj- ningsställen för receptfria läkemedel har tillkommit, är nätet av

The government formally announced on April 28 that it will seek a 15 percent across-the- board reduction in summer power consumption, a step back from its initial plan to seek a

Indien, ett land med 1,2 miljarder invånare där 65 procent av befolkningen är under 30 år står inför stora utmaningar vad gäller kvaliteten på, och tillgången till,

Det finns många initiativ och aktiviteter för att främja och stärka internationellt samarbete bland forskare och studenter, de flesta på initiativ av och med budget från departementet

Den här utvecklingen, att både Kina och Indien satsar för att öka antalet kliniska pröv- ningar kan potentiellt sett bidra till att minska antalet kliniska prövningar i Sverige.. Men

Av 2012 års danska handlingsplan för Indien framgår att det finns en ambition att även ingå ett samförståndsavtal avseende högre utbildning vilket skulle främja utbildnings-,

Det är detta som Tyskland så effektivt lyckats med genom högnivåmöten där samarbeten inom forskning och innovation leder till förbättrade möjligheter för tyska företag i