ArticlePDF Available

Diagnostic accuracy of blood B-cell subset profiling and autoimmunity markers in Sjögren’s syndrome

Authors:

Abstract and Figures

The aims of this study were to evaluate the diagnostic accuracy of blood B-cell subset profiling and immune-system activation marker assays in primary Sjogren's syndrome (pSS) and to assess whether adding these tools to the current laboratory item would improve the American-European Consensus Group (AECG) criteria. In a single-center cohort of patients with suspected pSS, we tested the diagnostic performance of anti-SSA, antinuclear antibody (ANA), rheumatoid factor (RF), gammaglobulins, IgG titers, and B-cell ratio defined as (Bm2 + Bm2[prime])/(eBm5 + Bm5), determined using flow cytometry. The reference standard was a clinical diagnosis of pSS established by a panel of experts. Of 181 patients included in the study, 77 had pSS. By logistic regression analysis, only ANA >=1:640 (sensitivity, 70.4%; specificity 83.2%) and B-cell ratio >=5 (sensitivity, 52.1%; specificity, 83.2%) showed independent associations with pSS, of similar strength. In anti-SSA-negative patients, presence of either of these two criteria had 71.0% sensitivity but only 67.3% specificity for pSS; whereas combining both criteria had 96.2% specificity but only 12.9% sensitivity. Adding either of these two criteria to the AECG criteria set increased sensitivity from 83.1% to 90.9% but decreased specificity from 97.1% to 85.6%, whereas adding both criteria in combination did not substantially modify the diagnostic performance of the criteria set. The adjunction of RF + ANA >= 1:320, as proposed in the new American College of Rheumatology (ACR) criteria, did not improve the diagnostic value of anti-SSA. Blood B-cell subset profiling is a simple test that has good diagnostic properties for pSS. However, adding this test, with or without ANA positivity, does not improve current classification criteria.
Content may be subject to copyright.
R E S E A R C H A R T I C L E Open Access
Diagnostic accuracy of blood B-cell subset
profiling and autoimmunity markers in Sjögrens
syndrome
Divi Cornec
1,2
, Alain Saraux
1,2
, Jacques-Olivier Pers
2
, Sandrine Jousse-Joulin
1,2
, Thierry Marhadour
1
,
Anne-Marie Roguedas-Contios
3
, Steeve Genestet
4
, Yves Renaudineau
2
and Valérie Devauchelle-Pensec
1,2*
Abstract
Introduction: The aims of this study were to evaluate the diagnostic accuracy of blood B-cell subset profiling and
immune-system activation marker assays in primary Sjögrens syndrome (pSS) and to assess whether adding these
tools to the current laboratory item would improve the American-European Consensus Group (AECG) criteria.
Methods: In a single-center cohort of patients with suspected pSS, we tested the diagnostic performance of
anti-SSA, antinuclear antibody (ANA), rheumatoid factor (RF), gammaglobulins, IgG titers, and B-cell ratio defined as
(Bm2 + Bm2)/(eBm5 + Bm5), determined using flow cytometry. The reference standard was a clinical diagnosis of
pSS established by a panel of experts.
Results: Of 181 patients included in the study, 77 had pSS. By logistic regression analysis, only ANA 1:640
(sensitivity, 70.4%; specificity 83.2%) and B-cell ratio 5 (sensitivity, 52.1%; specificity, 83.2%) showed independent
associations with pSS of similar strength. In anti-SSA-negative patients, presence of either of these two criteria had
71.0% sensitivity but only 67.3% specificity for pSS; whereas combining both criteria had 96.2% specificity but only
12.9% sensitivity. Adding either of these two criteria to the AECG criteria set increased sensitivity from 83.1% to
90.9% but decreased specificity from 97.1% to 85.6%, whereas adding both criteria in combination did not
substantially modify the diagnostic performance of the criteria set. The adjunction of RF + ANA 1:320, as proposed
in the new American College of Rheumatology (ACR) criteria, did not improve the diagnostic value of anti-SSA.
Conclusions: Blood B-cell subset profiling is a simple test that has good diagnostic properties for pSS. However,
adding this test, with or without ANA positivity, does not improve current classification criteria.
Introduction
Primary Sjögrens syndrome (pSS) is a chronic auto-
immune disorder that primarily affects the salivary and
lachrymal glands. B cells play a major role in the patho-
genesis of pSS [1]. Thus, biological markers for B-cell
activity and autoimmunity might help to establish the
diagnosis of pSS.
The main serological markers for pSS are autoanti-
bodies against Ro/SSA or La/SSB ribonucleoproteins.
These markers are the only biological item in the widely
used American-European Consensus Group (AECG)
classification criteria [2]. However, they are present in
only 50% to 75% of patients with pSS [3] and are
frequently encountered in other systemic autoimmune
diseases [4]. Other biological markers may thus be va-
luable for the diagnosis of pSS.
Recently published American College of Rheumatology
(ACR) classification criteria for pSS suggest that positiv-
ity for antinuclear antibodies (ANAs) and rheumatoid
factor (RF) should be considered in patients negative for
anti-Ro/SSA antibodies [5]. ANAs are present in 80% of
patients with pSS and RF in 40% [3]. However, the sensi-
tivity (Se) and specificity (Sp) of these tests for pSS com-
pared to controls with other causes of sicca syndrome
need to be assessed before accepting these as part of a
criteria set.
* Correspondence: valerie.devauchelle-pensec@chu-brest.fr
1
Service de Rhumatologie, Centre Hospitalier Régional et Universitaire de
Brest, Hôpital de la Cavale Blanche, BP 824, F-29609 Brest cedex, France
2
EA 2216 Immunologie et Pathologie, Université de Brest, SFR ScinBios,
Labex Imunotherapy, Graft, Oncology, BP 824, F-29609 Brest cedex, France
Full list of author information is available at the end of the article
© 2014 Cornec et al.; licensee BioMed Central Ltd. This is an open access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Cornec et al. Arthritis Research & Therapy 2014, 16:R15
http://arthritis-research.com/content/16/1/R15
B-cell activation may result in hypergammaglobu-
linemia, which is common in patients with pSS [3]. Im-
munoglobulin (Ig)A and IgG are often elevated and can
display RF activity [6].
Sound evidence indicates that the distribution of per-
ipheral-blood B-cell subsets is profoundly altered in pa-
tients with pSS. Memory B cells accumulate in target
epithelial organs, and their proportion is decreased in
peripheral blood [7]. On the other hand, the proportions
of transitional and naive B cells are increased in peri-
pheral blood [8]. IgD/CD38 staining was originally de-
signed for B cell subset study in tonsils, separating
important stages in B-cell development from naive to
memory B cells (Bm1 to Bm5) [9]. This classification is
helpful for studying blood B-cell subset alterations in
pSS: in peripheral blood, Bm2 (IgD+/CD38 low) and
Bm2(IgD+/CD38 high) populations include mainly
transitional and activated naïve B cells and are increased
in pSS patients, whereas eBm5 (IgD-/CD38 low) and
Bm5 (IgD-/CD38-) populations are memory B cells,
which are less represented in patients with pSS com-
pared to patients with rheumatoid arthritis or normal
control subjects [10]. We previously showed in a case
control study that these alterations in blood B-cell subset
distributionmayhaveaninterestingdiagnosticvaluefor
pSS. The better item to predict a diagnosis of pSS using
only fluorescence-activated cell sorting (FACS) analysis was
the B-cell ratio defined as (Bm2 + Bm2)/(eBm5 + Bm5),
which was strongly associated with pSS compared to
rheumatoid arthritis, systemic lupus erythematosus, and
healthy controls [11].
The primary aim of this study was to assess the
diagnostic value of B-cell subset profiling and other bio-
logical autoimmunity markers in a cross-sectional cohort
of patients with suspected pSS. We also evaluated the
diagnostic usefulness of these tools compared to anti-
SSA antibodies and other items of the AECG criteria set.
Methods
Study population
This prospective study was performed in a cohort of pa-
tients with suspected pSS recruited in Brittany, France,
between November 2006 and September 2011 [12]. In-
clusion criteria were subjective ocular or oral dryness,
recurrent or bilateral parotidomegaly, or extraglandular
symptoms suggestive of pSS. The study was approved by
the local medical ethics committee (Brest University
Hospital), and written informed consent was obtained
from all patients before study inclusion.
Clinical and laboratory evaluations
Each patient underwent a standardized assessment inclu-
ding a bilateral Schirmers test (abnormal if 5mm/5min
on at least one side) and unstimulated salivary flow
measurement (abnormal if <0.1 mL/min); a joint eva-
luation; and a general examination. Laboratory tests in-
cluded serum protein electrophoresis; assays of IgG, IgA,
and IgM; ANA on Hep2 cells, anti-SSA and anti-SSB anti-
bodies using commercial ELISAs, and RF using in-house
ELISA (IgM and IgA isotypes) [6]. Minor labial salivary
gland biopsy (SGB) was graded according to the semi-
quantitative Chisholm and Mason score [13].
Blood B-cell subset profiling
Flow cytometry was performed as previously published
[11]. The Bm2 + Bm2subset was defined as the IgD +
and CD38low/high population, and the eBm5 + Bm5
subset as the IgD- and CD38negative/low population
(Figure 1). The proportion of these two subsets in the
total CD19+ B-cell population was determined, and
the (Bm2 + Bm2)/(eBm5 + Bm5) ratio (B-cell ratio) was
computed.
Reference standard
The reference standard was a clinical diagnosis of pSS
performed by the evaluating rheumatologist, based on
the clinical interview and examination, standard biology
and salivary gland biopsy results. All doubtful cases were
reviewed by a panel of three experts, who were blinded
to the results of B-cell profiling. Other systemic diseases
were diagnosed according to published classification
criteria.
Statistical analysis
Statistical tests were performed using the Statistical
Package for the Social Sciences (SPSS 18.0, 2009, SPSS
Inc., Chicago, IL, USA). Quantitative variables are des-
cribed as means ± standard deviation and qualitative va-
riables as numbers (%). We compared patients who
fulfilled the reference standard (clinical diagnosis of pSS)
to those who did not, using MannWhitney and chi-
square tests as appropriate. We plotted receiver-operating
characteristic (ROC) curves to determine the optimal
cutoff associated with the best combination of sensitivity
(Se) and specificity (Sp) for each test.
To determine which laboratory tests were independently
associated with a diagnosis of pSS, we performed multiple
logistic regression with backward selection using the likeli-
hood ratio test. All items associated with a diagnosis of
pSS by univariate analysis with Pvalues <0.2 were in-
cluded in this analysis.
Results
Of the 181 patients included in the study (Table 1), 167
(92.2%) were women. Mean age was 56.1 ± 13.0 years and
mean symptom duration was 6.4 ± 6.9 years. Of the 77
patients given a clinical diagnosis of pSS, 64 (83.1%) ful-
filled AECG criteria. All AECG items were significantly
Cornec et al. Arthritis Research & Therapy 2014, 16:R15 Page 2 of 6
http://arthritis-research.com/content/16/1/R15
associated with a diagnosis of pSS except xerophthalmia
and xerostomia. No differences were found between pSS
and non-pSS patients for age, disease duration, or sex
ratio. Other diagnoses were: idiopathic sicca syndrome
(N = 51), other systemic autoimmune diseases (N = 29),
drug-induced sicca syndrome (N = 21), ill-controlled dia-
betes mellitus (N = 2), hepatitis C virus-related sicca syn-
drome (N = 1).
The mean B-cell ratio was significantly higher in the pSS
group than in the non-pSS group (7.4 ± 6.9 vs. 3.2 ± 2.3,
P<0.001). ROC curve analysis (Figure 2) identified 5as
the best cutoff, with 52.1% Se and 83.2% Sp. The highest
values of this ratio were strongly suggestive of pSS, with a
cutoff 6.5 having 92.6% Sp and a cutoff 9 97.9% Sp (with
42.3% and 28.2% Se, respectively). The correlation of
B-cell ratio 5 with anti-SSA and abnormal SGB findings,
estimated using Cohens kappa coefficient, was moderate
(kappa = 0.35 and 0.30, respectively).
Figure 2 shows the ROC curve analysis for the biological
markers. Their diagnostic value is shown in Table 2. Anti-
SSB antibodies were found in 25 (32.5%) patients with
pSS, all of whom also had anti-SSA antibodies. No patient
had anti-SSA without ANA. ACR 2012 criteria serological
item (anti-SSA/B or (ANA 1:320 + positive RF)) did not
perform better than anti-SSA/B alone, but gave more false
positive results.
Thirty patients were diagnosed as pSS but were anti-
SSA negative. Among them, three patients had a normal
SGB, and the diagnosis of pSS was made based on subject-
ive and objective ocular and mouth dryness without other
explanation, high-titer ANA, and suggestive extraglan-
dular manifestations (cutaneous vasculitis, peripheral
neuropathy, interstitial pneumonitis or cytopenia). In this
group of anti-SSA negative pSS patients, the frequency of
the different tests was: 56.7% for ANA 1:320; 40.0% for
ANA 1:640; 20.0% for IgM-RF; 10.0% for ACR 2012
criteria serological item (anti-SSA/B or (ANA 1:320 +
positive RF)); 10.0% for gammaglobulins 14 g/l; 16.7% for
IgG 14 g/l; and 40.0% for B-cell ratio 5.
By logistic regression analysis, only anti-SSA anti-
bodies, ANA 1:640 and B-cell ratio 5 showed inde-
pendent associations with pSS. Using both ANA 1:640
and B-cell ratio 5 in combination had 37.7% Se and
96.2% Sp in the overall population but only 12.9% Se in
the anti-SSA-negative subset. Using either ANA 1:640
or B-cell ratio 5 had 85.7% Se and 67.3% Sp in the
overall group and 71.0% Se in the anti-SSA-negative
subgroup.
Modifying the AECG criteria set by adding either
ANA 1:640 or B-cell ratio 5 increased Se from 83.1%
to 90.9%, but decreased Sp from 97.1% to 85.6%. Adding
both ANA 1:640 and B-cell ratio 5 did not signifi-
cantly modify the diagnostic performance of the criteria
set (Se 84.2% vs. 83.1% and Sp 96.1% vs. 97.1%).
Discussion
We assessed the diagnostic performance of autoimmu-
nity and B-cell-related markers for pSS. Although ANA
and RF positivity, hypergammaglobulinemia, IgG eleva-
tion, and altered peripheral-blood B-cell subset distri-
bution were far more common in the patients with pSS
compared to those with other causes of sicca symptoms,
these abnormalities correlated closely with anti-SSA
positivity. Only ANA 1:640 and B-cell ratio 5 were
Figure 1 Examples of typical blood B-cell subset profiling by
flow cytometry. All analyses are gated on CD19+ B cells. A ratio is
computed between Bm2 + Bm2(immunoglobulin (Ig)D+/CD38low-high
population) and eBm5 + Bm5 (IgD-/CD38low-negative population)
subset proportions. (a) Primary Sjögrens syndrome patient, with a ratio
of 17.9. (b) Idiopathic sicca patient, with a ratio of 2.2.
Cornec et al. Arthritis Research & Therapy 2014, 16:R15 Page 3 of 6
http://arthritis-research.com/content/16/1/R15
Table 1 Comparison of pSS and non-pSS patients
Overall pSS No pSS Pvalue
n = 181 n = 77 n = 104
Age (years, mean ± SD) 56.1 ± 13.0 56.3 ± 13.5 55.9 ± 12.7 0.89
Symptom duration (years, mean ± SD) 6.4 ± 6.9 7.0 ± 7.6 6.0 ± 6.3 0.48
Female, n (%) 167 (92.2) 70 (90.9) 97 (93.3) 0.56
Xerophthalmia, n (%) 156 (86.2) 70 (90.9) 86 (82.7) 0.11
Xerostomia, n (%) 166 (91.7) 73 (94.8) 93 (89.4) 0.19
Abnormal Schirmers test, n (%) 76 (42.0) 44 (57.1) 32 (30.8) <0.001
Decreased salivary flow, n (%) 80 (44.2) 48 (62.3) 32 (30.8) <0.001
Abnormal salivary gland biopsy, n (%) 75 (41.4) 62 (80.5) 13 (12.5) <0.001
Anti-SSA or -SSB positivity, n (%) 47 (26.0) 47 (61.0) 0 (0.0) <0.001
AECG criteria, n (%) 67 (37.0) 64 (83.1) 3 (2.9) <0.001
Anti-SSA positivity, n (%) 47 (26.0) 47 (61.0) 0 (0.0) <0.001
Anti-SSB positivity, n (%) 25 (13.8) 25 (32.5) 0 (0.0) <0.001
ANA 1:320, n (%) 105 (58.0) 62 (80.5) 43 (41.3) <0.001
ANA 1:640, n (%) 73 (40.3) 54 (70.1) 19 (18.3) <0.001
IgM-RF positivity, n (%) 56 (30.9) 35 (45.5) 21 (20.2) <0.001
IgA-RF positivity, n (%) 37 (20.4) 32 (41.6) 5 (4.8) <0.001
Gammaglobulins 14 g/L, n (%) 44 (24.3) 36 (46.8) 8 (7.7) <0.001
IgG 14 g/L, n (%) 47 (26.0) 38 (49.4) 9 (8.7) <0.001
B-cell ratio 5, n (%) 59 (32.6) 41 (53.2) 18 (17.3) <0.001
ANA 1:320 + IgM-RF, n (%) 39 (21.5) 32 (41.6) 7 (6.7) <0.001
ACR criteria serologic item, n (%) 56 (30.9) 49 (63.6) 7 (6.7) <0.001
pSS, primary Sjögrens syndrome; AECG, American-European Consensus Group; ANA, antinuclear antibodies; Ig, immunoglobulin; RF, rheumatoid factor; ACR criteria
serologic item: (anti-SSA/B or (ANA 1:320 + IgM-RF)). Xerophthalmia and xerostomia referred to subjective complaints by the patients. Schirmers test was considered
abnormal if 5 mm/5 min and unstimulated salivary flow if 0.1 mL/min. Salivary gland biopsies were graded according to Chisholm and Mason. MannWhitney and
chi-square tests were used as appropriate to compare pSS and non-pSS patients. P<0.05 was considered significant.
Figure 2 ROC curve analysis. The best cutoff for each test is chosen as the best combination of sensitivity and specificity. ROC, receiver-operating
characteristic; ANA, antinuclear antibodies; RF, rheumatoid factor; Ig, immunoglobulin.
Cornec et al. Arthritis Research & Therapy 2014, 16:R15 Page 4 of 6
http://arthritis-research.com/content/16/1/R15
associated with pSS independently from anti-SSA anti-
bodies. However, adding these two tests to the currently
used AECG classification criteria did not improve their
diagnostic performance.
Classification criteria are developed as research tools
for establishing homogeneous patient groups with well-
defined conditions and for comparing different studies,
but they are widely used in clinical practice for diagnos-
tic purposes. However, clinicians may diagnose a disease
in patients who do not meet the classification criteria,
especially those with recent-onset or mild symptoms.
We studied a cohort of patients who reflected the diag-
nostic conditions encountered in everyday clinical prac-
tice, since they were referred to a specialized center
for the evaluation of suspected pSS. The diagnosis was
established by consensus among three experts indepen-
dently from the classification criteria. This study design
allowed us to assess the diagnostic performance of vari-
ous tests and of the AECG criteria in the real-life setting.
We previously used this methodology to study the diag-
nostic performance of salivary gland ultrasonography for
pSS [12].
However, this methodology induces mandatorily a cer-
tain part of circular reasoning, since the experts in their
own minds gave probably more weight for their diagno-
sis to known and objective features of pSS, such as anti-
SSA antibodies or SGB biopsy results. If the experts
would have taken into account the results of B-cell ratio
(to which they were blinded) to make their diagnosis,
they would probably have considered some patients as
pSS due to a high B-cell ratio, which would have im-
proved the diagnostic value of the test in this study.
Flow cytometry is simple, reproducible, and widely avai-
lable. A high (Bm2 + Bm2)/(eBm5 + Bm5) ratio is far
more common in pSS both compared to other rheumatic
diseases, as reported in our previous study [11], and
compared to sicca syndromes not due to pSS, as shown
here. Higher ratio values are associated with a higher
probability of pSS. Thus, in the individual patient, this test
is valuable. However, its diagnostic weight seems small
compared to the other items of the AECG classification
criteria set.
The abnormal distribution of blood B-cell subsets dur-
ing pSS has previously been explored in regard to the
disease pathophysiology. The decrease in blood memory
B cells in pSS patients may be explained by their accu-
mulation within salivary glands. Indeed, this variation
partly reflects their migration into the exocrine glands of
the patients [7], as well as into their skin [14]. We had
previously observed that Bm2/Bm2cells express CD19
at 55% and 61% higher levels than the same cell subsets
in normal controls [8]. Such marked increases could
be related to pathogenesis, since modest increases in the
density of CD19 are sufficient to shift the balance bet-
ween tolerance and autoimmunity [15]. To address the
functional significance of the observed phenotype ano-
malies, the dynamic translocation of protein into lipid
rafts was also explored, since these domains are critical
for proximal B cell receptor (BCR) signal transduction.
The results revealed that the association of the BCR with
the lipid rafts was prolonged in pSS. This may be
accounted for by the overexpression of CD19, which
prolongs signaling or prevents the recruitment of nega-
tive regulators, whether such regulators are insufficient
(CD32) or altered (CD45) [8]. As such, changes in the
lipid raft dynamics might lead to an aberrant B-cell
response in pSS. Finally, these perturbations in B-cell
homeostasis in pSS are closely correlated with the dys-
regulation of various cytokines regulating B-cell survival
and activation, such as BAFF [8] and FLT3L [16].
ANA are frequent in patients with pSS. In the new
preliminary ACR classification criteria set proposed by
Shiboski et al. [5], the required ANA titer is 1:320, but
this cutoff was chosen by consensus among experts and
the article does not report a detailed Se and Sp analysis.
In our study, the higher ANA titer of 1:640 was re-
quired to obtain the best combination of Se and Sp, and
was associated with pSS independently from anti-SSA
positivity. RF, and especially IgA-RF, are far more fre-
quent in patients with pSS compared to controls. How-
ever, this test correlates strongly with anti-SSA positivity.
Considering ANA titer and RF positivity beside anti-SSA
for the diagnosis of pSS do not seem useful.
Conclusions
This study shows that biological evidence of auto-
immunity is common in patients with pSS but is closely
related to the presence of anti-SSA antibodies. Anti-SSA
remains the main serological tool for diagnosing pSS.
Other serological markers should be tested in cohorts of
Table 2 Diagnostic value of the biological items
Sensitivity Specificity PPV NPV
Anti-SSA or -SSB positivity 61.0% 100% 100% 79.9%
Anti-SSA positivity 61.0% 100% 100% 79.9%
Anti-SSB positivity 32.5% 100% 100% 66.7%
ANA 1:320 80.5% 58.7% 59.0% 80.3%
ANA 1:640 70.1% 81.7% 74.0% 78.7%
IgM-RF positivity 45.5% 79.8% 62.5% 66.4%
IgA-RF positivity 41.6% 95.2% 86.5% 68.8%
Gammaglobulins 14 g/L 46.8% 92.3% 81.8% 70.1%
IgG 14 g/L 49.4% 91.3% 80.9% 70.9%
B-cell ratio 5 53.2% 82.7% 69.5% 70.5%
ACR criteria serologic item 63.6% 93.7% 87.5% 77.6%
PPV, positive predictive value; NPV, negative predictive value; ANA, antinuclear
antibodies; Ig, immunoglobulin; RF, rheumatoid factor; American College of
Rheumatology (ACR) criteria serologic item: (anti-SSA/B or (ANA 1:320 + IgM-RF)).
Cornec et al. Arthritis Research & Therapy 2014, 16:R15 Page 5 of 6
http://arthritis-research.com/content/16/1/R15
patients with suspected pSS. The development of new
classification criteria for pSS has been the focus of an
international debate since the publication of the ACR
2012 criteria [17,18], and we believe that our cohort of
patients with suspected pSS should be useful to validate
such criteria.
Abbreviations
ACR: American College of Rheumatology; AECG: American-European
Consensus Group; ANA: antinuclear antibody; Ig: immunoglobulin;
pSS: primary Sjögrens syndrome; RF: rheumatoid factor; ROC: receiver-
operating characteristic; Se: sensitivity; SGB: salivary gland biopsy;
Sp: specificity.
Competing interests
The authors have no competing interests to declare concerning this work.
Authorscontributions
DC, AS and VDP designed the study. DC, AS, JOP, SJJ, TM, AMRC, SG, YR and
VDP participated in the management of the patients and their recruitment in
the cohort, and in the acquisition of data. JOP and YR performed the
biological assays. DC and AS performed the statistical analysis. DC and VDP
drafted the manuscript. AS, JOP, SJJ, TM, AMRC, SG and YR critically revised
the manuscript. All authors read and approved the final manuscript, and
agree to be accountable for all aspects of the work.
Author details
1
Service de Rhumatologie, Centre Hospitalier Régional et Universitaire de
Brest, Hôpital de la Cavale Blanche, BP 824, F-29609 Brest cedex, France.
2
EA
2216 Immunologie et Pathologie, Université de Brest, SFR ScinBios, Labex
Imunotherapy, Graft, Oncology, BP 824, F-29609 Brest cedex, France.
3
Service
de Dermatologie, Centre Hospitalier Régional et Universitaire de Brest,
Hôpital Morvan, BP 824, F-29609 Brest cedex, France.
4
Explorations
Fonctionnelles Neurologiques, Centre Hospitalier Régional et Universitaire de
Brest, Hôpital de la Cavale Blanche, BP 824, F-29609 Brest cedex, France.
Received: 29 July 2013 Accepted: 30 December 2013
Published: 17 January 2014
References
1. Cornec D, Devauchelle-Pensec V, Tobón GJ, Pers JO, Jousse-Joulin S, Saraux A:
B cells in Sjögrens syndrome: from pathophysiology to diagnosis and
treatment. JAutoimmun2012, 39:161167.
2. Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE,
Daniels TE, Fox PC, Fox RI, Kassan S: Classification criteria for Sjögrens
syndrome: a revised version of the European criteria proposed by the
American-European Consensus Group. Ann Rheum Dis 2002, 61:554558.
3. Fauchais AL, Martel C, Gondran G, Lambert M, Launay D, Jauberteau MO,
Hachulla E, Vidal E, Hatron PY: Immunological profile in primary Sjögren
syndrome: clinical significance, prognosis and long-term evolution to
other auto-immune disease. Autoimmun Rev 2010, 9:595599.
4. Peene I, Meheus L, Veys EM, De Keyser F: Diagnostic associations in a
large and consecutively identified population positive for anti-SSA
and/or anti-SSB: the range of associated diseases differs according to
the detailed serotype. Ann Rheum Dis 2002, 61:10901094.
5. Shiboski SC, Shiboski CH, Criswell LA, Baer AN, Challacombe S, Lanfranchi H,
Schiødt M, Umehara H, Vivino F, Zhao Y, Dong Y, Greenspan D, Heidenreich AM,
Helin P, Kirkham B, Kitagawa K, Larkin G, Li M, Lietman T, Lindegaard J,
McNamara N, Sack K, Shirlaw P, Sugai S, Vollenweider C, Whitcher J, Wu A,
Zhang S, Zhang W, Greenspan JS, et al:American College of Rheumatology
classification criteria for Sjögrenssyndrome:adatadriven, expert consensus
approach in the Sjögrens International Collaborative Clinical Alliance cohort.
Arthritis Care Res 2012, 64:475487.
6. Bendaoud B, Pennec YL, Lelong A, Le Noach JF, Magadur G, Jouquan J,
Youinou P: IgA-containing immune complexes in the circulation of
patients with primary Sjögrens syndrome. J Autoimmun 1991, 4:177184.
7. Hansen A, Odendahl M, Reiter K, Jacobi AM, Feist E, Scholze J, Burmester
GR, Lipsky PE, Dörner T: Diminished peripheral blood memory B cells and
accumulation of memory B cells in the salivary glands of patients with
Sjögrens syndrome. Arthritis Rheum 2002, 46:21602171.
8. DArbonneau F, Pers JO, Devauchelle V, Pennec Y, Saraux A, Youinou P:
BAFF-induced changes in B cell antigen receptor-containing lipid rafts in
Sjögrens syndrome. Arthritis Rheum 2006, 54:115126.
9. Pascual V, Liu YJ, Magalski A, de Bouteiller O, Banchereau J, Capra JD:
Analysis of somatic mutation in five B cell subsets of human tonsil.
J Exp Med 1994, 180:329339.
10. Bohnhorst JØ, Bjørgan MB, Thoen JE, Natvig JB, Thompson KM: Bm1-Bm5
classification of peripheral blood B cells reveals circulating germinal
center founder cells in healthy individuals and disturbance in the B cell
subpopulations in patients with primary Sjögrens syndrome. J Immunol
2001, 167:36103618.
11. Binard A, Le Pottier L, Devauchelle-Pensec V, Saraux A, Youinou P, Pers J-O:
Is the blood B-cell subset profile diagnostic for Sjogren syndrome?
Ann Rheum Dis 2009, 68:14471452.
12. Cornec D, Jousse-Joulin S, Pers J-O, Marhadour T, Cochener B, Boisramé-Gastrin S,
Nowak E, Youinou P, Saraux A, Devauchelle-Pensec V: Contribution of salivary
gland ultrasonography to the diagnosis of Sjögrens syndrome: toward new
diagnostic criteria? Arthritis Rheum 2013, 65:216225.
13. Chisholm DM, Mason DK: Labial salivary gland biopsy in Sjögrens disease.
J Clin Pathol 1968, 21:656660.
14. Roguedas AM, Pers JO, Lemasson G, Devauchelle V, Tobón GJ, Saraux A,
Misery L, Youinou P: Memory B-cell aggregates in skin biopsy are
diagnostic for primary Sjögrens syndrome. J Autoimmun 2010,
35:241247.
15. Sato S, Hasegawa M, Fujimoto M, Tedder TF, Takehara K: Quantitative
genetic variation in CD19 expression correlates with autoimmunity.
J Immunol 2000, 165:66356643.
16. Tobón GJ, Renaudineau Y, Hillion S, Cornec D, Devauchelle-Pensec V,
Youinou P, Pers JO: The Fms-like tyrosine kinase 3 ligand, a mediator of
B cell survival, is also a marker of lymphoma in primary Sjögrens
syndrome. Arthritis Rheum 2010, 62:34473456.
17. Vitali C, Bootsma H, Bowman SJ, Dorner T, Gottenberg J-E, Mariette X,
Ramos-Casals M, Ravaud P, Seror R, Theander E, Tzioufas AG: Classification
criteria for Sjogrens syndrome: we actually need to definitively resolve
the long debate on the issue. Ann Rheum Dis 2013, 72:476478.
18. Bootsma H, Spijkervet FKL, Kroese FGM, Vissink A: Toward new
classification criteria for Sjögrens syndrome? Arthritis Rheum 2013,
65:2123.
doi:10.1186/ar4442
Cite this article as: Cornec et al.:Diagnostic accuracy of blood B-cell
subset profiling and autoimmunity markers in Sjögrens syndrome.
Arthritis Research & Therapy 2014 16:R15.
Submit your next manuscript to BioMed Central
and take full advantage of:
Convenient online submission
Thorough peer review
No space constraints or color figure charges
Immediate publication on acceptance
Inclusion in PubMed, CAS, Scopus and Google Scholar
Research which is freely available for redistribution
Submit your manuscript at
www.biomedcentral.com/submit
Cornec et al. Arthritis Research & Therapy 2014, 16:R15 Page 6 of 6
http://arthritis-research.com/content/16/1/R15
... Binard studied the effect of adding a naiveto-mature B-cells' ratio to the 2002 AECG classification criteria, reporting an increase in their performance for the diagnosis 20 . However, in a later study, these parameters performed poorly in a distinct clinical setting 21 . ...
... It has been previously reported that a high (Bm2 + Bm2′)/(eBm5 + Bm5) ratio is more frequent in SjS both compared to RA, SLE, Sicca patients and healthy subjects, and could constitute a diagnostic tool 20,21 . However, Cornec et al 21 reported that evaluating this ratio, although valuable for the individual patient, had a small diagnostic weight compared to other items of AECG CC, as adding an item consisting of a (Bm2 + Bm2′)/ (eBm5 + Bm5) ratio ≥ 5 did not significantly modify the performance of the criteria. ...
... It has been previously reported that a high (Bm2 + Bm2′)/(eBm5 + Bm5) ratio is more frequent in SjS both compared to RA, SLE, Sicca patients and healthy subjects, and could constitute a diagnostic tool 20,21 . However, Cornec et al 21 reported that evaluating this ratio, although valuable for the individual patient, had a small diagnostic weight compared to other items of AECG CC, as adding an item consisting of a (Bm2 + Bm2′)/ (eBm5 + Bm5) ratio ≥ 5 did not significantly modify the performance of the criteria. ...
Article
Full-text available
Sjögren's Syndrome (SjS) is a chronic systemic immune-mediated inflammatory disease characterized by lymphocytic infiltration and consequent lesion of exocrine glands. SjS diagnosis and classification remains a challenge, especially at SjS onset, when patients may have milder phenotypes of the disease or uncommon presentations. New biomarkers are needed for the classification of SjS, thus, we aimed to evaluate the added-value of lymphocyte subpopulations in discriminating SjS and non-Sjögren Sicca patients. Lymphocyte subsets from 62 SjS and 63 Sicca patients were characterized by flow cytometry. The 2002 AECG and the 2016 ACR/EULAR SjS classification criteria were compared with clinical diagnosis. The added discriminative ability of joining lymphocytic populations to classification criteria was assessed by the area under the Receiver-Operating-Characteristic Curve (AUC). Considering clinical diagnosis as the gold-standard, we obtained an AUC = 0.952 (95% CI: 0.916–0.989) for AECG and an AUC = 0.921 (95% CI: 0.875–0.966) for ACR/EULAR criteria. Adding Tfh and Bm1 subsets to AECG criteria, performance increased, attaining an AUC = 0.985 (95% CI: 0.968–1.000) (p = 0.021). Th1/Breg-like CD24hiCD27⁺ and switched-memory B-cells maximized the AUC of ACR/EULAR criteria to 0.953 (95% CI: 0.916–0.990) (p = 0.043). Our exploratory study supports the potential use of lymphocyte subpopulations, such as unswitched memory B cells, to improve the performance of classification criteria, since their discriminative ability increases when specific subsets are added to the criteria.
... Regarding B cells, a profound imbalance between the different subsets of B cells has been described in pSS, with a decrease in the frequencies and absolute counts of memory B cells, and an increase in those of naïve B cells [16,[18][19][20][21][22][23]. Moreover, it has been proposed that the ratios between immature and mature B cells could be used for diagnostic purposes in pSS [24][25][26]. ...
... Given these imbalances in circulating B lymphocyte subsets, some authors have explored the accuracy of the blood B cell subsets profile for diagnostic purposes [24][25][26]. In our study we explored the potential utility of the complete lymphocyte profile to discriminate between pSS and sicca syndrome patients. ...
... In our study we explored the potential utility of the complete lymphocyte profile to discriminate between pSS and sicca syndrome patients. Related to B cells, we found that the best diagnostic parameter for differentiating the two diseases was the ratio between the percentage of naïve B cells and non-switched memory B cells, which showed a slightly poorer diagnostic accuracy than the analogous ratio (Bm2 þ Bm2 0 )/ (eBm5 þ Bm5) previously described [24,25]. However, the ratio of B NSM /CD4 ACT percentages not only exhibited better diagnostic accuracy for pSS diagnosis than the ratios between B cell subpopulations, but also allowed us to discriminate between seronegative pSS and sicca syndrome. ...
Article
Background: blood B cell profile has been proposed to have diagnostic utility in primary Sjögren syndrome (pSS), but the potential utility of advanced lymphocyte profiling to differentiate between pSS and Sicca syndrome has not been fully investigated. Methods: distribution of peripheral lymphocyte subpopulations was analysed by flow cytometry in 68 patients with pSS, 26 patients with Sicca syndrome and 23 healthy controls. The ability to discriminate between pSS and Sicca syndrome was analysed using the area under the curve (AUC) of the receiver operating characteristic curve of the different lymphocyte subsets. Results: the ratio between naïve/memory B cell proportions showed an AUC of 0.742 to differentiate pSS and Sicca syndrome, with a sensitivity of 76.6% and a specificity of 72% for a cut-off value of 3.4. The ratio of non-switched memory B cells to activated CD4+ T cells percentage (BNSM/CD4ACT) presented the highest AUC (0.840) with a sensitivity of 83.3% and specificity of 81.7% for a cut-off value < 4.1. To differentiate seronegative pSS patients from Sicca patients the BNSM/CD4ACT ratio exhibited an AUC of 0.742 (sensitivity 75%, specificity 66.7%, cut-off value < 4.4), and the number of naïve CD4 T cells had an AUC of 0.821 (sensitivity 76.9%, specificity 88.9%, cut-off value < 312/mm3). Conclusion: patients with pSS show a profound imbalance in the distribution of circulating T and B lymphocytes subsets. The ratio BNSM/CD4ACT is useful to discriminate between pSS and Sicca syndrome.
... To exclude a low titer ANA positivity, the ANA positivity was initially defined in our cohort of pSS patients as a 1:320 titer or higher due to the lack of specificity in the old people or subjects with other chronic diseases. Further, it was suggested that the 1:320 of ANA positivity and RF (particularly IgA type of RF) were related to the presence of anti-SSA antibodies 20 In our study, the high titer ANA positivity was common in the H. pylori-infected pSS patients. In this context, the ANA-positive subgroup showed augmented levels of IgG, RF, and LSGB, consistent with the previous reports. ...
Article
Full-text available
Objective Although infectious pathogens are predominant factors for inducing and maintaining immune system disorders, there exist few reports establishing the significant correlation between Helicobacter pylori ( H. pylori ) infection and Sjogren's syndrome. This study aims to demonstrate the correlation between Sjogren's syndrome and H. pylori infection in patients, highlighting various clinical characteristics and risk factors. Methods A single‐center retrospective observational study was conducted in patients ( n = 224) admitted from January 1, 2012, to February 10, 2021, in the First Affiliated Hospital of Wenzhou Medical University (Wenzhou, China). All the recruited subjects with Sjogren's syndrome and H. pylori infection were only included by validating the available medical records online. Results In this study, a total of 224 patients from January 1, 2012, to February 10, 2021, were diagnosed with Sjogren's syndrome. Among them, 94 patients (41.96%) with Sjogren's syndrome were infected with H. pylori . Accordingly, the clinical manifestations, serological and immunological characteristics, as well as gastroscopic biopsy outcomes of the recruited patients with primary Sjogren's syndrome (pSS) were reported. The multivariable analysis of the dry syndrome patients infected with H. pylori displayed hypergammaglobulinemia (odds ratio [OR], 0.354; 95% confidence interval [CI], 0.189‐0.663), total cholesterol (OR, 1.158; 95% CI, 0.856‐1.550), hypertension (OR, 0.227; 95% CI, 0.114‐0.455), Female sex (OR, 5.778; 95% CI, 1.458‐22.9), anti‐SSA/Ro60 positive (OR, 2.384; 95% CI, 233‐4.645), γ‐GT (OR, 0.99; 95% CI, 0.99‐1.00) and alkaline phosphatase (ALP, OR, 1.00; 95% CI, 0.99–1.00) levels. Conclusion Together, our findings demonstrated that hypergammaglobulinemia could be the independent risk factors of H. pylori infection in patients with Sjogren's syndrome, requiring the physician's advice in the future.
... A UWS ≤0.1 ml/min was deemed abnormal (1,2). Serum levels of anti-SSA/Ro antibodies, anti-Sjögren's syndrome B (SSB) antibodies, rheumatoid factor (RF), and antinuclear antibody (ANA) were assessed with ELISA tests (27) The parotid glands were scored using a simplified salivary gland ultrasonography (SGUS) scoring system (6). Parenchymal homogeneity in salivary glands was scored 0-3 (0, normal; 1, mild inhomogeneity interpreted as normal or unspecific; 2, several rounded; 3, numerous or confluent hypoechoic lesions) and a score of 2 or 3 indicated typical pSS. ...
Article
Full-text available
Purpose The study assessed the validity of impression cytology (IC) and in vivo confocal microscopy (IVCM) of lip mucosa compared with labial gland biopsy, anti-Sjögren’s syndrome A (SSA)/Ro antibody status, and classification criteria in suspected primary Sjögren’s syndrome (pSS) patients. Methods Clinically suspected pSS patients (n = 201) were enrolled consecutively and were divided into pSS (n = 56) and control (n = 145, only with dryness) groups according to the American College of Rheumatology-European League Against Rheumatism (ACR-EULAR) criteria. All patients underwent lip mucosa IC (inflammatory cell density) and IVCM (epithelium/intrinsic layer thickness and labial gland density/diameter) analyses. The associations between IC/IVCM parameters and clinical/laboratory results were analyzed. Results The absolute agreement between positive lip mucosal IC (≥50 cells/4 mm²) and the ACR-EULAR criteria (94.5%)/labial gland biopsy (95.5%) was good, with sensitivities of 82.1 and 85.2%, respectively, and a specificity of 99.3%. Compared with controls, IVCM revealed significant lip mucosal atrophy and glandular decreases in the pSS group (all P = 0.000). The sensitivities for diagnosing pSS corresponding to a lamina propria thickness ≤128 μm and a gland diameter ≤114 μm were 85.7 and 89.3%; the specificities were 90.3 and 95.9%, respectively. A combination of positive IC/IVCM and anti-SSA/Ro antibody results showed a high predictive value for diagnosing pSS. Conclusions IC and IVCM could detect distinctive cellular and morphological changes in the lip mucosa of patients with pSS. These noninvasive and easy-to-perform examinations may be an alternative to labial gland biopsy for diagnosing pSS.
... Bharaj et al. reported that the accumulation of CD20þ B cells and a decline in CD138þ plasma cells reflected the high inflammatory disease severity in the minor salivary gland (MSG) tissue as they may be recruited into glandular tissues as inflammation developed [37]. We found that memory B cells including unswitched and switched memory B cells experienced a considerable reduction in peripheral blood, reflecting their likely accumulation in exocrine glands and skins of pSS patients and a probable differentiating towards [38,39]. Considering the reduced proportion of B10 cells followed by reduced secretion of IL10, their failure to restrain type I interferons coupled with the hyperactivation of plasmacytoid dendritic cells (pDCs) might engage in the pathogenesis of pSS [40]. ...
Article
Full-text available
Objectives Detailed analysis targeting B cell subgroups was considered crucial in monitoring autoimmune diseases and treatment responses. Thus, precisely describing the phenotypes of B cell differentiation and their variation in primary Sjögren’s syndrome (pSS) is particularly needed. Methods To characterize the proportions and absolute counts of B cell subsets, peripheral blood from 114 healthy adults of China (age range: 19–73 years) and 55 patients with pSS were performed by flow cytometry and CD19, CD20, CD24, CD27, CD38 and IgD were used as surface markers to identify B cell mature process. Age- and gender-stratified analyses were then carried out to improve the interpretation of B cell subsets. Results The assessments from healthy adults showed that the proportion of naive B cells presented a significant increase with age. A reversal trend was noted that the percentage of B10 decreased markedly with age. In addition, analysis based on gender showed that the relative percentage and number of naive B cells were higher in females than in males whereas the proportions of switched memory B cells and B10 cells were decreased in female. Patients with pSS exhibited a significant expansion in naïve B cells and unswitched memory B cells, accompanied with decreased switched memory B cells and B10 cells, which were identified to be associated with autoantibody production. Conclusions Our study presented a reliable analysis by flow cytometry to cover the principal B cell subtypes. These different stages of B lymphocytes may have implications for evaluating the activation of pSS and other autoimmune diseases and treatment efficacy. KEY MESSAGES B cell subsets play a pivotal role in the pathogenesis of primary Sjögren’s syndrome (pSS) and other autoimmune diseases. A practical and accurate flow cytometry method to profile B cell phenotypes in peripheral blood of healthy adults is especially essential. Additionally, we presented reliable reference ranges for B cell subsets in regards to the local population. Age- and gender-related analyses are available to better understand their influence in immune status and treatment outcome. The distribution of B-cell subsets is found substantially altered in patients with pSS, bringing novel avenues for pSS research in the future.
... A recent study has demonstrated that activated naive B cells could generate highly polyclonal antibody-secreting cells and could be responsible for serum autoantibody repertoire in lupus (53). From several studies it has become clear that the decreased ratio of peripheral memory B cells may be interpreted by the upregulation of CXCR4 and CXCR3 chemokines which arrange their recruitment into the inflamed tissues of affected organs (3,(54)(55)(56)(57). Ectopic expression of chemokines, including CXCL13, CCL21, and CXCL12 not only induces the migration and survival of memory B cells and long-lived plasma cells, but supports lymphoid neogenesis in the pSS salivary glands (58). ...
Article
Full-text available
Since B-cell hyperactivity and pathologic antibody response are key features in the immunopathogenesis of primary Sjögren’s syndrome (pSS), the role of follicular T helper (TFH) cells as efficient helpers in the survival and differentiation of B cells has emerged. Our aim was to investigate whether a change in the balance of circulating (c)TFH subsets and follicular regulatory T (TFR) cells could affect the distribution of B cells in pSS. Peripheral blood of 38 pSS patients and 27 healthy controls was assessed for the frequencies of cTFH cell subsets, TFR cells, and certain B cell subpopulations by multicolor flow cytometry. Serological parameters, including anti-SSA, anti-SSB autoantibodies, immunoglobulin, and immune complex titers were determined as part of the routine diagnostic evaluation. Patients with pSS showed a significant increase in activated cTFH cell proportions, which was associated with serological results. Frequencies of cTFH subsets were unchanged in pSS patients compared to healthy controls. The percentages and number of cTFR cells exhibited a significant increase in autoantibody positive patients compared to patients with seronegative pSS. The proportions of transitional and naïve B cells were significantly increased, whereas subsets of memory B cells were significantly decreased and correlated with autoantibody production. Functional analysis revealed that the simultaneous blockade of cTFH and B cell interaction with anti-IL-21 and anti-CD40 antibodies decreased the production of IgM and IgG. Imbalance in TFH subsets and TFR cells indicates an ongoing over-activated humoral immune response, which contributes to the characteristic serological manifestations and the pathogenesis of pSS.
... Adding SGUS improved the sensitivity of the classification criteria for detecting pSS, with little or no loss of specificity. To our knowledge, no other item has been shown to improve the performance of pSS classification criteria sets in groups of patients [29]. ...
Article
Full-text available
Objective: Major Salivary gland ultrasonography (SGUS) is widely used for the diagnosis of primary Sjögren's syndrome (pSS). Our objective was to assess the contribution of SGUS compared to other items of the 2016 ACR/EULAR pSS classification criteria, based on expert opinion. Methods: A secure web-based relational database was used by 24 experts from 14 countries to assess 512 realistic vignettes developed from data of patients with suspected pSS. Each vignette provided classification criteria items and information on history, clinical symptoms, and SGUS findings. Each expert assessed 64 vignettes and each vignette was assessed by 3 experts. A diagnosis of pSS was defined according to at least 2 of 3 experts. Validation was performed in the independent French DiapSS cohort of patients with suspected pSS. Results: A criteria-based pSS diagnosis and SGUS findings were independently associated with an expert diagnosis of pSS (p<0.001) The derived diagnostic weights of individual items in the 2016 ACR/EULAR criteria including SGUS were : anti-SSA, 3; focus score ≥1, 3; SGUS score ≥2, 1; positive Schirmer's test, 1; dry mouth, 1; and salivary flow rate <0.1 mL/min, 1. The corrected C statistic area under the curve for the new weighted score was 0.96.Adding SGUS improve the sensitivity from 90.2 % to 95.6% with a quite similar specificity 84.1% versus 82.6%. Results were similar in the DiapSS cohort: adding SGUS improve the sensitivity from 87% to 93%. Conclusion: SGUS had similar weight compared to minor items and its addition improves the performance of the 2016 ACR/EULAR classification criteria.
Article
Full-text available
Primary Sjögren’s syndrome (pSS) is a systemic autoimmune epithelitis and recent advances in our comprehension of its pathophysiology strongly suggest a multi-step process that involves environmental factors (e.g. chronic viral infection, drugs), followed by deregulation of the epigenetic machinery (e.g. DNA demethylation, histone modifications, microRNAs), which in turn specifically affects lymphocytes and epithelial cells leading to an aberrant inflammation. This process is amplified in the case of genetic mutations. As a consequence, autoreactive lymphocytes and autoantigens are produced leading to the development of autoantibodies. Moreover, it was observed that epigenetic modifications in pSS could be reversed, thus providing arguments to suggest that therapeutic strategies targeting the epigenetic deregulation and in particular the PKC-delta/Erk/DNMT1 pathway would be effective in pSS.
Thesis
Les LB jouent un rôle central dans le syndrome de Sjögren primitif (SSp), cependant très peu d’études ont été réalisées sur les caractéristiques des LB auto-réactifs qui sécrètent des autoanticorps contre l'auto-antigène SSA.Nous avons cherché à caractériser les LB circulants spécifiques de la SSA (LB SSA+), afin de mieux comprendre les mécanismes conduisant à la rupture de la tolérance chez les patients atteints du SSp.En utilisant une méthode de cytométrie en flux, basée sur la spécificité du BCR, nous avons détecté et caractérisé phénotypiquement les LB SSA+ circulants chez des patients atteints de SSp et chez des contrôles sains (HCs). Nous avons testé leur capacité à sécréter des immunoglobulines spécifiques de la SSA in vitro après stimulation. Ensuite, nous avons réalisé une première expérience de profilage transcriptionnel de ces cellules par RNA seq unicellulaire afin de mieux comprendre le rôle des LB auto-réactifs chez les sujets sains.L'analyse du phénotype a montré une expansion d'un sous-ensemble de LB naïfs CD19+IgD+CD27-CD21low/-CD11c-SSA+ avec un BCR régulé à la baisse chez les patients atteints de SSp.Les LB SSA+ étaient également enrichis parmi les LB mémoires non commutés IgMlow chez les patients, ainsi que dans les LB mémoires DN (IgD-CD27-) et commutés des différents isotypes. Les LB SSA+ triés, provenant d'un sujet sain, étaient capables de sécréter in vitro des immunoglobulines IgM mais pas IgG anti-SSA, contrairement aux LB SSA+ provenant de patients atteints de SSp, qui sont capables de sécréter des autoanticorps IgG spécifiques de SSA. Sur le plan transcriptomtique, les DEG du cluster enrichi en LB SSA+ ont montré une régulation positive des gènes HLA impliqués dans la présentation de l'antigène ainsi qu’une régulation négative du BCR par la surexpression de CD72.Nos résultats suggèrent que les LB SSA+ auto-réactifs, chez les patients atteints de SSP, sont enrichis dans le compartiment mémoire, ce qui suggère qu'ils échappent aux différents points de contrôle de la tolérance périphérique et se différencient in vivo encellules sécrétant des anticorps anti-SSA.
Article
Full-text available
Signaling thresholds influence the balance between humoral immunity and autoimmunity. Cell surface CD19 regulates intrinsic and Ag receptor-induced B lymphocyte signaling thresholds, and transgenic mice that overexpress CD19 by 3-fold generate spontaneous autoantibodies in a genetic background not associated with autoimmunity. To quantify the extent that genetically determined differences in expression of a single cell surface molecule can influence autoantibody production, we have assessed autoimmunity in a C57BL/6-transgenic mouse line with subtle 15-29% increases in CD19 cell surface expression (CD19 trans- genic). Antinuclear Abs, especially anti-spindle pole Abs, rheumatoid factor, and autoantibodies for ssDNA, dsDNA, and histone were produced in these transgenic mice, but not littermate controls. This demonstrates that small changes in CD19 expression can induce autoantibody production. Remarkably, similar changes in CD19 expression were found on B cells from patients with systemic sclerosis, a multisystem disorder of connective tissue with autoantibody production. CD19 density on blood B cells from systemic sclerosis patients was significantly (;20%) higher compared with normal individuals, whereas CD20, CD22, and CD40 expression were normal. These results suggest that modest changes in the expression or function of regulatory molecules such as CD19 may shift the balance between tolerance and immunity to autoimmunity. Thereby autoimmune disease may result from a collection of subtle multigenic alterations that could include incremental density changes in cell surface signaling molecules. The Journal of Immunology, 2000, 165: 6635- 6643.
Article
A new approach for the classification of patients with Sjögren's syndrome (SS) has been recently proposed. Although these new criteria substantially differ from the American European Consensus Group criteria, which have represented the gold standard for the last decade, when compared with each other the two sets show a high statistical degree of agreement. However, the fact that two different criteria to classify patient with SS could be available may introduce some additional difficulties in the scientific communication, making cohorts of patients selected by using different methods less than completely equivalent, and the results of epidemiological studies and therapeutic trials not entirely comparable. Consequently, to reach a consensus agreement on universally accepted classification criteria for SS seems to be a very desirable objective.
Article
Background AECG classification criteria are largely used as diagnostic criteria for primary Sjögren’s syndrome (pSS) in the clinical setting, but their diagnostic value has not been studied in cohorts of patients with suspected pSS since their publication in 2002. They do not include salivary gland ultrasonography (SGUS), which is a non-invasive procedure to assess salivary glands involvement during pSS Objectives The objective of this work was to determine the accuracy of SGUS for diagnosing pSS and to suggest modifications of AECG classification criteria. Methods We conducted this cross-sectional study in a prospective cohort of patients with suspected pSS established between 2006 and 2011. Both parotid and submandibular glands were examined by ultrasonography, and the echostructure of each gland was rated between 0 and 4. We also evaluated the size of the glands, and parotid blood flow by Döppler waveform analysis. The reference standard was a clinical diagnosis of pSS by a group of experts blinded to the results of SGUS. Results Of 158 included patients, 78 patients were diagnosed by the experts as having pSS, including 61 (78.2%) who met AECG criteria. All AECG items were significantly associated with a pSS diagnosis except xerostomy and xerophtalmy. Döppler analysis and measurement of gland size did not display good diagnostic properties. By ROC curve analysis of echostructure scores, the best performance was for the maximal score of one of the four glands in each patient. Optimal cut-off was ≥2/4, with 62.8% sensitivity and 95.0% specificity. The diagnostic value of the echostructure score was similar when the patients were stratified according to the disease duration. A weighted score was constructed using the five variables selected by logistic regression analysis: (salivary flow*1.5) + Schirmer*1.5 + (salivary gland biopsy*3) + (SSA/SSB*4.5) + SGUS*2. By ROC curve analysis, a score ≥5/12.5 had 85.7% sensitivity and 94.9% specificity, compared to 77.9% and 98.7% for AECG criteria (figure). Internal validation through bootstrapping analysis showed good performance of this score. Conclusions SGUS has good diagnostic properties for pSS. Modifications of AECG criteria including the addition of an SGUS score notably improve their performance. Disclosure of Interest None Declared
Article
Primary Sjögren's syndrome (pSS) is a chronic autoimmune systemic disease, characterized by a lymphoplasmocytic infiltration and a progressive destruction of salivary and lachrymal glands, leading to ocular and mouth dryness. T cells were originally considered to play the initiating role in the autoimmune process, while B cells were restricted to autoantibody production. However, recent years have seen growing evidence that the roles of B cells in pSS pathophysiology are multiple, and that these cells may actually play a central role in the development of the disease. B cells are over-stimulated and produce excessive amounts of immunoglobulins and various autoantibodies. Peripheral blood and salivary-gland B-cell subset distribution is altered, leading to the constitution of ectopic germinal centers where auto-reactive clones may escape tolerance checkpoints. B cells control T-cell activation by different means: B effector cells guide Th1 or Th2 differentiation, whereas regulatory B cells inhibit T-cell proliferation. Several B-cell specific cytokines, such as BAFF or Flt-3L, are instrumental in the occurrence of B-cell dysfunction. Chronic and excessive stimulation of B cells may lead to the development of lymphoma in pSS patients. Autoantibodies and blood B-cell subset analysis are major contributors of a clinical diagnosis of pSS. These considerations led to the development of B-cell depletion therapies for the management of pSS. Rituximab, a monoclonal antibody to CD20, is the best studied biologics in pSS, but other treatments hold promise, targeting for example CD22 or BAFF. Thus, during the last 20 years, the understanding of the multifaceted roles of B cells in pSS has revolutionized the management of this complex disease.
Article
We propose new classification criteria for Sjögren's syndrome (SS), which are needed considering the emergence of biologic agents as potential treatments and their associated comorbidity. These criteria target individuals with signs/symptoms suggestive of SS. Criteria are based on expert opinion elicited using the nominal group technique and analyses of data from the Sjögren's International Collaborative Clinical Alliance. Preliminary criteria validation included comparisons with classifications based on the American–European Consensus Group (AECG) criteria, a model-based “gold standard”obtained from latent class analysis (LCA) of data from a range of diagnostic tests, and a comparison with cases and controls collected from sources external to the population used for criteria development. Validation results indicate high levels of sensitivity and specificity for the criteria. Case definition requires at least 2 of the following 3: 1) positive serum anti-SSA and/or anti-SSB or (positive rheumatoid factor and antinuclear antibody titer >1:320), 2) ocular staining score >3, or 3) presence of focal lymphocytic sialadenitis with a focus score >1 focus/4 mm2 in labial salivary gland biopsy samples. Observed agreement with the AECG criteria is high when these are applied using all objective tests. However, AECG classification based on allowable substitutions of symptoms for objective tests results in poor agreement with the proposed and LCA-derived classifications. These classification criteria developed from registry data collected using standardized measures are based on objective tests. Validation indicates improved classification performance relative to existing alternatives, making them more suitable for application in situations where misclassification may present health risks.
Article
There is a crucial need for reliable diagnostic criteria for SS. Our objective was to evaluate the frequency of xerosis in patients with primary Sjögren's syndrome (SS), and compare histopathology of cutaneous sweat glands and labial salivary glands (LSGs), with respect to their contribution to the diagnosis. Twenty-two patients with primary SS and 22 matched normal volunteers were invited to rate their skin dryness on a visual analog scale. The skin was dryer (58.3 ± 10.1 versus 38.9 ± 7.6, P < 0.01), and xerosis more frequent (9 of 22 versus 2 of 22, P < 0.02) in the patients than in the controls. The axilla skin was chosen for a 6-mm punch biopsy. Lymphocytic infiltration was seen in the skin of 8 of the 12 patients tested. Two of them had normal LSGs. Most interestingly, B cell infiltrates were identified in patients' skin infiltrates, so that their presence might be a clue to the diagnosis of primary SS. These cell aggregates associated memory CD10-/CD20+/CD27+/IgD- B lymphocytes and immature CD20+/CD24 + lymphocytes. These latter findings strongly suggest that skin biopsies warrant inclusion into the routine clinical care of patients suspected of suffering from primary SS.
Article
To determine if the Fms-like tyrosine kinase 3 ligand (Flt-3L), a cytokine implicated in B cell ontogenesis and proliferation in hematologic malignancies, might be responsible for the increased numbers of circulating Bm2 and Bm2′ B cell subsets in patients with primary Sjögren's syndrome (SS). Serum levels of Flt-3L were measured in 64 patients with primary SS and in 20 healthy controls matched for age and sex. Flt-3L and its receptor Flt-3 were quantified in circulating B cells and in salivary gland (SG) biopsy tissues by immunofluorescence analysis. The effect of Flt-3L on circulating B lymphocytes was then determined by coculture with cells of a human SG (HSG) epithelial cell line. Serum levels of Flt-3L were increased in patients with primary SS as compared with controls (mean ± SD 135.8 ± 5.5 versus 64.4 ± 4.5 pg/ml; P < 0.001). Serum levels of Flt-3L in primary SS patients correlated with the numbers of Bm2 and Bm2′ cells (r = 0.46, P < 0.0006), and Flt-3 was selectively expressed in Bm2 and Bm2′ cells. B cell culture experiments showed that Flt-3L potentiated the proliferative effect of anti-IgM stimulation. In SGs, we found that infiltrating B cells expressed Flt-3 and epithelial cells produced Flt-3L. Finally, Flt-3L levels were associated with high disease activity scores and increased risk of developing lymphoma. Serum levels of Flt-3L are elevated in patients with primary SS and correlate with abnormal B cell distribution. Flt-3 is mainly expressed by Bm2 and Bm2′ cells. Serum levels of Flt-3L might explain the clinical evolution of primary SS to B cell lymphoma that is observed in some patients, thus opening the possibility of new avenues for therapy.
Article
To study evolution of pSS immunological profile, impact on pSS activity and the long-term evolution of patients with atypical auto-antibodies in a bicentric cohort of patients with pSS (n=445, mean age 53.6+/-14years, mean follow-up 76.1+/-51months). 212 patients were SSA positive and 131 were both SSA and SSB positive. During follow-up, SSA antibodies disappear in 8 patients; 2 of them exhibit new systemic complications of pSS. 68 patients had cryoglobulinemia. 52 patients had other anti-nuclear antibodies (ANA) specificities: anti-RNP (n=12), anti-centromere (n=14), anti-DNA native (n=19), anti-Scl70 (n=3), anti-JO1 (n=3), anti-Sm (n=3) and anti-histone (n=1). Fourteen patients developed ANA-associated auto-immune disease during the follow-up: 5 polymyositis (mean apparition delay 78months), 6 systemic lupus erythematosus (mean occurrence delay 77months) and 2 systemic sclerosis (mean occurrence delay 133+/-64months). Among these 14 patients, only three presented atypical-ANA at pSS diagnosis. Cryoglobulinemia and anti-SSA and SSB antibodies at diagnosis were associated with new systemic involvements. Cryoglobulinemia and SSA/SSB positivity are associated with systemic activity after diagnosis in pSS. Although atypical ANA are found in 12% of the cases, long-term evolution to ANA associated auto-immune diseases concerned patients with active immunological profile and extra-glandular manifestations.