ArticlePDF Available

The Expansion of CD25 high IL-10 high FoxP3 high B Regulatory Cells Is in Association with SLE Disease Activity

Authors:

Abstract and Figures

B regulatory cells (Bregs) belong to a subgroup of activated B cells tasked with maintaining self-tolerance and preventing autoimmunity. While sharing similar regulatory mechanisms such as IL-10 dependency, they also defer in exhibiting their suppressive effects by expressing Fas-Ligand, TGF-beta, and PDL-1. In this study we show, for the first time, the expansion of CD25 high FoxP3 high Bregs in systemic lupus erythematosus (SLE) patients compared to healthy individuals (18.5 ± 3.052% versus 11.0 ± 1.654%, p < 0.001 , resp.). This expansion was also shown to correlate with SLE disease activity ( r = 0.75 ). In addition, CD25 high FoxP3 high Bregs were also IL-10 high expressing and further expanded when stimulated with semaphorin 3A. In sum we show that CD25 high FoxP3 high are an additional subtype of Bregs, involved in regulating SLE disease activity. Being IL-10 expressing, we may assume that they are one of the sources of increased serum IL-10 in SLE patients. Further studies are required in order to assess the relation between high serum IL-10 and CD25 high FoxP3 high Breg cells.
This content is subject to copyright. Terms and conditions apply.
Research Article
The Expansion of CD25highIL-10highFoxP3high B Regulatory Cells
Is in Association with SLE Disease Activity
Zahava Vadasz,1Regina Peri,1Nasren Eiza,1Gleb Slobodin,2
Alexandra Balbir-Gurman,3and Elias Toubi1
1Division of Allergy & Clinical Immunology, Bnai Zion Medical Center, Faculty of Medicine, Technion, 4940 Haifa, Israel
2Rheumatology Unit, Bnai Zion Medical Center, Faculty of Medicine, Technion, 4940 Haifa, Israel
3B. Shine Rheumatology Unit, Rambam Health Care Campus, Faculty of Medicine, Technion, 4940 Haifa, Israel
Correspondence should be addressed to Elias Toubi; elias.toubi@b-zion.org.il
Received  July ; Accepted September 
Academic Editor: Carlo Perricone
Copyright ©  Zahava Vadasz et al. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
B regulatory cells (Bregs) belong to a subgroup of activated B cells tasked with maintaining self-tolerance and preventing
autoimmunity. While sharing similar regulatory mechanisms such as IL- dependency, they also defer in exhibiting their
suppressive eects by expressing Fas-Ligand, TGF-beta, and PDL-. In this study we show, for the rst time, the expansion of
CDhighFoxPhigh Bregs in systemic lupus erythematosus (SLE) patients compared to healthy individuals (. ±.% versus
. ±.%, 𝑝 < 0.001, resp.). is expansion was also shown to correlate with SLE disease activity (𝑟 = 0.75). In addition,
CDhighFoxPhigh Bregs were also IL-high expressing and further expanded when stimulated with semaphorin A. In sum we
show that CDhighFoxPhigh are an additional subtype of Bregs, involved in regulating SLE disease activity. Being IL- expressing,
we may assume that they are one of the sources of increased serum IL- in SLE patients. Further studies are required in order to
assess the relation between high serum IL- and CDhighFoxPhigh Breg cells.
1. Introduction
Among the many immune mediated responses involved in
systemic lupus erythematosus (SLE) is the imbalance between
T-helper cells () subsets, namely, //, and both
T and B regulatory (reg) cells [].  proinammatory
cytokine levels such as IL-, IL-, and IFNs are usually
increased in association with SLE disease activity index
(SLEDAI).  related cytokines such as IL- and IL-
 are also reported to be enhanced and contribute to
inammatory processes in SLE and other rheumatic dis-
eases such as rheumatoid arthritis (RA) and psoriasis. 
relatedcytokines,thatis,IL-andIL-,areknownfor
their ability in driving humoral immune responses, B cell
overactivation, and the production of many specic autoan-
tibodies [–]. Many studies during the last decade have
reported on the failure of Treg cells to maintain self-tolerance,
allowing the development of many autoimmune diseases.
e failure in suppressing eector cell proliferation is
mainly considered to be IL- dependent (lower expression
and/or production of IL-) due to the altered expression of
FoxP and/or inhibitory molecules such as CTLA- in Treg
cells []. Breg cells are involved in regulating/suppressing
immune mediated inammation but act earlier than Treg
cells. ey use similar suppressive modalities, that is, IL-
, TGF-beta, and the expression of proapoptotic membrane
molecules which vary across dierent Breg subtypes [].
Among these dierent subtypes, CD19+CD24highCD38high
and CD19+CD25highCD86highCD1dhigh were both described
as being involved in suppressing autoimmune processes,
both in an IL- dependent way and with an altered
function in SLE [, ]. Breg cells have also been charac-
terized as CD5high ,FoxP3high , and Fas-Ligand expressing
cells. CD19+CD5highFoxP3high Breg cells were reported to
be involved in non-IgE-mediated food allergies, namely,
in maintaining tolerance to milk allergies []. In addi-
tion to this subtype, Breg cells were dened as being
Hindawi Publishing Corporation
Journal of Immunology Research
Volume 2015, Article ID 254245, 6 pages
http://dx.doi.org/10.1155/2015/254245
Journal of Immunology Research
CD19+CD5highFas-Lhigh ,alsocalled“killerBcells.”Numer-
ous researchers have reported that these cells participate
in the escape of viral infections from the ecient cyto-
toxic T cell response []. e similarities and dierences
between all the above-mentioned Breg cells are not suf-
ciently understood. Are they similar in their regulatory
eects? Do they express/produce similar amounts of IL-
and TGF-beta? How do they react to various stimuli? (see
[]). In previous studies, we and others showed that Breg
cell function was enhanced when stimulated by CpG and
CDL, increasing by this autologous Treg cell properties
following their coculture [, ]. When cocultured with
semaphorin A (semaA), IL- and TGF-beta expression
was enhanced in CD19+CD25high Breg cells, suggesting that
semaA is a frontier factor in improving Breg cell function
(unpublished data). Later, we reported on the ability of
semaA in enhancing Breg cell properties by increasing CD
(a regulatory molecule) expression on B cells []. Expecting
to nd lower serum levels of IL- in some autoimmune
diseases, namely, in SLE, the opposite was found. Paradoxi-
cally, serum IL- is reported to be increased in association
with increased SLEDAI and with high titers of anti-dsDNA
antibodies. e source of increased serum IL- in SLE is
yet undened, suggested to be overproduced by  and/or
by one of the Breg subtypes. In addition, the association of
Atg rs single nucleotide polymorphism (SNP) with
SLE susceptibility and IL- serum levels was analyzed. Here,
carriage of the rs T allele was associated with IL-
upregulation and clinical features of SLE, concluding that
suchmutatedalleleinuencedbothSLEsusceptibilityandIL-
production[].Inthisstudy,weaimtoevaluatethestatus
of CD19+CD25highFoxP3high Breg cells, namely, whether they
are IL- expressing. We will also assess the status of these
cells in SLE patients when compared to healthy individuals.
We speculate on their possible contribution to increased
serum IL- in SLE patients. Finally, we will evaluate the
response of this subtype of Breg cells to semaA, to see if this
coculture increases IL- expression as it does in other Breg
cells.
2. Patients and Methods
2.1. Patients Population. is study examined  SLE patients
( females and male; age range – years; mean
30.5 ± 9.2). All patients are routinely followed up by well-
trained rheumatologists and all fulll the ACR criteria for
the classication of SLE []. Clinical and serological data
(skin involvement; arthritis; renal involvement; full cell
blood count; serum complement levels; anti-dsDNA and
other extractable nuclear autoantibodies) were all available,
enabling the determination of SLEDAI. e serological work-
up was performed at the Bnai Zion Medical Center by a
single experienced technician to insure uniformity of all
analyses, utilizing identical kits. Patients in whom SLEDAI
was between and points were treated with hydroxychloro-
quine and in some patients prednisolone (.mg/daily) was
added. When SLEDAI was above points, azathioprine was
added, but only aer analyzing specic serology and purify-
ing B cells. When SLEDAI was above  points the addition
of cyclophosphamide or MMF was considered again, only
aer performing SLE serology and purifying B cells. Twenty
healthycontrols,sexandagematched,wereassessedand
analyzed for all above-mentioned parameters. is study was
approved by both the local Helsinki Committee of the Bnai
Zion Medical Center and the Rambam Health Care Campus,
Haifa, Israel.
2.2. B Cell Purication. B cells were puried from peripheral
bloodofhealthycontrolsandSLEpatients.Todoso,
peripheral blood mononuclear cells (PBMCs) were isolated
on Lymphoprep (Axis-Shield, Oslo, Norway), and B lympho-
cytes were then twice puried by positive selection using
CD microbeads ( 𝜇L/7cells; Miltenyi Biotec, Ber-
gisch Gladbach, Germany) according to the manufacturers
instructions, achieving by this >% purity.
2.3. FoxP3 and IL-10 Expression in CD19+CD25ℎ𝑖𝑔ℎ BCells.
e expression of FoxP and IL- in CD19+CD25high cells
(considered as Breg cells) from healthy controls and SLE
patients was initially assessed by staining puried B cells
aer  hours of activation with ODN-CpG and CDL.
e staining was performed by using monoclonal antibodies,
human anti-CD-BUV (BD Horizon, Becton Dickinson,
NJ, USA) and human anti-CD BUV (BD Horizon,
Becton Dickinson, NJ, USA) as outer membrane antibod-
ies, and FoxP PE\CF594 and IL- APC (BD Horizon,
Becton Dickinson, NJ, USA) as intracellular staining, using
a “Fix and Perm” kit (Invitrogen, NY, USA) according to
the manufacturer’s instructions. e staining was evaluated
using ow cytometry soware (FC and CXP soware,
Beckman Coulter, Brea, CA, USA, and Becton Dickinson,
NJ, USA). CD positive cells in the puried cell culture were
determined by using monoclonal CD PerCP-Cy. antibody
(BD Pharmingen, Becton Dickinson, NJ, USA) and analyzed
by Becton Dickinson FACS-Fortessa. e results are shown
as % of CD19+CD25high Breg cells expressing FoxP or IL-,
taking into consideration that the absolute number of Breg
cells in all groups was found to be comparable. Standard
deviation(STDEV)wasusedtoquantifytheamountof
variationofasetofdatavalues(e.g.,percentageofBregcells
expressing FoxP among the patients in each indicated group
of disease or normal control).
2.4. Semaphorin 3A Enhances FoxP3 Expression. Aiming to
evaluate the eect of semaA on FoxP expression, condition-
media from HEKcells, which were infected by NSPI-
CMV-FLAG lentivirus with or without human semaA
cDNA, a kind gi from Professor Gera Neufeld and Dr.
Ofra Kessler, Ruth and Bruce Rappaport Faculty of Medicine,
Technion, Israel, as previously described [], were added to
theabove-mentionedpuriedBcellsactivatedbyODN-CpG
and CDL and incubated for  hours. Aer incubation,
CD19+CD25high cellswereanalyzedforthepossiblechange
in FoxP expression using the above-mentioned specic
monoclonal antibodies and evaluated using an FC ow
Journal of Immunology Research
103
102
101
100
CD25 PC5
FoxP3Pe
28%48%
100101102103
(a)
100102
101103104105
0
SSC-A
Comp-PerCP-Cy5.5-A::CD3
250 K
200 K
150 K
100 K
50 K
CD3+0.33%
(b)
F : (a) A representative FACS analysis of puried B cells (expressing CD25high FoxP) following CpG-ODN and CDL activation. Of
note is that CD25dim-low B cells (upper le quadrant) do not express FoxP. However, CD25high B cells coexpress signicant amount of FoxP
(upper right quadrant). (b) A representative FACS analysis of puried activated B cells, showing that CD+T cell contamination (gated CD3+
T cells) is less than .%.
cytometer and Becton Dickinson FACS-Fortessa. e results
are shown as % of Breg cells expressing FoxP, taking into
consideration that the absolute number of Breg cells in all
groups was found to be comparable.
2.5. Clinical Correlation and Statistical Analysis. Comparison
of FoxP expression in B cells from SLE patients and healthy
controls was done using the unpaired Student 𝑡-test. e
correlation coecient (𝑟)ofclinicalcorrelationbetween
SLEDAI score and % of Breg cells expressing FoxP was
determined using the Pearson correlation test. A two-tailed
𝑝value of . or less was considered to be statistically
signicant.
3. Results
3.1. CD19+CD25ℎ𝑖𝑔ℎ Activated B Cells Are FoxP3ℎ𝑖𝑔ℎ.First, we
examined whether CD19+CD25high B regulatory cells are also
FoxP expressing cells. Puried resting B cells (immediately
following purication) were FoxP3dim (weakly detectable)
(data not shown). However, following their stimulation with
CpG-ODN and CDL for  h, CD19+CD25high Bcells
turned to become FoxP3high (Figure (a)). As also seen, there
are less than .% gated CD T cells and therefore B cell
contamination with CD is unlikely and FoxP expression in
CD25high B cells is very prominent (Figure (b)).
3.2. Activated CD19+CD25ℎ𝑖𝑔ℎ FoxP3ℎ𝑖𝑔ℎ Are Also IL-10ℎ𝑖𝑔ℎ.
Gating on activated CD25highFoxP3high one can see that most
Comp-APC-A::IL-10
Comp-PE-CF594-A::FoxP3
Q1
6.84%
Q4
2.91%
Q3
4.06%
Q2
86.2%
100102
101103104105
105
104
103
101
102
100
F : A demonstrative FACS analysis of puried B cells,
showing that activated CD19+CD25highFoxP3high are also IL-10high .
Of note is that CD25dim-low \FoxP3dim-low B cells express very little
IL-.
of these cells (>% of these cells) are IL-10high (Figure ) in
contrast to B cells that are FoxP3dim being also IL-10dim.
3.3. CD19+CD25ℎ𝑖𝑔ℎFoxP3ℎ𝑖𝑔ℎ in SLE. e percentage of Breg
cells (CD19+CD25high cells) in peripheral blood (highly
Journal of Immunology Research
0
5
10
15
20
25
30
Controls SLE
W/O sema3A
With sema3A
p < 0.002
p < 0.005
p < 0.001
Average % of CD19+CD25high expressing FoxP3
F : e percentage of CD19+CD25high FoxP3high Breg cells in
normal controls (𝑛=20)andinpatientssueringfromSLE(𝑛=
21). One can see that this subtype of B cells is signicantly increased
in SLE patients. In addition, the addition of semaA to these cells
increased signicantly the percentage of these cells.
expressing FoxP) was signicantly higher in SLE patients
when compared to that of healthy individuals (18.5%±3.052
versus 11.0 ± 1.654%, resp., 𝑝 < 0.005) (Figure ).
3.4. Semaphorin 3A Increases FoxP3 Expression in Breg Cells.
We then sought to determine if semaA increases the
expression of FoxP in these Breg cells. As is demonstrated
in Figure , semaA increases the percentage of Breg cells
(CD19+CD25high cells) in peripheral blood expressing FoxP,
in normal controls and to a higher extent in SLE patients
(in normal controls up to 13.6 ± 1.806%frombaseline,𝑝<
0.002, and in SLE patients up to 28.5 ± 3.506%, 𝑝 < 0.0001)
(Figure ).
3.5. FoxP3 Expression in B Cells Is Correlated with SLEDAI.
Figure demonstrates the correlation between the percentage
of CD19+CD25highFoxP3high cells of SLE patients and the
SLEDAI score of these patients. As can be seen, there is a
positive correlation with an 𝑟 Pearson coecient of ..
isresultisinlinewiththeknowncorrelationbetweenIL-
 level in SLE patients and their SLEDAI.
4. Discussion
In most autoimmune diseases, immune mediated inamma-
torydamageisalwaystheresultofanetbalancebetween
the overactivity of self-reactive cells (T and B eector cells)
and immune regulatory mechanisms (T and B regulatory
cells). Most B regulatory cells are dened as being IL-
 expressing/producing cells; however, they have dierent
subtypes, are heterogeneous, and have dierent mechanisms
in diseases in which they are involved. eir homology
to Treg subtypes, namely, Br cells (expressing IL-), Br
05 10152025
0
20
40
60
80
CD19+CD25highFoxP3high cells (%)
r = 0.75
SLEDAI
F : Clinical correlation between the percentage of
CD19+CD25highFoxP3high cells of SLE patients and the SLEDAI
score of these patients. e correlation was done using the Pearson
correlation test.
cells (mainly expressing TGF-beta), and B-FoxP positive
cells, was recently mentioned. In this case, Breg cells were
shown to initiate immune regulatory responses by facilitating
the recruitment of Tregs and then disappearing once Tregs
become dominant in the immune response []. As men-
tioned above, when CD19+CD24highCD38high Bcellswere
evaluatedinSLE,theyhadbothareducedabilitytoproduce
IL- and a reduced ability to suppress T cell cytokine pro-
duction, although it is unclear if this latter defect is a cause or
a consequence of SLE. In contrast to this nding, human IL-
 producing CD24highCD27high Breg cells (found to suppress
monocytes in an IL- dependent manner) were increased
in patients with rheumatoid arthritis, SLE, and multiple
sclerosis when compared to healthy individuals, suggesting
this increase to be compensatory, aiming (with little success)
to maintain self-tolerance []. e role of CD5highFasLhigh
“killer B cells” was assessed in lupus susceptible MRL/lpr
mice. Being cytotoxic to T cells they were found to be
increased, probably in attempt to suppress autoreactive T cells
in these mice []. Focusing on CD19+CD25highFoxP3high
Breg cells we rst assessed their status in healthy individuals.
Here,weshowforthersttimethatbothIL-andFoxP
expressions were noticed mainly in activated CD25high B
cells (activated with CpG and ODN) and that this expression
was enhanced when these B cells were stimulated with
add-onsemaA.InthiscaseCD25high FoxP3high Breg cells
were characterized by being IL-10high whereas FoxP3dim B
cells were IL-10dim aswell.WhenanalyzedinSLEpatients,
we found CD19+CD25high FoxP3high cells to be signicantly
increased as compared to healthy individuals. is was found
to be in positive correlation with increased SLEDAI and in
association with lupus nephritis. In a recent study and in line
with our nding, CD19+CD25highFoxP3high Bregulatorycells
were found to be increased in the cerebrospinal uid of active
patients suering from relapsing-remitting multiple sclerosis
(MS) when compared to that of nonclinically active MS. is
Journal of Immunology Research
expansion of B regulatory cells was attributed to the compen-
satory attempt of these cells to maintain immune regulatory
processes []. In contrast to this study, rheumatoid arthritis
patients had signicantly lower proportions of peripheral
blood CD19+FoxP3+B cells as compared to healthy controls,
particularly in patients with interstitial lung disease. is
nding suggests that Breg phenotypes may have dierent
functions in the pathogenesis of dierent rheumatic diseases
[]. e fact that serum IL- is increased in SLE and in
association with SLE disease activity has been established in
many previous studies. In one, increased IL- was shown to
exhibit a modulatory eect by suppressing the dierentiation
and function of monocyte-derived dendritic cells []. In
a recent study, increased IL- in the sera of SLE patients
was capable of inducing Fas and FasL expression on CD+
T cell surfaces, promoting apoptosis of this cell subset, thus
contributing to many other mechanisms of self-tolerance
[].However,westillneedtoexplainthemechanismsby
which serum IL- is increased in SLE. In this regard, the
expansion of IL- producing B cells was shown to be in
parttheresultofincreasedBcellactivatingfactor(BAFF).
Enhanced serum BAFF in SLE was described in many studies
as being associated with increased expression of TLR- and
other markers of B cell activation [, ]. is may explain
ourndingofincreasedIL-10high FoxP3high Bregcellsaswell
as increased serum IL- in SLE. Another signicance of
FoxP3high B cells being increased in SLE is the possibility
that by multiplying they also increase their IL- production
improving by this their regulatory function. When B cells
were cocultured with semaA they responded by increasing
their FoxP expression. is raises the possibility that if
provided with the proper stimulation Bregs may develop
higher regulatory properties and that by increasing their IL-
production they may induce a better regulatory mechanism
in SLE.
5. Conclusion
CD25highFoxP3high Bregs (highly expressing IL-) are sig-
nicantly increased in SLE, in correlation with SLEDAI.
Semaphorin A increases FoxP expression in Breg cells
improving by this their regulatory properties. We assume that
the expansion of these cells is the attempt of our regulatory
immune responses to maintain self-tolerance and to suppress
as much as possible SLE disease activity. Further studies are
required in order to better understand the role of this subset
of B cells in autoimmunity.
Conflict of Interests
e authors declare that there is no conict of interests
regarding the publication of this paper.
References
[]R.M.Talaat,S.F.Mohamed,I.H.Bassyouni,andA.A.
Raouf, “///Treg cytokine imbalance in systemic
lupus erythematosus (SLE) patients: correlation with disease
activity, Cytokine, vol. , no. , pp. –,  (Chinese).
[] K.Shah,W.-W.Lee,S.-H.Leeetal.,“Dysregulatedbalanceof
 and  cells in systemic lupus erythematosus, Arthritis
Research & erapy,vol.,no.,articleR,.
[] D.Li,B.Guo,H.Wu,L.Tan,C.Chang,andQ.Lu,“Interleukin-
 in systemic lupus erythematosus: a comprehensive review,
Autoimmunity,vol.,no.,pp.,.
[] B. Terrier, N. Costedoat-Chalumeau, M. Garrido et al., “Inter-
leukin  correlates with T cell and B cell subset alterations in
systemic lupus erythematosus, e Journal of Rheumatology,
vol.,no.,pp.,.
[]S.Futatsugi-Yumikura,K.Matsushita,A.Fukuokaetal.,
“Pathogenic -type follicular helper T cells contribute to
the development of lupus in Fas-decient mice, International
Immunology,vol.,no.,pp.,.
[]B.Yan,S.Ye,G.Chen,M.Kuang,N.Shen,andS.Chen,
“Dysfunctional CD+,CD+ regulatory T cells in untreated
active systemic lupus erythematosus secondary to interferon-𝛼-
producing antigen-presenting cells, Arthritis and Rheumatism,
vol.,no.,pp.,.
[]A.Ray,L.Wang,andB.N.Dittel,“IL--independentregu-
latory B-cell subsets and mechanisms of action, International
Immunology,.
[]P.A.Blair,L.Y.Nore
˜
na, F. Flores-Borja et al.,
“CD+CDhiCDhi B cells exhibit regulatory capacity
in healthy individuals but are functionally impaired in systemic
lupus erythematosus, Immunity,vol.,no.,pp.,
.
[] A. Kessel, T. Haj, R. Peri et al., “Human CD+CDhigh B
regulatory cells suppress proliferation of CD+Tcellsand
enhanceFoxpandCTLA-expressioninT-regulatorycells,
Autoimmunity Reviews, vol. , no. , pp. –, .
[] J. Noh and G. Noh, Allergen specic responses of CDhigh
and CDlow B cells in non-IgE mediated food allergy of late
eczematous reactions in atopic dermatitis: presence of IL- and
IL- producing regulatory B cells (Br & Br), Inammation
&Allergy-DrugTargets,vol.,no.,pp.,.
[] S. K. Lundy, Killer B lymphocytes: the evidence and the
potential, Inammation Research,vol.,no.,pp.,
.
[] F. Mion, S. Topnon, B. Tooletto, D. Cesselli, C. E. Pucillo, and
G. Vitale, “IL- production by B cells is dierentially regulated
by immune-mediated and infectious stimuli and requires p
activation, Molecular Immunology,vol.,no.,pp.,
.
[] K.Yanaba,J.-D.Bouaziz,T.Matsushita,T.Tsubata,andT.F.
Tedder, “e development and function of regulatory B cells
expressing IL- (B cells) requires antigen receptor diversity
and TLR signals, Journal of Immunology,vol.,no.,pp.
– , .
[]Z.Vadasz,T.Haj,A.Balbiretal.,“Aregulatoryrolefor
CD expression on B cells in systemic lupus erythematosus,
Seminars in Arthritis & Rheumatism,vol.,no.,pp.,
.
[] P. L´
opez, E. Alonso-P´
erez, J. Rodr´
ıguez-Carrio, and A. Su´
arez,
“Inuence of Atg mutation in SLE depends on functional IL-
genotype, PLoS ONE,vol.,no.,ArticleIDe,.
[] C. Bombardier, D. D. Gladman, M. B. Urovitz, D. Caron, and C.
H. Chang, “Derivation of the SLEDAI. A disease activity index
for lupus patients. e committee on prognosis studies in SLE,
Arthritis and Rheumatism,vol.,no.,pp.,.
Journal of Immunology Research
[] B. Kigel, A. Varshavsky, O. Kessler, and G. Neufeld, “Suc-
cessful inhibition of tumor development by specic class-
semaphorins is associated with expression of appropriate
semaphorin receptors by tumor cells, PLoS ONE,vol.,no.,
article e, .
[] J.-M. Berthelot, C. Jamin, K. Amrouche, B. Le Go, Y. Maugars,
and P. Youinou, “Regulatory B cells play a key role in immune
system balance, Joint Bone Spine,vol.,no.,pp.,.
[] Y. Iwata, T. Matsushita, M. Horikawa et al., Characterization
of a rare IL--competent B-cell subset in humans that parallels
mouse regulatory B cells, Blood,vol.,no.,pp.,
.
[] D. Bonardelle, K. Benihoud, N. Kiger, and P. Bob´
e, “B lym-
phocytes mediate Fas-dependent cytotoxicityin MRL/lpr mice,
JournalofLeukocyteBiology,vol.,no.,pp.,.
[] C. de Andr´
es, M. Tejera-Alhambra, B. Alonso et al., “New
regulatory CD+CD+B-cell subset in clinically isolated
syndrome and multiple sclerosis relapse. Changes aer gluco-
corticoids, Journal of Neuroimmunology,vol.,no.-,pp.
–, .
[] Y. Guo, X. Zhang, M. Qin, and X. Wang, “Changes in peripheral
CD+Foxp+and CD+TGF-𝛽+regulatory B cell populations
in rheumatoid arthritis patients with interstitial lung disease ,
Journal of oracic Disease,vol.,no.,pp.,.
[] Z. Sun, R. Zhang, H. Wang et al., “Serum IL- from systemic
lupus erythematosus patients suppresses the dierentiation
and function of monocyte-derived dendritic cells, Journal of
Biomedical Research, vol. , no. , pp. –, .
[] X. Yang, B. Sun, H. Wang, C. Yin, X. Wang, and X. Ji, “Increased
serum IL- in lupus patients promotes apoptosis of T cell
subsets via the caspase pathway initiated by Fas signaling,
JournalofBiomedicalResearch,vol.,no.,pp.,.
[] M. Yang, L. Sun, S. Wang et al., “Cutting edge: novel function
of B cell-activating factor in the induction of IL--producing
regulatory B cells,” e Journal of Immunology,vol.,no.,
pp. –, .
[] D. Yehudai, A. Snir, R. Peri et al., “B cell-activating factor
enhances interleukin- and interleukin- productionby ODN-
activated human B cells, Scandinavian Journal of Immunology,
vol. , no. , pp. –, .
... The use of Breg-based therapies should be based on changes in the number of Bregs and/or impaired regulatory function associated with the pathogenesis of SLE. However, the results of studies on the number of Bregs in active SLE patients and normal healthy people are quite contradictory; the frequency of Bregs in SLE patients is reduced or increased [8][9][10][11][12][13][14] . Importantly, the role of Bregs in SLE is also controversial. ...
... Then 16 duplicated articles were excluded, 15 articles were not case-control trials, 15 articles were not for SLE patients, 14 articles were conference abstract superseded by publications, 17 articles did not provide relevant data, and 18 articles could not obtain full-text information. Therefore, this meta-analysis included a total of seven articles [8][9][10][11][12][13][14] . ...
... The analysis included 301 active SLE patients and 218 controls from seven eligible articles. Of these articles, five were conducted in China [8,10,[12][13][14] , one in Israel [11] and one in Germany [9] . The diagnose criteria of SLE varied across studies. ...
Article
Full-text available
Background: The study of regulatory B cells (Bregs) in systemic lupus erythematosus (SLE) has been in full swing in recent years, but the number and function of Bregs in SLE patients have also present quite contradictory results. Therefore, we conducted a meta-analysis to verify the changes in Bregs in active SLE. Methods: We identified studies reporting the proportions of Bregs in SLE patients by searching Pubmed, Embase, Web of Science, Cochrane and CNKI. Due to the degree of heterogeneity is very high, we used a random effects model to assess the mean differences in percentages of Bregs between active SLE and controls. Then, sensitivity analysis and subgroup analysis were performed to verify potential sources of heterogeneity. Results: Seven eligible articles involving 301 active SLE patients and 218 controls were included in the meta-analysis. The pooled percentages of Bregs were found no significant difference between active SLE patients and healthy controls [0.259, (−1.150, 1.668), p = 0.719], with great heterogeneity ( I2 = 97.5%) . The result of sensitivity analysis showed that exclusion of any single study or single article did not materially resolve the heterogeneity, but after excluding the article conducted by Cai X and his colleagues, the percentages of Bregs were significantly higher in active SLE than those in controls [1.394, (0.114,2.675), p = 0.033]. The results of subgroup analysis revealed that when the disease activity was judged by SLEDAI score ≥ 5, the percentages of Bregs were significantly lower in the SLE groups than in the control groups[-1.99,(-3.241,-0.739), p = 0.002], but when the threshold of SLEDAI score ≥ 6 chosen for active SLE, the percentages of Bregs were significantly increased in the SLE groups[2.546,(1.333,3.759), p < 0.001]. Meanwhile, other subgroup analysis based on the different phenotypes of Bregs, diagnostic criteria, enrolled research countries, treatment status, and organ involvement did not differ in proportion of Bregs between SLE patients and controls. Conclusions: The study implies that Bregs may play a role in the pathogenesis of active SLE, and the thresholds of SLEDAI score to distinguish between active and inactive SLE patients are important factors affecting the percentages of Bregs.
... Using various techniques, we demonstrated the expression of both FOXP3 mRNA and protein in purified CLL B cells after exclusion of any Tregs or other immune cells, and using FOXP3-positive cells such as Tregs and CD4 + T cells as a control. FOXP3 expression has been described in tumour infiltrating Tregs in epithelial solid tumours such as pancreatic adenocarcinoma, in glioma cells and in Bregs present in systemic lupus erythematosus (SLE) [50][51][52]. Anti-tumour and oncogenic properties have been attributed to FOXP3 depending on the cellular context and interacting partners [53]. For example, FOXP3 interaction with NFAT or NFκB regulates anti-tumour immunity [54,55]. ...
... Moreover, FOXP3 and TGFβ1 are concomitant intermediates in Tregs/Th17 homeostasis. CLL B cells, also expressed CD25, another key marker and regulator of Tregs, which has been observed in IL10 + mature Breg cells with antigen specific suppressive functions in SLE patients [52]. Furthermore, FOXP3 associates with STAT3, which is constitutively phosphorylated at S 727 in CLL, to promote the expression of IL10 in a subset of Tregs. ...
Article
Full-text available
Chronic Lymphocytic Leukemia (CLL) is a heterogeneous B cell neoplasm ranging from indolent to rapidly progressive disease. Leukemic cell subsets with regulatory properties evade immune clearance; however, the contribution of such subsets during CLL progression is not completely elucidated. Here, we report that CLL B cells crosstalk with their immune counterparts, notably by promoting the regulatory T (Treg) cell compartment and shaping several helper T (Th) subsets. Among various constitutively- and BCR/CD40-mediated factors secreted, tumour subsets co-express two important immunoregulatory cytokines, IL10 and TGFβ1, both associated with a memory B cell phenotype. Neutralizing secreted IL10 or inhibiting the TGFβ signalling pathway demonstrated that these cytokines are mainly involved in Th- and Treg differentiation/maintenance. In line with the regulatory subsets, we also demonstrated that a CLL B cell population expresses FOXP3, a marker of regulatory T cells. Analysis of IL10, TGFβ1 and FOXP3 positive subpopulations frequencies in CLL samples discriminated 2 clusters of untreated CLL patients that were significantly different in Tregs frequency and time-to-treatment. Since this distinction was pertinent to disease progression, the regulatory profiling provides a new rationale for patient stratification and sheds light on immune dysfunction in CLL.
... Для наших досліджень привертають увагу отримані останнім часом дані щодо експресії на CD19+CD5+-В-кл маркеру Т-регуляторних Foxp3, який може виступати і як фактор транскрипції B-регуляторних, тому що CD5-B-л не експресують Foxp3 [23][24][25]. Подальші дослідження співвідношень CD19+CD5+-В-л як активованих клітин та В-л з маркерами регуляторних, а також Tim-1 дуже важливі та доцільні для жінок з ПЕ в зв»язку з даними про те, що Tim-1+IL-10+-B-кл пригнічують певні аутоімунні реакції [26], а тому і мають імуносупресивний потенціал для вагітних. ...
Article
Full-text available
The study of immune mechanisms during pregnancy is an important area of research aimed at understanding the immunogenesis of pregnancy and its complications. One such complication is nephropathy, which requires accurate prediction of its course and appropriate therapy. The present study aimed to assess the blood levels of T-lymphocytes (T-helper, T-suppressor/cytotoxic), B-lymphocytes, and markers of their activation in pregnant women, with a particular focus on patients with preeclampsia (PE). Methods. Using a cellular cytofluorimeter and the corresponding test systems, we determined the relative levels of immunocompetent blood cells such as CD3+, CD4+, CD8+, CD19+-l with the expression of HLA-DR, CD25, CD5 activation markers in 436 non-pregnant women and 514 pregnant women. We analyzed the characteristics of these indicators at different stages of pregnancy, specifically before (1-4 groups) and after (5 group) 20 weeks. In the second stage, we performed an analysis of the indicators in 107 women with PE, 14 women after PE, and 54 pregnant women in the reference group without complications at 20+ weeks. This analysis included T- and B-cell studies of these patient groups under in vitro conditions with autoserum and inactivated serum. Results. The first weeks of pregnancy were characterized by a high relative level (%) in the blood of T-l and subpopulations of T-h and Ts/c, as well as their activation according to the expression data of DR, CD25 (for CD4+-l), and CD5 (for CD19+-l). These indicators significantly decrease from 20 weeks of pregnancy and become comparable to the levels in non-pregnant women. The percentages of CD3+4+25+-l and CD19+5+-l are below the normal range, indicating a decrease in T-helper cell and B-cell activation during this period of pregnancy. In patients with PE after 20 weeks of pregnancy, the relative levels of T-l, B-l, T-h, and Ts/c did not differ from the reference group. Incubation of T- and B-l of women with PE with autoserum in vitro almost tripled the number of T- and B-l, and the blocking effect was greater than in women without PE, with a significant difference between them; the average ratio T-l/T-las exceeded the reference in the subgroup of active manifestations of PE. Conclusion. Studying the relative levels of T-l, T-h, T-s/c, and B-l in the blood, as well as markers of their activation, at different stages of pregnancy with in vitro detection of features of the humoral immune response as predictors of NP allows determining individual components of immunogenetic and prognostic markers for personalized therapy during pregnancy.
... When sensitive individuals exposed to allergens, the allergens would be processed by antigen-presenting cell (APC) and then the signal of antigen peptide could be presented to T cells, which makes the differentiation of Th initial cells shift and leads to the Th1 reaction switch to Th2 reaction. 48 The immune response of Th2>Th1 is the cause of AR. 49 Bregs can inhibit the immune response mediated by Th2 cells by secreting IL-10 and TGF-βcytokines, reducing the imbalance of Th1/Th2 ratio in patients with AR and preventing the occurrence of AR. 50,51 literature suggests that IL-35 is involved in immune tolerance, but the mechanism of Bregs-derived IL-35 in allergic reactions and SIT needs further study. ...
Article
Full-text available
Allergic rhinitis (AR) is a common otolaryngologic disease with frequent episodes of sneezing, clear nasal discharge flow and nasal congestion. The mechanisms of AR are complex and considered generally caused by the immune tolerance deficiency. Regulatory B cells (Bregs) are immunosuppressive cells that can modulate immune responses by the secretion of IL-10, IL-35, and tumor growth factor-β (TGF-β) and via the interaction of membrane surface molecules. However, Bregs are numerically deficient and/or dysfunctional in airway allergic diseases such as AR and allergic asthma, and the related mechanisms remain unclear. In this review, we summarize the role of Bregs in AR pathogenesis and highlight the importance of Bregs in maintaining immune tolerance. It is believed that further research on Bregs will contribute to developing new treatments and finding specific biomarkers that could help to predict disease progression.
... Indeed, individuals with two or more X chromosomes are at increased risk for SLE (7), suggesting that X-linked genes have a significant role in disease. Immunity-related genes are enriched on the X chromosome (8,9), and some of these genes are routinely overexpressed in SLE patient B cells (10)(11)(12)(13). In addition, mouse models with X-linked gene duplication [such as the BXSB-Yaa mouse model (14, 15)] or transgenic overexpression of either of the X-linked genes Tlr7 (16,17) or Btk (18,19) exhibit disease resembling human SLE, with production of double-stranded DNA autoantibodies. ...
Article
Full-text available
Significance Pathogenic autoantibodies are a feature of systemic lupus erythematosus (SLE), of which 85% of patients are women. Multiple X chromosomes increase the risk for SLE, suggesting an important role for X-linked gene expression for the female sex bias. X-chromosome inactivation (XCI) regulates X-linked gene expression on the inactive X. This study examines XCI maintenance across multiple human B cell subsets from healthy individuals and SLE patients. Importantly, we found that both pediatric and adult SLE patient B cells have significant reductions with epigenetic modifications on the inactive X and aberrant X-linked gene expression. Our findings will be instrumental for future investigations of disease mechanisms underlying the female bias of SLE and abnormal autoantibody production in B cells.
... Additionally, Breg cells exhibit other markers. While transcription factor Forkhead Box P3 (Foxp3) is known to be expressed on several immune cells and is commonly regarded as a marker for Tregs, it has also been identified as a transcription factor of Breg cells, namely CD19 + CD5 + B cells; in contrast, CD5 -B cells do not express Foxp3 (17,(23)(24)(25). Another transcription factor should be noticed is T cell immunoglobulin and mucin domain 1 (Tim-1), a member of Tim family. ...
Article
Full-text available
B cells, commonly regarded as proinflammatory antibody-producing cells, are detrimental to individuals with autoimmune diseases. However, in recent years, several studies have shown that regulatory B (Breg) cells, an immunosuppressive subset of B cells, may exert protective effects against autoimmune diseases by secretion of inhibitory cytokines such as IL-10. In practice, Breg cells are identified by their production of immune-regulatory cytokines, such as IL-10, TGF-β, and IL-35, however, no specific marker or Breg cell-specific transcription factor has been identified. Multiple phenotypes of Breg cells have been found, whose functions vary according to their phenotype. This review summarizes the discovery, phenotypes, development, and function of Breg cells and highlights their potential therapeutic value in kidney diseases.
... We also found that tolerized B cells have significant increased FoxP3 mRNA. Another study reported that the expansion of CD25 +hi CD5 + and FoxP3 + regulatory B cells is associated with SLE disease activity in humans (82). Similarly, the presence of FoxP3 + CD19 + CD5 + B cells in human peripheral blood mononuclear cells has also been reported (83). ...
Article
Full-text available
The regulation of autoimmunity and the molecular mechanisms by which different immune cells, including T cells, polymorphonuclear leukocytes (PMN-granulocytes), and B cells suppress autoimmune diseases is complex. We have shown previously that BWF1 lupus mice are protected from autoimmunity after i.v. injection or oral administration of tolerogenic doses of pCons, an artificial synthetic peptide based on sequences containing MHC class I and MHC class II determinants in the VH region of a J558-encoded BWF1 anti-DNA Ab. Several T cell subsets can transfer this tolerance. In this study, we determined the potential roles of granulocytes, B cells and regulatory T cells altered by pCons treatment in the BWF1 (NZB/NZW) mouse model of lupus. Immunophenotyping studies indicated that pCons treatment of BWF1 mice significantly increased CD4 ⁺ FoxP3 ⁺ T cells, reduced the percent of B cells expressing CD19 ⁺ CD5 ⁺ but increased the percent of CD19 ⁺ CD1d ⁺ regulatory B cells and increased the ability of the whole B cell population to suppress IgG anti-DNA production in vitro . pCons treatment significantly decreased the expression of CTLA-4 (cytotoxic T-lymphocyte-associated protein-4) in CD8 ⁺ T cells. In addition, peptide administration modified granulocytes so they became suppressive. We co-cultured sorted naïve B cells from mice making anti-DNA Ab (supported by addition of sorted naive CD4 ⁺ and CD8 ⁺ T cells from young auto-antibody-negative BWF1 mice) with sorted B cells or granulocytes from tolerized mice. Both tolerized granulocytes and tolerized B cells significantly suppressed the production of anti-DNA in vitro . In granulocytes from tolerized mice compared to saline-treated littermate controls, real-time PCR analysis indicated that expression of interferon-induced TNFAIP2 increased more than 2-fold while Ptdss2 and GATA1 mRNA were up-regulated more than 10-fold. In contrast, expression of these genes was significantly down-regulated in tolerized B cells. Further, another IFN-induced protein, Bcl2, was reduced in tolerized B cells as determined by Western blot analyses. In contrast, expression of FoxP3 was significantly increased in tolerized B cells. Together, these data suggest that B cells and granulocytes are altered toward suppressive functions by in vivo tolerization of BWF1 mice with pCons and it is possible these cell types participate in the clinical benefits seen in vivo .
... In 2002, Mizoguchi et al. (21,22) reported that interleukin 10 (IL-10)producing B cells can suppress intestinal inflammation progression in mouse models and first described these cells as Bregs. Currently, various Bregs have been identified, such as CD1d hi CD5 + Bregs (23), CD25 hi FoxP3 hi Bregs (24) and Tim-1 + Bregs (25). The potent regulatory functions of various Bregs have been identified in immune-related pathologies, including inflammation, autoimmunity, and transplantation (22,23,26). ...
Article
Full-text available
Balance of Tfh/Tfr cell is critically important for the maintenance of immune tolerance, as evidenced by the fact that T follicular helper (Tfh) cells are central to the autoantibodies generation through providing necessary help for germinal center (GC) B cells, whereas T follicular regulatory (Tfr) cells significantly inhibit autoimmune inflammation process through restraining Tfh cell responses. However, signals underlying the regulation of Tfh and Tfr cells are largely undefined. Regulatory B cells (Bregs) is a heterogeneous subpopulation of B cells with immunosuppressive function. Considerable advances have been made in their functions to produce anti‐inflammatory cytokines and to regulate Th17, Th1, and Treg cells in autoimmune diseases. The recent identification of their correlations with dysregulated Tfr/Tfh cells and autoantibody production makes Bregs an important checkpoint in GC response. Bregs exert profound impacts on the differentiation, function, and distribution of Tfh and Tfr cells in the immune microenvironment. Thus, unraveling mechanistic information on Tfh-Breg and Tfr-Breg interactions will inspire novel implications for the establishment of homeostasis and prevention of autoantibodies in diverse diseases. This review summarizes the dysregulation of Tfh/Tfr cells in autoimmune diseases with a focus on the emerging role of Bregs in regulating the balance between Tfh and Tfr cells. The previously unsuspected crosstalk between Bregs and Tfh/Tfr cells will be beneficial to understand the cellular mechanisms of autoantibody production and evoke a revolution in immunotherapy for autoimmune diseases.
Article
Uveitis is a disease resulting in the inflammation of uveal tracts, but the factors resulting in uveitis is still obscure. Previous studies have shown that miR-379-5p was involved in the pathogenesis of several diseases, however, the role and regulatory mechanism of miR-379-5p in uveitis were unclear. In our study, we established experimental autoimmune uveitis (EAU) mouse models to explore the role of miR-379-5p in uveitis. RT-qPCR identified that miR-379-5p level was increased in serum of EAU mice. In mechanism, SEMA3A 3’UTR was proven to be directly targeted by miR-379-5p and SEMA3A expression was negatively regulated by miR-379-5p in CD4⁺ T cells. Moreover, ELISA analysis revealed that knockdown of miR-379-5p suppressed the production of inflammation cytokines including IL-17, TNF-α and IL-β in vitro. These results were reversed by SEMA3A overexpression. In addition, the reduction of Th17 cells under miR-379-5p inhibitor was neutralised by SEMA3A knockdown in vitro. Furthermore, we demonstrated that knockdown of miR-379-5p significantly reversed the increased clinical scores and inflammatory response resulting from EAU treatment and this effect was further countervailed by SEMA3A silencing. Our study suggested that miR-379-5p aggravated uveitis in EAU mice via the regulation of SEMA3A, which may provide a novel insight for uveitis treatment.
Article
Nonspecific binding of conjugated antibodies represents a critical step which could significantly influence the results of immunostaining or flow cytometry. In this respect, various staining procedures and distinct cell types can alter the results obtained with different fluorochromes. In this study, we analysed nonspecific binding of R-phycoerythrin (R-PE)-conjugated antibodies to mouse mitogen-stimulated B and T lymphocytes. The cells were fixed, permeabilized and stained using isotype control antibodies conjugated with different fluorochromes and assessed by flow cytometry. R-PE-conjugated antibodies bound to LPS-stimulated B cells, in contrast to Con A-stimulated T cells, independently of their specificity. The percentage of R-PE positive B cells varied, according to the used antibodies or the fixation/permeabilization kit. Nevertheless, up to 30% of R-PE⁺ B cells after staining with R-PE-conjugated isotype control antibodies was detected. Furthermore, LPS-stimulated B cells bound nonspecifically, in a dose-dependent manner, unconjugated R-PE molecules. Con A-stimulated T cells slightly bound R-PE only in high concentrations. Similarly, the antibodies conjugated with other fluorochromes showed less than 1% of nonspecific binding independently of the manufacturer of antibodies or fixation/permeabilization kits. The data demonstrated that LPS-stimulated B cells, in contrast to Con A-stimulated T cells, bind R-PE nonspecifically following formaldehyde or paraformaldehyde fixation. Therefore, the results based on the use of R-PE-conjugated antibodies should be taken with a precaution.
Article
Full-text available
We sought to investigate the expression of Fas and FasL on T cell surface and caspase 8 involvement in T cell apoptosis promoted by serum IL-10 in systemic lupus erythematosus (SLE) patients. Cells and sera were obtained from 35 SLE patients. Apoptosis of T cells in patients with SLE was increased and associated with the SLE disease activity index (SLEDAI). Elevated expression of Fas and FasL on T cell surface contributed to increased apoptosis of T cells. Increased IL-10 in the sera of SLE patients was capable of inducing Fas and FasL expression on CD4(+)T cell surface, promoting apoptosis of this cell subset. Decreased IL-10 serum levels and low expression of Fas were found in 5 patients of the first follow-up group after 2-month treatment. In another group with one-year treatment, the SLEDAI declined to inactive scores. Serum IL-10 was decreased significantly, and expression of Fas and FasL on T cells was also reduced. Declined apoptosis was predominant only in CD4(+)T cell subset. When sera with high level of IL-10 were used to culture PBMCs from healthy controls, activated caspase 8 was elevated in CD3(+)T, CD4(+)T and CD8(+)T cells. The study showed that serum IL-10 induced apoptosis of T cell subsets via the caspase 8 pathway initiated by Fas signaling. Increased apoptosis of T cells contributes to autoantigen burden, which is pathogenic in the development of SLE.
Article
Full-text available
Although classically B cells are known to play important roles in immune protection via humoral immunity, recently their regulatory mechanisms have been best appreciated in the context of autoimmunity. Several studies have identified different subsets of regulatory B cells that vary not only in their phenotype, but also in their mechanism of action. Although the best-studied mechanism of B cell immune regulation is IL-10 production, other IL-10-independent mechanisms have been proposed. These include maintenance of CD4(+)Foxp3(+) T regulatory cells (Treg); production of TGF-β, IL-35, IgM or adenosine; or expression of PD-L1 (programmed death 1) or FasL (Fas ligand). Given that B cell-targeted therapy is being increasingly used in the clinic, a complete understanding of the mechanisms whereby B cells regulate inflammation associated with specific diseases is required for designing safe and effective immunotherapies targeting B cells. © The Japanese Society for Immunology. 2015. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.
Article
Background: Food allergies are important etiologic factors in atopic dermatitis. CD19 is a B-cell-specific cell-surface molecule, with a critical role in B-cell activation. Recently, B cells showed independent two subpopulations as CD19hi and CD19low. The allergen-specific responses of the CD19high and CD19low B-cell subpopulations were investigated in patients with non-IgE-mediated food allergy. Methods: Five milk-allergic subjects and eight milk-tolerant subjects were selected by a double-blind placebo-controlled food challenge. Peripheral blood mononuclear cells (PBMCs) were stimulated in vitro with casein or ovalbumin and stained with monoclonal antibodies to distinguish the B-cell subsets. Results: After allergen stimulation, CD19high B cells increased in the number and the fraction in PBMCs in the milk-tolerant group, whereas those remained unchanged in the milk-allergic group. These responses were constant, regardless of the kind of food allergen (milk or egg). The resulting CD19high/CD19low B-cell ratio increased markedly in the milk-tolerant group after allergen stimulation, but was unchanged in the milk-allergic group. IL-10, IL-17, IL-32 and TGF-p-producing regulatory B cells and Foxp3-expressing regulatory B cells were identified predominantly on CD19 low and CD5(+) B cells. Conclusions: The response of the CD19high B-cell subpopulation to allergen stimulation is decisive for immune tolerance of non-IgE-mediated food allergy in atopic dermatitis. CD19 high and CD5(+) B cells dominantly produce cytokines and express Foxp3. Especially, IL-17 and IL-32 expressing B cells (Br17 & Br32) are present. The exact immunological role of CD19 and cytokines including IL-17 and IL-32 around B cells in immune tolerance requires further investigation.
Article
Although the role of regulatory B cells (Bregs) in rheumatic disease has gained increasing attention, two lesser-known Breg subsets that express either Foxp3 or transforming growth factor beta (TGFβ) are rarely examined in studies of rheumatic disease. This study investigates the association between the relative proportions of CD19(+)Foxp3(+) and CD19(+)TGFβ(+) Bregs, and clinical indicators of disease severity in rheumatoid arthritis (RA) patients with or without interstitial lung disease (ILD). A total of 31 RA patients (14 with and 17 without ILD) and 26 healthy control subjects were included. All subjects did not have other autoimmune disease except RA, tumor, active infection, or a history of related drug administration. Peripheral blood mononuclear cells (PBMCs) were isolated and analyzed by flow cytometry (FCM). The relationship between the relative proportions of CD19(+)Foxp3(+) and CD19(+)TGFβ(+) Bregs and their associations to RA and ILD incidence, as well as disease severity assessed by common clinical indicators, were then examined. Our analyses revealed RA patients had significantly lower proportions of peripheral CD19(+)Foxp3(+) and CD19(+)TGFβ(+) Bregs as compared to healthy controls. While no association was observed between CD19(+)Foxp3(+) Bregs and ILD incidence, patients with ILD had a substantially lower percentage of CD19(+)TGFβ(+) Bregs compared to RA patients without ILD. In addition, CD19(+)Foxp3(+) Bregs were negatively correlated with erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels in RA patients, whereas CD19(+)TGFβ(+) Bregs were only correlated with CRP in RA patients with ILD. Furthermore, there was a negative association between CD19(+)Foxp3(+) Bregs and disease severity scores, which was not found in analyses with CD19(+)TGFβ(+) Bregs. The proportions CD19(+)Foxp3(+) and CD19(+)TGFβ(+) Bregs were significantly decreased in RA patients, particularly in those with ILD complications, suggesting that Breg phenotypes may have different functions in the pathogenesis of RA and ILD.
Article
Systemic lupus erythematosus (SLE) is a complicated autoimmune disease of multifactorial pathoaetiology. One of the most serious manifestations is lupus nephritis. The pathogenesis of SLE has not been well elucidated, but it has been reported that interleukin-17 (IL-17) and Th17 cells play important roles in the pathogenesis of SLE. IL-17A, a member of IL-17 family, amplifies the immune response by inducing the local production of chemokines and cytokines, recruiting neutrophils and monocytes, augmenting the production of autoantibodies, and aggravating the inflammation and damage of target organs such as the kidney in SLE. In recent years, several IL-17A pathway inhibitors have advanced into clinical trials, including the anti-IL-17A monoclonal antibody and the anti-17RA monoclonal antibody. Several agents have shown great success in Phase II trials in multiple autoimmune diseases such as psoriasis, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, and non-infectious uveitis, which has sparked the urgent need of anti-IL-17A as innovative therapeutic option in controlling disease activity of moderate-to-severe SLE. Here, we review and summarize current progress in IL-17A and SLE from in vitro studies, human expression studies, and animal models, providing novel insight into its therapeutic potential.
Article
Imbalance of T-helper-cell (TH) subsets (TH1/TH2/TH17) and regulatory T-cells (Tregs) is suggested to contribute to the pathogenesis of Systemic lupus erythematosus (SLE). Therefore, we evaluated their cytokine secretion profile in SLE patients and their possible association with disease activity. Sixty SLE patients, 24 rheumatoid arthritis (RA) patients and 24 healthy volunteers were included in this study. Demographic, clinical, disease activity and serological data were prospectively assessed. Plasma cytokines levels of TH1 (IL-12, IFN-γ), TH2 (IL-4, IL-6, IL-10), TH17 (IL-17, IL-23) and Treg (IL-10 and TGF-β) were measured by enzyme linked immunosorbent assays (ELISA). SLE patients were found to have significantly higher levels of IL-17 (p<0.001), IL-6 (p<0.01), IL-12 (p<0.001) and IL-10 (p<0.05) but comparable levels of IL-23 and IL-4 and slight reduction (but statistically insignificant) of TGF-β levels compared to controls. IL-6, IL-10 and IL-17 were significantly increased (p<0.05) with disease activity. The RA group exhibited significantly higher levels of plasma IL-4 (p<0.01), IL-6 (p<0.05), IL-17 (p<0.001), IL-23 (p<0.01) and TGF-β (p<0.5) and lower IFN-γ (p<0.001) and IL-10 (p<0.01) than those of healthy subjects. Our study showed a distinct profile of cytokine imbalance in SLE patients. Reduction in IFN-γ (TH1) and TGF-β1 (Treg) with the elevation in IL-6 and IL-17 (TH17) could imply skewing of T-cells toward TH17 cells. Breaking TH17/Treg balance in peripheral blood may play an important role in the development of SLE and could be responsible for an increased pro-inflammatory response especially in the active form of the disease. Copyright © 2015 Elsevier Ltd. All rights reserved.
Article
IL-10 is an immune suppressive cytokine with pleiotropic effects on B cell biology. IL-10 production has a pivotal role for the regulatory suppressive functions that B cells exert in many physiological and pathological settings. Several exogenous stimuli and endogenous immune mediators can trigger IL-10-producing B cell maturation. To clarify and gain a better insight into the mechanisms of IL-10 production by B cells, we first compared the effects of LPS, CpG, agonistic CD40 mAb and BAFF on IL-10 production, and then we investigated the signal transduction mechanisms responsible for these responses. While infectious/danger stimuli determine the rapid production and release of IL-10 by B cells, a limited subset of CD40-poised, IL-10-competent B cells produce IL-10 in response to a later antigenic or infectious signal. Although BAFF is able to induce a similar subset of IL-10-competent B cells, these cells do not similarly respond to the same antigenic or infectious signals. Importantly, by using specific inhibitors of the MAP kinase pathways, we found that while il-10 gene expression triggered by the TLR agonists LPS and CpG is strongly dependent on p38 activity, the induction of IL-10 competence in CD40-activated B cells does not depend on ERK1/2, p38 or JNK pathways.
Article
In multiple sclerosis (MS), the immune damage to the central nervous system results from the net balance between self-reactive and immunoregulatory cells, among other factors. We identified novel perforin-expressing regulatory B-cells (BReg) in patients with clinical isolated syndrome, significantly enriched within the cerebrospinal fluid when compared to peripheral blood, of memory B cell phenotype (CD19+CD25+, CD19+CD25+FoxP3+and CD19+FoxP3+, p = 0.007, p = 0.06 and p = 0.03, respectively). These BRegsubsets were also higher in relapsing-remittent MS during relapse symptoms than in non-clinically active MS patients. Suppressive effects by CD19+CD25+ hi BReg on CD4+ T cells proliferation seem to be mediated at least in part by perforin/granzyme pathway. To our knowledge, this is the first report that shows cytolytic perforin/granzyme granules storage in B cells; the interesting point is its involvement on BRegcells immunosuppressive mechanisms, similarly to that in TRegcells. Our data may extend the understanding of pathophysiological processes in MS immunoregulation.
Article
Background B regulatory cells and their regulatory products/markers, such us semaphorin 3A (sema3A) and its receptor NP-1, FcγIIB, IL-10, and others, act at the very base of self-tolerance maintenance and prevention of autoimmune disease development. Objectives The aim of the present study was to assess the involvement of CD72, a regulatory receptor on B cells, in systemic lupus erythematosus (SLE). In addition, the potential of soluble sema3A in enhancing the expression of CD72 on B cells of SLE patients was investigated. Results CD72 expression on activated B cells of SLE patients was significantly lower than that of normal controls. This lower expression of CD72 in SLE patients correlated inversely with SLE disease activity and was associated with lupus nephritis, the presence of anti-dsDNA antibodies and with low levels of complement. Co-culture of purified B cells from healthy controls with condition-media containing recombinant sema3A resulted in significant enhancement of CD72. Similar enhancement of CD72 on activated B cells from SLE patients, though significant, was still lower than in normal individuals. Conclusions The lower expression of CD72 on activated B cells from SLE patients correlates with SLE disease activity, lupus nephritis, the presence of anti-dsDNA antibodies, and low levels of complement. The improvement of CD72 expression following the addition of soluble semaphorin 3A suggests that CD72 may be useful as a biomarker to be followed during the treatment of SLE.
Article
Fas mutant mice are well recognized as autoimmune mouse models, which develop symptoms similar to human systemic lupus erythematosus. Although disease severity in Fas mutant mice is greatly affected by the genetic background, the mechanisms affecting pathological heterogeneity among different strains of Fas mutant mice are poorly understood. In this study, we examined the phenotypic differences between Fas-deficient (Fas(-/-)) mice on the BALB/c- and C57BL/6-backgrounds to gain insight into the etiological and pathological heterogeneity of monogenic autoimmune diseases. Fas(-/-) mice on the BALB/c background (BALB/c-Fas(-/-)) developed more severe autoimmune disease with high serum autoantibodies and renal disease compared with those on the C57BL/6 background (C57BL/6-Fas(-/-)). Splenic B cells were highly activated, and germinal center formation was enhanced in BALB/c-Fas(-/-) but not in C57BL/6-Fas(-/-) mice. Follicular helper T (Tfh) cells were equally abundant in the spleens from both strains of Fas(-/-) mice. However, Tfh cells from BALB/c-Fas(-/-) mice produced much higher amounts of B cell-activating cytokines including IL-4 and IL-10, a phenotype reminiscent of Th2-type Tfh cells described in human studies. Our results revealed a qualitative difference in Tfh cells between the two strains of Fas(-/-) mice. We propose that the pathogenic Th2-type Tfh cells in BALB/c-Fas(-/-) mice contribute to the excessive activation of B cells, resulting in high serum immunoglobulin levels and the severe lupus phenotype, which may account for the differential outcomes of human monogenic autoimmune diseases.