Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Clinical and Experimental Immunology, 2022, 210, 201–216

https://doi.org/10.1093/cei/uxac104
Advance access publication 7 January 2023
Review

Review
Human B-cell subset identification and changes in
inflammatory diseases
Rebekah L. Velounias1 and Thomas J. Tull2,*,
1
Department of Immunobiology, King’s College London, Guy’s Hospital Campus, London, UK

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


2
St John’s Institute of Dermatology, King’s College London, Guy’s Hospital Campus, London, UK
Correspondence: Thomas Tull, St John’s Institute of Dermatology, King’s college London, London, UK. Email: thomas.tull@kcl.ac.uk
*

Summary
Our understanding of the B-cell subsets found in human blood and their functional significance has advanced greatly in the past decade. This has
been aided by the evolution of high dimensional phenotypic tools such as mass cytometry and single-cell RNA sequencing which have revealed
heterogeneity in populations that were previously considered homogenous. Despite this, there is still uncertainty and variation between studies
as to how B-cell subsets are identified and named. This review will focus on the most commonly encountered subsets of B cells in human blood
and will describe gating strategies for their identification by flow and mass cytometry. Important changes to population frequencies and function
in common inflammatory and autoimmune diseases will also be described.

Keywords: B cell, systemic lupus erythematosus, autoimmunity, immune checkpoints


Abbreviations: ABC: age associated B cell; aNAV: activated naïve; ASC: antibody secreting cell; BAFF: B cell activating factor; BCR: B cell receptor; Breg: B
regulatory cell; CSM: class switched memory; DN: double negative; IgV: immunoglobulin variable; HCD: healthy control donor; GALT: gut associated lymphoid
tissue; IFN: interferon; MFI: median fluorescence intensity; MS: multiple sclerosis; MTG: mitotracker; MZB: marginal zone B cell; MZP: marginal zone precursor;
RA: rheumatoid arthritis; RH: rhodamine; SLE: systemic lupus erythematosus; SPADE: spanning tree progression of density normalized events; SS: Sjogren’s
syndrome; SSc: systemic sclerosis; TLR: toll like receptor; TNF: tumour necrosis factor.

Introduction cytokines as well as antigen presentation [13–20]. Their


A unified and consistent approach to the identification and pathogenic role is supported by the response of a variety of
description of B-cell subsets is essential to ensure reproduci- inflammatory conditions to B-cell directed therapies such
bility of scientific studies and that analogous conclusions can as rituximab and B-cell activating factor (BAFF) antagon-
be drawn from them. Although many recent advances have ists [21–25]. Immune mediated inflammatory diseases such
been made in B-cell biology, significant areas of uncertainty as systemic lupus erythematosus (SLE), Sjogren’s syndrome
remain regarding the nature of certain B-cell subsets and their (SS), rheumatoid arthritis (RA), and COVID-19 are associ-
role in homeostasis and inflammatory disease. This is likely to ated with significant changes in B-cell subset frequency which
have arisen due to a number of reasons. Firstly, whilst murine is likely driven by inflammatory cytokines and pro-survival
models are a valuable tool to understand B-cell biology, there factors as well as changes in B-cell differentiation and homing
are significant differences between murine and human B-cell [26–28].
subsets which mean that it can be difficult to draw analogous In this review we will describe the main subsets of B cells in
conclusions from them [1–4]. Secondly, the evolution of high peripheral blood and how these can be identified using flow
dimensional phenotypic techniques has identified many novel and mass cytometry, as well as important changes in B-cell
subsets but further work is needed to expand on their func- subsets in inflammatory immune mediated conditions. B1
tional and pathogenic roles [5–8]. Thirdly, B cells in blood cells will not be discussed in this review due to uncertainties
often represent a developmental continuum and overlap regarding their status and because they represent a rare subset
phenotypically without sharply demarcated gates to differen- in adult human blood [29–31].
tiate them [5, 9–11]. And finally, there remain significant gaps
in our understanding of human B-cell maturation and there-
fore the developmental trajectories that link human B-cell Identification of B cells in peripheral blood
subsets to one another [12]. The surface expression of both CD19 and CD20 are induced
B cells are known to play important roles in a variety of early in B-cell development in the bone marrow [11, 32].
inflammatory diseases via the production of antibodies and They are both expressed on B-cell subsets in blood but CD20

Received 4 August 2022; Revised 19 October 2022; Accepted for publication 15 November 2022
© The Author(s) 2023. Published by Oxford University Press on behalf of the British Society for Immunology. All rights reserved. For permissions, please
e-mail: journals.permissions@oup.com
202 Velounias and Tull

expression is lost during terminal B-cell differentiation into CD24, CD38, and CD10 are downregulated and these are
antibody secreting cells (ASCs) [33, 34]. CD19 therefore al- commonly utilized markers to distinguish transitional B-cell
lows identification of both B cells and ASCs. A gating strategy subsets. T1 and T2 B cells share expression of CD10 with im-
using CD19 for the identification of both B cells and ASCs mature B cells in the bone marrow, but this is not expressed
in blood by flow cytometry is illustrated in Fig. 1A. An anti- by T3 cells [37, 45]. T1 and T2 cells are often distinguished
body panel of 9 ‘key’ markers can be used to identify the main from one another based on high and intermediate expression
B-cell subsets in blood and these are summarized in Table 1. of CD24 and CD38 (Fig. 2A). This approach is subjective as
Additional markers are also given in this table which can aid the definition of high and intermediate expression can differ
gating and the identification of further subsets. between individuals and experiments given technical factors
Mass cytometry utilizes the ionic signal from antibodies such as fluorescent signal intensity. T1 cells are known to have
tagged to rare earth metals and therefore is not limited by the low expression of CD21 and the use of this marker in con-
spectral overlap from fluorochrome-conjugated antibodies junction with CD24 and CD38 can be useful to guide the T1
that are used for flow cytometry [80]. Mass cytometry there- gate (Fig. 2A) [35].
fore allows single cells to be labelled with ~40 metal tagged The use of additional markers can also help to accurately
antibodies that can be directed to cell surface or intracel- identify T1, T2, and T3 subsets and are represented in Fig. 2B

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


lular targets. A gating strategy to identify live single B cells and C as SPADE trees. T1 cells have relatively high expression
by mass cytometry is displayed in Fig. 1B. Further identi- of β1 integrin, which is likely to be important for interaction
fication of B-cell subsets can then be performed by biaxial with bone marrow stromal cells [85]. T1 cells also lack ex-
gating or dimensional reduction and clustering methods. pression of CD62L and CCR7 (Fig. 2C), which is likely to
Biaxial gating of mass cytometry data can be challenging as result in a lack of homing capability as seen in studies of both
the expression of B-cell subset markers often lack distinct murine and human transitional B cells [9, 86, 87]. IgM was
pinch point gates and metal minus one control samples that traditionally thought to be downregulated through T1–T3 de-
guide the accurate placement of gates are impractical due to velopment but T2 and T3 populations are both heterogeneous
cost. Dimensionality reduction and clustering techniques that in their expression of this marker (Fig. 2D). IgMhi T2 cells
identify subsets based on the expression of multiple markers have high expression of β7 integrin and are enriched in gut
can therefore be advantageous over biaxial gating. ViSNE, a associated lymphoid tissue (Fig. 2E) [9]. Furthermore, IgMhi
visualization tool for high-dimensional single-cell data based T2 cells align with cells undergoing marginal zone B (MZB)
on the t-Distributed Stochastic Neighbour Embedding algo- cell development in terms of surface and transcriptomic fea-
rithm and uniform manifold approximation and projection tures [9]. This suggests that a gut homing T2 MZP population
(UMAP) are common dimensionality reduction tools used to exists in humans as has been demonstrated in mice [88–90].
visualize mass cytometry data [81, 82]. These algorithms can T3 cells are a late transitional B-cell subset and are thought
be run using all panel markers or select B-cell subset markers to represent an intermediate stage of development between
and allow the two-dimensional projection of B cells arranged T2 and naïve B cells. In keeping with this, T3 cells appear in
in multidimensional space. Representative viSNE plots of the blood after T2 cells during B-cell repopulation post B-cell
CD19+ B cells from a healthy control donor (HCD) gener- ablation therapy and differentiate into naïve B cells in vitro
ated by running the algorithm on lineage markers are dis- [44, 45]. Transitional B cells can be distinguished from naïve
played in Fig. 1C. In these viSNE plots, memory B cells appear B cells as they lack expression of the ABCB1 cotransporter,
as distinct islands but significant continuity exists between which as a result prevents the extrusion of dyes such as rhoda-
CD10+CD24hiCD38hi transitional and CD10−CD24intCD38int mine (RH) or mitotracker (MTG) [43]. T3 cells can therefore
naïve B-cell subsets and manually gating such plots can be gated as CD27−IgD+CD10−RHhi/MTGhi (Fig. 3A and B).
therefore be challenging (Fig. 1C). For this reason, span- The distinction between RH low and high populations can
ning tree progression of density normalized events (SPADE) be guided by the signal obtained from CD27+ memory B cells
run on ViSNE coordinates can be a useful tool to separate which also lack to ABCB1 cotransporter and therefore are
subpopulations existing in phenotypic continuity (Fig. 1D) RHhi (Fig. 3C). The expression of CD45RB in T3 cell popu-
[9, 10]. With SPADE, B-cell populations that are phenotyp- lations is heterogeneous (Fig. 3D) and CD45RBhi cells also
ically similar to one another are displayed as nodes which have higher expression of IgM and CD1c and align with cells
are interlinked by branches to form trees. Nodes representing undergoing MZB cell differentiation (Fig. 3E) [9, 46]. The
B-cell subsets can then be manually grouped together to form use of dye extrusion is essential for the distinction of T3 cells
bubbles representing that population (Fig. 1D). Whilst this from naïve B cells as they otherwise share an overlapping sur-
approach is in part directed it has the advantage over biaxial face phenotype (Fig. 3F).
gating in that multiple markers can be used simultaneously to Multiple abnormalities in transitional B-cell subsets have
define B-cell subsets. been reported in inflammatory diseases. Transitional B cells
harbour a high proportion of autoreactive cells which are re-
moved through sequential maturation into naïve B cells in
Transitional B cells healthy individuals and this checkpoint is defective in SLE
Transitional B cells are defined as recent emigrant B cells from [91]. The mechanisms that underlie this checkpoint failure
the bone marrow and represent the most immature B-cell are not known but are may be secondary to aberrant tran-
subset in peripheral blood [12]. Studies of B-cell repopulation sitional B-cell maturation or migration into lymphoid tissue.
following bone marrow stem cell transplantation and B-cell Transitional B cells from patients with SLE have been reported
depletion with rituximab have identified sequential matur- to have lower expression of the gut homing receptor β7 in-
ation of transitional B cells through T1, T2, and T3 phases tegrin suggestive of defective gut homing capacity and β7hi
[35, 36, 45, 83, 84]. This maturational sequence is associated T2 cells are depleted in lupus nephritis [9, 39]. Transitional
with the acquisition and loss of various markers. Notably, B cells have been shown to be increased in frequency in SLE
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 203

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023

Figure 1: identification of B cells in peripheral blood. (A) Flow cytometry plots showing a gating strategy used to identify CD19+ B cells and
CD19+CD27hiCD38hi ASCs. Data are from PBMCs from a representative healthy control donor (HCD). (B) A gating strategy of mass cytometry data
to identify live CD19+ B cells. Antibodies to CD3 and CD14 were conjugated to the same metal and therefore created a ‘dump’ channel which allows
exclusion of T cells and cells of myeloid origin. Data are from a female representative healthy control donor. (C) ViSNE plots generated using the
following markers; CD10, CD24, CD38, CD27, CD45RB IgD, IgM, IgA, and IgG. The plots are from a female HCD and depict expression of lineage
markers used to identify B-cell subsets. (D) SPADE on viSNE plots demonstrating expression of B-cell lineage markers used to classify CD19+ B cells
into transitional (TS), naïve, activated naive (aNAV), marginal zone B cell (MZB), and their precursors (MZP) and IgG+ and IgA+ memory subsets. Data
adapted from Tull TJ et al [9]
204 Velounias and Tull

Table 1: Lineage markers to define B-cell subsets with expected frequencies in healthy peripheral blood and studies reporting altered frequency or
function in autoimmune rheumatic diseases and COVID-19

Subset Key markers Additional markers and subsets Frequency Disease associations
(% CD19+)

T1 CD27−IgD+IgM+CD10+ β1hiβ7loCCR7loCD62Llo [9, 37] 1−4 [37, 38] SLE [38, 39]
CD21loCD24+++CD38+++ [35–37] SSc [40]
COVID [41, 42]
T2 CD27−IgD+IgM+CD10+ β1hi/intCD62Lhi/int 2−10 [28, 38] SLE [9, 28, 38, 39]
CD21hiCD24++CD38++ [35–37] IgMhiT2: IgMhiCCR7hiβ7hi [9] SSc [40]
IgMloT2: IgMloCCR7loβ7lo [9] COVID [41, 42]
T3 CD27−IgD+IgM+CD10−CD24+ IgMhiT3: IgMhiCD1chiβ7hiCD45RBint/hi [9, 46] 9−13 [9, 45] SLE [9, 44]
CD38+RHhi/MTGhi [43–45] IgMloT3: IgMloCD1cloβ7loCD45RBlo/int [9]
Naive CD27−IgD+IgM+CD10−CD24+ 40−60 [9, 45] SLE [47, 48]

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


CD38+RHlo/MTGlo [43, 44] SSc [49]
Naïve MZP CD27−IgD+IgMhiCD10 CD1chi [9, 46] 0.5−3 [9, 46] SLE [9]

CD24+CD38+RHlo/
MTGloCD45RBhi [46, 50]
Activated CD27−IgD+CD10−CD21lo FRCL5+, CXCR5+, T-bet+ [26] 0.1−3 [9] SLE [26, 27, 51]
naive CD24loCD38lo [26, 27] COVID [42]
MZB CD27+IgD+IgM+ [10] CD1chi [52] 2−30 [53, 54] SLE [54, 55]
MZB1: β7hiCCR7hi [6] SS [56, 57]
MZB2: β7loCCR7lo [6] SSc [58]
RA [59]
COVID [42, 60]
IgM only CD27+IgD−IgM+ [61, 62] CD45RB+ [5] 10 [63]
CSM CD27+IgD−IgM− [5, 61] CD45RB+, IgG, IgA or IgE+ [64] 5−40 [26, 53] SLE [54, 65]
SS [66]
RA [67, 68]
ANCA vasc [69]
DN CD27−IgD− [70, 71] DN1: CD21int/hi, CXCR5+ [26, 41] 1−10 [26, 71] SLE [26, 27, 74]
DN2: CD21lo, FRCL5+, CXCR5−, T-bet+, RA [75–77]
CD11chi [26, 41] SSc [49]
DN3: CD21−CD11c−CXCR5− [72] COVID [41, 42, 78, 79]
DN4: CD21+CD11c+ [73]
ANCA vasc = ANCA-associated vasculitis; B1 = B1 integrin; B7 = B7 integrin; CSM = class switched memory; DN = double negative; MZP = marginal zone
precursor; SLE = systemic lupus erythematosus; SS = Sjorgen’s syndrome; SSc = Systemic sclerosis;.

and this has been correlated with serum BAFF levels [28, capacity to inhibit CD4 T-cell cytokine production [100].
38, 92]. The important role of BAFF as a survival factor is Induction of Bregs by plasmacytoid dendritic cells has also
also supported by the increased frequency of transitional B been reported to be defective in SLE which is thought to be
cells in patients with multiple sclerosis that are treated with mediated by type I interferon [102]. Breg dysfunction in SLE
fingolimod and interferon-β which are both known to in- may also be mediated by defective B cell homing or matur-
crease serum BAFF levels [93]. Transitional B cells in SLE ation in GALT, which is known to be important for Breg
have also been shown to be hyperresponsive to B-cell receptor induction [9, 39, 103]. Reduced numbers of CD24hiCD38hi
(BCR) signalling which is likely secondary to type I interferon B cells have been identified in patients with RA and they
[94, 95]. Increased frequencies of transitional B cells have also failed to repress Th17 responses but maintained their ability
been reported in a number of other autoimmune diseases such to suppress Th1 responses [104]. Furthermore, treatment of
as juvenile dermatomyositis, primary SS (pSS), and systemic RA with tumour necrosis factor alpha (TNF-α) antagonists
sclerosis (SSc) [40, 96, 97]. Altered T1:T2 cell ratios have also has been demonstrated to increase the number of CD27+
been identified in patients with renal transplant rejection and Bregs [105]. Primary SS has been associated with increased
are postulated to be an important predictor of graft loss [98, frequency of CD24hiCD38hi B cells but these cells fail to sup-
99]. Finally, a reduction in transitional B cells has been seen in press T cell TNF-α or IFN-γ secretion [97]. Furthermore,
COVID-19 infection which correlates with infection severity adoptive transfer of CD24hiCD38hi B cells from patients with
[41, 42]. pSS into mice with experimental SS demonstrated failure of
Transitional B cells are enriched in B regulatory cells suppression of T follicular cell expansion [106]. Reduced
(Bregs) that are best described as B cells that can suppress Breg frequency or defective Breg function has also been re-
T-cell responses mainly via the production of IL-10 [100, ported in MS, SSc, pemphigus, and myasthenia gravis [107–
101]. Breg dysfunction has been identified in a number of 111]. Breg dysfunction has therefore been identified in a
autoimmune diseases. CD24hiCD38hi B cells in SLE have less number of autoimmune diseases but further work is required
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 205

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023

Figure 2: transitional B-cell subsets and phenotype. (A) Flow cytometry plots showing a gating strategy used for identification of T1 cells (CD27−IgD+
CD10+CD24+++CD38+++) and T2 cells (CD27−IgD+CD10+CD24++CD38++). Data are from PBMCs from a representative HCD. (B) Schema highlighting the
transitional B-cell population from the SPADE on viSNE plot in Fig. 1D. (C) SPADE on viSNE plots of TS B cells exported from TS SPADE bubble from
Fig 1D demonstrating marker expression and classification of transitional B-cell populations. T1 cells were defined as CD10+CD24+++CD38+++CD21lo, T2
as CD10+CD24++CD38++CD21hi, and T3 as CD10 − CD24+CD38+CD21hi. Data are from a representative female HCD. (D) SPADE on viSNE plot displaying
IgM expression. IgMhi T2 and T3 populations are shown by dotted red and light blue lines, respectively. IgMlo T2 and T2 populations are shown by dotted
green and purple lines, respectively. (E) Flow cytometry plot showing gating strategy of T2 B cells into T2 IgMhi and T2 IgMlo subpopulations and β7
integrin median fluorescence intensity (MFI) in these populations. Data are from six HCD
206 Velounias and Tull

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


Figure 3: T3 and naïve B-cell subsets. (A) Flow cytometry plots showing a gating strategy used to identify CD27−IgD+CD10− B cells. (B) Flow cytometry
plots demonstrating gating strategies to identify naïve (CD27−IgD+CD10−RH123lo) and T3 (CD27−IgD+CD10−RH123hi) populations. (C) Flow cytometry plot
demonstrating the placement of the RH123hi gate from the signal from CD27+ memory B cells. Data are from a representative HCD. (D) Flow cytometry
plot depicting a gating strategy used to identify variants of T3 cells based on CD45RB. (E) Comparative histograms taken from flow cytometry data
displaying IgM and CD1c MFI in T3 CD45RBlo T3 CD45RBhi populations. (F) Comparative histograms taken from flow cytometry data displaying MFI of
CD24, CD21, CD38, CD5, IgM, and IgD between T3 and naïve B cells. (G) Comparative histograms taken from flow cytometry data displaying MFI of
IgM and CD1c between MZP and naive B cells. (H) Gating strategy of flow cytometry data to identify T3 and aNAV cells based on their expression of
CD21, CD24, and CD38

to identify the pathogenic significance and the mechanisms the distinction between T3 and naïve B cell, to our knowledge
that underlie this. this is not currently commercially available.
Naïve B cells are a heterogeneous population and a number
of naïve subsets have been identified including marginal zone
Naïve B cells precursor cells (MZP), activated naïve (aNAV), and an IgMlo
Naïve B cells represent B cells that have not encountered anergic subset [26, 46, 47]. Naïve MZP cells are characterized
antigen and have not undergone germinal centre matur- by high expression of CD45RB and also highly express IgM
ation. They therefore lack CD27 expression and have a low and CD1c have been shown to undergo differentiation into
level of somatic mutations within their immunoglobulin MZB like B cells following NOTCH ligation (Fig. 3G) [46].
variable (IgV) genes [63]. Naïve B cells represent 40–60% A T3 population with a similar surface phenotype has also
of peripheral blood B cells and as discussed above can only been identified and is thought to be in developmental con-
be distinguished from T3 cells by their ability to extrude tinuity with naïve MZP cells [9, 30]. Naïve B cells with high-
RH or MTG dyes due to their expression of the ABCB1 surface expression of CD45RB has been described as an early
cotransporter [43]. Naïve B cells can therefore be gated as memory population and found to have more somatic muta-
CD27−IgD+CD10−RHhiCD45RBlo (Fig. 3A). Distinction by tions than CD45RB− naïve B cells [5]. The same study com-
dye extrusion is not possible using mass cytometry although mented on an anergic profile of naïve B cells as evidenced by
a metal tagged antibody to ABCB1 would theoretically allow lower expression of intracellular transport proteins suggesting
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 207

that this subset would be less responsive to stimulation. Other human transitional and naïve MZP have an IgMhi gut homing
functional studies have, however, demonstrated robust prolif- phenotype suggestive of a role of GALT in their induction [9].
erative responses of naïve B cells to IgM crosslinking as well Furthermore, MZB are observed to occupy a peri-germinal
as CD40 and TLR9 agonists [37, 44, 45, 112]. centre niche and to diversify their BCR within GALT [10].
Naïve B cells with low IgM expression have been reported Functionally, MZB undergo T cell independent responses and
as having anergic properties as evidence by lower intracel- are important for immunity against encapsulated bacteria
lular calcium influxes and expression of activation markers [52, 129].
following IgM crosslinking [47]. This subset was also found Uncertainty regarding the developmental origin and func-
to express higher levels of the inhibitory receptor CD22. The tional status of MZB cells is reflected by the plethora of
identity of anergic B cells as IgMloIgD+ is supported by a names assigned to this subset in the literature. These include
number of murine studies using the hen egg lysozyme model unswitched memory [56, 130, 131], pre-switched memory
in which the majority of B cells recognize this epitope. When [58, 119, 132], non-switched [133, 134], natural effector
HEL is expression was induced endogenously the majority of [64, 135], and IgM memory [136, 137] B cells. MZB have a
B cells adopted this phenotype, suggesting anergy can be in- CD27+IgD+IgM+ surface phenotype and have high expression
duced, and is a key tolerance mechanism [113–115]. of CD1c, CD21, and CD45RB (Fig. 4A) [63]. MZB have also

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


ANAV are a subset of CD27−IgD+ naïve cells with low ex- been reported to have low levels of CD23 and high expres-
pression of CD21, CD23, CD24, and CD38 but high expres- sion of the inhibitory receptor CD32b [138, 139]. Recently,
sion of FCRL5, CD11c, and T-bet [26, 27] (Fig. 3H). This two distinct populations of MZB have been described that
subset also does not express the ABCB1 cotransporter and is differ in cell surface properties, transcriptional factors, and
therefore RH or MTG high following dye extrusion assays distribution in lymphoid tissues [6]. These have been termed
[116]. ANAV were first identified in SLE as being clonally re- MZB1 and MZB2, MZB1 having higher expression of β7 in-
lated to ASC populations and are hyperresponsive to TLR7 tegrin, CCR7, CD24, and CD27 than MZB2 (Fig. 4B and C).
stimulation [26, 27]. The aNAV subset is therefore thought to MZB1 and MZB2 were not found to be clonally related and
undergo extrafollicular maturation into ASC in SLE and are NOTCH induced genes were only upregulated in MZB2 sug-
primed by interferon and TLR7 agonists. gesting independent maturational pathways.
Inflammatory disorders in which extrafollicular B-cell Significant changes in MZB frequency have been reported
maturation is thought to occur include severe SLE and in association with inflammatory diseases. As seen with MZP
COVID-19 and are associated with an increase in aNAV fre- subsets, severe depletion of MZB is observed in severe SLE
quency alongside increased frequencies of ASCs [26, 41, 51]. and this predominantly affects the MZB1 subset (Fig. 4D)
Conversely, MZP and naïve B cells are reduced in severe SLE [6, 9]. As well as SLE, reduced frequencies of MZB have also
[9]. Furthermore, inhibition of BAFF with belimumab results been reported in SS, SSc, and COVID-19 [9, 55, 56, 58–60,
in a lower frequency of naïve B cells, suggesting the import- 135]. The factors that result in this depletion in blood are
ance of BAFF for their survival [117, 118]. Of note, numerous not understood and could represent reduced cell survival or
studies of B-cell subset abnormalities in inflammatory disease genesis, altered tissue distribution or increased differentiation
have gated naïve B cells as solely CD27− or CD27−IgD+ and into ASCs. The depletion of MZB alongside their putative
therefore included double negative (DN) and transitional B precursors in severe SLE suggests defective MZB differenti-
cells, respectively in these gates [119–122]. ation. The reciprocal reduction of MZB cells and the increase
of ASCs in SLE could also support their differentiation into
MZB cells short-lived ASCs that are characteristic of SLE disease flares
The splenic marginal zone refers to a microanatomical area [27]. Also, as MZB depletion in COVID and SLE has been
on the periphery of B-cell follicles in the white pulp [123]. shown to be reversible following resolution of the infection
MZB cells are termed as such as they share similar surface and with treatment respectively, inflammatory factors can re-
phenotype to the subset of cells found in the splenic marginal sult in significant plasticity in this population [55, 135]. The
zone that express low levels of IgD and high levels of CD1c functional significance of depletion in MZB is unknown but
[52, 124]. In mice, MZB cells are confined to the spleen and could underly the increased susceptibility of patients to in-
do not have IgV gene mutations and therefore arise independ- fection with encapsulated bacteria or defective B regulatory
ently of germinal centre responses. In humans, MZB cells responses [140, 141].
circulate in blood and lymphoid tissue and have IgV gene mu-
tations, although to a lower degree when compared to class- CD27+ memory and IgM only B cells
switched memory (CSM) B cells [10, 52, 63, 125]. The finding Memory B cells are formed from mature naïve B cells that have
that MZB have IgV mutations in patients with CD40L defi- encountered antigen and undergone T dependent immune re-
ciency who lack germinal centres suggest they may mature in- sponses and germinal centre maturation [2, 142]. They can be
dependent of germinal centre responses as in mice [64, 125]. distinguished from naïve B cells by a much higher degree of
This is supported by studies that demonstrate little overlap IgV gene mutations, the gain of expression of CD27, and the
between MZB and CSM B-cell clones, suggesting that MZB loss of IgD expression [1, 63, 128, 143, 144]. Rounds of pro-
represent a distinct entity rather than just a precursor to CSM liferation and diversification within germinal centres create
[10, 61]. Germinal centre independent maturation of MZB is, clones of memory B cells that produce high-affinity immuno-
however, challenged by the finding that 20% of blood MZB globulin and differentiate into long-lived ASCs [145–147].
carry BCL6 gene mutations and that human MZB have been Functionally, memory B cells proliferate rapidly in response
reported to share clones with GC delivered IgG memory B to stimulation with CD40L and interleukin (IL) 2 and IL21
cells [126–128]. Similarly, whilst a specific MZB maturational [148, 149].
pathway exists in mice, the developmental origins of human Distinct subsets of CD27+IgD− memory B cells can be
MZB remain incompletely understood. As described above, identified by the expression of IgM, IgA, or IgG (Fig. 5A).
208 Velounias and Tull

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


Figure 4: human MZB cells. (A) Flow cytometry plot showing a gating strategy used to identify MZB cells as CD27+IgD+ and histograms showing MFI
of markers that demonstrate that MZB cells are IgM+CD1c+CD45RB+CD21+. (B) Flow cytometry plots demonstrating the identification of two marginal
zone subsets, which can be distinguished based on expression of CCR7 and β7 integrin. MZB1 are CD27+IgD+IgM+CD1c+CCR7+β7hi and MZB2 are
CD27+IgD+IgM+CD1c+CCR7-β7lo. (C) SPADE on viSNE plot showing events exported from the MZB SPADE bubble in Fig. 1D. Higher expression of
CCR7 and β7 integrin is seen in MZB1 and low expression in MZB2. (D) Comparative flow cytometry plots showing the CD19+ compartment in a
HCD and a patient with SLE. Plots show reduction in CD27+IgD+ MZB cells in SLE compared to HCD [9]. (E) Quantification of flow cytometry data
demonstrating a significant reduction in CD27+IgD+ cells in SLE compared to HCD [9]

CD27+ memory cells also share high expression of CD24 are not well understood in humans and other than IgM ex-
and CD45RB and relatively low expression of CD38 (Fig. pression they share a similar surface phenotype (Fig. 5D)
5B). CD27 and CD38 are also important in distinguishing [150]. Of interest is a recent study of BK polyoma virus re-
memory B cells from ASCs (Fig. 5C). IgM only B cells are sponses demonstrated different antiviral specificities of IgM
CD27+IgD−IgM+ and have been identified to have a higher and IgG memory B cells which suggests a distinct function
frequency of IgV gene mutations than MZB cells and to share of IgM memory B cells [151]. CSM B cells consist of IgA+,
clones with CSM B cells, suggesting that these cells are GC de- IgG+, and IgE+ subsets although the latter is a rare subset in
rived [10, 61]. The functional differences of human IgM only peripheral blood. CSM have a high frequency of IgV gene mu-
and CSM B cells in terms of recall responses and longevity tations and differentiate into ASCs that produce high-affinity
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 209

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023

Figure 5: CD27+ memory B cells and IgM only B cells. (A) Flow cytometry plots showing a gating strategy used to identify memory (CD27+IgD−IgM−
and IgG+/IgA+) and IgM only (CD27+IgD−IgM+) B cells. Data are from a representative healthy control donor. (B) Comparative histogram plots from flow
cytometry data showing differences in expression of CD45RB, CD27, and CD38 in CD27+IgD− cells compared to naïve (CD27−IgD+) cells. (C) Histogram
plots from flow cytometry data showing lower expression of CD38 and CD27 and higher CD24 in CSM compared to ASCs. (D) Heatmap from mass
cytometry data from Fig. 1D showing median expression of lineage markers on naïve, CSM and IgM only B cells

immunoglobulin that are essential for humoral immunity and CD95 or Fas is a pro-apoptotic receptor which is mainly ex-
the basis of effective vaccine responses [127]. Heterogeneity pressed by CSM, ASCs, and CD27− memory B cells which is
within memory B-cell populations has been identified through elevated in SLE and RA and thought to indicate an activated
differential expression of CD73 and CD95. Memory B cells memory B-cell phenotype [5, 152–154].
can be CD73 positive or negative, its expression is inversely Memory B cells play a critical role in autoimmune, inflam-
correlated with IgM and is mainly seen in CSM B cells [5]. matory and allergic diseases as they can differentiate into
210 Velounias and Tull

ASCs that produce pathogenic autoantibodies. Autoreactive with lupus nephritis in whom they have been identified within
memory B-cell clones arise due to checkpoint failure, where the nephrotic kidney [26, 163]. Extrafollicular B-cell matur-
autoreactive naïve B cells are allowed to undergo germinal ation has been correlated with the severity of COVID-19 and
centre or extrafollicular responses. This checkpoint has been similar to severe SLE, increased frequencies of aNAV, DN2
shown to be defective in a variety of diseases including SLE cells are observed alongside depletion of MZB cells [9, 41,
and SS, although the mechanisms that result in this are still 60, 78, 116]. The correlation between the severity of infec-
unclear [57, 155]. B cells in SLE are known to express greater tion and B-cell subset abnormalities suggests a pivotal role of
levels of CD40 and blockade of germinal centre responses with cytokines such as Type II interferon. CD11c positive B cells
CD40L (CD154) antagonists has therapeutic promise [156– have been reported in patients with MS, RA, and SSc and
158]. Variable frequencies of CSM B cells have been observed to accumulate with age in female patients with RA [164].
in SLE which is likely to represent heterogeneity in disease ac- Autoreactive CD27−CD19hiCD21lo B cells akin to DN2 cells
tivity, duration, and treatment [54, 65, 66]. Interestingly, un- have also been reported in SS [165]. Increased frequency of
like naïve and MZB subsets, CSM frequency is not affected by DN B cells has also been correlated with improved clinical
BAFF inhibition with belimumab [118]. Variable frequencies responses to rituximab of patients with RA whilst reduced
of CSM B cells have also been observed in RA with a lower numbers have been reported to predict response to IL-6 in-

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


proportion of CXCR3+CD27+ cells which correlated inversely hibition with tocilizumab [77, 166]. Further work is needed
with disease activity [67, 68]. Increased frequency of switched to investigate the clinical significance of these findings. DN
memory B cells have also been observed in other autoimmune cells may be of pathogenic significance through the produc-
conditions including ANCA vasculitis [69]. B-cell depleting tion of inflammatory cytokines or differentiation into ASCs.
agents such as rituximab have long-lasting effects of memory Alternatively, altered DN cell frequency may be indicative of
B cells, which may underlie their efficacy in treating auto- certain inflammatory environments that affect treatment out-
immune disease [159]. comes.

DN memory B cells Conclusion


CD27− memory B cells were first identified as IgG+ cells in The evolution of high dimensional phenotypic tools has
blood with a lower level of somatic IgV gene mutations than greatly advanced our understanding of B-cell subsets in blood
CSM [70]. DN B cells expressing IgM, IgA, and IgA were then and the surface markers and transcriptomic properties that
identified and found to be expanded in SLE [71]. They are define them. However, significant gaps remain in our under-
termed DN B cells as they lack both CD27 and IgD expres- standing of the functional roles of these subsets, their develop-
sion. Their accumulation in chronic infections, autoimmune mental origins and pathogenic roles in disease. By addressing
diseases and with advanced age has also led to them being these areas of uncertainty, we will advance our knowledge
termed exhausted memory B cells and age associated B cells of human B-cell development and the role these cells play in
(ABC) [160–162]. ABC share features of DN cells such as normal homeostasis and in the pathogenesis of autoimmune
upregulation of T-bet and low expression of CD21 but have and inflammatory disease.
been described using a variety of markers and likely repre-
sent a heterogeneous population which are not entirely syn-
Materials and methods for data used in
onymous with DN cells [34, 162].
figures and figure generation
Since their first description a number of DN B-cell subsets
have been identified. Initially DN B cells were split into DN1 Experimental subjects
(CD21+CXCR5+) and DN2 B cells (CD21−CXCR5−) [26]. Blood samples were donated by adult HCD and patients with
Similar to aNAV, DN2 also have low expression of CD24 SLE with regional ethics committee (REC) approval and in-
(Fig. 6A). DN2 cells were found to be a rare subset in health formed consent (REC reference 11/LO/1433).
but are found in much higher frequencies in SLE [26]. DN2
B cells also highly express CD11c as well as the transcription Isolation and storage of PBMCs
factor T-bet but lack FCRL5 expression (Fig. 6B and C) [26, PBMCs were obtained from whole blood by Ficoll-Hypaque
116]. Functionally, DN2 cells are hyperresponsive to TLR7 density gradient centrifugation as previously described [9].
signalling and can undergo rapid ASC differentiation in re- Isolated PBMCs were then centrifuged and cryopreserved in
sponse to IL21 and IFN-γ and are therefore felt to be pivotal ice-cold freezing medium (FCS with 10% DMSO) to give a
in extrafollicular B-cell responses [116, 163]. DN2 cells have cell concentration of 5–10 × 106 cells/ml.
been identified to be clonally related to aNAV B cells and ASC
with a lower level of IgV gene mutations and are enriched in Mass cytometry staining and analysis
autoreactive cells [27]. More recently DN3 and DN4 subsets The staining protocol and antibody panel for the mass
have been reported, the DN3 cluster being CD11c−CXCR5− cytometry data displayed in Figs. 1 and 2 is described by Tull
and the DN4 subset being enriched in IgE expressing cells and TJ et al. [9]. FCS files were normalized using Nolan Laboratory
to highly express transcripts for IL4R and CD40 [72, 73]. Software (https://github.com/nolanlab/beadnormalization).
SLE disease flares are associated with dramatic increases Single live B cells from 10 HCD (5 male and 5 female, mean
in ASCs that secrete pathogenic autoantibodies that are de- age 31.8 years) are gated as displayed in Fig. 1B. ViSNE plots
rived from extrafollicular B-cell responses [27]. This is asso- displayed in Fig. 1C are generated using Cytobank software
ciated with expansion of aNAV and DN2 B-cell populations (https://mrc.cytobank.org/) using equal numbers of B cells
(Fig. 6D) which are proposed to differentiate into ASCs via (n = 35 000) from 10 HCD. SPADE was then run on the
extrafollicular B-cell responses [116]. DN2 B cells are more viSNE coordinates and nodes corresponding to B-cell subsets
frequent in SLE patients of African–American ethnicity and are grouped as displayed in Fig. 1D. Transitional B cells were
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 211

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


Figure 6. DN B cells. (A) Flow cytometry plots demonstrating a gating strategy used to identify DN1 and DN2 DN B cells. DN1 cells are
CD27−IgD−CD21+CD24+ and DN2 cells are CD27−IgD−CD21−CD24−. Data are from a representative HCD. (B) SPADE on viSNE plot from mass
cytometry data showing expression of CD21, CD11c, and CXCR5 used to identify DN1 (CD21+CD11c−CXCR5+), DN2 (CD21−CD11c+CXCR5−), DN3
(CD21−CD11c−CXCR5−), and DN4 (CD21+CD11c+CXCR5+). (C) Heatmap demonstrating marker expression by DN B-cell subsets from mass cytometry
data. (D) Comparative SPADE on viSNE plots demonstrating expansion of DN2 cells in SLE compared to health (Tull TJ et al. [9])

identified as CD27−IgD+CD24+++/++CD38+++/++. Events within then exported and DN nodes were re-clustered using CD11c,
this transitional cell bubble were then exported and a fur- CD21, CXCR5, and CD24 to identify DN subsets.
ther viSNE run using equal events (n = 3535) and all panel
markers except CD45, CD3, CD14, and class-switched Flow cytometry staining and analysis
isotypes IgA and IgG, which are not expressed by transitional Cryopreserved PBMCs were stained with the fluorochrome-
B cells. The SPADE plots in Fig. 1C are generated from these conjugated antibodies as previously described [9]. The fol-
viSNE coordinates. SPADE on viSNE plots in Fig. 4C are gen- lowing antibodies clones were used: CD27 APC (M-T271),
erated by exporting events within the MZB SPADE bubble in IgD APC-Cy7 (IA6-2), CD19 PerCP-Cy5.5 (HIB19), CD10
Fig. 1D and re-clustered using CD45RB, IgD, CD21, integrin BV421 (HI10a), CD24 BV605 (ML5), IgM BV711 (MHM-
beta 7, CD27, CD24, IgM, HLA-DR, and CCR7. The DN 88), CD1c PE (L161), and CD45RB PE-TexasRed (MEM-
SPADE on viSNE was generated using data from 8 HCD and 55). CD21 BUV737 (HB7) and CD38 BUV395 (B-ly4). DAPI
8 patients with active severe SLE as described by Tull TJ et al. was used for live dead staining. Flow cytometry data were
[9]. CD19+ cells were gated and SPADE on viSNE plots cre- obtained by running samples on a Fortessa flow cytometer.
ated as in Fig. 1D. Events within the DN SPADE bubble were FCS files obtained were analysed using FlowJo software.
212 Velounias and Tull

Acknowledgements synovial tissues by integrating single-cell transcriptomics and mass


cytometry. Nat Immunol 2019, 20, 928–42.
Ms Louise Nel, Prof. David D’Cruz, and Dr Mike Robson 8. Papalexi E, Satija R. Single-cell RNA sequencing to explore im-
for sample collection. The Guy’s and St Thomas’ NHS Trust mune cell heterogeneity. Nat Rev Immunol 2018, 18, 35–45.
BRC flow and mass cytometry cores for sample processing 9. Tull TJ, Pitcher MJ, Guesdon W, Siu JHY, Lebrero-Fernandez C,
and data generation. Zhao Y, et al. Human marginal zone B cell development from early
T2 progenitors. J Exp Med 2021, 218(4), e20202001.
10. Zhao Y, Uduman M, Siu JHY, Tull TJ, Sanderson JD, Wu YB, et al.
Conflict of interest Spatiotemporal segregation of human marginal zone and memory
None declared. B cell populations in lymphoid tissue. Nat Commun 2018, 9, 3857.
11. Bendall SC, Davis KL, Amir el AD, Tadmor MD, Simonds EF, Chen
TJ, et al. Single-cell trajectory detection uncovers progression and
Funding regulatory coordination in human B cell development. Cell 2014,
157, 714–25.
This work was funded by the Medical Research Council of 12. Bemark M. Translating transitions - how to decipher peripheral
Great Britain (MR/R000964/1) and the St Thomas’ Lupus human B cell development. J Biomed Res 2015, 29, 264–84.
Trust. 13. Bao Y, Cao X. The immune potential and immunopathology

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


of cytokine-producing B cell subsets: a comprehensive review. J
Autoimmun 2014, 55, 10–23. doi:10.1016/j.jaut.2014.04.001
Data availability 14. Duddy ME, Alter A, Bar-Or A. Distinct profiles of human B cell
Raw mass cytometry data can be requested from the corres- effector cytokines: a role in immune regulation?. J Immunol 2004,
172, 3422–7. doi:10.4049/jimmunol.172.6.3422
ponding author.
15. Bar-Or A, Fawaz L, Fan B, Darlington PJ, Rieger A, Ghorayeb C, et
al. Abnormal B-cell cytokine responses a trigger of T-cell-mediated
disease in MS?. Ann Neurol 2010, 67, 452–61.
Author contributions 16. Canny SP, Jackson SW. B cells in systemic lupus erythematosus:
T.J.T.: Manuscript generation and figure review. R.L.V.: from disease mechanisms to targeted therapies. Rheum Dis Clin
Figure generation and manuscript review. North Am 2021, 47, 395–413.
17. Ghosh D, Jiang W, Mukhopadhyay D, Mellins ED. New insights
into B cells as antigen presenting cells. Curr Opin Immunol 2021,
70, 129–37. doi:10.1016/j.coi.2021.06.003
Permission to reproduce 18. Rodriguez-Pinto D. B cells as antigen presenting cells. Cell Immunol
Granted within terms set out by the licensing agreement. 2005, 238, 67–75.
19. Chong AS. B cells as antigen-presenting cells in transplantation re-
jection and tolerance. Cell Immunol 2020, 349, 104061.
Clinical trial registration 20. Xiao ZX, Miller JS, Zheng SG. An updated advance of
Not applicable. autoantibodies in autoimmune diseases. Autoimmun Rev 2021, 20,
102743. doi:10.1016/j.autrev.2020.102743
21. Hassan RI, Gaffo AL. Rituximab in ANCA-associated vasculitis.
Curr Rheumatol Rep 2017, 19, 6.
The animal research adheres to the ARRIVE 22. Tandan R, Hehir MK 2nd, Waheed W, Howard DB. Rituximab
guidelines treatment of myasthenia gravis: a systematic review. Muscle Nerve
Not applicable. 2017, 56, 185–96. doi:10.1002/mus.25597
23. Chisari CG, Sgarlata E, Arena S, Toscano S, Luca M, Patti F.
Rituximab for the treatment of multiple sclerosis: a review. J
Neurol 2022, 269, 159–83.
References 24. Wise LM, Stohl W. Belimumab and rituximab in systemic lupus er-
1. Weisel NM, Joachim SM, Smita S, Callahan D, Elsner RA, Conter ythematosus: a tale of two B cell-targeting agents. Front Med 2020,
LJ, et al. Surface phenotypes of naive and memory B cells in mouse 7, 303.
and human tissues. Nat Immunol 2022, 23, 135–45. 25. Alvarez-Rivas N, Sang-Park H, Diaz Del Campo P, Fernandez-
2. Cancro MP, Tomayko MM. Memory B cells and plasma cells: the Castro M, Corominas H, Andreu JL, et al. Efficacy of belimumab
differentiative continuum of humoral immunity. Immunol Rev in Primary Sjogren’s syndrome: a systematic review. Reumatol Clin
2021, 303, 72–82. doi:10.1111/imr.13016 (Engl Ed) 2021, 17, 170–4.
3. Benitez A, Weldon AJ, Tatosyan L, Velkuru V, Lee S, Milford TA, 26. Jenks SA, Cashman KS, Zumaquero E, Marigorta UM, Patel AV,
et al. Differences in mouse and human nonmemory B cell pools. J Wang X, et al. Distinct effector B cells induced by unregulated toll-
Immunol 2014, 192, 4610–9. doi:10.4049/jimmunol.1300692 like receptor 7 contribute to pathogenic responses in systemic lupus
4. Reynaud CA, Descatoire M, Dogan I, Huetz F, Weller S, Weill JC. erythematosus. Immunity 2018, 49, 725–39 e6.
IgM memory B cells: a mouse/human paradox. Cell Mol Life Sci 27. Tipton CM, Fucile CF, Darce J, Chida A, Ichikawa T, Gregoretti
2012, 69, 1625–34. doi:10.1007/s00018-012-0971-z I, et al. Diversity, cellular origin and autoreactivity of antibody-
5. Glass DR, Tsai AG, Oliveria JP, Hartmann FJ, Kimmey SC, Calderon secreting cell population expansions in acute systemic lupus erythe-
AA, et al. An integrated multi-omic single-cell atlas of human matosus. Nat Immunol 2015, 16, 755–65. doi:10.1038/ni.3175
B cell identity. Immunity 2020, 53, 217–32.e5. doi:10.1016/j. 28. Landolt-Marticorena C, Wither R, Reich H, Herzenberg A, Scholey
immuni.2020.06.013 J, Gladman DD, et al. Increased expression of B cell activation
6. Siu JHY, Pitcher MJ, Tull TJ, Velounias RL, Guesdon W, Montorsi factor supports the abnormal expansion of transitional B cells in
L, et al. Two subsets of human marginal zone B cells resolved by systemic lupus erythematosus. J Rheumatol 2011, 38, 642–51.
global analysis of lymphoid tissues and blood. Sci Immunol 2022, doi:10.3899/jrheum.100214
7, eabm9060. 29. Perez-Andres M, Grosserichter-Wagener C, Teodosio C, van
7. Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S, et Dongen JJ, Orfao A, van Zelm MC. The nature of circulating
al. Defining inflammatory cell states in rheumatoid arthritis joint CD27+CD43+ B cells. J Exp Med 2011, 208, 2565–6.
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 213

30. Descatoire M, Weill JC, Reynaud CA, Weller S. A human equivalent lupus erythematosus. J Immunol 2000, 165, 5970–9. doi:10.4049/
of mouse B-1 cells?. J Exp Med 2011, 208, 2563–4. doi:10.1084/ jimmunol.165.10.5970
jem.20112232 49. Simon D, Balogh P, Bognar A, Kellermayer Z, Engelmann P, Nemeth
31. Suo C, Dann E, Goh I, Jardine L, Kleshchevnikov V, Park JE, et al. P, et al. Reduced non-switched memory B cell subsets cause imbal-
Mapping the developing human immune system across organs. Sci- ance in B cell repertoire in systemic sclerosis. Clin Exp Rheumatol
ence 2022, 376, eabo0510. 2016, 34, 30–6.
32. Scheuermann RH, Racila E. CD19 antigen in leukemia and lym- 50. Bemark M, Friskopp L, Saghafian-Hedengren S, Koethe S, Fasth
phoma diagnosis and immunotherapy. Leuk Lymphoma 1995, 18, A, Abrahamsson J, et al. A glycosylation-dependent CD45RB epi-
385–97. doi:10.3109/10428199509059636 tope defines previously unacknowledged CD27(-)IgM(high) B cell
33. Mahevas M, Patin P, Huetz F, Descatoire M, Cagnard N, Bole- subpopulations enriched in young children and after hematopoietic
Feysot C, et al. B cell depletion in immune thrombocytopenia stem cell transplantation. Clin Immunol 2013, 149, 421–31.
reveals splenic long-lived plasma cells. J Clin Invest 2013, 123, 51. Wangriatisak K, Thanadetsuntorn C, Krittayapoositpot T,
432–42. Leepiyasakulchai C, Suangtamai T, Ngamjanyaporn P, et al. The
34. Sanz I, Wei C, Jenks SA, Cashman KS, Tipton C, Woodruff MC, expansion of activated naive DNA autoreactive B cells and its as-
et al. Challenges and opportunities for consistent classification of sociation with disease activity in systemic lupus erythematosus
human B cell and plasma cell populations. Front Immunol 2019, patients. Arthritis Res Ther 2021, 23, 179.
10, 2458. 52. Weller S, Braun MC, Tan BK, Rosenwald A, Cordier C, Conley ME,

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


35. Suryani S, Fulcher DA, Santner-Nanan B, Nanan R, Wong M, Shaw et al. Human blood IgM “memory” B cells are circulating splenic
PJ, et al. Differential expression of CD21 identifies developmen- marginal zone B cells harboring a prediversified immunoglobulin
tally and functionally distinct subsets of human transitional B cells. repertoire. Blood 2004, 104, 3647–54.
Blood 2010, 115, 519–29. doi:10.1182/blood-2009-07-234799 53. Feng R, Zhao J, Sun F, Miao M, Sun X, He J, et al. Comparison of
36. Anolik JH, Friedberg JW, Zheng B, Barnard J, Owen T, Cushing E, the deep immune profiling of B cell subsets between healthy adults
et al. B cell reconstitution after rituximab treatment of lymphoma and Sjogren’s syndrome. Ann Med 2022, 54, 472–83. doi:10.1080/
recapitulates B cell ontogeny. Clin Immunol 2007, 122, 139–45. 07853890.2022.2031272
37. Sims GP, Ettinger R, Shirota Y, Yarboro CH, Illei GG, Lipsky PE. 54. Zhu L, Yin Z, Ju B, Zhang J, Wang Y, Lv X, et al. Altered frequencies
Identification and characterization of circulating human transi- of memory B cells in new-onset systemic lupus erythematosus
tional B cells. Blood 2005, 105, 4390–8. doi:10.1182/blood-2004- patients. Clin Rheumatol 2018, 37, 205–12.
11-4284 55. Zhang W, Wang YF, Hu FL, Lu FA, Wu T, Feng YL, et al. Dys-
38. Dieudonne Y, Gies V, Guffroy A, Keime C, Bird AK, Liesveld J, function of CD27(+)IgD(+) B cells correlates with aggravated sys-
et al. Transitional B cells in quiescent SLE: an early checkpoint temic lupus erythematosus. Clin Rheumatol 2022, 41, 1551–9.
imprinted by IFN. J Autoimmun 2019, 102, 150–8. doi:10.1016/j. doi:10.1007/s10067-022-06051-z
jaut.2019.05.002 56. Roberts ME, Kaminski D, Jenks SA, Maguire C, Ching K, Burbelo
39. Vossenkamper A, Blair PA, Safinia N, Fraser LD, Das L, Sanders PD, et al. Primary Sjogren’s syndrome is characterized by dis-
TJ, et al. A role for gut-associated lymphoid tissue in shaping tinct phenotypic and transcriptional profiles of IgD+ unswitched
the human B cell repertoire. J Exp Med 2013, 210, 1665–74. memory B cells. Arthritis Rheumatol 2014, 66, 2558–69.
doi:10.1084/jem.20122465 57. Corsiero E, Sutcliffe N, Pitzalis C, Bombardieri M. Accumulation
40. Taher TE, Ong VH, Bystrom J, Hillion S, Simon Q, Denton CP, et of self-reactive naive and memory B cell reveals sequential defects
al. Association of defective regulation of autoreactive interleukin- in B cell tolerance checkpoints in Sjogren’s syndrome. PLoS One
6-producing transitional B lymphocytes with disease in patients 2014, 9, e114575e114575. doi:10.1371/journal.pone.0114575
with systemic sclerosis. Arthritis Rheumatol 2018, 70, 450–61. 58. Gernert M, Tony HP, Schwaneck EC, Gadeholt O, Frohlich M,
doi:10.1002/art.40390 Portegys J, et al. Lymphocyte subsets in the peripheral blood are
41. Woodruff M, Ramonell R, Cashman K, Nguyen D, Ley A, Kyu S, et disturbed in systemic sclerosis patients and can be changed by im-
al. Critically ill SARS-CoV-2 patients display lupus-like hallmarks munosuppressive medication. Rheumatol Int 2022, 42(8), 1373–
of extrafollicular B cell activation. medRxiv 2020, preprint: not 81. doi:10.1007/s00296-021-05034-8
peer reviewed. doi:10.1101/2020.04.29.20083717 59. Hu F, Zhang W, Shi L, Liu X, Jia Y, Xu L, et al. Impaired CD27(+)
42. Sosa-Hernandez VA, Torres-Ruiz J, Cervantes-Diaz R, Romero- IgD(+) B cells with altered gene signature in rheumatoid arthritis.
Ramirez S, Paez-Franco JC, Meza-Sanchez DE, et al. B cell subsets Front Immunol 2018, 9, 626.
as severity-associated signatures in COVID-19 patients. Front 60. Laing AG, Lorenc A, Del Molino Del Barrio I, Das A, Fish M,
Immunol 2020, 11, 611004. Monin L, et al. A dynamic COVID-19 immune signature includes
43. Wirths S, Lanzavecchia A. ABCB1 transporter discriminates human associations with poor prognosis. Nat Med 2020, 26(10), 1623–35.
resting naive B cells from cycling transitional and memory B cells. doi:10.1038/s41591-020-1038-6
Eur J Immunol 2005, 35, 3433–41. doi:10.1002/eji.200535364 61. Bagnara D, Squillario M, Kipling D, Mora T, Walczak AM, Da
44. Lee J, Kuchen S, Fischer R, Chang S, Lipsky PE. Identification and Silva L, et al. A reassessment of IgM memory subsets in humans. J
characterization of a human CD5+ pre-naive B cell population. J Immunol 2015, 195, 3716–24. doi:10.4049/jimmunol.1500753
Immunol 2009, 182, 4116–26. doi:10.4049/jimmunol.0803391 62. Klein U, Kuppers R, Rajewsky K. Evidence for a large compart-
45. Palanichamy A, Barnard J, Zheng B, Owen T, Quach T, Wei C, et ment of IgM-expressing memory B cells in humans. Blood 1997,
al. Novel human transitional B cell populations revealed by B cell 89, 1288–98.
depletion therapy. J Immunol 2009, 182, 5982–93. doi:10.4049/ 63. Klein U, Rajewsky K, Kuppers R. Human immunoglobulin (Ig)
jimmunol.0801859 M+IgD+ peripheral blood B cells expressing the CD27 cell surface
46. Descatoire M, Weller S, Irtan S, Sarnacki S, Feuillard J, Storck S, et antigen carry somatically mutated variable region genes: CD27 as
al. Identification of a human splenic marginal zone B cell precursor a general marker for somatically mutated (memory) B cells. J Exp
with NOTCH2-dependent differentiation properties. J Exp Med Med 1998, 188, 1679–89. doi:10.1084/jem.188.9.1679
2014, 211, 987–1000. doi:10.1084/jem.20132203 64. Berkowska MA, Driessen GJ, Bikos V, Grosserichter-Wagener C,
47. Quach TD, Manjarrez-Orduno N, Adlowitz DG, Silver L, Yang H, Stamatopoulos K, Cerutti A, et al. Human memory B cells originate
Wei C, et al. Anergic responses characterize a large fraction of human from three distinct germinal center-dependent and -independent
autoreactive naive B cells expressing low levels of surface IgM. J maturation pathways. Blood 2011, 118, 2150–8.
Immunol 2011, 186, 4640–8. doi:10.4049/jimmunol.1001946 65. Korganow AS, Knapp AM, Nehme-Schuster H, Soulas-Sprauel
48. Odendahl M, Jacobi A, Hansen A, Feist E, Hiepe F, Burmester GR, P, Poindron V, Pasquali JL, et al. Peripheral B cell abnormalities
et al. Disturbed peripheral B lymphocyte homeostasis in systemic in patients with systemic lupus erythematosus in quiescent
214 Velounias and Tull

phase: decreased memory B cells and membrane CD19 expression. single-cell data and reveals phenotypic heterogeneity of leukemia.
J Autoimmun 2010, 34, 426–34. doi:10.1016/j.jaut.2009.11.002 Nat Biotechnol 2013, 31, 545–52.
66. Szabo K, Papp G, Szanto A, Tarr T, Zeher M. A comprehensive 83. Marie-Cardine A, Divay F, Dutot I, Green A, Perdrix A, Boyer O,
investigation on the distribution of circulating follicular T helper et al. Transitional B cells in humans: characterization and insight
cells and B cell subsets in primary Sjogren’s syndrome and systemic from B lymphocyte reconstitution after hematopoietic stem cell
lupus erythematosus. Clin Exp Immunol 2016, 183, 76–89. transplantation. Clin Immunol 2008, 127, 14–25.
67. Souto-Carneiro MM, Mahadevan V, Takada K, Fritsch-Stork R, 84. Bemark M, Holmqvist J, Abrahamsson J, Mellgren K. Transla-
Nanki T, Brown M, et al. Alterations in peripheral blood memory B tional Mini-Review Series on B cell subsets in disease. Reconstitu-
cells in patients with active rheumatoid arthritis are dependent on tion after haematopoietic stem cell transplantation - revelation of
the action of tumour necrosis factor. Arthritis Res Ther 2009, 11, B cell developmental pathways and lineage phenotypes. Clin Exp
R84. doi:10.1186/ar2718 Immunol 2012, 167, 15–25.
68. Sellam J, Rouanet S, Hendel-Chavez H, Abbed K, Sibilia J, Tebib J, 85. Mraz M, Zent CS, Church AK, Jelinek DF, Wu X, Pospisilova
et al. Blood memory B cells are disturbed and predict the response S, et al. Bone marrow stromal cells protect lymphoma B-cells
to rituximab in patients with rheumatoid arthritis. Arthritis Rheum from rituximab-induced apoptosis and targeting integrin
2011, 63, 3692–701. doi:10.1002/art.30599 alpha-4-beta-1 (VLA-4) with natalizumab can overcome this
69. Elmer E, Smargianaki S, Pettersson A, Skattum L, Ohlsson S, resistance. Br J Haematol 2011, 155, 53–64. doi:10.1111/j.1365-
Hellmark T, et al. Increased frequencies of switched memory B cells 2141.2011.08794.x

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


and plasmablasts in peripheral blood from patients with ANCA- 86. Loder F, Mutschler B, Ray RJ, Paige CJ, Sideras P, Torres R, et al.
associated vasculitis. J Immunol Res 2020, 2020, 8209737. B cell development in the spleen takes place in discrete steps and is
70. Fecteau JF, Cote G, Neron S. A new memory CD27-IgG+ B cell determined by the quality of B cell receptor-derived signals. J Exp
population in peripheral blood expressing VH genes with low fre- Med 1999, 190, 75–89.
quency of somatic mutation. J Immunol 2006, 177, 3728–36. 87. Allman D, Lindsley RC, DeMuth W, Rudd K, Shinton SA, Hardy
71. Wei C, Anolik J, Cappione A, Zheng B, Pugh-Bernard A, Brooks RR. Resolution of three nonproliferative immature splenic B
J, et al. A new population of cells lacking expression of CD27 cell subsets reveals multiple selection points during peripheral B
represents a notable component of the B cell memory compartment cell maturation. J Immunol 2001, 167, 6834–40. doi:10.4049/
in systemic lupus erythematosus. J Immunol 2007, 178, 6624–33. jimmunol.167.12.6834
doi:10.4049/jimmunol.178.10.6624 88. Srivastava B, Quinn WJ 3rd, Hazard K, Erikson J, Allman D. Char-
72. Woodruff MC, Ramonell RP, Nguyen DC, Cashman KS, Saini AS, acterization of marginal zone B cell precursors. J Exp Med 2005,
Haddad NS, et al. Extrafollicular B cell responses correlate with 202, 1225–34. doi:10.1084/jem.20051038
neutralizing antibodies and morbidity in COVID-19. Nat Immunol 89. Pillai S, Cariappa A, Moran ST. Positive selection and lineage com-
2020, 21, 1506–16. doi:10.1038/s41590-020-00814-z mitment during peripheral B-lymphocyte development. Immunol
73. Stewart A, Ng JC, Wallis G, Tsioligka V, Fraternali F, Dunn-Walters Rev 2004, 197, 206–18. doi:10.1111/j.0105-2896.2003.097.x
DK. Single-cell transcriptomic analyses define distinct peripheral B 90. Evans JG, Chavez-Rueda KA, Eddaoudi A, Meyer-Bahlburg A,
cell subsets and discrete development pathways. Front Immunol Rawlings DJ, Ehrenstein MR, et al. Novel suppressive function of
2021, 12, 602539. transitional 2 B cells in experimental arthritis. J Immunol 2007,
74. You X, Zhang R, Shao M, He J, Chen J, Liu J, et al. Double negative 178, 7868–78. doi:10.4049/jimmunol.178.12.7868
B cell is associated with renal impairment in systemic lupus erythe- 91. Yurasov S, Wardemann H, Hammersen J, Tsuiji M, Meffre E,
matosus and acts as a marker for nephritis remission. Front Med Pascual V, et al. Defective B cell tolerance checkpoints in systemic
2020, 7, 85. lupus erythematosus. J Exp Med 2005, 201, 703–11. doi:10.1084/
75. Wang Y, Lloyd KA, Melas I, Zhou D, Thyagarajan R, Lindqvist jem.20042251
J, et al. Rheumatoid arthritis patients display B-cell dysregulation 92. Wang H, Liu C, Chen W, Ding G. The skewed frequency of B-cell
already in the naive repertoire consistent with defects in B-cell tol- subpopulation CD19(+) CD24 (hi) CD38 (hi) cells in peripheral
erance. Sci Rep 2019, 9, 19995. blood mononuclear cells is correlated with the elevated serum
76. Moura RA, Quaresma C, Vieira AR, Goncalves MJ, Polido-Pereira sCD40L in patients with active systemic lupus erythematosus. J
J, Romao VC, et al. B-cell phenotype and IgD-CD27- memory B Cell Biochem 2019. doi:10.1002/jcb.28427
cells are affected by TNF-inhibitors and tocilizumab treatment in 93. Smets I, Prezzemolo T, Imbrechts M, Mallants K, Mitera T,
rheumatoid arthritis. PLoS One 2017, 12, e0182927. doi:10.1371/ Humblet-Baron S, et al. Treatment-induced BAFF expression and B
journal.pone.0182927 cell biology in multiple sclerosis. Front Immunol 2021, 12, 676619.
77. Mahmood Z, Muhammad K, Schmalzing M, Roll P, Dorner T, 94. Chang NH, Li TT, Kim JJ, Landolt-Marticorena C, Fortin PR,
Tony HP. CD27-IgD-memory B cells are modulated by in vivo in- Gladman DD, et al. Interferon-alpha induces altered transitional
terleukin-6 receptor (IL-6R) blockade in rheumatoid arthritis. Ar- B cell signaling and function in Systemic Lupus Erythematosus. J
thritis Res Ther 2015, 17, 61. Autoimmun 2015, 58, 100–10. doi:10.1016/j.jaut.2015.01.009
78. Cervantes-Diaz R, Sosa-Hernandez VA, Torres-Ruiz J, Romero- 95. Klarquist J, Cantrell R, Lehn MA, Lampe K, Hennies CM, Hoebe K,
Ramirez S, Canez-Hernandez M, Perez-Fragoso A, et al. Severity of et al. Type I IFN drives experimental systemic lupus erythematosus
SARS-CoV-2 infection is linked to double-negative (CD27(-) IgD(- by distinct mechanisms in CD4 T cells and B cells. Immunohorizons
)) B cell subset numbers. Inflamm Res 2022, 71, 131–40. 2020, 4, 140–52. doi:10.4049/immunohorizons.2000005
79. Oliviero B, Varchetta S, Mele D, Mantovani S, Cerino A, Perotti 96. Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S,
CG, et al. Expansion of atypical memory B cells is a prominent fea- Duurland CL, et al. CD19(+)CD24(hi)CD38(hi) B cells are ex-
ture of COVID-19. Cell Mol Immunol 2020, 17, 1101–3. panded in juvenile dermatomyositis and exhibit a pro-inflammatory
80. Bodenmiller B, Zunder ER, Finck R, Chen TJ, Savig ES, Bruggner phenotype after activation through toll-like receptor 7 and
RV, et al. Multiplexed mass cytometry profiling of cellular states interferon-alpha. Front Immunol 2018, 9, 1372.
perturbed by small-molecule regulators. Nat Biotechnol 2012, 30, 97. Lin W, Jin L, Chen H, Wu Q, Fei Y, Zheng W, et al. B cell subsets
858–67. doi:10.1038/nbt.2317 and dysfunction of regulatory B cells in IgG4-related diseases and
81. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, et primary Sjogren’s syndrome: the similarities and differences. Ar-
al. Dimensionality reduction for visualizing single-cell data using thritis Res Ther 2014, 16, R118. doi:10.1186/ar4571
UMAP. Nat Biotechnol 2018. doi:10.1038/nbt.4314 98. Cherukuri A, Salama AD, Carter CR, Landsittel D, Arumugakani
82. Amir el AD, Davis KL, Tadmor MD, Simonds EF, Levine JH, G, Clark B, et al. Reduced human transitional B cell T1/
Bendall SC, et al. viSNE enables visualization of high dimensional T2 ratio is associated with subsequent deterioration in renal
Clinical and Experimental Immunology, 2022, Vol. 210, No. 3 215

allograft function. Kidney Int 2017, 91, 183–95. doi:10.1016/j. autoreactive B cells in systemic lupus erythematosus patients. JCI
kint.2016.08.028 Insight 2018, 3(17), e122525.
99. Burton H, Dorling A. Transitional B cell subsets-a convincing pre- 118. Ramskold D, Parodis I, Lakshmikanth T, Sippl N, Khademi
dictive biomarker for allograft loss?. Kidney Int 2017, 91, 18–20. M, Chen Y, et al. B cell alterations during BAFF inhibition
doi:10.1016/j.kint.2016.10.028 with belimumab in SLE. EBioMedicine 2019, 40, 517–27.
100. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, doi:10.1016/j.ebiom.2018.12.035
Ehrenstein MR, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit 119. Yagi-Numata N, Matsushita T, Takehara K, Hamaguchi Y.
regulatory capacity in healthy individuals but are functionally im- Increased expression levels of FcgammaRIIB on naive and double-
paired in systemic Lupus Erythematosus patients. Immunity 2010, negative memory B cells in patients with systemic sclerosis. Clin
32, 129–40. doi:10.1016/j.immuni.2009.11.009 Exp Rheumatol 2019, 37, 23–31.
101. Mauri C, Menon M. Human regulatory B cells in health and 120. Sato S, Fujimoto M, Hasegawa M, Takehara K. Altered blood
disease: therapeutic potential. J Clin Invest 2017, 127, 772–9. B lymphocyte homeostasis in systemic sclerosis: expanded naive
doi:10.1172/jci85113 B cells and diminished but activated memory B cells. Arthritis
102. Menon M, Blair PA, Isenberg DA, Mauri C. A regulatory feed- Rheum 2004, 50, 1918–27. doi:10.1002/art.20274
back between plasmacytoid dendritic cells and regulatory B cells 121. von Borstel A, Abdulahad WH, Sanders JS, Rip J, Neys SFH,
is aberrant in systemic lupus erythematosus. Immunity 2016, 44, Hendriks RW, et al. Evidence for enhanced Bruton’s tyrosine
683–97. doi:10.1016/j.immuni.2016.02.012 kinase activity in transitional and naive B cells of patients with

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


103. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, granulomatosis with polyangiitis. Rheumatology 2019, 58, 2230–
et al. Regulatory B cells are induced by gut microbiota-driven 9.
interleukin-1beta and interleukin-6 production. Nat Med 2014, 122. Hansen A, Odendahl M, Reiter K, Jacobi AM, Feist E, Scholze J, et
20, 1334–9. doi:10.1038/nm.3680 al. Diminished peripheral blood memory B cells and accumulation
104. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg of memory B cells in the salivary glands of patients with Sjogren’s
DA, et al. CD19+CD24hiCD38hi B cells maintain regulatory T syndrome. Arthritis Rheum 2002, 46, 2160–71. doi:10.1002/
cells while limiting TH1 and TH17 differentiation. Sci Transl Med art.10445
2013, 5, 173ra23. 123. Mebius RE, Kraal G. Structure and function of the spleen. Nat
105. Banko Z, Pozsgay J, Gati T, Rojkovich B, Ujfalussy I, Sarmay G. Rev Immunol 2005, 5, 606–16. doi:10.1038/nri1669
Regulatory B cells in rheumatoid arthritis: alterations in patients 124. Weill JC, Weller S, Reynaud CA. Human marginal zone B cells.
receiving anti-TNF therapy. Clin Immunol 2017, 184, 63–9. Annu Rev Immunol 2009, 27, 267–85. doi:10.1146/annurev.
106. Lin X, Wang X, Xiao F, Ma K, Liu L, Wang X, et al. IL-10- immunol.021908.132607
producing regulatory B cells restrain the T follicular helper cell 125. Weller S, Faili A, Garcia C, Braun MC, Le Deist FF, de Saint Basile
response in primary Sjogren’s syndrome. Cell Mol Immunol 2019, GG, et al. CD40-CD40L independent Ig gene hypermutation
16, 921–31. suggests a second B cell diversification pathway in humans. Proc
107. Cencioni MT, Ali R, Nicholas R, Muraro PA. Defective Natl Acad Sci U S A 2001, 98, 1166–70.
CD19+CD24(hi)CD38(hi) transitional B-cell function in patients 126. Seifert M, Kuppers R. Molecular footprints of a germinal center
with relapsing-remitting MS. Mult Scler 2021, 27, 1187–97. derivation of human IgM+(IgD+)CD27+ B cells and the dynamics
108. Matsushita T, Hamaguchi Y, Hasegawa M, Takehara K, Fujimoto of memory B cell generation. J Exp Med 2009, 206, 2659–69.
M. Decreased levels of regulatory B cells in patients with systemic doi:10.1084/jem.20091087
sclerosis: association with autoantibody production and disease 127. Budeus B, Schweigle de Reynoso S, Przekopowitz M, Hoffmann
activity. Rheumatology 2016, 55, 263–7. D, Seifert M, Kuppers R. Complexity of the human memory B-cell
109. Zhu HQ, Xu RC, Chen YY, Yuan HJ, Cao H, Zhao XQ, et al. Im- compartment is determined by the versatility of clonal diversifi-
paired function of CD19(+) CD24(hi) CD38(hi) regulatory B cells cation in germinal centers. Proc Natl Acad Sci U S A 2015, 112,
in patients with pemphigus. Br J Dermatol 2015, 172, 101–10. E5281–9.
110. Sheng JR, Rezania K, Soliven B. Impaired regulatory B cells in my- 128. Seifert M, Kuppers R. Human memory B cells. Leukemia 2016,
asthenia gravis. J Neuroimmunol 2016, 297, 38–45. doi:10.1016/j. 30, 2283–92. doi:10.1038/leu.2016.226
jneuroim.2016.05.004 129. Cerutti A, Cols M, Puga I. Marginal zone B cells: virtues of innate-
111. Lin Y, Chang T, Lin J, Sun C, Wei C, Zhao J, et al. Regulatory B like antibody-producing lymphocytes. Nat Rev Immunol 2013,
cells are decreased and functionally impaired in myasthenia gravis 13, 118–32. doi:10.1038/nri3383
patients. Front Neurol 2022, 13, 808322. 130. Nagafuchi Y, Shoda H, Sumitomo S, Nakachi S, Kato R, Tsuchida
112. Guerrier T, Youinou P, Pers JO, Jamin C. TLR9 drives the devel- Y, et al. Enhanced gut homing receptor expression of unswitched
opment of transitional B cells towards the marginal zone pathway memory B cells in rheumatoid arthritis. Clin Exp Rheumatol
and promotes autoimmunity. J Autoimmun 2012, 39, 173–9. 2017, 35, 354–5.
doi:10.1016/j.jaut.2012.05.012 131. Newell KL, Clemmer DC, Cox JB, Kayode YI, Zoccoli-Rodriguez
113. Bell SE, Goodnow CC. A selective defect in IgM antigen receptor V, Taylor HE, et al. Switched and unswitched memory B cells
synthesis and transport causes loss of cell surface IgM expres- detected during SARS-CoV-2 convalescence correlate with limited
sion on tolerant B lymphocytes. EMBO J 1994, 13, 816–26. symptom duration. PLoS One 2021, 16, e0244855. doi:10.1371/
doi:10.1002/j.1460-2075.1994.tb06324.x journal.pone.0244855
114. Sabouri Z, Perotti S, Spierings E, Humburg P, Yabas M, Bergmann 132. Karlsen M, Hansen T, Nordal HH, Brun JG, Jonsson R, Appel S.
H, et al. IgD attenuates the IgM-induced anergy response in tran- Expression of Toll-like receptor -7 and -9 in B cell subsets from
sitional and mature B cells. Nat Commun 2016, 7, 13381. patients with primary Sjogren’s syndrome. PLoS One 2015, 10,
115. Brink R, Goodnow CC, Crosbie J, Adams E, Eris J, Mason DY, et e0120383. doi:10.1371/journal.pone.0120383
al. Immunoglobulin M and D antigen receptors are both capable 133. Si R, Zhao P, Yu Z, Qu Z, Sun W, Li T, et al. Increased non-
of mediating B lymphocyte activation, deletion, or anergy after in- switched memory B cells are associated with plasmablasts, serum
teraction with specific antigen. J Exp Med 1992, 176, 991–1005. IL-6 levels and renal functional impairments in IgAN patients.
doi:10.1084/jem.176.4.991 Immunol Invest 2020, 49, 178–90.
116. Jenks SA, Cashman KS, Woodruff MC, Lee FE, Sanz I. 134. Simon Q, Pers JO, Cornec D, Le Pottier L, Mageed RA, Hillion
Extrafollicular responses in humans and SLE. Immunol Rev 2019, S. In-depth characterization of CD24(high)CD38(high) transi-
288, 136–48. tional human B cells reveals different regulatory profiles. J Al-
117. Huang W, Quach TD, Dascalu C, Liu Z, Leung T, Byrne-Steele lergy Clin Immunol 2016, 137, 1577–84.e10. doi:10.1016/j.
M, et al. Belimumab promotes negative selection of activated jaci.2015.09.014
216 Velounias and Tull

135. Carter MJ, Fish M, Jennings A, Doores KJ, Wellman P, Seow J, et 152. Manea ME, Mueller RB, Dejica D, Sheriff A, Schett G, Herrmann
al. Peripheral immunophenotypes in children with multisystem in- M, et al. Increased expression of CD154 and FAS in SLE patients’
flammatory syndrome associated with SARS-CoV-2 infection. Nat lymphocytes. Rheumatol Int 2009, 30, 181–5. doi:10.1007/
Med 2020, 26, 1701–7. s00296-009-0933-4
136. Seifert M, Przekopowitz M, Taudien S, Lollies A, Ronge V, Drees 153. Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch
B, et al. Functional capacities of human IgM memory B cells in A, et al. Activated memory B cell subsets correlate with disease ac-
early inflammatory responses and secondary germinal center tivity in systemic lupus erythematosus: delineation by expression
reactions. Proc Natl Acad Sci U S A 2015, 112, E546–55. of CD27, IgD, and CD95. Arthritis Rheum 2008, 58, 1762–73.
137. Wu YC, Kipling D, Leong HS, Martin V, Ademokun AA, Dunn- doi:10.1002/art.23498
Walters DK. High-throughput immunoglobulin repertoire analysis 154. Wang J, Shan Y, Jiang Z, Feng J, Li C, Ma L, et al. High
distinguishes between human IgM memory and switched memory frequencies of activated B cells and T follicular helper cells
B-cell populations. Blood 2010, 116, 1070–8. doi:10.1182/blood- are correlated with disease activity in patients with new-onset
2010-03-275859 rheumatoid arthritis. Clin Exp Immunol 2013, 174, 212–20.
138. Trend S, Leffler J, Teige I, Frendeus B, Kermode AG, French MA, doi:10.1111/cei.12162
et al. FcgammaRIIb expression is decreased on naive and mar- 155. Pugh-Bernard AE, Silverman GJ, Cappione AJ, Villano ME, Ryan
ginal zone-like B cells from females with multiple sclerosis. Front DH, Insel RA, et al. Regulation of inherently autoreactive VH4-34
Immunol 2020, 11, 614492. B cells in the maintenance of human B cell tolerance. J Clin Invest

Downloaded from https://academic.oup.com/cei/article/210/3/201/6974682 by guest on 17 May 2023


139. Liu Y, Feng Y, Tang S, Zhang L, Huang Z, Shi X, et al. Aber- 2001, 108, 1061–70. doi:10.1172/jci200112462
rant expression of inhibitory receptors on B cells in patients with 156. Desai-Mehta A, Lu L, Ramsey-Goldman R, Datta SK.
Graves’ disease. Hum Immunol 2022, 83, 144–52. doi:10.1016/j. Hyperexpression of CD40 ligand by B and T cells in human lupus
humimm.2021.12.001 and its role in pathogenic autoantibody production. J Clin Invest
140. Danza A, Ruiz-Irastorza G. Infection risk in systemic lupus erythe- 1996, 97, 2063–73. doi:10.1172/jci118643
matosus patients: susceptibility factors and preventive strategies. 157. Grammer AC, Slota R, Fischer R, Gur H, Girschick H, Yarboro
Lupus 2013, 22, 1286–94. doi:10.1177/0961203313493032 C, et al. Abnormal germinal center reactions in systemic lupus
141. Doyon-Laliberte K, Aranguren M, Poudrier J, Roger M. Marginal erythematosus demonstrated by blockade of CD154-CD40
zone B-cell populations and their regulatory potential in the con- interactions. J Clin Invest 2003, 112, 1506–20. doi:10.1172/
text of HIV and other chronic inflammatory conditions. Int J Mol jci200319301
Sci 2022, 23(6), 3372. 158. Ramanujam M, Steffgen J, Visvanathan S, Mohan C, Fine JS,
142. Weisel F, Shlomchik M. Memory B cells of mice and humans. Putterman C. Phoenix from the flames: rediscovering the role
Annu Rev Immunol 2017, 35, 255–84. doi:10.1146/annurev- of the CD40-CD40L pathway in systemic lupus erythematosus
immunol-041015-055531 and lupus nephritis. Autoimmun Rev 2020, 19, 102668102668.
143. Maurer D, Holter W, Majdic O, Fischer GF, Knapp W. CD27 ex- doi:10.1016/j.autrev.2020.102668
pression by a distinct subpopulation of human B lymphocytes. 159. Roll P, Mahmood Z, Muhammad K, Feuchtenberger M, Dorner
Eur J Immunol 1990, 20, 2679–84. doi:10.1002/eji.1830201223 T, Tony HP. Long-term repopulation of peripheral B-cell subsets
144. Tangye SG, Liu YJ, Aversa G, Phillips JH, de Vries JE. Identifica- after single and repeated rituximab infusions in patients with
tion of functional human splenic memory B cells by expression of rheumatoid arthritis. Clin Exp Rheumatol 2015, 33, 347–53.
CD148 and CD27. J Exp Med 1998, 188, 1691–703. doi:10.1084/ 160. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, et al.
jem.188.9.1691 Evidence for HIV-associated B cell exhaustion in a dysfunctional
145. Tarlinton D, Good-Jacobson K. Diversity among memory B cells: memory B cell compartment in HIV-infected viremic individuals. J
origin, consequences, and utility. Science 2013, 341, 1205–11. Exp Med 2008, 205, 1797–805. doi:10.1084/jem.20072683
doi:10.1126/science.1241146 161. Liechti T, Kadelka C, Braun DL, Kuster H, Boni J, Robbiani M,
146. Good-Jacobson KL, Shlomchik MJ. Plasticity and heterogeneity et al. Widespread B cell perturbations in HIV-1 infection afflict
in the generation of memory B cells and long-lived plasma cells: naive and marginal zone B cells. J Exp Med 2019, 216, 2071–90.
the influence of germinal center interactions and dynamics. J doi:10.1084/jem.20181124
Immunol 2010, 185, 3117–25. doi:10.4049/jimmunol.1001155 162. Cancro MP. Age-associated B cells. Annu Rev Immunol 2020, 38,
147. Lanzavecchia A, Sallusto F. Progressive differentiation and se- 315–40. doi:10.1146/annurev-immunol-092419-031130
lection of the fittest in the immune response. Nat Rev Immunol 163. Wang S, Wang J, Kumar V, Karnell JL, Naiman B, Gross PS, et
2002, 2, 982–7. doi:10.1038/nri959 al. IL-21 drives expansion and plasma cell differentiation of
148. Tangye SG, Avery DT, Deenick EK, Hodgkin PD. Intrinsic autoreactive CD11c(hi)T-bet(+) B cells in SLE. Nat Commun
differences in the proliferation of naive and memory human B 2018, 9, 1758.
cells as a mechanism for enhanced secondary immune responses. J 164. Rubtsov AV, Rubtsova K, Fischer A, Meehan RT, Gillis JZ, Kappler
Immunol 2003, 170, 686–94. doi:10.4049/jimmunol.170.2.686 JW, et al. Toll-like receptor 7 (TLR7)-driven accumulation of a
149. Good KL, Bryant VL, Tangye SG. Kinetics of human B cell be- novel CD11c(+) B-cell population is important for the develop-
havior and amplification of proliferative responses following stim- ment of autoimmunity. Blood 2011, 118, 1305–15. doi:10.1182/
ulation with IL-21. J Immunol 2006, 177, 5236–47. doi:10.4049/ blood-2011-01-331462
jimmunol.177.8.5236 165. Saadoun D, Terrier B, Bannock J, Vazquez T, Massad C, Kang I,
150. Weill JC, Reynaud CA. IgM memory B cells: specific effectors of et al. Expansion of autoreactive unresponsive CD21-/low B cells
innate-like and adaptive responses. Curr Opin Immunol 2019, 63, in Sjogren’s syndrome-associated lymphoproliferation. Arthritis
1–6. Rheum 2013, 65, 1085–96. doi:10.1002/art.37828
151. Lindner JM, Cornacchione V, Sathe A, Be C, Srinivas H, Riquet 166. Tony HP, Roll P, Mei HE, Blumner E, Straka A, Gnuegge L, et
E, et al. Human memory B cells harbor diverse cross-neutralizing al. Combination of B cell biomarkers as independent predictors
antibodies against BK and JC polyomaviruses. Immunity 2019, of response in patients with rheumatoid arthritis treated with
50, 668–76.e5. doi:10.1016/j.immuni.2019.02.003 rituximab. Clin Exp Rheumatol 2015, 33, 887–94.

You might also like