ArticlePDF Available

Predominant selection of T cells specific for the glycosylated collagen type II epitope (263–270) in humanized transgenic mice and in rheumatoid arthritis

Authors:

Abstract and Figures

Rheumatoid arthritis (RA) is associated with certain MHC class II alleles and is characterized by a chronic autoimmune response in the joints. Using transgenic mice expressing human DR4 (DRB1*0401) and human CD4, but lacking endogenous MHC class II, we show that posttranslational glycosylation of type II collagen (CII) influences the level of T cell tolerance to this candidate cartilage-specific autoantigen. In such mice, the expression of human CII resulted in a tolerized murine T cell response to human CII. However, tolerance induction remained incomplete, preferentially deleting responses to the nonmodified CII 263-270 epitope, whereas T cell recognition of a glycosylated variant of this epitope was affected to a lesser degree. A similar dominance of T cell responses to CII-glycopeptides was recorded in a cohort of severely affected RA-patients (n = 14). Thus, RA T cells predominantly recognize the immunodominant CII peptide in its glycosylated form and may explain why previously it has been difficult to detect T cell responses to CII in RA patients.
Content may be subject to copyright.
Predominant selection of T cells specific for the
glycosylated collagen type II epitope (263–270)
in humanized transgenic mice and in
rheumatoid arthritis
Johan Ba
¨cklund
†‡
, Stefan Carlsen
†‡
, Torsten Ho
¨ger
§
, Bjo
¨rn Holm
, Lars Fugger
, Jan Kihlberg
, Harald Burkhardt
§
,
and Rikard Holmdahl
**
Section of Medical Inflammation Research, So¨ lvegatan 19, I11 BMC, Lund University, SE-221 84 Lund, Sweden; §Department of Internal Medicine III and
Institute of Clinical Immunology, Friedrich-Alexander-University Erlangen-Nu¨ rnberg, D-91054 Erlangen, Germany; Department of Chemistry, Umeå
University, SE-901 87 Umeå, Sweden; and Department of Clinical Immunology, Aarhus University Hospital, Skejby Sygehus, 8200N Aarhus, Denmark
Communicated by N. Avrion Mitchison, University College London, London, United Kingdom, April 29, 2002 (received for review February 25, 2002)
Rheumatoid arthritis (RA) is associated with certain MHC class II
alleles and is characterized by a chronic autoimmune response
in the joints. Using transgenic mice expressing human DR4
(DRB1*0401) and human CD4, but lacking endogenous MHC class
II, we show that posttranslational glycosylation of type II collagen
(CII) influences the level of T cell tolerance to this candidate
cartilage-specific autoantigen. In such mice, the expression of
human CII resulted in a tolerized murine T cell response to human
CII. However, tolerance induction remained incomplete, preferen-
tially deleting responses to the nonmodified CII 263–270 epitope,
whereas T cell recognition of a glycosylated variant of this epitope
was affected to a lesser degree. A similar dominance of T cell
responses to CII-glycopeptides was recorded in a cohort of severely
affected RA-patients (n14). Thus, RA T cells predominantly
recognize the immunodominant CII peptide in its glycosylated form
and may explain why previously it has been difficult to detect T cell
responses to CII in RA patients.
Rheumatoid arthritis (RA) is an autoimmune disease that
primarily affects peripheral joints with cartilage destruction
and subsequent bone erosion. The role of T cells in RA is
supported by the large number of activated CD4
cells reported
in the synovium of affected joints and by the association of RA
to certain MHC (HLA) class II genes [e.g., DRB1*0401 (in DR4)
and DRB1*0101 (in DR1)] that encode a specific peptide
binding pocket, the so-called shared epitope.
Collagen type II (CII), the main constituent of hyaline carti-
lage, has been proposed as one possible autoantigen in RA
because CII-specific antibodies are frequently found in RA
patients and because an RA-like disease can be induced in
certain mouse strains after immunization with CII. However,
reports on T cell immunity to CII in RA patients as well as in
healthy individuals are inconclusive, and the role of CII, and
even T cells, in RA is still argued and remains to be proven.
Previous studies perfor med in DR4- and DR1-expressing mice
have located the immunodominant T cell epitope to position
263–270 in CII by using synthetic peptides (1–3), but studies in
RA patients have in general failed to identify DR4DR1-
restricted T cells specific for the same epitope (4, 5). One
potential reason for this failure is that these studies did not
directly address that CII can become posttranslationally modi-
fied. Within CII263–270, the lysines at positions 264 and 270 can
be hydroxylated and further glycosylated with mono- or disac-
charides, i.e., with a
-D-galactopyranosyl or an
-D-
glucopyranosyl-(1-2)-
-D-galactopyranosyl residue. Such modi-
fications have previously been shown to be of importance in the
development of collagen-induced arthritis (CIA) in A
q
-
expressing mice (6, 7). Although the A
q
-molecule is of a DQ
isotype, its peptide-binding groove shows more similarity to the
shared epitope variant of the DR4-molecule, because it binds
almost the same CII epitope and presents the same amino acid
side chains to T cells (3, 8–11). Furthermore, CIA is most
commonly induced with heterologous CII, which is believed to
induce a heteroreactive T cell response, followed by a B cell
response, which in contrast to the T cell response is highly
crossreactive to mouse CII (12, 13). Thus, earlier CIA experi-
ments in DR4-expressing mice have not addressed T cell toler-
ance to self-CII. Therefore, to get a better understanding of
autoimmunity to cartilage-derived proteins, we need to consider
both posttranslational modifications and the aspect of T cell
tolerance in these models. To achieve this result, we used a
humanized mouse model expressing HLA-DRB1*0401
DRA1*0101, human CD4, and human CII (huCII) on a back-
ground deficient of murine class II expression (14, 15). In these
mice, T cell responses to huCII peptides and the impact of
posttranslational modification on the induction of CII-specific T
cell tolerance was investigated.
Materials and Methods
Mice. Crossing of different transgenic mice generated two groups of
mice that were used for experiments: (i) DR4, expressing transgenic
DRB*0401 and human CD4 but no murine class II molecule; and
(ii) huCIIDR4, expressing DRB*0401, human CD4 and huCII but
no murine class II molecule. To get these groups, huCII transgenic
mice (15), mice expressing HLA-DR4 together with human CD4
(14), and mice lacking H-2 class II (16) were crossed as follows.
First, HLA-DR4huCD4 on a B10 background was introduced
with H-2
plus/
on a B6 background and backcrossed twice to B10
and subsequently intercrossed. HLA-DR4huCD4, H-2
/
mice
were then crossed with huCII mice on a C3H background. Off-
spring were backcrossed three generations to HLA-DR4huCD4,
H-2
/
on the B10 background. Finally, mice were intercrossed
twice and selected for homozygosity in H-2
/
. Mice were bred and
kept in the animal facility of Medical Inflammation Research
(http:兾兾net.inflam.lu.se).
Antigens and
in Vitro
T Cell Assays. HuCII was extracted from hip
joints (obtained from replacement surgery) after pepsin diges-
tion and purified as described (17). The following nonmodified
and glycosylated CII peptides were synthesized as described (8,
18, 19): K264270 (nonmodified CII261–275 with a lysine resi-
due at position 264 and 270); Gal264 (CII261–278, glycosylated
Abbreviations: CII, type II collagen; CIA, collagen-induced arthritis; huCII, human type II
collagen; PBMC, peripheral blood mononucleated cells; RA, rheumatoid arthritis; mDR, T
cell clones from mouse CII and DR4; hDR, T cell clones from huCIIDR4.
See commentary on page 9611.
J.B. and S.C. contributed equally to the present work.
**To whom reprint requests should be addressed. E-mail: rikard.holmdahl@inflam.lu.se.
9960–9965
PNAS
July 23, 2002
vol. 99
no. 15 www.pnas.orgcgidoi10.1073pnas.132254199
with a
-D-galactopyranose residue on L-hydroxylysine exclu-
sively at position 264); Gal270 (CII261278, glycosylated exclu-
sively at position 270); and Gal264270 (CII259278, glycosy-
lated at position 264 and 270). The following modified forms of
the Gal264-glycopeptide, named deoxy-glycopeptides, were also
synthesized (B.H., unpublished data): 2-deoxyGal (CII259273,
where the hydroxy-group at position 2 on the
-D-galactopyr-
anosyl moiety has been eliminated); 3-deoxyGal (CII259 273,
missing the hydroxy-group at position 3 on galactose); and
4-deoxyGal (CII259273, missing the hydroxy-group at position
4 on galactose). As control for the deoxy-peptides, a shortened
form of the Gal264-peptide (CII259273) was used. Mice were
immunized with huCII in complete Freunds adjuvant, and 10
days later cells from draining lymph nodes were stimulated in
vitro for determination of antigen-specific proliferation and
IFN-
production as described (20). Establishment of T cell
hybridoma clones and determination of antigen specificity was
performed as described (8), with the exception that Gal264
270-peptide was used for immunization and for the first in vitro
restimulation (20
gml).
Analysis of Human T Cell Responses. Fourteen patients fulfilling the
RA-classification criteria of the American College of Rheuma-
tology (21) were recruited for the study. The study protocol was
approved by the review board of the Friedrich-Alexander-
University Erlangen-Nuremberg, and informed consent was
obtained from all individuals before entering the study. All
patients had a severe course of the disease so that insufficient
response to conventional therapy required an intensified treat-
ment with TNF
-blocking antibodies [D2E7 (Knoll AG-BASF
Pharma, Ludwigshafen, Germany) or infliximab (Essex Pharma,
Munich, Germany]. The mean age of the patients (2 male and
12 female) was 63.6 6.5 (SD) years, and the disease duration
10.9 8.1 (SD) years. In 9 patients, information about the
expression of HLA-DRB1*-alleles was available and is included
in the result section. Peripheral blood mononucleated cells
(PBMC) from the patients were separated by using Histoprep
(BAG, Lich, Germany). For antigenic stimulation of 10
6
PBMC,
10
g of CII-peptide and 1
g anti-human CD28 (Becton
Dickinson) were added per ml of culture medium. T cell
receptor-specific responses were controlled in parallel by using
culture conditions that either omitted any stimulation or only
exposed the cells to the costimulatory anti-CD28 antibody
overnight in the absence of antigen. T cell responsiveness to a
common recall antigen was tested in parallel cultures of PBMC
by using 10
gml tetanus toxoid (Calbiochem-Nova Biochem)
and anti-CD28 for stimulation. Monensin (2.5 mM; Sigma-
Aldrich) was added to the overnight cultures, and the cells were
incubated for additional 4 h before harvesting. Subsequently, the
cells were washed twice in PBS and fixed in 4% paraformalde-
hydePBS solution for 7 min at 37°C, followed by a repeated
washing procedure in PBS. A permeabilization step was per-
formed for 10 min with 0.5% saponin1% BSA0.1% NaN
3
in
PBS; afterwards, the cells were washed twice with PBS1% BSA.
Cells were stained with 0.2
g rat anti-human IL-2-PE (Becton
Dickinson) and 3
l CD4-FITC or CD3-FITC (Beckman
Coulter) for 20 min at 4°C. Fluorescence intensities were deter-
mined by using a Coulter Epics XL-MCL flow cytometer and
SYSTEM-II software. Large activated lymphocytes (blasts) were
gated according to forward and side scatter as described previ-
ously (22, 23). Cells not treated with saponin were used to
exclude background staining of anti-IL-2 antibody.
Results
Strong, but Incomplete, Negative Selection of the Autoreactive T Cell
Population. Previous studies have identified the immunodomi-
nant T cell epitope in bovine and human CII in DR4-transgenic
mice, and they have also shown that these mice are susceptible
to CIA (13). However, because bovine and human CII both
differ from mouse CII within the identified epitope (at position
266, glutamic acid in heterologous CII, compared with aspartic
acid in mouse CII), these reports did not include the aspect of
T cell tolerance to self-CII. To generate an animal model of RA
that would reflect the situation in humans more accurately, we
crossed DR4human CD4-transgenic and mouse MHC class
II-deficient mice with huCII-transgenic mice to generate two
lines of mice: DR4 mice (DR4human CD4-transgenic and
mouse MHC class II-deficient mice, expressing mouse CII in
cartilage) and huCIIDR4 mice (DR4human CD4-transgenic
and mouse MHC class II-deficient mice, expressing huCII in
cartilage). These mice were then immunized with huCII to
examine the degree of T cell tolerance to endogenously ex-
pressed CII.
The heterologous immune response in DR4 mice to CII was
biased toward the nonmodified peptide (K264270), followed by
a weaker response to peptides glycosylated with
-D-
galactopyranosyl moieties on hydroxylysine residues at either
position 264 (Gal264) or 270 (Gal270) (Fig. 1). The response to
huCII protein was also weak but significantly above background
level when measuring the production of IFN-
. In sharp contrast,
the response in huCIIDR4 mice was severely reduced against all
CII antigens as compared with the response in DR4 mice,
showing that strong tolerance to self-CII is present (Pvalue less
than 0.05 against all CII antigens except the Gal264270-specific
IFN-
production). Most importantly, however, tolerance was
not complete because huCIIDR4 mice were able to mount a
significant response above background level against the Gal264-
peptide, when the proliferation (P0.023) as well as IFN-
production (P0.041) was measured (Fig. 1). In contrast, the
response against the nonmodified K-peptide in huCIIDR4 mice
was not significant above the background response (P0.05,
Fig. 1).
To further evaluate the shift in epitope selection between DR4
mice (with a dominant response to the K264270-peptide) and
huCIIDR4 mice (with a significant response against the
Fig. 1. Strong but incomplete tolerance to glycosylated CII in humanized
mice. Recall in vitro response of lymph node cells from DR4 and huCIIDR4
mice immunized 10 days earlier with human CII (huCII). Cells were restimu-
lated with huCII and the following CII-peptides: nonglycosylated (K264270),
glycosylated at hydroxylysine 264 (Gal264), glycosylated at hydroxylysine 270
(Gal270), or glycosylated at both hydroxylysine residues (Gal264270) with
-D-galactopyranosyl residues. Ten animals (5 males and 5 females) of each
mouse line were investigated. As a positive control, the recall response to
mycobacteria antigen PPD (puried protein derivate, present in complete
Freunds adjuvant) was measured. The response in huCIIDR4 mice was sig-
nicantly reduced against all CII antigens (P0.05) as compared with DR4
mice, except for Gal264270-specic IFN-
production. *, Signicant responses
above background levels for huCIIDR4 mice. *,P0.05, MannWhitney U
test; bars represent mean SEM. Antigen-specic proliferation and IFN-
production: cpm and Uml respectively (response with antigenresponse
in the absence of antigen).
Ba¨cklund et al. PNAS
July 23, 2002
vol. 99
no. 15
9961
IMMUNOLOGY
Gal264-peptide alone), we made a pairwise comparison of the
response to the K- and the Gal264-peptide (Fig. 2). The response
to the K-peptide was stronger than to the Gal264-peptide in all
DR4 mice at both concentrations tested (10 and 50
gml, P
0.0051). In contrast, a relatively stronger response against the
Gal264-peptide than to the K-peptide was noted in 8 of 10
huCIIDR4 mice when cells were stimulated with the higher
antigen concentration (P0.0218) and in all mice when
stimulated with the lower antigen concentration (P0.0051).
Hence, CII-specific T cells are strongly tolerized in DR4 mice
expressing huCII in cartilage. However, glycopeptide-specific T
cells appear to be less affected by tolerance than T cells specific
for the nonmodified epitope.
Different T Cell Fine Specificity of Auto- and Heteroreactive T Cell
Clones. To confirm our finding that DR4-restricted CII recog-
nition includes T cells specific for glycosylated epitopes, hybrid-
oma clones from Gal264270-peptide-immunized DR4 and hu-
CIIDR4 mice were established and characterized for
comparison. Despite the weak in vitro response to glycosylated
CII antigens seen earlier in DR4 and huCIIDR4 mice after CII
immunization, glycopeptide-specific hybridomas were success-
fully established from both lines of transgenic mice (Table 1).
Specific recognition of the glycopeptide was evident because
all clones responded to the Gal264270-peptide but not to the
nonglycosylated peptide. The majority of T cell clones from DR4
mice (named mDR for mouse CII and DR4) recognized the
Gal264-peptide (8 of 10, group I and II), whereas only 2 clones,
originally collected from the same subcloning, specifically re-
sponded to the Gal270-peptide (group III). Gal264-specific
clones could further be divided in two groups where clones that
displayed a higher sensitivity to the glycopeptide also had a
strong response toward huCII (group I), whereas clones that
responded weakly to the Gal264-peptide also responded weakly
to huCII (group II). In line with this finding, the Gal270-specific
clone(s) responded weakly to the Gal270-peptide and not at all
to huCII.
Interestingly, T cell clones from huCIIDR4 mice (named
hDR, for huCII and DR4) differed from mDR clones in their
response to CII-peptides: (i) The biased recognition of the
Gal264-peptide was not observed for hDR-clones (Table 1).
(ii) Among the Gal264-specific clones (group I and II), the
response was somewhat weaker, compared with the correspond-
ing mDR clones. This difference in antigen sensitivity was even
more evident in the response to intact huCII (Fig. 3 and Table
1). (iii) Gal270-specific hDR clones also responded to huCII
(group III), although high antigen concentration was required
(Table 1).
To confirm that it was the posttranslational modification of
Fig. 2. Comparative analysis of the individual response to the nonmodied
and the galactosylated T cell epitope in DR4 and huCIIDR4 mice. Recall in vitro
response of lymph node cells from DR4 and huCIIDR4 mice immunized 10
days earlier with human CII. Cells were restimulated with either 10 or 50
gml
of the K264270- or the Gal264-peptide. A ratio above 1 indicates a stronger
response against the K264270-peptide whereas a ratio below 1 indicates a
stronger response to the Gal264-peptide. The response of individual DR4 mice
was stronger to the K264-peptide whereas the responses of huCIIDR mice
were stronger to the Gal264-peptide (Wilcoxon signed rank test).
Table 1. DR4-restricted T cell hybridoma responses to the CII259273 epitope and human CII
Group Clone
Glycopeptide specicity Galactose ne-specicity
K264270* Gal264* Gal270* Gal264270* huCII
Gal264* 2-deoxyGal* 3-deoxyGal* 4-deoxyGal*
I mDR-1.1 0 5 0 5 5 5 0 4 0
mDR-2.2 0 4 0 4 5 4 0 3 0
mDR-8.4 0 4 0 4 5 4 0 4 0
mDR-14.1 0 5 0 5 6 5 0 4 0
mDR-16.2 0 5 0 5 6 4 0 3 0
II mDR-6.3 0 3 0 4 1 3 0 0 0
mDR-15.2 0 3 0 3 1 2 0 0 0
mDR-17.1 0 5 0 3 3 5 0 0 0
III mDR-4.1 0 0 3 3 0
mDR-4.2 0 0 3 3 0
I hDR-2.3 0 4 0 2 1 4 0 0 4
hDR-9.1 0 4 0 2 2 3 0 1 3
hDR-11.2 0 4 0 2 1 4 0 1 4
II hDR-13.1 0 3 0 4 0
hDR-13.2 0 3 0 3 0 2 0 4 0
III hDR-1.1 0 0 4 4 2
hDR-4.5 0 0 4 4 2
hDR-6.1 0 0 4 3 1
hDR-12.3 0 0 4 4 1
IV hDR-3.3 0 0 0 4 0
Semiquantitative scoring of T cell hybridoma response following stimulation with glycosylated or nonglycosylated collagen peptides (see Fig. 1). Shaded boxes
highlight specic responses for the different groups of clones.
*Sensitivity of the T cell hybridomas was determined by the amount of peptide required for a CTLL response 1000 CPM above background; 0, no response; 1
50
gml; 2 10
gml; 3 2
gml; 4 0.4
gml; 5 0.08
gml; 6 0.016
gml.
Sensitivity of the T cell hybridomas to CII protein was determined as for the CII-peptides, but number indicates a 5 times higher concentration of antigen, i.e.,
1250
gml; 2 50
gml; 3 10
gml; 4 2
gml; 5 0.4
gml; 6 0.08
gml.
9962
www.pnas.orgcgidoi10.1073pnas.132254199 Ba¨cklund et al.
CII that was specifically recognized by the obtained clones,
Gal264-specific hybridomas were also tested against three dif-
ferently modified glycopeptides, where one of the hydroxy-
groups on the galactose moiety had been selectively removed at
either position 2, 3, or 4, thereby generating three mono-deoxy
glycopeptides. The two groups among the Gal264-specific mDR
clones were confirmed in that they, apart from having different
sensitivity to the Gal264-peptide, also recognized the sugar
residue differently (Table 1). Similarly, the two groups of
Gal264-specific hDR clones were also confirmed by the use of
the deoxy-glycopeptides. Notably, differences in deoxy-
glycopeptide responses between mDR and hDR clones indicated
that T cells were differently selected if huCII was expressed. For
example, all mDR clones were stringently dependent on the
hydroxy-group at the C4-carbon of galactose, whereas the re-
sponses of 3 of 4 hDR clones did not depend on the C4-hydroxy-
group.
Glycopeptide-Specific T Cells Dominate the CII Response in RA Pa-
tients. Thus far, results from our humanized animal model
showed that T cells specifically recognizing the glycostructure
remain after tolerance induction and become dominant by the
introduction of autologous huCII, whereas T cells with other fine
specificities to the CII epitope seem to be functionally impaired
or deleted to a greater extent. We argued that these findings
could explain earlier reported difficulties in identifying CII
specific T cell clones from joints or blood of RA patients, and
therefore we tested a total number of 14 RA patients for T cell
responses against the different glycosylated peptides. In the
investigated cohort of RA patients, cytokine flow cytometry of
in vitro-stimulated PBMC revealed specific IL-2 responses to the
glycosylated CII-peptides. Using the increase in the percentage
of IL-2-producing cells within the CD3 or CD4 positive T cell
population above baseline of negative control as a parameter of
specific T cell stimulation, 7 patients could be identified as
CII-peptide responsive (Table 2). Interestingly, all CII-
responding patients recognized the glycopeptides, but only 2 of
these responded also to the nonglycosylated peptide. This result
shows that T cell recognition was confined to the glycosylated
CII-variants in 5 patients, representing more than 30% of the
entire cohort. With one exception (patient 10) the baseline
population of IL-2-producing T cells was low in all patients
investigated (Table 2). However, also in patient 10, a specific rise
in the percentage of IL-2-producing CD4
T cells from 0.20 to
0.92% on stimulation with Gal264270 was detectable. The
individual glycospecific responses of patients were heteroge-
nous. In some patients, the response was biased to one of the
glycosylation sites, whereas other patients had equal responses to
both sites. In addition, in some patients, the response was
restricted to one of the glycosylation sites, whereas the response
to the peptide with glycosylation at both positions was weak or
nonexistent. This latter response pattern was also seen in mice
expressing both human DR4 and human CII (see Fig. 1). We also
Fig. 3. An example of different responses to glycosylated CII between DR4
and huCIIDR4 mice. T cell hybridomas clones mDR-16.2 and hDR-9.1 were
obtained from DR4 and huCIIDR4 mice, respectively, after immunization with
the Gal264270-peptide. Cells were stimulated with titrated amounts of huCII
or CII-peptides (see Fig. 1) and investigated for production of IL-2.
Table 2. Analysis of T cell recognition of CII-peptide 259273 in RA patients
Patient
no. No antigen* TT* K264270* Gal264* Gal270* Gal264270* HLA
CII-peptide
response
FACS staining
1 0.04 0.42 0.05 0.02 0.03 0 ND CD3IL-2
2 0.03 0.07 0.06 0.04 0.04 0.03 ND CD3IL-2
3
§
0.07 1.35 0.09 0.24 0 0.12 DRB1*0404;11 () CD3IL-2
4 0.02 0.04 0.02 0.79 0.99 0.89 DRB1*0401;14 ⫹⫹⫹ CD3IL-2
5 0.02 0.07 0.29 0.29 0.65 0 DRB1*0401;08 ⫹⫹⫹ CD3IL-2
6 0.02 0.09 0.03 0.02 0.02 0.02 ND CD3IL-2
7 0.04 0.20 0.02 0.03 0.01 0.03 DRB1*08; 14 CD3IL-2
8 0.01 0.02 0.01 0.01 0 0.01 DRB1*07; 11 CD4IL-2
9
§
0.04 0.37 0.01 0.12 0.04 0.05 DRB1*0401;11 () CD4IL-2
10
§
0.20 0.28 0.26 0.14 0.25 0.92 DRB1*0401;03 ⫹⫹ CD4IL-2
11 0.01 0.38 0.31 0.17 0.18 0.14 DRB1*0102;13 CD4IL-2
12 0.05 0.61 0.07 0.41 0.48 0.07 DRB1*0404;15 CD4IL-2
13 0.04 0.21 0.02 0.04 0.02 0.03 ND CD4IL-2
14 0.02 0.11 0.06 0.04 0.01 0.01 ND CD4IL-2
Percentage of IL-2-producing T cells after in vitro culture of PBMC without antigen (No antigen) or following stimulation with tetanus toxoid (TT1), or
following stimulation with CII-peptides (see Fig. 1). IL-2 production was analyzed for the entire CD3T cell population in patients 1 to 7 or within the CD4subset
in patients 8 to 14.
*Percentage of double-positive T cells.
DRB1-alleles containing the ‘‘shared epitope’’ QKRAA or QRRAA in amino acid positions 7074 are highlighted in shaded cells and bold italics; subtyping of
DRB1* was performed only on 04 and 01 haplotypes. ND, Not determined.
Semiquantitative scoring of T cell response to any of the CII-peptides (% of IL-2-producing T cells): ,0.1% or 3(no antigen control); (), 3(no antigen
control); ,4(no antigen control); ⫹⫹,5(no antigen control); ⫹⫹⫹,10(no antigen control). Shaded cells and bold gures in the table highlight
CII-peptide-specic responses fullling at least the () criterion.
§Patients where repetitive analyses were performed. Patient 3 elicited a similar response exclusively to the Gal264-peptide in a second analysis performed 1 wk
later (see Fig. 4). For patient 9, in a second study performed 1 mo later, 0.1% of CD3cells produced IL-2 upon stimulation with the Gal264-peptide, compared
with 0.04% in the absence of antigen and 0.02% after stimulation with the K264270- or Gal270-peptide. Patient 10 failed to respond to any CII-peptide when
investigated 6 mo later.
Ba¨cklund et al. PNAS
July 23, 2002
vol. 99
no. 15
9963
IMMUNOLOGY
performed repetitive analysis of T cell responses to CII in 3 of
the 14 patients. In the initial investigation, patient 3 responded
exclusively to the Gal264-peptide because 0.24% of CD3
cells
produced IL-2 on stimulation with this peptide, compared with
0.07% in the absence of antigen. One week later, the same
patient elicited an almost identical response (Fig. 4) where the
two-color staining for CD4 and IL-2 show an increase in
the double-positive T cell population on stimulation with the
Gal264-peptide to 0.22% whereas exposure to the K264270-
peptide did not result in an increased IL-2 production. Similarly,
patient 9 initially responded to the Gal264-peptide (Table 2),
and this exclusive response was also noted when the patient was
investigated 1 mo later (see footnotes in Table 2). However, in
patient 10, the initially recorded response to the Gal264270-
peptide (Table 2) was undetectable 6 mo after the first assess-
ment (see footnotes in Table 2). Thus, although CII-specific T
cell responses may vary with time in some patients, possibly
because of complex influences by treatment or spontaneous
variations in the immune response of a characteristically chronic
relapsing disease, the results obtained in a small cohort of RA
patients with established diseases and severe course show the
dominant targeting of glycosylated variants of the CII-peptide by
the autoreactive T cell response. Finally, the available HLA-
typing information on the patients reveals that all patients
exhibiting T cell responses to the respective CII-peptide variants
express at least one of the HLA-DRB1*-alleles containing the
amino acid consensus motif QK(R)RAA in position 7074. This
motif constitutes the so-called ‘‘shared epitope’’ (24, 25), and is
crucial for the binding of the CII-peptides to the respective class
II molecules during antigen presentation (26).
Discussion
Posttranslationally glycosylated peptides from CII are presented
by the RA-associated DR4 molecules as shown in human
DR4-transgenic mice. In DR4-transgenic mice expressing huCII,
and most likely also in humans, T cells recognizing the nongly-
cosylated 263270 epitope are strongly tolerized, or even de-
leted, whereas T cells specific for the different glycosylated
peptides persist. This is a potentially crucial finding because it
provides an explanation not only for the role of the DR4 in the
presentation of joint derived peptides, but also for the difficulties
in providing evidence for T cells recognizing autologous CII.
The relevance of the findings made in mice expressing huCII
and DR4 is evident because 30% of the investigated RA patients
exhibited a predominant response to the glycosylated forms of
the CII263270 epitope. Thus, our data show that the physio-
logical posttranslational modification of variable carbohydrate
attachment converts the immunodominant naked self-peptide,
which is tolerogenic, into several cryptic self-determinants that
remain immunogenic.
In addition to reports showing that transgenic expression of
DR4 and DR1 in mice permits development of CIA (2, 3, 27),
there are several investigations describing T cell immunity to CII
in either healthy individuals or RA patients or both (5, 2836).
However, less is known regarding the determinants recognized
by CII-specific T cells in humans, and the interpretations of the
reported findings are not all in agreement (4, 5, 34, 35). For
example, one recent study failed to identify CII-specific T cells
in RA patients when CII259272 loaded DR4-tetramers were
used (4), despite the earlier defined immunodominance of this
peptide in DR4-transgenic mice (1).
By including the aspect of T cell tolerance to self-CII and
recognition of glycosylated CII-epitopes in our humanized an-
imal model of RA, we can now provide a possible explanation
for the ambiguous results concerning CII recognition in RA
patients. Our finding that T cells specific for glycosylated CII
appeared less tolerized by endogenous expression of huCII than
T cells specific for the nonmodified epitope is predictable from
an earlier finding in A
q
-expressing mice (15, 37). Mice with
transgenic expression of either rat or human CII displayed strong
T cell tolerance to the immunodominant CII epitope and were
also partially protected from CIA when immunized with rat and
human CII, respectively (15, 37). Moreover, mice transgenic for
the A
q
-restricted immunodominant T cell epitope CII256270,
present on heterologous CII, displayed relatively stronger tol-
erance against the nonglycosylated variant (20). Whether per-
sistence of glycopeptide-specific T cells in huCIIDR4 mice
depends on the possibility that the glycopeptides bind with lower
affinity to DR4 than the nonmodified peptide, as was observed
for A
q
(10), was not addressed, but could be considered as a
reasonable explanation. It should be emphasized, however, that
the glycosylated side chain is more likely to be oriented toward
the T cell receptor rather than the MHC, as we show here that
glycopeptide-specific T cell responses are critically dependent on
the galactose moiety. This assumption is further supported from
earlier studies using the A
q
-molecule (10) and is also indicated
from other studies using glycopeptide-specific T cells (38). As an
alternative or complementary explanation for a biased toler-
ance, it has been shown that relatively small differences in
availability or levels of autoantigens in vivo have great impact on
the size or status of the autoreactive T cell repertoire (39, 40).
Thus, different expression levels of the posttranslational forms
of the CII263270 epitope in endogenous CII might modulate
the repertoire selection of CII-specific T cells. It should be
emphasized, however, that also glycopeptide-specific T cells
were influenced by endogenous huCII, although to a lesser
degree than nonglycosylated CII-specific T cells. The response to
the individual glycopeptides was only slightly weaker in huCII
DR4 mice than in DR4 mice, but the reduction was more obvious
when comparing the response to the intact CII protein, indicat-
ing anergized or low affinity T cells. In addition, with the use of
deoxy-peptides, we found that T cells from huCIIDR4 mice
recognized the glycopeptide differently, indicating also that
huCII influences T cell repertoire selection.
Fig. 4. Specic recognition of CII-glycopeptides by human T cells. The
two-color ow-cytometry of in vitro-stimulated T cells from an RA-patient
shows uorescence intensities for surface-binding of a FITC-labeled anti-CD4
antibody on the xaxis whereas the yaxis represents signal intensities for
intracellular staining with a phycoerythrin-labeled anti-IL-2 antibody. PBMCs
were cultured overnight without antigen, with tetanus toxoid, the K264270-
peptide, or the Gal264-peptide. The gures in the upper right quadrant of the
different panels represent the percentage of double-positive cells.
9964
www.pnas.orgcgidoi10.1073pnas.132254199 Ba¨cklund et al.
The mice used in this report are complex, because they carry
multiple genetic modifications on a mixed genetic background,
and translation of our in vitro findings to the human system
should be done with some caution. Nevertheless, despite the
complexity of the experimental set-up, our data on T cell
tolerance in huCIIDR4 mice are intact and support earlier
findings (15, 20, 37, 41). Collectively, these data strongly suggest
that endogenous CII is physiologically exposed to the immune
system but does not lead to complete tolerance of a subset of
CII-specific T cells.
In summary, the results in a humanized animal model of RA
and the functional assays on cellular autoimmunity in RA
patients provide convincing evidence for incomplete T cell
tolerance to a set of closely related self-determinants that are
physiologically modified by posttranslational glycosylation. It
should be stressed, however, that our data do not prove that the
remaining CII-specific T cells are causative or even involved in
the pathogenesis of RA. Despite this fact, our findings are
interesting because they may constitute the missing link between
the CII-specific humoral response, observed in a substantial
fraction of RA patients, and the difficulties in the identification
of CII-specific T cells in RA patients. Furthermore, T cell
recognition of glycosylated CII in RA may be useful as a disease
progression marker and for classification of this heterogenous
disease.
We thank Carlos Palestro for taking good care of the animals, as well as
Alexandra and Caroline Treschow for critically reading the manuscript.
The work was supported by grants from the King Gustaf Vs 80-Year
Foundation, Kocks Foundations, O
¨sterlunds foundation, the Swedish
Association against Rheumatism, the Swedish Medical Research Coun-
cil, the Swedish Foundation for Strategic Research, the Go¨ran Gustafs-
son Foundation, the Deutsche Forschungsgemeinschaft (SFB 263,
project C3), the Bundesministerium fu¨r Bildung und Forschung (Med-
Net Entzu¨ndlich rheumatische Erkrankungen, project C 2.1; BMBF,
01GI9948), the European Commission (Bio4-98-0479), and the Karen
Elise Jensen and Novo Nordisk Foundations in Denmark.
1. Fugger, L., Rothbard, J. B. & Sonderstrup McDevitt, G. (1996) Eur. J. Im-
munol. 26, 928933.
2. Rosloniec, E. F., Brand, D. D., Myers, L. K., Esaki, Y., Whittington, K. B.,
Zaller, D. M., Woods, A., Stuart, J. M. & Kang, A. H. (1998) J. Immunol. 160,
25732578.
3. Andersson, E. C., Hansen, B. E., Jacobsen, H., Madsen, L. S., Andersen, C. B.,
Engberg, J., Rothbard, J. B., McDevitt, G. S., Malmstro¨m, V., Holmdahl, R.,
et al. (1998) Proc. Natl. Acad. Sci. USA 95, 75747579.
4. Kotzin, B. L., Falta, M. T., Crawford, F., Rosloniec, E. F., Bill, J., Marrack, P.
& Kappler, J. (2000) Proc. Natl. Acad. Sci. USA 97, 291296.
5. Kim, H. Y., Kim, W. U., Cho, M. L., Lee, S. K., Youn, J., Kim, S. I., Yoo,
W. H., Park, J. H., Min, J. K., Lee, S. H., et al. (1999) Arthritis Rheum. 42,
20852093.
6. Michae¨lsson, E., Malmstro¨m, V., Reis, S., Engstro¨m, Å., Burkhardt, H. &
Holmdahl, R. (1994) J. Exp. Med. 180, 745749.
7. Corthay, A., Ba¨cklund, J., Broddefalk, J., Michae¨lsson, E., Goldschmidt, T. J.,
Kihlberg, J. & Holmdahl, R. (1998) Eur. J. Immunol. 28, 25802590.
8. Michae¨lsson, E., Andersson, M., Engstrom, Å. & Holmdahl, R. (1992) Eur.
J. Immunol. 22, 18191825.
9. Brunsberg, U., Gustafsson, K., Jansson, L., Michae¨lsson, E., Ahrlund Richter,
L., Pettersson, S., Mattsson, R. & Holmdahl, R. (1994) Eur. J. Immunol. 24,
16981702.
10. Kjellen, P., Brunsberg, U., Broddefalk, J., Hansen, B., Vestberg, M., Ivarsson,
I., Engstrom, Å., Svejgaard, A., Kihlberg, J., Fugger, L. & Holmdahl, R. (1998)
Eur. J. Immunol. 28, 755767.
11. Dessen, A., Lawrence, C. M., Cupo, S., Zaller, D. M. & Wiley, D. C. (1997)
Immunity 7, 473481.
12. Holmdahl, R., Andersson, M., Goldschmidt, T. J., Gustafsson, K., Jansson, L.
& Mo, J. A. (1990) Immunol. Rev. 118, 193232.
13. Holmdahl, R., Vingsbo, C., Mo, J. A., Michae¨lsson, E., Malmstro¨m, V.,
Jansson, L. & Brunsberg, U. (1995) Immunol. Rev. 144, 109135.
14. Fugger, L., Michie, S. A., Rulifson, I., Lock, C. B. & McDevitt, G. S. (1994)
Proc. Natl. Acad. Sci. USA 91, 61516155.
15. Malmstro¨m, V., Ho, K. K., Lun, J., Tam, P. P., Cheah, K. S. & Holmdahl, R.
(1997) Scand. J. Immunol. 45, 670677.
16. Cosgrove, D., Gray, D., Dierich, A., Kaufman, J., Lemeur, M., Benoist, C. &
Mathis, D. (1991) Cell 66, 10511066.
17. Andersson, M. & Holmdahl, R. (1990) Eur. J. Immunol. 20, 10611066.
18. Broddefalk, J., Ba¨cklund, J., Almqvist, F., Johansson, M., Holmdahl, R. &
Kihlberg, J. (1998) J. Am. Chem. Soc. 120, 76767683.
19. Holm, B., Broddefalk, J., Flodell, S., Wellner, E. & Kihlberg, J. (2000)
Tetrahedron 56, 15791586.
20. Malmstro¨m, V., Ba¨cklund, J., Jansson, L., Kihlberg, J. & Holmdahl, R. (2000)
Arthritis Res. 2, 315326.
21. Arnett, F. C., Edworthy, S. M., Bloch, D. A., McShane, D. J., Fries, J. F.,
Cooper, N. S., Healey, L. A., Kaplan, S. R., Liang, M. H., Luthra, H. S., et al.
(1988) Arthritis Rheum. 31, 315324.
22. Assenmacher, M., Schmitz, J. & Radbruch, A. (1994) Eur. J. Immunol. 24,
10971101.
23. Assenmacher, M., Lohning, M., Scheffold, A., Manz, R. A., Schmitz, J. &
Radbruch, A. (1998) Eur. J. Immunol. 28, 15341543.
24. Stastny, P. (1978) N. Engl. J. Med. 298, 869871.
25. Gregersen, P. K., Silver, J. & Winchester, R. J. (1987) Arthritis Rheum. 30,
12051213.
26. Diab, B. Y., Lambert, N. C., LFaqihi, F. E., Loubet-Lescoulie, P., de Preval,
C. & Coppin, H. (1999) Immunogenetics 49, 3644.
27. Rosloniec, E. F., Brand, D. D., Myers, L. K., Whittington, K. B., Gu-
manovskaya, M., Zaller, D. M., Woods, A., Altmann, D. M., Stuart, J. M. &
Kang, A. H. (1997) J. Exp. Med. 185, 11131122.
28. Trentham, D. E., Dynesius, R. A., Rocklin, R. E. & David, J. R. (1978) N. Engl.
J. Med. 299, 327332.
29. Stuart, J. M., Postlethwaite, A. E., Townes, A. S. & Kang, A. H. (1980) Am. J.
Med. 69, 1318.
30. Solinger, A. M., Bhatnagar, R. & Stobo, J. D. (1981) Proc. Natl. Acad. Sci . USA
78, 38773881.
31. Londei, M., Savill, C. M., Verhoef, A., Brennan, F., Leech, Z. A., Duance,
V., Maini, R. N. & Feldmann, M. (1989) Proc. Natl. Acad. Sci. USA 86,
636640.
32. Ofosu-Appiah, W. A., Warrington, R. J. & Wilkins, J. A. (1989) Clin. Immunol.
Immunopathol. 50, 264271.
33. Lacour, M., Rudolphi, U., Schlesier, M. & Peter, H. H. (1990) Eur. J. Immunol.
20, 931934.
34. Yan, T., Burkhardt, H., Ritter, T., Broker, B., Mann, K. H., Bertling, W. M.,
von der Mark, K. & Emmrich, F. (1992) Eur. J. Immunol. 22, 5156.
35. Snowden, N., Reynolds, I., Morgan, K. & Holt, L. (1997) Arthritis Rheum. 40,
12101218.
36. Berg, L., Ronnelid, J., Sanjeevi, C. B., Lampa, J. & Klareskog, L. (2000)
Arthritis Res. 2, 7584.
37. Malmstro¨m, V., Michae¨lsson, E., Burkhardt, H., Mattsson, R., Vuorio, E. &
Holmdahl, R. (1996) Proc. Natl. Acad. Sci. USA 93, 44804485.
38. Rudd, P. M., Elliott, T., Cresswell, P., Wilson, I. A. & Dwek, R. A. (2001)
Science 291, 23702376.
39. Cibotti, R., Kanellopoulos, J. M., Cabaniols, J. P., Halle-Panenko, O., Kos-
matopoulos, K., Sercarz, E. & Kourilsky, P. (1992) Proc. Natl. Acad. Sci. USA
89, 416420.
40. Akkaraju, S., Ho, W. Y., Leong, D., Canaan, K., Davis, M. M. & Goodnow,
C. C. (1997) Immunity 7, 255271.
41. Malmstro¨m, V., Kjellen, P. & Holmdahl, R. (1998) J. Autoimmun. 11, 213221.
Ba¨cklund et al. PNAS
July 23, 2002
vol. 99
no. 15
9965
IMMUNOLOGY
... T cells against COL2, specific for relevant COL2 peptides, are activated in most RA patients (14,15). Lymphocytes reactive with COL2 are of significant interest as COL2 is not only a major tissue-specific protein expressed in joint cartilage but is also expressed in bone marrow and thymus (6,7). ...
... Immunization of mice, using strong adjuvants, can break this tolerance and induce arthritis, but in the normal state COL2 T cell reactivity might be predominantly of regulatory importance in both mice and humans. It is thus of interest that, out of numerous autoantigenspecific T cells detected in RA, T cells reactive with the galactosylated COL2259-273 peptide frequently occur (14,15). The COL2259-273 peptide binds well to both the mouse A q molecule, associated with the development of CIA, and the human DR*0401 molecule, associated with RA (10,13,15). ...
Article
Full-text available
A longstanding goal has been to find an antigen-specific preventive therapy, i.e., a vaccine, for autoimmune diseases. It has been difficult to find safe ways to steer the targeting of natural regulatory antigen. Here, we show that the administration of exogenous mouse major histocompatibility complex class II protein bounding a unique galactosylated collagen type II (COL2) peptide (Aq-galCOL2) directly interacts with the antigen-specific TCR through a positively charged tag. This leads to expanding a VISTA-positive nonconventional regulatory T cells, resulting in a potent dominant suppressive effect and protection against arthritis in mice. The therapeutic effect is dominant and tissue specific as the suppression can be transferred with regulatory T cells, which downregulate various autoimmune arthritis models including antibody-induced arthritis. Thus, the tolerogenic approach described here may be a promising dominant antigen-specific therapy for rheumatoid arthritis, and in principle, for autoimmune diseases in general.
... Notably, glycosylation of immunopeptides has also been shown to be important for the specificity of some human MHC-IIrestricted T-cells (13)(14)(15). Forming a foundation of this present study, we have recently found that dendritic cells (DCs) phagocytose and process the highly N-glycosylated SARS-CoV-2 spike protein and prevalently present MHC-II-bound immunopeptides carrying both under-and hyper-processed (remodelled) spike N-glycans on their surfaces including oligomannosidic (Man 5-9 GlcNAc 2 ) and paucimannosidic (Man 1-3 GlcNAc 2 Fuc 0-1 ) N-glycans for T-cell recognition, starkly contrasting the glycophenotype of the native spike protein that was shown to be rich in elongated complex-type N-glycans (16). While the unusual glycosylation features of these immunopeptides provide clues regarding how glycans decorating exogenous viral glycoproteins are perceived and altered by our immune system, it remains unknown if endogenous (human) immunopeptides also carry remodelled N-glycans that differ from their native cellular glycoproteins, details that are important to enable a better understanding of the glycobiology underpinning immunopeptide presentation and recognition. ...
Article
Full-text available
Immunopeptidomics, the study of peptide antigens presented on the cell surface by the major histocompatibility complex (MHC), offers insights into how our immune system recognises self/non-self in health and disease. We recently discovered that hyper-processed (remodelled) N -glycans are dominant features decorating viral spike immunopeptides presented via MHC-class II (MHC-II) molecules by dendritic cells pulsed with SARS-CoV-2 spike protein, but it remains unknown if endogenous immunopeptides also undergo N -glycan remodelling. Taking a multi-omics approach, we here interrogate published MHC-II immunopeptidomics datasets of cultured monocyte-like (THP-1) and breast cancer-derived (MDA-MB-231) cell lines for overlooked N -glycosylated peptide antigens, which we compare to their source proteins in the cellular glycoproteome using proteomics and N -glycomics data from matching cell lines. Hyper-processed chitobiose core and paucimannosidic N- glycans alongside under-processed oligomannosidic N -glycans were found to prevalently modify MHC-II-bound immunopeptides isolated from both THP-1 and MDA-MB-231, while complex/hybrid-type N -glycans were (near-)absent in the immunopeptidome as supported further by new N -glycomics data generated from isolated MHC-II-bound peptides derived from MDA-MB-231 cells. Contrastingly, the cellular proteomics and N -glycomics data from both cell lines revealed conventional N -glycosylation rich in complex/hybrid-type N -glycans, which, together with the identification of key lysosomal glycosidases, suggest that MHC-II peptide antigen processing is accompanied by extensive N -glycan trimming. N -glycan remodelling appeared particularly dramatic for cell surface-located glycoproteins while less remodelling was observed for lysosomal-resident glycoproteins. Collectively, our findings indicate that both under- and hyper-processed N -glycans are prevalent features of endogenous MHC-II immunopeptides, an observation that demands further investigation to enable a better molecular-level understanding of immune surveillance.
... The most common post-translational modification of the major immunodominant T-cell epitope CII259-273 is hydroxylation and subsequent glycosylation at position K264. The T-cell response in RA patients and in mouse models of rheumatoid arthritis (collagen-induced arthritis, CIA) is directed and predominates against this modified form of the recognizable epitope [34][35][36][37]. This was the reason to include this peptide in the present study. ...
Article
Full-text available
Citrullinated proteins and anti-citrullinated protein antibodies (ACPAs) play an important role in the pathogenesis of rheumatoid arthritis (RA). It has been suggested that during inflammation or dysbiosis, bacteria could initiate production of ACPAs. Most patients with RA are seropositive for ACPAs, but these antibodies have overlapping reactivity to different posttranslational modifications (PTMs). For initiation and development of RA, T lymphocytes and T cell epitopes are still required. In this study, we evaluated the ability of bacterial L-asparaginase to modify RA-related T cell epitopes within type II collagen (CII259-273 and CII311-325), as well as whether these modified epitopes are recognized by ACPAs from RA patients. We included 12 patients with early RA and 11 healthy subjects selected according to predefined specific criteria. LC-MS/MS analyses revealed that the bacterial L-asparaginase can modify investigated T cell epitopes. ELISA tests showed cross-reactivity of ACPA positive sera from early RA patients towards the enzymatically modified immunodominant T cell epitopes within type II collagen (CII), but not to the modified irrelevant peptides. These data suggest that the cross-reactive ACPAs recognize the “carbonyl-Gly-Pro” motif in CII. Moreover, the T cell recognition of the modified major immunodominant T cell epitope Gal264-CII259-273 was not affected. This epitope was still able to activate autoreactive T cells from early RA patients. It is likely that such modifications are the missing link between the T cell priming and the development of anti-modified protein antibodies (AMPAs). Our results provide additional information on the etiology and pathogenesis of RA.
... Our model also adds another functional layer that could motivate the need for antigen-specific autoreactive B cells. The escape of COL2-reactive T cells from intrathymic selection (Raposo et al., 2018) and their association with autoimmune arthritis (Bäcklund et al., 2002a(Bäcklund et al., , 2002b begs this question: What peripheral mechanism governs antigen-specific tolerance? The COL2-specific T cell tolerance underlined by C1-B cells may provide the answer. ...
Article
Full-text available
B cells undergo several rounds of selection to eliminate potentially pathogenic autoreactive clones, but in contrast to T cells, evidence of positive selection of autoreactive B cells remains moot. Using unique tetramers, we traced natural autoreactive B cells (C1-B) specific for a defined triple-helical epitope on collagen type-II (COL2), constituting a sizeable fraction of the physiological B cell repertoire in mice, rats, and humans. Adoptive transfer of C1-B suppressed arthritis independently of IL10, separating them from IL10-secreting regulatory B cells. Single-cell sequencing revealed an antigen processing and presentation signature, including induced expression of CD72 and CCR7 as surface markers. C1-B presented COL2 to T cells and induced the expansion of regulatory T cells in a contact-dependent manner. CD72 blockade impeded this effect suggesting a new downstream suppressor mechanism that regulates antigen-specific T cell tolerization. Thus, our results indicate that autoreactive antigen-specific naïve B cells tolerize infiltrating T cells against self-antigens to impede the development of tissue-specific autoimmune inflammation.
Article
Full-text available
Transgenic mice expressing human major histocompatibility complex class II (MHCII) risk alleles are widely used in autoimmune disease research, but limitations arise due to non‐physiologic expression. To address this, physiologically relevant mouse models are established via knock‐in technology to explore the role of MHCII in diseases like rheumatoid arthritis. The gene sequences encoding the ectodomains are replaced with the human DRB1*04:01 and 04:02 alleles, DRA, and CD74 (invariant chain) in C57BL/6N mice. The collagen type II (Col2a1) gene is modified to mimic human COL2. Importantly, DRB1*04:01 knock‐in mice display physiologic expression of human MHCII also on thymic epithelial cells, in contrast to DRB1*04:01 transgenic mice. Humanization of the invariant chain enhances MHCII expression on thymic epithelial cells, increases mature B cell numbers in spleen, and improves antigen presentation. To validate its functionality, the collagen‐induced arthritis (CIA) model is used, where DRB1*04:01 expression led to a higher susceptibility to arthritis, as compared with mice expressing DRB1*04:02. In addition, the humanized T cell epitope on COL2 allows autoreactive T cell‐mediated arthritis development. In conclusion, the humanized knock‐in mouse faithfully expresses MHCII, confirming the DRB1*04:01 alleles role in rheumatoid arthritis and being also useful for studying MHCII‐associated diseases.
Article
Full-text available
It has been shown that synovial fibroblasts (SF) play a key role in the initiation of inflammation and joint destruction, leading to arthritis progression. Fibroblasts may express major histocompatibility complex class II region (MHCII) molecules, and thus, they could be able to process and present antigens to immunocompetent cells. Here we examine whether different types of fibroblasts (synovial, dermal, and thymic murine fibroblasts, destructive LS48 fibroblasts, and noninvasive NIH/3T3 fibroblasts) may be involved in the initiation of rheumatoid arthritis (RA) pathogenesis and can process and present type II collagen (COL2)—an autoantigen associated with RA. Using a panel of MHCII/Aq-restricted T-cell hybridoma lines that specifically recognize an immunodominant COL2 epitope (COL2259–273), we found that NIH/3T3 fibroblasts activate several T-cell clones that recognize the posttranslationally glycosylated or hydroxylated COL2259–273 epitope. The HCQ.3 hybridoma, which is specific for the glycosylated immunodominant COL2 epitope 259–273 (Gal264), showed the strongest response. Interestingly, NIH/3T3 cells, but not destructive LS48 fibroblasts, synovial, dermal, or thymic fibroblasts, were able to stimulate the HCQ.3 hybridoma and other COL2-specific T-cell hybridomas. Our experiments revealed that NIH/3T3 fibroblasts are able to activate COL2-specific T-cell hybridomas even in the absence of COL2 or a posttranslationally modified COL2 peptide. The mechanism of this unusual activation is contact-dependent and involves the T-cell receptor (TCR) complex.
Article
Full-text available
Collagen type II (COL2), the main structural protein of hyaline cartilage, is considerably affected by autoimmune responses associated with the pathogenesis of rheumatoid arthritis (RA). Posttranslational modifications (PTMs) play a significant role in the formation of the COL2 molecule and supramolecular fibril organization, and thus, support COL2 function, which is crucial for normal cartilage structure and physiology. Conversely, the specific PTMs of the protein (carbamylation, glycosylation, citrullination, oxidative modifications and others) have been implicated in RA autoimmunity. The discovery of the anti-citrullinated protein response in RA, which includes anti-citrullinated COL2 reactivity, has led to the development of improved diagnostic assays and classification criteria for the disease. The induction of immunological tolerance using modified COL2 peptides has been highlighted as a potentially effective strategy for RA therapy. Therefore, the aim of this review is to summarize the recent knowledge on COL2 posttranslational modifications with relevance to RA pathophysiology, diagnosis and treatment. The significance of COL2 PTMs as a source of neo-antigens that activate immunity leading to or sustaining RA autoimmunity is discussed.
Article
Rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), and celiac disease (CD), are strongly associated with susceptible HLA class II haplotypes. The peptide-binding pockets of these molecules are polymorphic, thus each HLA class II protein presents a distinct set of peptides to CD4+ T cells. Peptide diversity is increased through post-translational modifications, generating non-templated sequences that enhance HLA binding and/or T cell recognition. The high-risk HLA-DR alleles that confer susceptibility to RA are notable for their ability to accommodate citrulline, promoting responses to citrullinated self-antigens. Likewise, HLA-DQ alleles associated with T1D and CD favor the binding of deamidated peptides. In this review, we discuss structural features that promote modified self-epitope presentation, provide evidence supporting the relevance of T cell recognition of such antigens in disease processes, and make a case that interrupting the pathways that generate such epitopes and reprogramming neoepitope-specific T cells are key strategies for effective therapeutic intervention.
Article
In RA patients' synovial sites, citrullinated RA-related antigens such as type II collagens, fibrin (ogen), vimentin, and α-enolase could be targeted by ACCPAs. Since ACCPA production can be initiated a long time before RA sign appearance, primary auto-immunization against these citrullinated proteins can be originated from extra-articular sites. It has been shown that there is a significant association between P. gingivalis periodontitis, anti- P. gingivalis antibodies, and RA. P. gingivalis gingipains (Rgp, Kgp) can degrade proteins such as fibrin and α-enolase into some peptides in the form of Arg in the C-terminal which is converted to citrulline by PPAD. Also, PPAD can citrullinate type II collagen and vimentins (SA antigen). P. gingivalis induces inflammation and chemoattraction of immune cells such as neutrophils and macrophages through the increase of C5a (gingipain C5 convertase-like activity) and SCFA secretion. Besides, this microorganism stimulates anoikis, a special type of apoptosis, and NETosis, an antimicrobial form of neutrophil death, leading to the release of PAD1-4, α-enolase, and vimentin from apoptotic cells into the periodontal site. In addition, gingipains can degrade macrophages CD14 and decrease their ability in apoptotic cell removal. Gingipains also can cleave IgGs in the Fc region and transform them into rheumatoid factor (RF) antigens. In the present study, the effects of P. gingivalis on rheumatoid arthritis autoimmune response have been reviewed, which could attract practical insight both in bench and clinic.
Article
Full-text available
Although associations between the expression of particular HLA genes and the susceptibility to specific autoimmune diseases has been known for some time, the role that these HLA molecules play in the autoimmune response is unclear. Through the establishment of a chimeric HLA-DR/I-E transgene, we have examined the function of the rheumatoid arthritis (RA) susceptibility allele HLA-DR4 (DRB1*0401) in presenting antigenic peptides derived from the model Ag, type II collagen (CII), and in mediating an autoimmune response. As a transgene, the chimeric DR4 molecule conferred susceptibility to an autoimmune arthritis induced by immunization with human CII or bovine CII. These mice developed an inflammatory, autoimmune arthritis that was similar both histologically and in severity to that previously described for the collagen-induced arthritis model. The DR4-mediated autoimmune arthritis was accompanied by T cell and B cell responses to both the immunogen and the autoantigen, murine CII. The DR4-restricted T cell response to human CII was focused on an immunodominant determinant within CII263–270 and a minor determinant within CII286–300, the same CII determinants recently identified for yet another RA susceptibility allele, HLA-DR1 (DRB1*0101). Thus these data demonstrate that, like HLA-DR1, HLA-DR4 is capable of binding peptides derived from human CII and therefore probably plays a role in the autoimmune response to human CII observed in RA patients.
Article
Full-text available
Rheumatoid arthritis (RA) is an autoimmune disease associated with the HLA-DR4 and DR1 alleles. The target autoantigen(s) in RA is unknown, but type II collagen (CII) is a candidate, and the DR4- and DR1-restricted immunodominant T cell epitope in this protein corresponds to amino acids 261–273 (CII 261–273). We have defined MHC and T cell receptor contacts in CII 261–273 and provide strong evidence that this peptide corresponds to the peptide binding specificity previously found for RA-associated DR molecules. Moreover, we demonstrate that HLA-DR4 and human CD4 transgenic mice homozygous for the I-Abβ0 mutation are highly susceptible to collagen-induced arthritis and describe the clinical course and histopathological changes in the affected joints.
Article
A critical event in an immune response is the T cell recognition of peptides bound to major histocompatibility complex (MHC) molecules on the surface of an antigen presenting cell (APC). Although the majority of eukaryotic proteins are glycosylated, it has not yet been shown that T cell recognition of such proteins involves recognition of the bound carbohydrates. Type II collagen (CII), the major protein constituent of joint cartilage, is posttranslationally modified by hydroxylation and glycosylation of lysines. In this report we show that posttranslational modifications of the immunodominant peptide CII(256-270) generate a structural determinant that is distinct from the determinant represented by the corresponding synthetic peptide. Elimination of carbohydrates, present on CII, by two different biochemical methods revealed that the carbohydrates, O-linked to the hydroxylysines within the CII(256-270) determinant, were crucial for the reactivity towards the posttranslationally modified peptide. Furthermore, a T cell hybridoma specific for the glycosylated determinant was stimulated by tryptic CII-peptides presented by fixed APCs, thus showing that the carbohydrates are involved in the trimolecular complex T cell receptor/peptide/MHC. Finally, the importance of the bound carbohydrates for the arthritogenicity of CII was investigated by comparing the development of arthritis after immunization with carbohydrate-depleted and glycosylated CII, respectively. Incidence, time of onset, and severity of the disease were significantly affected by the elimination of carbohydrates, whereas no significant difference in anti-CII antibody titers was seen.
Article
The revised criteria for the classification of rheumatoid arthritis (RA) were formulated from a computerized analysis of 262 contemporary, consecutively studied patients with RA and 262 control subjects with rheumatic diseases other than RA (non-RA). The new criteria are as follows: 1) morning stiffness in and around joints lasting at least 1 hour before maximal improvement; 2) soft tissue swelling (arthritis) of 3 or more joint areas observed by a physician; 3) swelling (arthritis) of the proximal interphalangeal, metacarpophalangeal, or wrist joints; 4) symmetric swelling (arthritis); 5) rheumatoid nodules; 6) the presence of rheumatoid factor; and 7) radiographic erosions and/or periarticular osteopenia in hand and/or wrist joints. Criteria 1 through 4 must have been present for at least 6 weeks. Rheumatoid arthritis is defined by the presence of 4 or more criteria, and no further qualifications (classic, definite, or probable) or list of exclusions are required. In addition, a “classification tree” schema is presented which performs equally as well as the traditional (4 of 7) format. The new criteria demonstrated 91–94% sensitivity and 89% specificity for RA when compared with non-RA rheumatic disease control subjects.
Article
Almost all of the key molecules involved in the innate and adaptive immune response are glycoproteins. In the cellular immune system, specific glycoforms are involved in the folding, quality control, and assembly of peptide-loaded major histocompatibility complex (MHC) antigens and the T cell receptor complex. Although some glycopeptide antigens are presented by the MHC, the generation of peptide antigens from glycoproteins may require enzymatic removal of sugars before the protein can be cleaved. Oligosaccharides attached to glycoproteins in the junction between T cells and antigen-presenting cells help to orient binding faces, provide protease protection, and restrict nonspecific lateral protein-protein interactions. In the humoral immune system, all of the immunoglobulins and most of the complement components are glycosylated. Although a major function for sugars is to contribute to the stability of the proteins to which they are attached, specific glycoforms are involved in recognition events. For example, in rheumatoid arthritis, an autoimmune disease, agalactosylated glycoforms of aggregated immunoglobulin G may induce association with the mannose-binding lectin and contribute to the pathology.
Article
Even though most eucaryotic proteins are glycosylated, very little is known on if, or how, the glycans influence essential immunological events such as antigen processing, major histocompatibility complex (MHC) restricted presentation, and recognition by T cells. We have used synthetic glycopeptides to elucidate the specificity of T cell hybridomas, obtained by immunization with the glycoprotein type II collagen in a mouse model for rheumatoid arthritis. To enable these studies, glycosylated and suitably protected derivatives of (5R)-5-hydroxy-l-lysine, and the similar 5-hydroxy-l-norvaline, were prepared and then used in Fmoc solid-phase synthesis of glycopeptides related to the immunodominant fragment from type II collagen, CII(256−270). Evaluation of the synthetic glycopeptides provided evidence that antigen-presenting cells can indeed process glycoproteins to glycopeptides, which elicit a T cell response when presented by class II MHC molecules. A glycopeptide carrying a single β-d-galactosyl residue attached to hydroxylysine at position 264 in the center of the CII(256−270) peptide was recognized by most of the hybridomas in a way involving specific contacts between the carbohydrate and the T cell receptor. The results suggest an explanation for the recent observation that glycosylated type II collagen induces more severe forms of arthritis in the mouse than deglycosylated type II collagen and provide additional knowledge on how rheumatoid arthritis may occur also in humans.
Article
Upon primary activation, T helper (Th) cell populations express different cytokines transiently and with different kinetics. Stimulation of naive murine splenic Th cells with the bacterial superantigen Staphylococcus aureus enterotoxin B (SEB) in vitro results in expression of IL-2, IFN-γ and IL-10 with fast, intermediate and slow kinetics, respectively. This first report of a functional analysis of cells separated alive according to cytokine expression shows that these cytokines are not produced by different Th cell subpopulations, but can be expressed sequentially by individual Th cells. Th cells, activated with SEB for 1 day and isolated according to expression of IL-2, using the cellular affinity matrix technology, upon continued stimulation with SEB later secrete most of the IFN-γ and IL-10. Likewise, after 2 days of SEB culture, cells expressing IFN-γ, separated according to specific surface-associated IFN-γ as detected by magnetofluorescent liposomes, 1 day later secrete IL-10. Thus, individual Th1 cells can contribute to the control of their own IFN-γ expression by sequential expression of first IL-2, supporting their proliferation, and later IL-10, down-regulating the production of IFN-γ-inducing monokines and limiting the pro-inflammatory effects of IFN-γ.
Article
Objective To determine the presence of specific immune recognition of type II collagen (CII) and its immunodominant epitope CII (255–274) in patients with rheumatoid arthritis (RA).MethodsT cell proliferative responses to bovine CII and a synthetic peptide encompassing CII (255–274) in peripheral blood mononuclear cells (PBMC) and synovial fluid mononuclear cells (SFMC) from RA patients, and in PBMC from osteoarthritis (OA) patients and healthy controls were assayed by mixed lymphocyte culture.ResultsThe stimulation index (SI) and the number of positive (SI ≥2) T cell responses to CII were higher in RA patients (n = 106) than in OA patients (n = 26) and healthy controls (n = 34). T cell responses to CII (255–274) were also enhanced in RA patients and correlated well with those to CII. In SFMC, positive responses to CII or CII (255–274) were detected in 61.9% of 42 RA patients. T cell responses to CII in SFMC were stronger and more prevalent than peripheral responses. The SI and positive responses to CII were higher in early RA than in late RA. Levels of IgG antibodies to CII in synovial fluid inversely correlated with T cell responses to CII.ConclusionT cell responses to CII or CII (255–274) were enhanced in RA, especially in early disease. Synthetic peptide CII (255–274), as well as native CII, could be recognized as immunogenic antigens by T cells, particularly in the synovial fluid. These observations suggest that CII-reactive T cells play an important role in the pathogenesis of RA. Peripheral tolerance induction using CII (255–274) might be useful in the treatment of RA.
Article
Different protective groups for (5R)-5-hydroxy-l-lysine were investigated in silver silicate promoted glycosylations with acetobromogalactose as glycosyl donor. Best results were obtained with Fmoc-Hyl(Cbz)-OAll, which was glycosylated in 80% yield. Removal of the allyl group gave a β-d-galactosylated building block which was used in solid-phase synthesis of a glycopeptide from type II collagen. Such glycopeptides are required for studies of rheumatoid arthritis in a mouse that is transgenic for HLA-DR4, i.e. the class II MHC molecule associated with rheumatism in humans.
Article
The major histocompatibility complex (MHC) class II region is assumed to influence autoimmune diseases such as rheumatoid arthritis. In the mouse, the H-2q haplotype is associated with susceptibility to collagen-induced arthritis, while the H-2p haplotype is not. The class II A molecules of these haplotypes differ by only four amino acids in the first domain of the β chain. To test if this difference accounts for the MHC influence on susceptibility to collagen-induced arthritis, H-2p mice were made transgenic with an Abp gene altered to resemble the Abq gene. The transgenic Aβ chain hybridized with the Aαp chain and was shown to be physiologically expressed by testing antigen-presentation capacity to Aq-restricted T cell hybridomas and with FACS analyses. These transgenic mice developed an autoimmune response to type II collagen and also collagen-induced arthritis. The data unequivocally suggest the Ab gene as a major genetic susceptibility locus for autoimmune collagen-induced arthritis.