ArticlePDF Available

Abstract and Figures

B-1 cells constitute a unique subset of B cells identified in several species including mice and humans. B-1 cells are further subdivided into B-1a and B-1b subsets as the former but not the later express CD5. The B-1a subset contributes to innate type of immune responses while the B-1b B cell subset contributes to adaptive responses. B-1 cell responses to B cell receptor (BCR) as well as Toll-like receptor (TLR) ligation are tightly regulated due to the cross-reactivity of antigen specific receptors on B-1 cells to self-antigens. B-1 cells are elevated in several autoimmune diseases. CD5 plays a major role in down regulation of BCR responses in the B-1a cell subset. Reduced amplification of BCR induced signals via CD19 and autoregulation of BCR and TLR responses by B-1 cell produced IL-10 appear to have a role in regulation of both B-1a and B-1b B cell responses. Siglec G receptors and Lyn kinase also regulate B-1 cell responses but their differential role in the two B-1 cell subsets is unknown.
This content is subject to copyright.
MINI REVIEW ARTICLE
published: 17 December 2012
doi: 10.3389/fimmu.2012.00372
Multiple regulatory mechanisms control B-1 B cell
activation
Vishal J. Sindhava1and Subbarao Bondada1,2*
1Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, USA
2Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, USA
Edited by:
Thomas L. Rothstein,The Feinstein
Institute for Medical Research, USA
Reviewed by:
Louis Justement, University of
Alabama at Birmingham, USA
Masaki Hikida, Kyoto University,
Japan
*Correspondence:
Subbarao Bondada, Department of
Microbiology, Immunology and
Molecular Genetics, University of
Kentucky College of Medicine, 303
Combs Cancer Building, Lexington,
KY 40536-0096, USA.
e-mail: bondada@uky.edu
Present address:
Vishal J. Sindhava, Department of
Pathology and Laboratory Medicine,
University of Pennsylvania,
Philadelphia, PA 19104, USA.
B-1 cells constitute a unique subset of B cells identified in several species including mice
and humans. B-1 cells are further subdivided into B-1a and B-1b subsets as the former but
not the later express CD5.The B-1a subset contributes to innate type of immune responses
while the B-1b B cell subset contributes to adaptive responses. B-1 cell responses to B
cell receptor (BCR) as well as Toll-like receptor (TLR) ligation are tightly regulated due to
the cross-reactivity of antigen specific receptors on B-1 cells to self-antigens. B-1 cells are
elevated in several autoimmune diseases. CD5 plays a major role in down regulation of
BCR responses in the B-1a cell subset. Reduced amplification of BCR induced signals via
CD19 and autoregulation of BCR and TLR responses by B-1 cell produced IL-10 appear to
have a role in regulation of both B-1a and B-1b B cell responses. Siglec G receptors and
Lyn kinase also regulate B-1 cell responses but their differential role in the two B-1 cell
subsets is unknown.
Keywords: B lymphocyte, B-1 cell, B cell receptor,Toll-like receptor, CD5, SHP-1, CD19, IL-10
INTRODUCTION
B cells are heterogeneous in their surface phenotypes, anatomical
localization, capacity for self-renewal, and functional properties.
The two major subsets of the B cells are B-2 and B-1 B cells, which
were initially defined by differential expression of a classical T cell
specific differentiation antigen, CD5 (expressed on B-1 cells). The
B cell receptors (BCRs) on B-1 cells exhibit polyreactivity enabling
them to respond to conserved epitopes on microbes, but that also
leads to cross-reactivity with self-antigens. In this review we sum-
marize the mechanisms involvedin regulation of BCR and Toll-like
receptor (TLR) mediated B-1 cell activation.
B-2 CELLS
B-2 cells are produced in bone marrow from hematopoietic stem
cells and migrate to secondary lymphoid organs as immature B
cells. Transitional B cells are the most immature B cells in the
spleen and are the crucial link between bone marrow immature
and peripheral mature B cells (Chung et al., 2003). These tran-
sitional B cells are called the T1 subset when they first emerge
from bone marrow, which mature into T2 subset mainly in the
spleen. The T1 subset is a stage of negative selection against
self-reactive B cells that have escaped central tolerance mech-
anisms in the bone marrow (Cancro et al., 1998). The T2 B
cells further mature and differentiate into follicular and mar-
ginal zone B cells (Miller et al., 2006). B cell-activating factor,
BAFF (a.k.a. B Lymphocyte Stimulator, BLyS) is essential for
the survival of mature follicular and marginal zone B cells. In
both BAFF receptor mutant and BAFF knockout mice, most
transitional B cells fail to differentiate into follicular B cells, and
the few follicular B cells formed have a short lifespan (Lentz
et al., 1996;Gross et al., 2001;Schiemann et al., 2001;Gavin
et al., 2005). BAFF signaling mainly promotes B cell survival,
as enforced expression of anti-apoptotic factors Bcl-2 or Bcl-xL
restored splenic B lymphocyte development in BAFF-R mutant
mice (Amanna et al., 2003;Tardivel et al., 2004). Upon exposure
to antigen, follicular B cells undergo clonal expansion, Ig class
switching, and differentiation into plasma and memory B cells
(McHeyzer-Williams and McHeyzer-Williams, 2005). The tran-
sitional, follicular, and marginal zone B cells comprise the B-2
subset.
B-1 CELLS
B-1 cells were first identified on the basis of expression of CD5,
a pan T cell marker, on a subset of B cells (Manohar et al., 1982;
Hayakawa et al., 1983). B-1 cells are mainly present in peritoneal
cavities, pleural cavities, and various parts of intestine and con-
stitute only a small fraction of B cells in the spleen (Kroese et al.,
1992). B-1 cell origin and development occurs primarily during
fetal and perinatal life. There have been two different models pro-
posed for the origin of the B-1 cells, lineage model, and selection
model. The lineage model supports the existence of distinct prog-
enitors for B-1 and B-2 cells (Hardy and Hayakawa, 1991;Kantor
et al., 1992;Herzenberg and Tung, 2006). Thus transfer of fetal
liver cells reconstituted both B-1 and B-2 cell populations,whereas
adult bone marrow transfer reconstituted conventional B (B-2)
cells but not B-1 cells. In contrast, the selection model proposes a
www.frontiersin.org December 2012 | Volume 3 | Article 372 | 1
Sindhava and Bondada B-1 B cell regulation
common progenitor for both B-1 and B-2 cells and antigen selec-
tion (antigenic stimuli) determines the development of B cells
with B-1 or B-2 characteristics (Lam and Rajewsky, 1999;Berland
and Wortis, 2002). Studies showing that CD5 expression can be
induced by BCR signaling in the presence of certain cytokines and
BCRs specific to some antigens support antigen selection mod-
els. Lam and Rajewsky (1999) showed that co-expression of non
B-1 specific BCR (VHB-1-8 or VHglD42) along with B-1 specific
BCR (VH12) on B cells leads to development of B-2 but not B-
1 cells. They proposed that expression of non B-1 specific BCR
dilutes out VH12-containing BCR complexes on the cell surface
and presumably acts in a dominant-negative” manner and may
not provide sufficient signals for the development of B-1 phe-
notype. This can perhaps best be explained by postulating that
signaling via a BCR of a certain specificity, expressed at the cell
surface at high density, is required to drive the differentiation
of B cells into the B-1 subset (Lam and Rajewsky, 1999). Both
the models are well supported by evidence and are the subject
of considerable debate. However, a recent study from Dorshkind
and colleagues identified specific B-1 cell restricted progenitors
(LinCD45Rlo-negCD19+cells) in bone marrow, which prefer-
entially reconstituted functional B-1 B cells, but not B-2 B cells,
in vivo, providing strong support to the lineage model (Montecino-
Rodriguez et al., 2006). Moreover,using single hematopoietic stem
cells for reconstitution Ghosn et al. (2012) demonstrated that B-1a
cell lineage derives from a precursor that is distinct from other B
cell lineages.
B-1 CELL SUBSETS
Along with the presence of CD5 on the surface, B-1 cells are further
differentiated from B-2 cells by surface expression of CD11b (Mac-
1), high levels of IgM, and low levels of IgD (Tung et al., 2004).
B-1 cells are subdivided into three different subsets, B-1a, B-1b,
and B-1c, on the basis of CD5 and CD11b expression. B-1a cells
are CD11b+CD5+, B-1b cells are CD11b+CD5, and the newly
described rare subpopulation of B-1c cells is CD11bCD5+(Tung
et al., 2004;Hastings et al., 2006). B-1a cells have a role in innate
immunity via their contribution to natural antibodies, while B-
1b cells are critical in development of IgM memory cells (Berland
and Wortis, 2002). The functional properties of B-1c are essentially
similar to those of B-1a and B-1b B cells (Hastings et al., 2006). The
unique markers for B-1 cells in the human were unclear since CD5
was expressed by activated human B cells. More recently, Griffin
et al. (2011) have characterized human B-1 cell surface phenotype
and function, which resembled the properties of murine B-1 cells
very closely (Griffin et al., 2011;Griffin and Rothstein, 2012).
B-1 cells are important in protection against certain bacterial
infections such as S. pneumoniae,Borrelia hermsii as well as in the
early IgM response against viruses such as influenza (Baumgarth
et al., 2000;Alugupalli et al., 2003;Haas et al., 2005). Despite their
role in protection against infection, B-1 cell antibodies have been
found to be poly-reactive and as such are reactive to self-antigens
such as those on red blood cells, thy 1.2, single stranded DNA
(Berland and Wortis, 2002). Moreover, B-1 cells have been found
to be elevated in autoimmune diseases both in mouse and human.
In mouse models, elimination of B-1 cells by genetic deficiency
reduced autoimmunity (Duan and Morel, 2006).
BCR SIGNALING IN B-1 CELLS
B cell receptor signaling plays a critical role in B-1 cell devel-
opment, survival, or expansion. Transgenic mice or mice with
mutations that disrupt BCR signaling have a decrease in B-1 cell
numbers, and mutations that enhance BCR signaling result in
increased B-1 cell compartment (Berland and Wortis, 2002). How-
ever, the cross-reactivity of B-1 BCRs with self-antigens raised
the question of how B-1 cells are prevented from activation via
self-antigens in the absence of overt infection. Studies of BCR sig-
naling have demonstrated distinct differences between B-1 and
B-2 cells. Engagement of BCR on B-2 cells leads to robust intra-
cellular calcium mobilization and proliferation, while in B-1 cells,
BCR ligation induces modest calcium mobilization, little or no
proliferation, and increased apoptosis (Murakami et al., 1992;
Morris and Rothstein, 1993;Bikah et al., 1996;Sen et al., 1999).
Here we summarized the key molecules that negatively regulate
BCR and TLR signaling in B-1 cells and have a role in B-1 cell
hypo-responsiveness to BCR ligation.
Negative regulatory role of CD5 in B-1a cells
CD5 is a 67-kDa monomeric type 1 transmembrane glycoprotein,
historically also known as Lyt-1 or Ly-1. Extracellular domains
of CD5 are characterized by the presence of the highly conserved
scavenger receptor cysteine-rich domain. CD5 expression was first
identified on T cells (Boyse et al., 1968) and subsequently shown
to be expressed on B cells (Manohar et al., 1982;Okumura et al.,
1982;Hardy et al., 1983;Hayakawa et al., 1983). CD5+B cells,
later termed B-1a cells, have unique function of “spontaneous”
IgM secretion that contributes to natural antibodies (Hayakawa
et al., 1983). Also, B-1 cells have a limited BCR repertoire with
dominant cross-reactivity to self-antigens, but expansion of these
poly-reactive B-1 cells is limited (Berland and Wortis, 2002). This
limited expansion of self-reactive B-1 cells may be in part due to
the presence of various mechanisms that negatively regulate BCR
signaling.
Various studies identified CD5 as one of the negative regulators
of BCR signaling, similar to its ability to inhibit T cell function
(Tarakhovsky et al., 1995). In B cells CD5 associates with mIgM
upon BCR stimulation (Lankester et al., 1994). However, CD5 is
shown to be constitutively associated with mIgM in peritoneal B-1
cells (Sen et al., 1999). Bikah et al. (1996) demonstrated for the first
time that CD5 negatively regulates BCR signaling in peritoneal B-1
cells. B-1 cells from both wild type (WT) and CD5 KO mice pro-
liferated comparably in response to anti-CD40 and LPS. However,
only CD5 KO B-1 cells, but not WT B-1 cells, proliferated to anti-
IgM stimulation. This involved sustained calcium mobilization
and increased nuclear localization of NF-κB following BCR liga-
tion in CD5 KO compared to WT peritoneal B-1 cells. Additionally,
blocking of CD5 association with mIgM rescued the proliferative
defect of B-1 cells upon BCR ligation (Bikah et al., 1996). Using a
novel fusion protein containing the extracellular and transmem-
brane domains of FcγRIIB and the cytoplasmic region of CD5
Gary-Gouy et al. (2000, 2002) showed that co-cross-linking of
BCR with the chimeric protein induced tyrosine phosphoryla-
tion in CD5 cytoplasmic tail along with rapid inhibition of BCR
induced calcium transients and extracellular regulated kinase-2
(ERK2) activation. Subsequent they showed that Y429, residue
Frontiers in Immunology | B Cell Biology December 2012 | Volume 3 | Article 372 | 2
Sindhava and Bondada B-1 B cell regulation
outside the putative immune receptor tyrosine based inhibitory
motif (ITIM) of CD5 cytoplasmic domain is responsible for the
inhibition of BCR induced calcium response, Akt relocalization
(Gary-Gouy et al., 2002), activation of the Ras/ERK2 pathway as
well as IL-2 production (Gary-Gouy et al., 2002).
Using the well-known transgenic mouse model of anti-hen
egg lysozyme (HEL) crossed to transgenic mice expressing sol-
uble HEL (Goodnow et al., 1988), Hippen and Behrens showed
that anergic B cells expressed significant surface levels of CD5,
though lower than those expressed on typical B-1a cells. This
suggested that a low level of CD5 induction on B cells upon stim-
ulation through auto-antigen might be sufficient to induce an
anergic state in self-reactive B cells and thus limiting production
of autoantibodies. Consistent with such a concept, CD5 KO mice,
but not CD5+mice, that are transgenic for both HEL specific BCR
and soluble lysozyme produced antibody to the self-antigen, HEL
(Hippen et al., 2000). Also, the CD5 KO HEL specific B cells that
are no longer anergic showed enhanced proliferative responses
and calcium mobilization upon BCR ligation. Together, data from
Bikah et al. (1996) and by Hippen et al. (2000) suggested that CD5
negatively regulates BCR signaling and limits self-reactive B cell
responses. Similar to anergic B cells in the transgenic model, con-
stitutive expression of CD5 on B-1a cells might also play a role in
limiting the expansion of auto-reactive B-1a cells.
Lyn, SHP-1, CD22 and Siglec G
B cell receptor signal transduction occurs via activation of sev-
eral protein tyrosine kinases (PTK), including members of the Src
family kinases (SFKs; Cambier et al., 1994). In addition to taking
part in activation of the BCR signaling, Lyn, an SFK, negatively
regulates BCR signaling by phosphorylating the ITIM motifs in
B cell co-receptors (DeFranco et al., 1998). Phosphorylation of
ITIM motif with Lyn induces the recruitment of negative mole-
cular switches, like protein tyrosine phosphatases (PTP; Thomas,
1995). Dasu et al. (2009) also made the surprising observation that
B-1 cells have a constitutively active Lyn. Lyn appears to have dual
roles in B-1 cells such that high doses provide negative signals,
whereas small amounts of Lyn were essential for B-1 cell activa-
tion as demonstrated by rescue of both proliferation and calcium
responses at low doses of Lyn kinase inhibitors (Dasu et al., 2009).
Several PTPs like, tyrosine-protein phosphatase non-receptor
type 6 (SHP-1/PTPN6), SHP-2, and inositol polyphosphate 50
phosphatase are involved in the inhibition of BCR signaling
(Thomas, 1995;Long, 1999). In B cells, SHP-1 associates with
inhibitory receptors like FcR, CD22, and paired Ig-like receptor,
PIR-B (Doody et al., 1995;Pani et al., 1995;Long, 1999). Mothe-
aten and the viable motheaten mice with mutations in the SHP-1
enzyme exhibit autoimmunity and accelerated mortality due to
the presence of hyper-responsive B-1 cells (Cyster and Goodnow,
1995). Moreover, B cell specific deletion of SHP-1 leads to an
expansion of B-1 cells, rescue of BCR induced calcium response
and autoimmunity characterized by anti-DNA antibodies (Pao
et al., 2007).
Cytoplasmic domain of CD5 contains an amino acid sequence
(LAY378KKL), with excellent homology to ITIMs of inhibitory
receptors (Perez-Villar et al., 1999) and thus can interact with SHP-
1. There is a constitutive association of the BCR with SHP-1 in both
B-1 and B-2 cells (Sen et al., 1999). Upon BCR ligation association
of SHP-1 is decreased in splenic B-2 cells, but not in peritoneal B-1
cells. The persistent BCR-SHP-1 association is mediated by CD5 in
peritoneal B-1 cells and is lost in CD5 KO peritoneal B-1 cells or in
wild type B-1 cells when CD5 is sequestered away from BCR (Sen
et al., 1999). These data suggest that CD5 may negatively regulate
BCR-mediated growth signals by recruiting SHP-1 into the BCR
complex in B-1 cells.
The B cell surface molecules CD22 and CD72 can also asso-
ciate with SHP-1, but were not found to have a critical role in
the anergic state of B-1 cells (Ochi and Watanabe, 2000;Lajau-
nias et al., 2002). However, Siglec G, another member of the
CD22 family of sialic acid binding proteins, is important for both
B-1 cell development and for regulating BCR induced calcium
responses, but not BCR induced B-1 cell proliferation (Hoffmann
et al., 2007). Interestingly, CD22/Siglec G/double knock-
out mice had even greater elevation of BCR induced calcium
responses in B-1 cells suggesting a role for both molecules in regu-
lating BCR responses in B-1 cells (Jellusova et al., 2010a,b). These
double knockouts develop anti-nuclear antibodies and glomerular
nephritis (Jellusova et al., 2010b).
CD19
The inability of B-1a cells to respond well to BCR signaling with a
role for negative regulation by CD5 and SHP-1 is described above.
B-1b B cells, which are CD5 negative by definition, are equally
defective in proliferation induced by BCR cross-linking raising
the possibility that additional mechanisms may exist to down-
regulate BCR responses in B-1 cells (Sen et al., 1999). CD19, a
co-receptor expressed by all B cell subsets, serves as a positive
regulator of BCR signaling and is critical for B cell development
and activation (Cambier et al., 1994;Tedder et al., 1994). CD19
is shown to be involved in the development and self-renewal of
B-1 cells (Krop et al., 1996;Sato et al., 1996). Signals from CD19
and BCR act in synergy to induce robust calcium mobilization in
splenic B-2 cells (Carter et al., 1991;Fujimoto et al., 2001). Using
this criterion, Sen et al. (2002) found that both B-1a and B-1b
B cells are equally hypo-responsive to synergistic calcium mobi-
lization obtained by co-cross-linking BCR and CD19 compared
to B-2 cells. CD19 cross-linking amplifies BCR signaling in part
by recruitment of Vav, a guanine nucleotide exchange factor for
the Rho, Rac, and Cdc42 family of small GTPases (O’Rourke et al.,
1998). Vav binds to phosphorylated tyrosine-391 of CD19 to medi-
ate a sustained increase in intracellular Ca2+concentration and
activation of the mitogen-activated protein kinase, JNK (O’Rourke
et al., 1998). Association of CD19 with Vav is reduced in B-1 cells
(Sen et al., 2002). Similarly BCR dependent proliferation as well as
CD19 mediated augmentation of BCR induced calcium elevation
were deficient in B-1a and B-1b B cell subsets from both spleen
and peritoneal cavity (Dasu et al., 2009). The defects in calcium
response were mainly in the mobilization of extracellular calcium
by both B-1a and B-1b B cells stimulated via CD19 and BCR.
IL-10 AND TLR SIGNALING IN B-1 CELLS
Toll-like receptors are pattern recognition receptors that recognize
pathogen-associated molecular patterns. So far eleven functional
www.frontiersin.org December 2012 | Volume 3 | Article 372 | 3
Sindhava and Bondada B-1 B cell regulation
TLRs (TLR8 and TLR10 genes do not encode functional pro-
teins) have been described in the mouse (Browne, 2012). Several
B cell subsets express TLRs and can be activated via TLR ligands
that result in robust proliferation and antibody secretion, even
in the absence of dendritic cell or T cell help (Genestier et al.,
2007;Gururajan et al., 2007). TLR mediated signals synergize
with self-antigen-mediated BCR signals to stimulate activation
of self-reactive B cells (Leadbetter et al., 2002), and B cell acti-
vation was severely reduced when the mice were deficient in
TLR signaling (Leadbetter et al., 2002;Browne, 2012). B-1 cells
express various TLRs (TLR1, 2, 3, 4, 7, 8, and 9; Gururajan et al.,
2007). B-1 cells are more prone to terminal plasma cell differ-
entiation than B-2 cells upon TLR stimulation (Genestier et al.,
2007). B-1 cell activation and B-1 cell mediated auto-antibody
production are augmented upon stimulation via TLR4 or TLR9
(Murakami et al., 1994;Kubo et al., 2009). Also, TLR signaling
in B-1 cells plays an important role in the clearance of various
pathogens such as influenza virus, pneumococcus, and Borre-
lia spp. (Baumgarth et al., 2000;Alugupalli et al., 2003;Haas
et al., 2005). Sindhava et al. (2010) showed that peritoneal B-1
cells are hypo-responsive to various TLR ligands compared to B-2
cells.
Peritoneal B-1 cells were the first B cell subset to be reported as
being high IL-10 producers (O’Garra et al., 1992). Recently human
CD11b+B-1 cells were also found to spontaneously secrete IL-
10 (Griffin and Rothstein, 2012). IL-10 is a potent regulator of
immune function through its ability to inhibit antigen presenta-
tion, pro-inflammatory cytokine production, T cell proliferation,
and acts as a key effector molecule for certain types of regulatory
T and B cells. As such, IL-10 production by B-1 cells raises the
possibility that B-1 cells may regulate their own function and/or
the function of other immunocompetent cells. Sindhava et al.
(2010) showed that among different B cell subsets from spleen
and peritoneum, B-1 cells from the peritoneum are the major IL-
10 producers. Peritoneal B-1a cells produced highest level of IL-10
followed by B-1b cells, whereas IL-10 production by peritoneal B-2
cells was minimal both constitutively and upon TLR stimulation
(Sindhava et al., 2010). The high levels of IL-10 produced by peri-
toneal B-1 cells regulated the TLR as well as BCR (Sindhava et al.,
unpublished results) induced B-1 cell proliferation in an autocrine
fashion, as B-1 cells from IL-10 knockout mice proliferated signif-
icantly more than WT B-1 cells both in vitro and in vivo. IL-10
regulated B-1 cell response to TLR by inhibiting classical NF-κB
signaling. Furthermore, IL-10 produced by peritoneal B-1 cells
FIGURE 1 | Regulation of B-1 B cell activation. (1 and 2) CD5 mediated
regulation CD5 acts as an anchor for SHP-1 recruitment on cell surface
near BCR signaling complex, which in turn inhibits BCR signaling. (3)
CD19 mediated regulation B-1 cells have defective Vav recruitment to
CD19 leading to reduced Ca2+mobilization and cell activation upon BCR
co-stimulation. (4) Src family kinase mediated regulation Src family
kinase, Lyn, plays an essential role in phosphorylation of CD5 and
subsequent recruitment of SHP-1 on CD5. (5) IL-10 mediated
regulation B-1 cells make high levels of IL-10 uponTLR and BCR
stimulation, which work in an autocrine manner and inhibit B-1 cell
responses by blocking degradation of IκBαand RelA translocation to the
nucleus.
Frontiers in Immunology | B Cell Biology December 2012 | Volume 3 | Article 372 | 4
Sindhava and Bondada B-1 B cell regulation
limits the clearance of B. hermsii infection (Sindhava et al., 2010).
Thus, similar to BCR signaling, TLR signaling is also regulated in
peritoneal B-1 cells, which might prevent excessive activation of
self-reactive B cells via TLR stimulation.
CONCLUDING REMARKS
B-1 cells express poly-reactive BCRs with cross-reactivity to self-
antigens. Accidental activation by self-antigens is prevented by
multiple mechanisms that keep B-1 cells in anergic state. Lyn, a
major negative regulator of BCR signaling phosphorylates ITIMs
on inhibitory receptors (CD5, Siglecs, etc.) leading to recruitment
of PTPs, which antagonize the BCR mediated activation of PTKs.
Anatomical location of B-1 cells makes them prone to activation
through microbial TLR ligands that might result in auto-antibody
production. IL-10 mediated autoregulation plays a key role in con-
trolling expansion of self-reactive B-1 cells. However, signals from
CD40 and high dose TLR ligands can overcome the anergic state
of B-1 cells enabling their activation during infection. Defects in
the negative regulatory mechanisms may account for elevation of
B-1 cells and autoantibodies in lupus like autoimmune diseases.
Various molecules that negatively regulate B-1 cell activation are
summarized in Figure 1.
ACKNOWLEDGMENTS
This work was supported in part by NIH grants AI21490, AG05731,
CA09357, and AI076956,a grant from Vice President for Research,
University of Kentucky and a grant from Edward P. Evans Founda-
tion to Subbarao Bondada. The authors acknowledge the contri-
butions of Drs. G. Bikah, C. Venkataraman, G. Sen, R. L. Chelvara-
jan, Hsin-Jung Wu, Murali Gururajan, and T. Dasu who were pre-
vious members of the laboratory. Also, the authors acknowledge
Dr. M. E. Woodman and Dr. B. Stevenson for their contributions.
REFERENCES
Alugupalli, K. R., Gerstein, R. M., Chen,
J., Szomolanyi-Tsuda, E., Wood-
land, R. T., and Leong, J. M.
(2003). The resolution of relaps-
ing fever borreliosis requires IgM
and is concurrent with expansion of
B1b lymphocytes. J. Immunol. 170,
3819–3827.
Amanna, I. J., Dingwall, J. P., and
Hayes, C. E. (2003). Enforced bcl-xL
gene expression restored splenic B
lymphocyte development in BAFF-
R mutant mice. J. Immunol. 170,
4593–4600.
Baumgarth, N., Chen, J., Herman, O.
C., Jager, G. C., and Herzenberg,
L. A. (2000). The role of B-1
and B-2 cells in immune protec-
tion from influenza virus infection.
Curr. Top. Microbiol. Immunol. 252,
163–169.
Berland, R., and Wortis, H. H. (2002).
Origins and functions of B-1 cells
with notes on the role of CD5. Annu.
Rev. Immunol. 20, 253–300.
Bikah, G., Carey, J., Ciallella, J. R.,
Tarakhovsky, A., and Bondada, S.
(1996). CD5-mediated negative reg-
ulation of antigen receptor-induced
growth signals in B-1 B cells. Science
274, 1906–1909.
Boyse, E. A., Miyazawa, M.,Aoki, T., and
Old, L. J. (1968). Ly-A and Ly-B: two
systems of lymphocyte isoantigens
in the mouse. Proc. R. Soc. Lond. B
Biol. Sci. 170, 175–193.
Browne, E. P. (2012). Regulation of B-
cell responses by Toll-like receptors.
Immunology 136, 370–379.
Cambier, J. C., Pleiman, C. M., and
Clark, M. R. (1994). Signal trans-
duction by the B cell antigen recep-
tor and its coreceptors. Annu. Rev.
Immunol. 12, 457–486.
Cancro, M. P., Allman, D. M., Hayes, C.
E., Lentz, V. M., Fields, R. G., Sah,
A. P., et al. (1998). B cell matura-
tion and selection at the marrow-
periphery interface. Immunol. Res.
17, 3–11.
Carter, R. H., Tuveson, D. A., Park,
D. J., Rhee, S. G., and Fearon, D.
T. (1991). The CD19 complex of
B lymphocytes. Activation of phos-
pholipase C by a protein tyro-
sine kinase-dependent pathway that
can be enhanced by the mem-
brane IgM complex. J. Immunol.147,
3663–3671.
Chung, J. B., Silverman, M., and Mon-
roe, J. G. (2003). Transitional B
cells: step by step towards immune
competence. Trends Immunol. 24,
343–349.
Cyster, J. G., and Goodnow, C.
C. (1995). Protein tyrosine phos-
phatase 1C negatively regulates anti-
gen receptor signaling in B lym-
phocytes and determines thresholds
for negative selection. Immunity 2,
13–24.
Dasu, T.,Sindhava, V.,Clarke, S. H., and
Bondada, S. (2009). CD19 signal-
ing is impaired in murine peritoneal
and splenic B-1 B lymphocytes. Mol.
Immunol. 46, 2655–2665.
DeFranco,A. L., Chan,V.W., and Lowell,
C. A. (1998). Positive and negative
roles of the tyrosine kinase Lyn in
B cell function. Semin. Immunol. 10,
299–307.
Doody, G. M., Justement, L. B.,
Delibrias, C. C., Matthews, R.
J., Lin, J., and Thomas, M. L.
(1995). A role in B cell activation
for CD22 and the protein tyro-
sine phosphatase SHP. Science 269,
242–244.
Duan, B., and Morel, L. (2006). Role of
B-1a cells in autoimmunity. Autoim-
mun. Rev. 5, 403–408.
Fujimoto, M., Poe, J. C., Hasegawa,
M., and Tedder, T. F. (2001).
CD19 amplification of B lympho-
cyte Ca2+ responses: a role for Lyn
sequestration in extinguishing neg-
ative regulation. J. Biol. Chem. 276,
44820–44827.
Gary-Gouy, H., Bruhns, P., Schmitt,
C., Dalloul, A., Daeron, M., and
Bismuth, G. (2000). The pseudo-
immunoreceptor tyrosine-based
activation motif of CD5 mediates
its inhibitory action on B-cell
receptor signaling. J. Biol. Chem.
275, 548–556.
Gary-Gouy, H., Harriague, J., Dalloul,
A., Donnadieu, E., and Bismuth, G.
(2002). CD5-negative regulation of
B cell receptor signaling pathways
originates from tyrosine residue
Y429 outside an immunoreceptor
tyrosine-based inhibitory motif. J.
Immunol. 168, 232–239.
Gavin, A. L., Duong, B., Skog, P., Ait-
Azzouzene, D., Greaves, D. R., Scott,
M. L., et al. (2005). deltaBAFF, a
splice isoform of BAFF,opposes full-
length BAFF activity in vivo in trans-
genic mouse models. J. Immunol.
175, 319–328.
Genestier, L., Taillardet, M., Mondiere,
P., Gheit, H., Bella, C., and Defrance,
T. (2007). TLR agonists selectively
promote terminal plasma cell differ-
entiation of B cell subsets specialized
in thymus-independent responses. J.
Immunol. 178, 7779–7786.
Ghosn, E. E., Yamamoto, R., Hamanaka,
S., Yang, Y., Herzenberg, L. A.,
Nakauchi, H., et al. (2012). Dis-
tinct B-cell lineage commitment
distinguishes adult bone mar-
row hematopoietic stem cells.
Proc. Natl. Acad. Sci. U.S.A. 109,
5394–5398.
Goodnow, C. C., Crosbie, J., Adel-
stein, S., Lavoie, T. B., Smith-
Gill, S. J., Brink, R. A. (1988).
Altered immunoglobulin expression
and functional silencing of self-
reactive B lymphocytes in transgenic
mice. Nature 334, 676–682.
Griffin, D. O., Holodick, N. E.,
and Rothstein, T. L. (2011).
Human B1 cells in umbilical
cord and adult peripheral blood
express the novel phenotype
CD20+CD27+CD43+CD70.J.
Exp. Med. 208, 67–80.
Griffin, D. O., and Rothstein, T.
L. (2012). Human orchestrator”
CD11b(+) B1 cells spontaneously
secrete IL-10 and regulate T cell
activity. Mol. Med. 18, 1003–1008.
Gross, J. A., Dillon, S. R., Mudri,
S., Johnston, J., Littau, A., Roque,
R., et al. (2001). TACI-Ig neutral-
izes molecules critical for B cell
development and autoimmune dis-
ease. Impaired B cell maturation in
mice lacking BLyS. Immunity 15,
289–302.
Gururajan, M., Jacob, J., and Pulen-
dran, B. (2007). Toll-like receptor
expression and responsiveness of
distinct murine splenic and mucosal
B-cell subsets. PLoS ONE 2:e863.
doi:10.1371/journal.pone.0000863
Haas, K. M., Poe, J. C., Steeber, D. A.,
and Tedder, T. F. (2005). B-1a and B-
1b cells exhibit distinct developmen-
tal requirements and have unique
functional roles in innate and adap-
tive immunity to S. pneumoniae.
Immunity 23, 7–18.
Hardy, R. R., and Hayakawa, K. (1991).
A developmental switch in B lym-
phopoiesis. Proc. Natl. Acad. Sci.
U.S.A. 88, 11550–11554.
Hardy, R. R., Hayakawa, K., Parks, D.
R., and Herzenberg, L. A. (1983).
Demonstration of B-cell matura-
tion in X-linked immunodeficient
mice by simultaneous three-colour
immunofluorescence. Nature 306,
270–272.
www.frontiersin.org December 2012 | Volume 3 | Article 372 | 5
Sindhava and Bondada B-1 B cell regulation
Hastings, W. D., Gurdak,S. M., Tumang,
J. R., and Rothstein, T. L. (2006).
CD5+/Mac-1- peritoneal B cells: a
novel B cell subset that exhibits char-
acteristics of B-1 cells. Immunol.
Lett. 105, 90–96.
Hayakawa, K., Hardy, R. R., Parks, D.
R., and Herzenberg, L. A. (1983).
The “Ly-1 B” cell subpopulation
in normal immunodefective, and
autoimmune mice. J. Exp. Med. 157,
202–218.
Herzenberg, L. A., and Tung, J. W.
(2006). B cell lineages: documented
at last! Nat. Immunol. 7, 225–226.
Hippen, K. L., Tze, L. E., and Behrens, T.
W. (2000). CD5 maintains tolerance
in anergic B cells. J. Exp. Med. 191,
883–890.
Hoffmann, A., Kerr, S., Jellusova, J.,
Zhang, J., Weisel, F., Wellmann, U.,
et al. (2007). Siglec-G is a B1
cell-inhibitory receptor that con-
trols expansion and calcium signal-
ing of the B1 cell population. Nat.
Immunol. 8, 695–704.
Jellusova, J., Duber, S., Guckel, E.,
Binder, C. J., Weiss, S., Voll, R.
(2010a). Siglec-G regulates B1 cell
survival and selection. J. Immunol.
185, 3277–3284.
Jellusova, J., Wellmann, U., Amann,
K., Winkler, T. H., and Nitschke,
L. (2010b). CD22 x Siglec-G
double-deficient mice have mas-
sively increased B1 cell numbers and
develop systemic autoimmunity. J.
Immunol. 184, 3618–3627.
Kantor, A. B., Stall, A. M., Adams,
S., and Herzenberg, L. A. (1992).
Differential development of prog-
enitor activity for three B-cell lin-
eages. Proc. Natl. Acad.Sci. U.S.A. 89,
3320–3324.
Kroese, F. G., Ammerlaan, W. A., and
Deenen, G. J. (1992). Location and
function of B-cell lineages. Ann. N.
Y. Acad. Sci. 651, 44–58.
Krop, I., de Fougerolles, A. R., Hardy, R.
R., Allison, M., Schlissel, M. S., and
Fearon, D. T. (1996). Self-renewal
of B-1 lymphocytes is dependent on
CD19. Eur. J. Immunol. 26,238–242.
Kubo, T., Uchida, Y., Watanabe, Y.,
Abe, M., Nakamura, A., Ono, M.,
et al. (2009). Augmented TLR9-
induced Btk activation in PIR-B–
deficient B-1 cells provokes exces-
sive autoantibody production and
autoimmunity. J. Exp. Med. 206,
1971–1982.
Lajaunias, F., Nitschke, L., Moll, T.,
Martinez-Soria, E., Semac, I.,
Chicheportiche, Y. (2002). Differ-
entially regulated expression and
function of CD22 in activated B-1
and B-2 lymphocytes. J. Immunol.
168, 6078–6083.
Lam, K. P., and Rajewsky, K. (1999). B
cell antigen receptor specificity and
surface density together determine
B-1 versus B-2 cell development. J.
Exp. Med. 190, 471–477.
Lankester, A. C., van Schijndel, G. M.,
Cordell, J. L., van Noesel, C. J., and
van Lier, R. A. (1994). CD5 is asso-
ciated with the human B cell antigen
receptor complex. Eur. J. Immunol.
24, 812–816.
Leadbetter, E. A., Rifkin, I. R.,
Hohlbaum, A. M., Beaudette,
B. C., Shlomchik, M. J., and
Marshak-Rothstein, A. (2002).
Chromatin-IgG complexes activate
B cells by dual engagement of IgM
and Toll-like receptors. Nature 416,
603–607.
Lentz, V. M., Cancro, M. P., Nashold, F.
E., and Hayes, C. E. (1996). Bcmd
governs recruitment of new B cells
into the stable peripheral B cell pool
in the A/WySnJ mouse. J. Immunol.
157, 598–606.
Long, E. O. (1999). Regulation
of immune responses through
inhibitory receptors. Annu. Rev.
Immunol. 17, 875–904.
Manohar, V., Brown, E., Leiserson, W.
M., and Chused, T. M. (1982).
Expression of Lyt-1 by a subset of
B lymphocytes. J. Immunol. 129,
532–538.
McHeyzer-Williams, L. J., and
McHeyzer-Williams, M. G. (2005).
Antigen-specific memory B cell
development. Annu. Rev. Immunol.
23, 487–513.
Miller,J. P.,Stadanlick, J. E., and Cancro,
M. P. (2006). Space, selection, and
surveillance: setting boundaries with
BLyS. J. Immunol. 176, 6405–6410.
Montecino-Rodriguez, E., Leathers, H.,
and Dorshkind, K. (2006). Identifi-
cation of a B-1 B cell-specified prog-
enitor. Nat. Immunol. 7, 293–301.
Morris, D. L., and Rothstein, T. L.
(1993). Abnormal transcription fac-
tor induction through the surface
immunoglobulin M receptor of B-
1 lymphocytes. J. Exp. Med. 177,
857–861.
Murakami, M., Tsubata, T., Okamoto,
M., Shimizu, A., Kumagai, S., Imura,
H., et al. (1992). Antigen-induced
apoptotic death of Ly-1 B cells
responsible for autoimmune dis-
ease in transgenic mice. Nature 357,
77–80.
Murakami, M., Tsubata, T., Shinkura,
R., Nisitani, S., Okamoto, M., Yosh-
ioka, H., et al. (1994). Oral adminis-
tration of lipopolysaccharides acti-
vates B-1 cells in the peritoneal cav-
ity and lamina propria of the gut and
induces autoimmune symptoms in
an autoantibody transgenic mouse.
J. Exp. Med. 180, 111–121.
Ochi, H., and Watanabe,T. (2000). Neg-
ative regulation of B cell receptor-
mediated signaling in B-1 cells
through CD5 and Ly49 co-receptors
via Lyn kinase activity. Int.Immunol.
12, 1417–1423.
O’Garra, A., Chang, R., Go, N., Hast-
ings, R., Haughton, G., and Howard,
M. (1992). Ly-1 B (B-1) cells are
the main source of B cell-derived
interleukin 10. Eur. J. Immunol. 22,
711–717.
Okumura, K., Hayakawa, K., and
Tada, T. (1982). Cell-to-cell interac-
tion controlled by immunoglobulin
genes. Role of Thy-1-, Lyt-1+, Ig+
(B0) cell in allotype-restricted anti-
body production. J. Exp. Med. 156,
443–453.
O’Rourke, L. M., Tooze, R., Turner,
M., Sandoval, D. M., Carter, R. H.,
Tybulewicz,V.L., et al. (1998). CD19
as a membrane-anchored adaptor
protein of B lymphocytes: costim-
ulation of lipid and protein kinases
by recruitment of Vav. Immunity 8,
635–645.
Pani, G., Kozlowski, M., Cambier, J.
C., Mills, G. B., and Siminovitch,
K. A. (1995). Identification of the
tyrosine phosphatase PTP1C as a B
cell antigen receptor-associated pro-
tein involved in the regulation of
B cell signaling. J. Exp. Med. 181,
2077–2084.
Pao, L. I., Lam, K. P., Henderson,
J. M., Kutok, J. L., Alimzhanov,
M., Nitschke, L. (2007). B cell-
specific deletion of protein-tyrosine
phosphatase Shp1 promotes B-1a
cell development and causes sys-
temic autoimmunity. Immunity 27,
35–48.
Perez-Villar, J. J., Whitney, G. S., Bowen,
M. A., Hewgill, D. H., Aruffo, A. A.,
and Kanner, S. B. (1999). CD5 neg-
atively regulates the T-cell antigen
receptor signal transduction path-
way: involvementof SH2-containing
phosphotyrosine phosphatase SHP-
1. Mol. Cell. Biol. 19, 2903–2912.
Sato, S., Ono, N., Steeber, D. A., Piset-
sky, D. S., and Tedder, T. F. (1996).
CD19 regulates B lymphocyte sig-
naling thresholds critical for the
development of B-1 lineage cells
and autoimmunity. J. Immunol. 157,
4371–4378.
Schiemann, B., Gommerman, J. L.,Vora,
K., Cachero,T. G., Shulga-Morskaya,
S., Dobles, M., et al. (2001). An
essential role for BAFF in the nor-
mal development of B cells through
a BCMA-independent pathway. Sci-
ence 293, 2111–2114.
Sen, G., Bikah, G., Venkataraman, C.,
and Bondada, S. (1999). Nega-
tive regulation of antigen receptor-
mediated signaling by constitutive
association of CD5 with the SHP-
1 protein tyrosine phosphatase in
B-1 B cells. Eur. J. Immunol. 29,
3319–3328.
Sen, G., Wu, H. J., Bikah, G., Venkatara-
man, C., Robertson, D. A., Snow, E.
C., et al. (2002). Defective CD19-
dependent signaling in B-1a and B-
1b B lymphocyte subpopulations.
Mol. Immunol. 39, 57–68.
Sindhava, V., Woodman, M. E., Steven-
son, B., and Bondada, S. (2010).
Interleukin-10 mediated autoreg-
ulation of murine B-1 B-cells
and its role in Borrelia herm-
sii infection. PLoS ONE 5:e11445.
doi:10.1371/journal.pone.0011445
Tarakhovsky, A., Kanner, S. B., Hom-
bach, J., Ledbetter, J. A., Muller, W.,
Killeen, N., et al. (1995). A role for
CD5 in TCR-mediated signal trans-
duction and thymocyte selection.
Science 269, 535–537.
Tardivel, A., Tinel, A., Lens, S., Steiner,
Q. G., Sauberli, E., Wilson, A., et
al. (2004). The anti-apoptotic fac-
tor Bcl-2 can functionally substitute
for the B cell survival but not for the
marginal zone B cell differentiation
activity of BAFF. Eur. J. Immunol. 34,
509–518.
Tedder, T. F., Zhou, L. J., and Engel,
P. (1994). The CD19/CD21 signal
transduction complex of B lympho-
cytes. Immunol. Today 15, 437–442.
Thomas, M. L. (1995). Positive and
negative regulation of leukocyte
activation by protein tyrosine
phosphatases. Semin. Immunol. 7,
279–288.
Tung, J. W., Parks, D. R., Moore, W.
A., and Herzenberg, L. A. (2004).
Identification of B-cell subsets: an
exposition of 11-color (Hi-D) FACS
methods. Methods Mol. Biol. 271,
37–58.
Conflict of Interest Statement: The
authors declare that the research was
conducted in the absence of any com-
mercial or financial relationships that
could be construed as a potential con-
flict of interest.
Received: 29 September 2012; accepted:
21 November 2012; published online: 17
December 2012.
Citation: Sindhava VJ and Bon-
dada S (2012) Multiple regulatory
mechanisms control B-1 B cell acti-
vation. Front. Immun. 3:372. doi:
10.3389/fimmu.2012.00372
This article was submitted to Frontiers in
B Cell Biology, a specialty of Frontiers in
Immunology.
Copyright © 2012 Sindhava and Bon-
dada. This is an open-access article dis-
tributed under the terms of the Creative
Commons Attribution License, which
permits use, distribution and reproduc-
tion in other forums, provided the original
authors and source are credited and sub-
ject to any copyright notices concerning
any third-party graphics etc.
Frontiers in Immunology | B Cell Biology December 2012 | Volume 3 | Article 372 | 6
... Other mechanisms have also been implicated in controlling B-1 cell activation and antibody release. To produce antibodies, B-1 cells require simultaneous toll-like receptor (TLR) and BCR signaling (Sindhava et al., 2010;Sindhava and Bondada, 2012;Alhakeem et al., 2015). Only certain TLR signaling pathways collaborate with precise B-1-cell BCRs (Genestier et al., 2007;Savage et al., 2019). ...
... The spontaneous release of IL-10 by peritoneal B-1 cells is not shared by splenic B-2 cells and has thus been extensively studied. Unlike B-2 cells, separate TLR or BCR signaling promotes IL-10 release (Sindhava et al., 2010;Sindhava and Bondada, 2012;Alhakeem et al., 2015). IL-10 signaling prevents B-1 cell activation, blocks the cell cycle, and prevents IgM release (Alhakeem et al., 2015;O'Garra et al., 1990). ...
Article
Full-text available
Since their discovery nearly 40 years ago, B-1 cells have continued to challenge the boundaries between innate and adaptive immunity, as well as myeloid and lymphoid functions. This B-cell subset ensures early immunity in neonates before the development of conventional B (B-2) cells and respond to immune injuries throughout life. B-1 cells are multifaceted and serve as natural- and induced-antibody-producing cells, phagocytic cells, antigen-presenting cells, and anti-/pro-inflammatory cytokine-releasing cells. This review retraces the origin of B-1 cells and their different roles in homeostatic and infectious conditions before focusing on pollutants comprising contact-sensitivity-inducing chemicals, endocrine disruptors, aryl hydrocarbon receptor (AHR) ligands, and reactive particles.
... There are some supporting evidence about the involvement of B-1 cells in MS, as Torring et al. reported the elevated frequency of B1 cells in the remiting phase of MS condition [53]. It is reported that the frequency of B1 cells are elevated in autoimmune disorders in human and mouse model and genetic depletion of this cell population in mouse models led to ameliorate autoimmunity [17,54]. Due to the paucity of studies, drawing a precise conclusion about the disparity of B1 cell in neuro autoimmune diseases, particulary NMOSD needs furthur studies. ...
... CD5 marker of B1 cells is contributor to IL-10 production through a non-selective calcium ion channel named transient receptor potential channel 1. Upon this Ca 2+ dependent pathway, mitogenactivated protein kinase is activated, which leads to phosphorylation of extracellularly-regulated kinase-1 and -2 Erk1/2 and IL-10 procuction [58]. Moreover, IL-10 is highly engaged in inhibition of auto-reactive B1 cells via a negative feedback [54,59]. Various subsets of leukocytes are invovled in IL-10 production including Th1, Treg, TCD8 + , B cells, macrophages, dendritic cells, neutrophils and eosinophils [60]. ...
Article
Full-text available
Abstract Background Multiple sclerosis (MS) and neuromyelitis optica syndrome disease (NMOSD) are inflammatory diseases of the central nervous system. The pathogenesis and treatments for these two conditions are very different. Natural killer (NK) and natural killer T (NKT) cells are immune cells with an important role in shaping the immune response. B cells are involved in antigen presentation as well as antibody and cytokine production. There is conflicting evidence of the roles of NK, NKT, and B cells in the two conditions. We aimed to compare the frequency of CD3−CD16+CD56+NK, CD3+ CD56+ NKT, and CD5+CD19+ B cells in the peripheral blood and serum Interleukin-10 (IL-10) in patients with MS and NMOSD. Methods CD19+CD5+ B, CD3− CD16+CD56+ NK, and CD3+CD56+ NKT cells were quantitated by flow cytometry in 15 individuals with Interferon-Beta (IFN-β) treated relapsing–remitting MS (RRMS), 15 untreated RRMS, and 15 NMOSD patients as well as 30 healthy controls (HC). Serum IL-10 was measured using an enzyme-linked immunosorbent assay (ELISA). Results The percentage of CD3−CD56+CD16+ NK cells in the peripheral blood of IFN-treated MS (1.81 ± 0.87) was significantly lower than for untreated RRMS (4.74 ± 1.80), NMOSD (4.64 ± 1.26) and HC (5.83 ± 2.19) (p
... While NAPc2 treatment did not influence CD19 + /CD43 + B1 cells abundance (Fig. S3A, B), NAPc2 increased the proportion of peritoneal B1 cells expressing CD5, which is a negative regulator of B cell receptor signaling. 56 In contrast, NAPc2 did not alter B1 phenotypes in the bone marrow (Fig. S3C), indicating that NAPc2 primarily altered immune pathology in the periphery. ...
Article
Antiphospholipid antibodies (aPL) in primary or secondary antiphospholipid syndrome (APS) are a major cause for acquired thrombophilia, but specific interventions preventing autoimmune aPL development are an unmet clinical need. While autoimmune aPL cross-react with various coagulation regulatory proteins, lipid-reactive and COVID-19 patient-derived aPL recognize the endo-lysosomal phospholipid lysobisphosphatidic acid (LBPA) presented by the cell surface expressed endothelial protein C receptor (EPCR). This specific recognition leads to complement-mediated activation of tissue factor (TF) dependent proinflammatory signaling and thrombosis. Here we show that specific inhibition of the TF coagulation initiation complex with nematode anticoagulant protein c2 (NAPc2) prevents the prothrombotic effects of COVID-19 patient-derived aPL in mice and the aPL-induced proinflammatory and prothrombotic activation of monocytes. The induction of experimental APS is dependent on the NAPDH oxidase complex and NAPc2 suppresses monocyte endosomal reactive oxygen species production requiring the TF cytoplasmic domain and IFNa secretion from dendritic cells. Latent infection with murine cytomegalovirus virus causes TF cytoplasmic domain-dependent development of persistent aPL and circulating phospholipid-reactive B1 cells, which is prevented by short term intervention with NAPc2 during acute viral infection. In addition, treatment of lupus prone MRL-lpr mice with NAPc2, but not with heparin, suppresses dendritic cell activation in the spleen, aPL production and circulating phospholipid-reactive B1 cells and attenuates lupus pathology. These data demonstrate a convergent TF-dependent mechanism of aPL development in latent viral infection and autoimmune disease and provide initial evidence that specific targeting of the TF initiation complex has therapeutic benefits beyond currently used clinical anticoagulant strategies.
... 26 Many studies have analyzed the function of CD5+CD19+ B1 cells in hepatic ischemia and cerebral), 27 intestinal, and other systemic inflammatory disorders, which may open novel research topics. (5) Regarding nIgM, numerous investigations have concentrated on finding novel molecules secreted by B1 cells to reduce and control the inflammatory state. 28 According to some studies, mice with low levels of nIgM can produce IgG autoantibodies and are predisposed to developing severe MS. ...
Article
Full-text available
Chronic lymphocytic leukemia (CLL) is the clonal expansion of mature CD5+ B cells and the most common lymphoproliferative disease in adults (B1-CLL). B1 cells' anti-inflammatory effects include the production of natural IgM (nIgM) by the spleen and bone marrow, decreased inflammatory cytokines as a primary response to maintaining tissue homeostasis, and enhanced release of transforming growth factor β (TGFβ). We used the flow cytometry technique in peripheral blood from patients with CLL and multiple sclerosis (MS) to immunophenotype B cells and their subpopulations. Whole blood from CLL and MS patients, as well as healthy controls, was used to detect nIgM using the VH4-34 gene copy number and real-time RT-PCR. We found that the proportion of CD5+ B cells was significantly lower in MS patients than in the control group and that CD5+ B lymphocytes were significantly higher in CLL patients than in the control group. Compared to the control group, CLL patients had significantly higher levels of the VH4-34 gene copy number. On the contrary, MS patients had significantly lower VH4-34 gene copy number levels compared to the control group. As the number of antibodies in CLL patients increases due to the high number of B1 cells, we propose a new way to treat MS by extracting this natural antibody from the sera of CLL patients and injecting it into MS patients.
... 51 Therefore, butyrate regulates the capacity of B cells to produce IL-10 and induces pTregs. 52 These results demonstrate that microbially derived metabolites can control the balance between regulatory and mature B cell subsets. ...
Article
Full-text available
Short-chain fatty acids (SCFAs) play an important role in the host system. Among SCFAs, butyrate has received particular attention for its large effect on host immunity, particularly in supplying energy to enterocytes and producing immune cells. Butyrate enters the cells through the Solute Carrier Family 5 Member 8 (SLC5A8) transporters, then works as a histone deacetylase inhibitor (HDAC) that inhibits the activation of Nuclear factor-κB (NF-κB), which down-regulates the expression of IL-1β, IL-6, TNF-α. Meanwhile, butyrate acts as a ligand to activate G protein-coupled receptors GPR41, GPR43, and GPR109, promoting the expression of anti-inflammatory factors. Besides, it inhibits the proinflammatory factors. Further, it can also suppress the expression of chemokines and reduce inflammation to maintain host homeostasis. This paper reviews the research progress highlighting the potential function of butyrate as a factor impacting intestinal health, obesity and brain disorders.
Article
Full-text available
B cells have been long studied for their role and function in the humoral immune system. Apart from generating antibodies and an antibody-mediated memory response against pathogens, B cells are also capable of generating cell-mediated immunity. It has been demonstrated by several groups that B cells can activate antigen-specific CD4 and CD8 T cells, and can have regulatory and cytotoxic effects. The function of B cells as professional antigen presenting cells (APCs) to activate T cells has been largely understudied. This, however, requires attention as several recent reports have demonstrated the importance of B cells within the tumor microenvironment, and B cells are increasingly being evaluated as cellular therapies. Antigen presentation through B cells can be through antigen-specific (B cell receptor (BCR) dependent) or antigen non-specific (BCR independent) mechanisms and can be modulated by a variety of intrinsic and external factors. This review will discuss the pathways and mechanisms by which B cells present antigens, and how B cells differ from other professional APCs.
Article
Full-text available
The outbreak and persistence of coronavirus disease 2019 (COVID-19) threaten human health. B cells play a vital role in fighting the infections caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Despite many studies on the immune responses in COVID-19 patients, it is still unclear how B cell receptor (BCR) constituents, including immunoglobulin heavy (IGHs) and light chains (IGLs), respond to SARS-CoV-2 in patients with varying symptoms. In this study, we conducted complementarity-determining region 3 (CDR3) sequencing of BCR IGHs and IGLs from the peripheral blood of COVID-19 patients and healthy donors. The results showed significantly reduced clonal diversity, more expanded clones, and longer CDR3 lengths of IGH and IGL in COVID-19 patients than those in healthy individuals. The IGLs had a much higher percentage of VJ skew usage (47.83% IGLV and 42.86% IGLJ were significantly regulated) than the IGHs (12.09% IGHV and 0% IGHJ) between the healthy individuals and patients, which indicated the importance of BCR light chains. Furthermore, we found a largely expanded IGLV3-25 gene cluster mostly pairing with IGLJ1 and ILGJ2 in COVID-19 patients and a newly identified upregulated IGLJ1 gene and IGLJ2+IGLV13-21 recombination, both of which are potential sources of SARS-CoV-2-targeting antibodies. Our findings on specific immune B-cell signatures associated with COVID-19 have clinical implications for vaccine and biomarker development for disease diagnosis.
Chapter
B cells have a central and dual role in the physio-pathological mechanisms of periodontitis. They take part in the elimination of the periodontal germs, the induction of tissue destructions and the regulation of the immune response. B cells play an essential role in the destruction of alveolar bone in periodontitis by immunomodulation, rather than by production of antibodies. In the periodontal cell network, B cells are in constant interaction with other immune cells and mesenchymal cells. Periodontitis is characterized by a cellular conversion from a dominant T-cell lesion to a dominant B-cell lesion, particularly enriched in plasma cells. This evolution results from abnormal interactions between B and T cells in periodontitis. Moreover, B cells are at the crossroads of the immune and the bone systems and are involved in the autoimmune mechanisms described in periodontitis. Different subsets of B cells are involved in periodontal destruction, in particular memory B cells, plasma cells and B1 cells. Effector memory B cells strongly express mRANKL in periodontitis and constitute the precursors of plasma cells. B1 cells are also involved in tissue destruction but also in the mechanisms of regulation, in particular via the natural secretion of IL-10 by CD11b+ B1 cells which form a part of the B10 cells that regulate the inflammatory response. As such, periodontitis seems to be associated with a deficit in regulation. In peripheral blood, B cells can also be systemic markers of infection and periodontal inflammation: circulating memory B cells are increased in periodontitis while circulating CD11b+ B1 cells are decreased. The study of B cells in periodontitis is constantly evolving for a better knowledge of the periodontitis setting, the evaluation and the follow-up of periodontitis, but also for the design of new therapeutic targets that may be promising in the management of severe periodontitis.
Article
Atypical cytokine production and immune cell subset ratios, particularly those that include high proportions of macrophages, characterize tumor microenvironments (TMEs). TMEs can be modeled by culturing peritoneal cavity (PerC) cells which have a high macrophage to lymphocyte ratio. With TCR or BCR ligation, PerC lymphocyte proliferation is tempered by macrophages. However, PHA (T cells) and anti-CD40 (B cells) are activators that induce proliferation. Herein, we report that ligating IgD, in contrast to IgM, triggers PerC B cell proliferation. IL-4 addition enhanced the IgD response for BALB/c PerC B cells but suppressed that of C57BL/6 mice. Intriguingly, concurrent ligation of IgD and CD3ε rescued a PerC T cell proliferative response. These results serve to expand the list of targets for promoting cellular and humoral immunity in conditions that model macrophage-rich TMEs.
Chapter
Prostate apoptosis response-4 (Par-4) was identified as a tumor suppressor protein that is silenced by promoter methylation in various cancers and has been shown to induce apoptosis selectively in cancer cells but not in normal cells. Par-4 interacts with a variety of partners in cells to mediate various cellular responses and appears to have a pro-apoptotic role in non-hematological tumors. Here, we summarize the literature on the role of Par-4 in hematological cells that is in contrast to its classic pro-apoptotic role. Par-4 is expressed basally in various hematopoietic cells and malignancies at the mRNA and protein level, but is predominant in the early stages of B-cell maturation and specifically in chronic lymphocytic leukemia (CLL). CLL B cells express higher levels of Par-4 than normal B-cell subsets and constitutively active B-cell receptor signaling (BCR) maintains high Par-4 levels in these cells, suggesting a novel regulation of Par-4 through BCR signaling. CLL cell growth is dependent on BCR signaling-mediated Par-4 expression, which is in part due to downregulation of p21 by Par-4. Bcl2 and NF-κB pathways cause differential regulation of apoptotic genes in contrast to non-hematological cancers, and Par-4 may also play a significant role in tumor microenvironment. Thus, Par-4 appears to have unique roles in hematological malignancies.
Article
Full-text available
Recent data implicating loss of PTP1C tyrosine phosphatase activity in the genesis of the multiple hemopoietic cell defects found in systemic autoimmune/immunodeficient motheaten (me) and viable motheaten (mev) mice suggest that PTP1C plays an important role in modulating intracellular signaling events regulating cell activation and differentiation. To begin elucidating the role for this cytosolic phosphatase in lymphoid cell signal transduction, we have examined early signaling events and mitogenic responses induced by B cell antigen receptor (BCR) ligation in me and mev splenic B cells and in CD5+ CH12 lymphoma cells, which represent the lymphoid population amplified in motheaten mice. Despite their lack of functional PTP1C, me and mev B cells proliferated normally in response to LPS. However, compared with wild-type B cells, cells from the mutant mice were hyperresponsive to normally submitogenic concentrations of F(ab')2 anti-Ig antibody, and they exhibited reduced susceptibility to the inhibitory effects of Fc gamma IIRB cross-linking on BCR-induced proliferation. Additional studies of unstimulated CH12 and wild-type splenic B cells revealed the constitutive association of PTP1C with the resting BCR complex, as evidenced by coprecipitation of PTP1C protein and phosphatase activity with BCR components and the depletion of BCR-associated tyrosine phosphatase activity by anti-PTP1C antibodies. These results suggest a role for PTP1C in regulating the tyrosine phosphorylation state of the resting BCR complex components, a hypothesis supported by the observation that PTP1C specifically induces dephosphorylation of a 35-kD BCR-associated protein likely representing Ig-alpha. In contrast, whereas membrane Ig cross-linking was associated with an increase in the tyrosine phosphorylation of PTP1C and an approximately 140-kD coprecipitated protein, PTP1C was no longer detected in the BCR complex after receptor engagement, suggesting that PTP1C dissociates from the activated receptor complex. Together these results suggest a critical role for PTP1C in modulating BCR signaling capacity, and they indicate that the PTP1C influence on B cell signaling is likely to be realized in both resting and activated cells.
Article
Full-text available
A subset of B lymphocytes present primarily in the peritoneal and pleural cavities is defined by the expression of CD5 and is elevated in autoimmune diseases. Upon signaling through membrane immunoglobulin M (mIgM), splenic B lymphocytes (B-2) proliferate, whereas peritoneal B cells (B-1) undergo apoptosis. However, in CD5-deficient mice, B-1 cells responded to mIgM crosslinking by developing a resistance to apoptosis and entering the cell cycle. In wild-type B-1 cells, prevention of association between CD5 and mIgM rescued their growth response to mIgM crosslinking. Thus the B cell receptor-mediated signaling is negatively regulated by CD5 in normal B-1 cells.
Article
Full-text available
Immune regulation produced by B cells has been attributed to production and secretion of interleukin (IL)-10, which is a characteristic of mouse B1 cells. In view of the widespread clinical use of B-cell depletion therapies in autoimmune and malignant diseases, it is important to monitor the function and fate of regulatory B cells. However, there is no consensus regarding the phenotypic identity of human IL-10(+) B cells. Here we show that human CD11b(+) B1 cells, one of two recently described subpopulations of B1 cells, spontaneously produce IL-10 and suppress T-cell activation. In view of the capacity of these B cells to either stimulate T-cell proliferation or suppress T-cell activation, CD11b(+) B1 cells are considered to be capable of orchestrating elements of immune responsiveness and thus are termed "orchestrator B1 cells," or "B1orc," whereas CD11b(-) B1 cells that primarily secrete antibody are termed "secretor B1 cells," or "B1sec."
Article
CD19 and CD21 are B-cell surface molecules that associate with each other and with CD81 and Leu-13 to generate a signal transduction complex that is independent of the antigen receptor. Current studies, reviewed here by Thomas Tedder, Liang-Ji Zhou and Pablo Engel, indicate an important biological role for this protein complex in the regulation of B-cell development and activation.
Article
A small subpopulation of normal murine splenic B cells carrying all of the classic B cells markers (IgM, IgD, Ia, and ThB) also carries Ly-1, one of the major T cell surface molecules. This "Ly-1 B" subpopulation (identified and characterized by multiparameter FACS analyses) consists of relatively large, high IgM/low-IgD/low-Ly-1 lymphocytes that represent approximately 2% of the spleen cells in normal animals and, generally, 5-10% of spleen cells in NZB mice. Ly-1 B are clearly detectable in all normal mouse strains tested as well as NZB, CBA/N, other X-id mice and nude (nu/nu) mice. They are found primarily in the spleen; are either absent or very poorly represented in lymph node, bone marrow, and thymus; appear early during ontogeny, and comprise about a third of the small number of lymphocytes present in 5-d-old mice. NZB and (NZB x NZW)F1 mice have more Ly-1 B than all other strains and, furthermore, have a unique Ly-1 B population that secretes IgM when cultured under usual conditions in the absence of added antigen. The IgM secretion by these Ly-1 B cells accounts for the previously reported "spontaneous" IgM secretion by NZB spleen cells in culture. Studies with FACS-sorted cells show that the presence of Ly-1 on these IgM-secreting cells distinguishes them from the (Ly-1 negative) IgM-secreting "direct" plaque-forming cells generated in NZB mice after stimulation with sheep erythrocytes.
Article
BLyS and APRIL have similar but distinct biological roles, mediated through two known TNF receptor family members, TACI and BCMA. We show that mice treated with TACI-Ig and TACI-Ig transgenic mice have fewer transitional T2 and mature B cells and reduced levels of circulating immunoglobulin. TACI-Ig treatment inhibits both the production of collagen-specific Abs and the progression of disease in a mouse model of rheumatoid arthritis. In BLyS-deficient mice, B cell development is blocked at the transitional T1 stage such that virtually no mature B cells are present, while B-1 cell numbers are relatively normal. These findings further elucidate the roles of BLyS and APRIL in modulating B cell development and suggest that BLyS is required for the development of most but not all mature B cell populations found in the periphery.
Article
CD5, a membrane-associated glycoprotein, has been shown to negatively regulate antigen receptor-mediated growth responses in peritoneal B lymphocytes, thymocytes and mature T cells. The CD5-expressing peritoneal B cells (B-1) that are normally unresponsive to B cell receptor (BCR)-mediated growth signals mount a proliferative response to BCR cross-linking if the CD5 gene is deleted or if the CD5 molecule is sequestered away from the BCR. SHP-1, a cytosolic protein tyrosine phosphatase, has also been implicated in the negative regulation of antigen receptor-mediated signaling. The present study shows that SHP-1 is constitutively associated with the BCR in B-1 cells. This association is mediated in part by CD5, as it is reduced substantially after antigen receptor ligation in CD5– / – B-1 cells, and upon sequestration of CD5 from the antigen receptor complexes in wild-type B-1 cells. Prior cross-linking of CD5 also restores a normal calcium mobilization response as well as NF-κB activation in B-1 cells. These data support a model whereby CD5 negatively regulates antigen receptor-mediated growth signals by recruiting SHP-1 into the BCR complex in B-1 cells.
Article
The B-1 subset of B lymphocytes is maintained by self-renewal of mature cells, and this process may involve signaling through membrane immunoglobulin (mIg). We determined whether CD19, a membrane protein that co-stimulates B cells by mIg, has a role in this process. Pre-natal treatment of mice with 1D3, a rat anti-mouse CD19 monoclonal antibody, down-regulated CD19 expression and reduced by sixfold the number of B-1a cells at birth; B-2 cells were relatively unaffected. Prolonged treatment of adult mice with 1D3 caused the loss of approximately 2% per day of peritoneal B-1a cells, without diminishing the recovery of splenic B-2 cells. The loss of B-1a cells was associated with inhibition of their replication rather than with accelerated turnover. Therefore, CD19 is involved in the development and self-renewal of B-1a cells, perhaps through its ability to amplify signaling through mIgM.
Article
B1 cells are an important cell population for the production of natural antibodies and for antibacterial immunoglobulin responses. Here we identified the mouse protein Siglec-G as a B1 cell inhibitory receptor. Siglec-G was expressed in a B cell–restricted way, with large amounts present in B1 cells. When overexpressed, Siglec-G inhibited B cell receptor–mediated calcium signaling. Siglec-G-deficient mice had massive expansion of the B1a cell population, which began early in development and was B cell intrinsic. Siglec-G-deficient mice had higher titers of natural IgM antibodies but not a higher penetrance of IgG autoantibodies. Siglec-G-deficient B1 cells showed a strongly enhanced calcium signaling. Our results demonstrate that Siglec-G-dependent negative regulation exists in B1 cells, which may explain the naturally muted signaling response of B1 cells.
Article
The discovery of host-encoded gene products that sense molecular patterns in infectious microbes, and the demonstration of their role in triggering innate and adaptive immune responses, has been a key milestone in our understanding of immunology. Twenty-three years after Janeway first outlined the fundamental concepts of the 'pattern recognition' model, and 15 years since the identification of Toll-like receptors (TLRs) as pattern recognition receptors (PRRs), new insights continue to be revealed, and questions remain. For example, innate immune responses to microbes that are mediated by PRRs have historically been viewed as the domain of innate immune cell populations such as dendritic cells and macrophages. New evidence, however, has pointed to the role of B-cell-intrinsic TLR activation in shaping antibody responses. These studies have revealed that TLRs regulate a complex transcriptional network that controls multiple steps in the development of antigen-specific antibodies. This review covers these recent developments regarding the role of TLRs in B-cell gene expression and function in vitro and in vivo, and highlights the remaining challenges in the field, with particular emphasis on the role of TLRs in antibody responses to viral infection. A more complete understanding of how TLRs regulate antibody responses will lead to improved vaccine design.