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
This review describes gating strategies to identify B-cell subsets in blood and the changes that occur in these subsets with inflammatory and autoimmune diseases.
Graphical Abstract
Graphical Abstract.
Introduction
A unified and consistent approach to the identification and description of B-cell subsets is essential to ensure reproducibility of scientific studies and that analogous conclusions can be drawn from them. Although many recent advances have been made in B-cell biology, significant areas of uncertainty remain regarding the nature of certain B-cell subsets and their role in homeostasis and inflammatory disease. This is likely to have arisen due to a number of reasons. Firstly, whilst murine models are a valuable tool to understand B-cell biology, there are significant differences between murine and human B-cell subsets which mean that it can be difficult to draw analogous conclusions from them [1–4]. Secondly, the evolution of high dimensional phenotypic techniques has identified many novel subsets but further work is needed to expand on their functional and pathogenic roles [5–8]. Thirdly, B cells in blood often represent a developmental continuum and overlap phenotypically without sharply demarcated gates to differentiate them [5, 9–11]. And finally, there remain significant gaps in our understanding of human B-cell maturation and therefore the developmental trajectories that link human B-cell subsets to one another [12].
B cells are known to play important roles in a variety of inflammatory diseases via the production of antibodies and cytokines as well as antigen presentation [13–20]. Their pathogenic role is supported by the response of a variety of inflammatory conditions to B-cell directed therapies such as rituximab and B-cell activating factor (BAFF) antagonists [21–25]. Immune mediated inflammatory diseases such as systemic lupus erythematosus (SLE), Sjogren’s syndrome (SS), rheumatoid arthritis (RA), and COVID-19 are associated with significant changes in B-cell subset frequency which is likely driven by inflammatory cytokines and pro-survival factors as well as changes in B-cell differentiation and homing [26–28].
In this review we will describe the main subsets of B cells in peripheral blood and how these can be identified using flow and mass cytometry, as well as important changes in B-cell subsets in inflammatory immune mediated conditions. B1 cells will not be discussed in this review due to uncertainties regarding their status and because they represent a rare subset in adult human blood [29–31].
Identification of B cells in peripheral blood
The surface expression of both CD19 and CD20 are induced early in B-cell development in the bone marrow [11, 32]. They are both expressed on B-cell subsets in blood but CD20 expression is lost during terminal B-cell differentiation into antibody secreting cells (ASCs) [33, 34]. CD19 therefore allows identification of both B cells and ASCs. A gating strategy using CD19 for the identification of both B cells and ASCs in blood by flow cytometry is illustrated in Fig. 1A. An antibody panel of 9 ‘key’ markers can be used to identify the main B-cell subsets in blood and these are summarized in Table 1. Additional markers are also given in this table which can aid gating and the identification of further subsets.
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]
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 (% CD19+) | Disease associations |
---|---|---|---|---|
T1 | CD27−IgD+IgM+CD10+ CD21loCD24+++CD38+++ [35–37] |
β1hiβ7loCCR7loCD62Llo [9, 37] | 1−4 [37, 38] | SLE [38, 39] SSc [40] COVID [41, 42] |
T2 | CD27−IgD+IgM+CD10+ CD21hiCD24++CD38++ [35–37] |
β1hi/intCD62Lhi/int IgMhiT2: IgMhiCCR7hiβ7hi [9] IgMloT2: IgMloCCR7loβ7lo [9] |
2−10 [28, 38] | SLE [9, 28, 38, 39] SSc [40] COVID [41, 42] |
T3 | CD27−IgD+IgM+CD10−CD24+CD38+RHhi/MTGhi [43–45] | IgMhiT3: IgMhiCD1chiβ7hiCD45RBint/hi [9, 46] IgMloT3: IgMloCD1cloβ7loCD45RBlo/int [9] |
9−13 [9, 45] | SLE [9, 44] |
Naive | CD27−IgD+IgM+CD10−CD24+CD38+RHlo/MTGlo [43, 44] | 40−60 [9, 45] | SLE [47, 48] SSc [49] |
|
Naïve MZP | CD27−IgD+IgMhiCD10−CD24+CD38+RHlo/MTGloCD45RBhi [46, 50] | CD1chi [9, 46] | 0.5−3 [9, 46] | SLE [9] |
Activated naive | CD27−IgD+CD10−CD21lo CD24loCD38lo [26, 27] | FCRL5+, CXCR5+, T-bet+ [26] | 0.1−3 [9] | SLE [26, 27, 51] COVID [42] |
MZB | CD27+IgD+IgM+ [10] | CD1chi [52] MZB1: β7hiCCR7hi [6] MZB2: β7loCCR7lo [6] |
2−30 [53, 54] | SLE [54, 55] SS [56, 57] 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] DN2: CD21lo, FCRL5+, CXCR5−, T-bet+, CD11chi [26, 41] DN3: CD21−CD11c−CXCR5− [72] DN4: CD21+CD11c+ [73] |
1−10 [26, 71] | SLE [26, 27, 74] RA [75–77] SSc [49] COVID [41, 42, 78, 79] |
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;.
Mass cytometry utilizes the ionic signal from antibodies tagged to rare earth metals and therefore is not limited by the spectral overlap from fluorochrome-conjugated antibodies that are used for flow cytometry [80]. Mass cytometry therefore allows single cells to be labelled with ~40 metal tagged antibodies that can be directed to cell surface or intracellular targets. A gating strategy to identify live single B cells by mass cytometry is displayed in Fig. 1B. Further identification of B-cell subsets can then be performed by biaxial gating or dimensional reduction and clustering methods. Biaxial gating of mass cytometry data can be challenging as the expression of B-cell subset markers often lack distinct pinch point gates and metal minus one control samples that guide the accurate placement of gates are impractical due to cost. Dimensionality reduction and clustering techniques that identify subsets based on the expression of multiple markers can therefore be advantageous over biaxial gating. ViSNE, a visualization tool for high-dimensional single-cell data based on the t-Distributed Stochastic Neighbour Embedding algorithm and uniform manifold approximation and projection (UMAP) are common dimensionality reduction tools used to visualize mass cytometry data [81, 82]. These algorithms can be run using all panel markers or select B-cell subset markers and allow the two-dimensional projection of B cells arranged in multidimensional space. Representative viSNE plots of CD19+ B cells from a healthy control donor (HCD) generated by running the algorithm on lineage markers are displayed in Fig. 1C. In these viSNE plots, memory B cells appear as distinct islands but significant continuity exists between CD10+CD24hiCD38hi transitional and CD10−CD24intCD38int naïve B-cell subsets and manually gating such plots can therefore be challenging (Fig. 1C). For this reason, spanning tree progression of density normalized events (SPADE) run on ViSNE coordinates can be a useful tool to separate subpopulations existing in phenotypic continuity (Fig. 1D) [9, 10]. With SPADE, B-cell populations that are phenotypically similar to one another are displayed as nodes which are interlinked by branches to form trees. Nodes representing B-cell subsets can then be manually grouped together to form bubbles representing that population (Fig. 1D). Whilst this approach is in part directed it has the advantage over biaxial gating in that multiple markers can be used simultaneously to define B-cell subsets.
Transitional B cells
Transitional B cells are defined as recent emigrant B cells from the bone marrow and represent the most immature B-cell subset in peripheral blood [12]. Studies of B-cell repopulation following bone marrow stem cell transplantation and B-cell depletion with rituximab have identified sequential maturation of transitional B cells through T1, T2, and T3 phases [35, 36, 45, 83, 84]. This maturational sequence is associated with the acquisition and loss of various markers. Notably, CD24, CD38, and CD10 are downregulated and these are commonly utilized markers to distinguish transitional B-cell subsets. T1 and T2 B cells share expression of CD10 with immature B cells in the bone marrow, but this is not expressed by T3 cells [37, 45]. T1 and T2 cells are often distinguished from one another based on high and intermediate expression of CD24 and CD38 (Fig. 2A). This approach is subjective as the definition of high and intermediate expression can differ between individuals and experiments given technical factors such as fluorescent signal intensity. T1 cells are known to have low expression of CD21 and the use of this marker in conjunction with CD24 and CD38 can be useful to guide the T1 gate (Fig. 2A) [35].
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
The use of additional markers can also help to accurately identify T1, T2, and T3 subsets and are represented in Fig. 2B and C as SPADE trees. T1 cells have relatively high expression of β1 integrin, which is likely to be important for interaction with bone marrow stromal cells [85]. T1 cells also lack expression of CD62L and CCR7 (Fig. 2C), which is likely to result in a lack of homing capability as seen in studies of both murine and human transitional B cells [9, 86, 87]. IgM was traditionally thought to be downregulated through T1–T3 development but T2 and T3 populations are both heterogeneous in their expression of this marker (Fig. 2D). IgMhi T2 cells have high expression of β7 integrin and are enriched in gut associated lymphoid tissue (Fig. 2E) [9]. Furthermore, IgMhi T2 cells align with cells undergoing marginal zone B (MZB) cell development in terms of surface and transcriptomic features [9]. This suggests that a gut homing T2 MZP population exists in humans as has been demonstrated in mice [88–90].
T3 cells are a late transitional B-cell subset and are thought to represent an intermediate stage of development between T2 and naïve B cells. In keeping with this, T3 cells appear in the blood after T2 cells during B-cell repopulation post B-cell ablation therapy and differentiate into naïve B cells in vitro [44, 45]. Transitional B cells can be distinguished from naïve B cells as they lack expression of the ABCB1 cotransporter, which as a result prevents the extrusion of dyes such as rhodamine (RH) or mitotracker (MTG) [43]. T3 cells can therefore be gated as CD27−IgD+CD10−RHhi/MTGhi (Fig. 3A and B). The distinction between RH low and high populations can be guided by the signal obtained from CD27+ memory B cells which also lack to ABCB1 cotransporter and therefore are RHhi (Fig. 3C). The expression of CD45RB in T3 cell populations is heterogeneous (Fig. 3D) and CD45RBhi cells also have higher expression of IgM and CD1c and align with cells undergoing MZB cell differentiation (Fig. 3E) [9, 46]. The use of dye extrusion is essential for the distinction of T3 cells from naïve B cells as they otherwise share an overlapping surface phenotype (Fig. 3F).
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
Multiple abnormalities in transitional B-cell subsets have been reported in inflammatory diseases. Transitional B cells harbour a high proportion of autoreactive cells which are removed through sequential maturation into naïve B cells in healthy individuals and this checkpoint is defective in SLE [91]. The mechanisms that underlie this checkpoint failure are not known but are may be secondary to aberrant transitional B-cell maturation or migration into lymphoid tissue. Transitional B cells from patients with SLE have been reported to have lower expression of the gut homing receptor β7 integrin suggestive of defective gut homing capacity and β7hi T2 cells are depleted in lupus nephritis [9, 39]. Transitional B cells have been shown to be increased in frequency in SLE and this has been correlated with serum BAFF levels [28, 38, 92]. The important role of BAFF as a survival factor is also supported by the increased frequency of transitional B cells in patients with multiple sclerosis that are treated with fingolimod and interferon-β which are both known to increase serum BAFF levels [93]. Transitional B cells in SLE have also been shown to be hyperresponsive to B-cell receptor (BCR) signalling which is likely secondary to type I interferon [94, 95]. Increased frequencies of transitional B cells have also been reported in a number of other autoimmune diseases such as juvenile dermatomyositis, primary SS (pSS), and systemic sclerosis (SSc) [40, 96, 97]. Altered T1:T2 cell ratios have also been identified in patients with renal transplant rejection and are postulated to be an important predictor of graft loss [98, 99]. Finally, a reduction in transitional B cells has been seen in COVID-19 infection which correlates with infection severity [41, 42].
Transitional B cells are enriched in B regulatory cells (Bregs) that are best described as B cells that can suppress T-cell responses mainly via the production of IL-10 [100, 101]. Breg dysfunction has been identified in a number of autoimmune diseases. CD24hiCD38hi B cells in SLE have less capacity to inhibit CD4 T-cell cytokine production [100]. Induction of Bregs by plasmacytoid dendritic cells has also been reported to be defective in SLE which is thought to be mediated by type I interferon [102]. Breg dysfunction in SLE may also be mediated by defective B cell homing or maturation in GALT, which is known to be important for Breg induction [9, 39, 103]. Reduced numbers of CD24hiCD38hi B cells have been identified in patients with RA and they failed to repress Th17 responses but maintained their ability to suppress Th1 responses [104]. Furthermore, treatment of RA with tumour necrosis factor alpha (TNF-α) antagonists has been demonstrated to increase the number of CD27+ Bregs [105]. Primary SS has been associated with increased frequency of CD24hiCD38hi B cells but these cells fail to suppress T cell TNF-α or IFN-γ secretion [97]. Furthermore, adoptive transfer of CD24hiCD38hi B cells from patients with pSS into mice with experimental SS demonstrated failure of suppression of T follicular cell expansion [106]. Reduced Breg frequency or defective Breg function has also been reported in MS, SSc, pemphigus, and myasthenia gravis [107–111]. Breg dysfunction has therefore been identified in a number of autoimmune diseases but further work is required to identify the pathogenic significance and the mechanisms that underlie this.
Naïve B cells
Naïve B cells represent B cells that have not encountered antigen and have not undergone germinal centre maturation. They therefore lack CD27 expression and have a low level of somatic mutations within their immunoglobulin variable (IgV) genes [63]. Naïve B cells represent 40–60% of peripheral blood B cells and as discussed above can only be distinguished from T3 cells by their ability to extrude RH or MTG dyes due to their expression of the ABCB1 cotransporter [43]. Naïve B cells can therefore be gated as CD27−IgD+CD10−RHloCD45RBlo (Fig. 3A). Distinction by dye extrusion is not possible using mass cytometry although a metal tagged antibody to ABCB1 would theoretically allow the distinction between T3 and naïve B cell, to our knowledge 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 precursor cells (MZP), activated naïve (aNAV), and an IgMlo anergic subset [26, 46, 47]. Naïve MZP cells are characterized by high expression of CD45RB and also highly express IgM and CD1c have been shown to undergo differentiation into MZB like B cells following NOTCH ligation (Fig. 3G) [46]. A T3 population with a similar surface phenotype has also been identified and is thought to be in developmental continuity with naïve MZP cells [9, 30]. Naïve B cells with high-surface expression of CD45RB has been described as an early memory population and found to have more somatic mutations than CD45RB− naïve B cells [5]. The same study commented on an anergic profile of naïve B cells as evidenced by lower expression of intracellular transport proteins suggesting that this subset would be less responsive to stimulation. Other functional studies have, however, demonstrated robust proliferative responses of naïve B cells to IgM crosslinking as well as CD40 and TLR9 agonists [37, 44, 45, 112].
Naïve B cells with low IgM expression have been reported as having anergic properties as evidence by lower intracellular calcium influxes and expression of activation markers following IgM crosslinking [47]. This subset was also found to express higher levels of the inhibitory receptor CD22. The identity of anergic B cells as IgMloIgD+ is supported by a number of murine studies using the hen egg lysozyme model in which the majority of B cells recognize this epitope. When HEL is expression was induced endogenously the majority of B cells adopted this phenotype, suggesting anergy can be induced, and is a key tolerance mechanism [113–115].
ANAV are a subset of CD27−IgD+ naïve cells with low expression of CD21, CD23, CD24, and CD38 but high expression of FCRL5, CD11c, and T-bet [26, 27] (Fig. 3H). This subset also does not express the ABCB1 cotransporter and is therefore RH or MTG high following dye extrusion assays [116]. ANAV were first identified in SLE as being clonally related to ASC populations and are hyperresponsive to TLR7 stimulation [26, 27]. The aNAV subset is therefore thought to undergo extrafollicular maturation into ASC in SLE and are primed by interferon and TLR7 agonists.
Inflammatory disorders in which extrafollicular B-cell maturation is thought to occur include severe SLE and COVID-19 and are associated with an increase in aNAV frequency alongside increased frequencies of ASCs [26, 41, 51]. Conversely, MZP and naïve B cells are reduced in severe SLE [9]. Furthermore, inhibition of BAFF with belimumab results in a lower frequency of naïve B cells, suggesting the importance of BAFF for their survival [117, 118]. Of note, numerous studies of B-cell subset abnormalities in inflammatory disease have gated naïve B cells as solely CD27− or CD27−IgD+ and therefore included double negative (DN) and transitional B cells, respectively in these gates [119–122].
MZB cells
The splenic marginal zone refers to a microanatomical area on the periphery of B-cell follicles in the white pulp [123]. MZB cells are termed as such as they share similar surface phenotype to the subset of cells found in the splenic marginal zone that express low levels of IgD and high levels of CD1c [52, 124]. In mice, MZB cells are confined to the spleen and do not have IgV gene mutations and therefore arise independently of germinal centre responses. In humans, MZB cells circulate in blood and lymphoid tissue and have IgV gene mutations, although to a lower degree when compared to class-switched memory (CSM) B cells [10, 52, 63, 125]. The finding that MZB have IgV mutations in patients with CD40L deficiency who lack germinal centres suggest they may mature independent of germinal centre responses as in mice [64, 125]. This is supported by studies that demonstrate little overlap between MZB and CSM B-cell clones, suggesting that MZB represent a distinct entity rather than just a precursor to CSM [10, 61]. Germinal centre independent maturation of MZB is, however, challenged by the finding that 20% of blood MZB carry BCL6 gene mutations and that human MZB have been reported to share clones with GC delivered IgG memory B cells [126–128]. Similarly, whilst a specific MZB maturational pathway exists in mice, the developmental origins of human MZB remain incompletely understood. As described above, human transitional and naïve MZP have an IgMhi gut homing phenotype suggestive of a role of GALT in their induction [9]. Furthermore, MZB are observed to occupy a peri-germinal centre niche and to diversify their BCR within GALT [10]. Functionally, MZB undergo T cell independent responses and are important for immunity against encapsulated bacteria [52, 129].
Uncertainty regarding the developmental origin and functional status of MZB cells is reflected by the plethora of names assigned to this subset in the literature. These include unswitched memory [56, 130, 131], pre-switched memory [58, 119, 132], non-switched [133, 134], natural effector [64, 135], and IgM memory [136, 137] B cells. MZB have a CD27+IgD+IgM+ surface phenotype and have high expression of CD1c, CD21, and CD45RB (Fig. 4A) [63]. MZB have also been reported to have low levels of CD23 and high expression of the inhibitory receptor CD32b [138, 139]. Recently, two distinct populations of MZB have been described that differ in cell surface properties, transcriptional factors, and distribution in lymphoid tissues [6]. These have been termed MZB1 and MZB2, MZB1 having higher expression of β7 integrin, CCR7, CD24, and CD27 than MZB2 (Fig. 4B and C). MZB1 and MZB2 were not found to be clonally related and NOTCH induced genes were only upregulated in MZB2 suggesting independent maturational pathways.
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]
Significant changes in MZB frequency have been reported in association with inflammatory diseases. As seen with MZP subsets, severe depletion of MZB is observed in severe SLE and this predominantly affects the MZB1 subset (Fig. 4D) [6, 9]. As well as SLE, reduced frequencies of MZB have also been reported in SS, SSc, and COVID-19 [9, 55, 56, 58–60, 135]. The factors that result in this depletion in blood are not understood and could represent reduced cell survival or genesis, altered tissue distribution or increased differentiation into ASCs. The depletion of MZB alongside their putative precursors in severe SLE suggests defective MZB differentiation. The reciprocal reduction of MZB cells and the increase of ASCs in SLE could also support their differentiation into short-lived ASCs that are characteristic of SLE disease flares [27]. Also, as MZB depletion in COVID and SLE has been shown to be reversible following resolution of the infection and with treatment respectively, inflammatory factors can result in significant plasticity in this population [55, 135]. The functional significance of depletion in MZB is unknown but could underly the increased susceptibility of patients to infection with encapsulated bacteria or defective B regulatory responses [140, 141].
CD27+ memory and IgM only B cells
Memory B cells are formed from mature naïve B cells that have encountered antigen and undergone T dependent immune responses and germinal centre maturation [2, 142]. They can be distinguished from naïve B cells by a much higher degree of IgV gene mutations, the gain of expression of CD27, and the loss of IgD expression [1, 63, 128, 143, 144]. Rounds of proliferation and diversification within germinal centres create clones of memory B cells that produce high-affinity immunoglobulin and differentiate into long-lived ASCs [145–147]. Functionally, memory B cells proliferate rapidly in response to stimulation with CD40L and interleukin (IL) 2 and IL21 [148, 149].
Distinct subsets of CD27+IgD− memory B cells can be identified by the expression of IgM, IgA, or IgG (Fig. 5A). CD27+ memory cells also share high expression of CD24 and CD45RB and relatively low expression of CD38 (Fig. 5B). CD27 and CD38 are also important in distinguishing memory B cells from ASCs (Fig. 5C). IgM only B cells are CD27+IgD−IgM+ and have been identified to have a higher frequency of IgV gene mutations than MZB cells and to share clones with CSM B cells, suggesting that these cells are GC derived [10, 61]. The functional differences of human IgM only and CSM B cells in terms of recall responses and longevity are not well understood in humans and other than IgM expression they share a similar surface phenotype (Fig. 5D) [150]. Of interest is a recent study of BK polyoma virus responses demonstrated different antiviral specificities of IgM and IgG memory B cells which suggests a distinct function of IgM memory B cells [151]. CSM B cells consist of IgA+, IgG+, and IgE+ subsets although the latter is a rare subset in peripheral blood. CSM have a high frequency of IgV gene mutations and differentiate into ASCs that produce high-affinity immunoglobulin that are essential for humoral immunity and the basis of effective vaccine responses [127]. Heterogeneity within memory B-cell populations has been identified through differential expression of CD73 and CD95. Memory B cells can be CD73 positive or negative, its expression is inversely correlated with IgM and is mainly seen in CSM B cells [5]. CD95 or Fas is a pro-apoptotic receptor which is mainly expressed by CSM, ASCs, and CD27− memory B cells which is elevated in SLE and RA and thought to indicate an activated memory B-cell phenotype [5, 152–154].
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
Memory B cells play a critical role in autoimmune, inflammatory and allergic diseases as they can differentiate into ASCs that produce pathogenic autoantibodies. Autoreactive memory B-cell clones arise due to checkpoint failure, where autoreactive naïve B cells are allowed to undergo germinal centre or extrafollicular responses. This checkpoint has been shown to be defective in a variety of diseases including SLE and SS, although the mechanisms that result in this are still unclear [57, 155]. B cells in SLE are known to express greater levels of CD40 and blockade of germinal centre responses with CD40L (CD154) antagonists has therapeutic promise [156–158]. Variable frequencies of CSM B cells have been observed in SLE which is likely to represent heterogeneity in disease activity, duration, and treatment [54, 65, 66]. Interestingly, unlike naïve and MZB subsets, CSM frequency is not affected by BAFF inhibition with belimumab [118]. Variable frequencies of CSM B cells have also been observed in RA with a lower proportion of CXCR3+CD27+ cells which correlated inversely with disease activity [67, 68]. Increased frequency of switched memory B cells have also been observed in other autoimmune conditions including ANCA vasculitis [69]. B-cell depleting agents such as rituximab have long-lasting effects of memory B cells, which may underlie their efficacy in treating autoimmune disease [159].
DN memory B cells
CD27− memory B cells were first identified as IgG+ cells in blood with a lower level of somatic IgV gene mutations than CSM [70]. DN B cells expressing IgM, IgA, and IgA were then identified and found to be expanded in SLE [71]. They are termed DN B cells as they lack both CD27 and IgD expression. Their accumulation in chronic infections, autoimmune diseases and with advanced age has also led to them being termed exhausted memory B cells and age associated B cells (ABC) [160–162]. ABC share features of DN cells such as upregulation of T-bet and low expression of CD21 but have been described using a variety of markers and likely represent a heterogeneous population which are not entirely synonymous with DN cells [34, 162].
Since their first description a number of DN B-cell subsets have been identified. Initially DN B cells were split into DN1 (CD21+CXCR5+) and DN2 B cells (CD21−CXCR5−) [26]. Similar to aNAV, DN2 also have low expression of CD24 (Fig. 6A). DN2 cells were found to be a rare subset in health but are found in much higher frequencies in SLE [26]. DN2 B cells also highly express CD11c as well as the transcription factor T-bet highly express CD1c and FCRL5 as well as the transcription factor T-bet (Fig. 6B and C) [26, 116]. Functionally, DN2 cells are hyperresponsive to TLR7 signalling and can undergo rapid ASC differentiation in response to IL21 and IFN-γ and are therefore felt to be pivotal in extrafollicular B-cell responses [116, 163]. DN2 cells have been identified to be clonally related to aNAV B cells and ASC with a lower level of IgV gene mutations and are enriched in autoreactive cells [27]. More recently DN3 and DN4 subsets have been reported, the DN3 cluster being CD11c−CXCR5− and the DN4 subset being enriched in IgE expressing cells and to highly express transcripts for IL4R and CD40 [72, 73].
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])
SLE disease flares are associated with dramatic increases in ASCs that secrete pathogenic autoantibodies that are derived from extrafollicular B-cell responses [27]. This is associated with expansion of aNAV and DN2 B-cell populations (Fig. 6D) which are proposed to differentiate into ASCs via extrafollicular B-cell responses [116]. DN2 B cells are more frequent in SLE patients of African–American ethnicity and with lupus nephritis in whom they have been identified within the nephrotic kidney [26, 163]. Extrafollicular B-cell maturation has been correlated with the severity of COVID-19 and similar to severe SLE, increased frequencies of aNAV, DN2 cells are observed alongside depletion of MZB cells [9, 41, 60, 78, 116]. The correlation between the severity of infection and B-cell subset abnormalities suggests a pivotal role of cytokines such as Type II interferon. CD11c positive B cells have been reported in patients with MS, RA, and SSc and to accumulate with age in female patients with RA [164]. Autoreactive CD27−CD19hiCD21lo B cells akin to DN2 cells have also been reported in SS [165]. Increased frequency of DN B cells has also been correlated with improved clinical responses to rituximab of patients with RA whilst reduced numbers have been reported to predict response to IL-6 inhibition with tocilizumab [77, 166]. Further work is needed to investigate the clinical significance of these findings. DN cells may be of pathogenic significance through the production of inflammatory cytokines or differentiation into ASCs. Alternatively, altered DN cell frequency may be indicative of certain inflammatory environments that affect treatment outcomes.
Conclusion
The evolution of high dimensional phenotypic tools has greatly advanced our understanding of B-cell subsets in blood and the surface markers and transcriptomic properties that define them. However, significant gaps remain in our understanding of the functional roles of these subsets, their developmental origins and pathogenic roles in disease. By addressing these areas of uncertainty, we will advance our knowledge of human B-cell development and the role these cells play in normal homeostasis and in the pathogenesis of autoimmune and inflammatory disease.
Materials and methods for data used in figures and figure generation
Experimental subjects
Blood samples were donated by adult HCD and patients with SLE with regional ethics committee (REC) approval and informed consent (REC reference 11/LO/1433).
Isolation and storage of PBMCs
PBMCs were obtained from whole blood by Ficoll-Hypaque density gradient centrifugation as previously described [9]. Isolated PBMCs were then centrifuged and cryopreserved in ice-cold freezing medium (FCS with 10% DMSO) to give a cell concentration of 5–10 × 106 cells/ml.
Mass cytometry staining and analysis
The staining protocol and antibody panel for the mass cytometry data displayed in Figs. 1 and 2 is described by Tull TJ et al. [9]. FCS files were normalized using Nolan Laboratory Software (https://github.com/nolanlab/beadnormalization). Single live B cells from 10 HCD (5 male and 5 female, mean age 31.8 years) are gated as displayed in Fig. 1B. ViSNE plots displayed in Fig. 1C are generated using Cytobank software (https://mrc.cytobank.org/) using equal numbers of B cells (n = 35 000) from 10 HCD. SPADE was then run on the viSNE coordinates and nodes corresponding to B-cell subsets are grouped as displayed in Fig. 1D. Transitional B cells were identified as CD27−IgD+CD24+++/++CD38+++/++. Events within this transitional cell bubble were then exported and a further viSNE run using equal events (n = 3535) and all panel markers except CD45, CD3, CD14, and class-switched isotypes IgA and IgG, which are not expressed by transitional B cells. The SPADE plots in Fig. 1C are generated from these viSNE coordinates. SPADE on viSNE plots in Fig. 4C are generated by exporting events within the MZB SPADE bubble in Fig. 1D and re-clustered using CD45RB, IgD, CD21, integrin beta 7, CD27, CD24, IgM, HLA-DR, and CCR7. The DN SPADE on viSNE was generated using data from 8 HCD and 8 patients with active severe SLE as described by Tull TJ et al. [9]. CD19+ cells were gated and SPADE on viSNE plots created as in Fig. 1D. Events within the DN SPADE bubble were then exported and DN nodes were re-clustered using CD11c, CD21, CXCR5, and CD24 to identify DN subsets.
Flow cytometry staining and analysis
Cryopreserved PBMCs were stained with the fluorochrome-conjugated antibodies as previously described [9]. The following antibodies clones were used: CD27 APC (M-T271), IgD APC-Cy7 (IA6-2), CD19 PerCP-Cy5.5 (HIB19), CD10 BV421 (HI10a), CD24 BV605 (ML5), IgM BV711 (MHM-88), CD1c PE (L161), and CD45RB PE-TexasRed (MEM-55). CD21 BUV737 (HB7) and CD38 BUV395 (B-ly4). DAPI was used for live dead staining. Flow cytometry data were obtained by running samples on a Fortessa flow cytometer. FCS files obtained were analysed using FlowJo software.
Acknowledgements
Ms Louise Nel, Prof. David D’Cruz, and Dr Mike Robson for sample collection. The Guy’s and St Thomas’ NHS Trust BRC flow and mass cytometry cores for sample processing and data generation.
Glossary
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
Contributor Information
Rebekah L Velounias, Department of Immunobiology, King’s College London, Guy’s Hospital Campus, London, UK.
Thomas J Tull, St John’s Institute of Dermatology, King’s College London, Guy’s Hospital Campus, London, UK.
Conflict of interest
None declared.
Funding
This work was funded by the Medical Research Council of Great Britain (MR/R000964/1) and the St Thomas’ Lupus Trust.
Data availability
Raw mass cytometry data can be requested from the corresponding author.
Author contributions
T.J.T.: Manuscript generation and figure review. R.L.V.: Figure generation and manuscript review.
Permission to reproduce
Granted within terms set out by the licensing agreement.
Clinical trial registration
Not applicable.
The animal research adheres to the ARRIVE guidelines
Not applicable.
References
- 1. Weisel NM, Joachim SM, Smita S, Callahan D, Elsner RA, Conter LJ, et al. Surface phenotypes of naive and memory B cells in mouse and human tissues. Nat Immunol 2022, 23, 135–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Cancro MP, Tomayko MM.. Memory B cells and plasma cells: the differentiative continuum of humoral immunity. Immunol Rev 2021, 303, 72–82. doi: 10.1111/imr.13016 [DOI] [PubMed] [Google Scholar]
- 3. Benitez A, Weldon AJ, Tatosyan L, Velkuru V, Lee S, Milford TA, et al. Differences in mouse and human nonmemory B cell pools. J Immunol 2014, 192, 4610–9. doi: 10.4049/jimmunol.1300692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Reynaud CA, Descatoire M, Dogan I, Huetz F, Weller S, Weill JC.. IgM memory B cells: a mouse/human paradox. Cell Mol Life Sci 2012, 69, 1625–34. doi: 10.1007/s00018-012-0971-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Glass DR, Tsai AG, Oliveria JP, Hartmann FJ, Kimmey SC, Calderon AA, et al. An integrated multi-omic single-cell atlas of human B cell identity. Immunity 2020, 53, 217–32.e5. doi: 10.1016/j.immuni.2020.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Siu JHY, Pitcher MJ, Tull TJ, Velounias RL, Guesdon W, Montorsi L, et al. Two subsets of human marginal zone B cells resolved by global analysis of lymphoid tissues and blood. Sci Immunol 2022, 7, eabm9060. [DOI] [PubMed] [Google Scholar]
- 7. Zhang F, Wei K, Slowikowski K, Fonseka CY, Rao DA, Kelly S, et al. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat Immunol 2019, 20, 928–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Papalexi E, Satija R.. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol 2018, 18, 35–45. [DOI] [PubMed] [Google Scholar]
- 9. Tull TJ, Pitcher MJ, Guesdon W, Siu JHY, Lebrero-Fernandez C, Zhao Y, et al. Human marginal zone B cell development from early T2 progenitors. J Exp Med 2021, 218(4), e20202001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zhao Y, Uduman M, Siu JHY, Tull TJ, Sanderson JD, Wu YB, et al. Spatiotemporal segregation of human marginal zone and memory B cell populations in lymphoid tissue. Nat Commun 2018, 9, 3857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Bendall SC, Davis KL, Amir el AD, Tadmor MD, Simonds EF, Chen TJ, et al. Single-cell trajectory detection uncovers progression and regulatory coordination in human B cell development. Cell 2014, 157, 714–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Bemark M. Translating transitions - how to decipher peripheral human B cell development. J Biomed Res 2015, 29, 264–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Bao Y, Cao X.. The immune potential and immunopathology of cytokine-producing B cell subsets: a comprehensive review. J Autoimmun 2014, 55, 10–23. doi: 10.1016/j.jaut.2014.04.001 [DOI] [PubMed] [Google Scholar]
- 14. Duddy ME, Alter A, Bar-Or A.. Distinct profiles of human B cell effector cytokines: a role in immune regulation?. J Immunol 2004, 172, 3422–7. doi: 10.4049/jimmunol.172.6.3422 [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PubMed] [Google Scholar]
- 16. Canny SP, Jackson SW.. B cells in systemic lupus erythematosus: from disease mechanisms to targeted therapies. Rheum Dis Clin North Am 2021, 47, 395–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 [DOI] [PubMed] [Google Scholar]
- 18. Rodriguez-Pinto D. B cells as antigen presenting cells. Cell Immunol 2005, 238, 67–75. [DOI] [PubMed] [Google Scholar]
- 19. Chong AS. B cells as antigen-presenting cells in transplantation rejection and tolerance. Cell Immunol 2020, 349, 104061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Xiao ZX, Miller JS, Zheng SG.. An updated advance of autoantibodies in autoimmune diseases. Autoimmun Rev 2021, 20, 102743. doi: 10.1016/j.autrev.2020.102743 [DOI] [PubMed] [Google Scholar]
- 21. Hassan RI, Gaffo AL.. Rituximab in ANCA-associated vasculitis. Curr Rheumatol Rep 2017, 19, 6. [DOI] [PubMed] [Google Scholar]
- 22. Tandan R, Hehir MK 2nd, Waheed W, Howard DB.. Rituximab treatment of myasthenia gravis: a systematic review. Muscle Nerve 2017, 56, 185–96. doi: 10.1002/mus.25597 [DOI] [PubMed] [Google Scholar]
- 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Wise LM, Stohl W.. Belimumab and rituximab in systemic lupus erythematosus: a tale of two B cell-targeting agents. Front Med 2020, 7, 303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Alvarez-Rivas N, Sang-Park H, Diaz Del Campo P, Fernandez-Castro M, Corominas H, Andreu JL, et al. Efficacy of belimumab in Primary Sjogren’s syndrome: a systematic review. Reumatol Clin (Engl Ed) 2021, 17, 170–4. [DOI] [PubMed] [Google Scholar]
- 26. Jenks SA, Cashman KS, Zumaquero E, Marigorta UM, Patel AV, Wang X, et al. Distinct effector B cells induced by unregulated toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus. Immunity 2018, 49, 725–39 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Tipton CM, Fucile CF, Darce J, Chida A, Ichikawa T, Gregoretti I, et al. Diversity, cellular origin and autoreactivity of antibody-secreting cell population expansions in acute systemic lupus erythematosus. Nat Immunol 2015, 16, 755–65. doi: 10.1038/ni.3175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Landolt-Marticorena C, Wither R, Reich H, Herzenberg A, Scholey J, Gladman DD, et al. Increased expression of B cell activation factor supports the abnormal expansion of transitional B cells in systemic lupus erythematosus. J Rheumatol 2011, 38, 642–51. doi: 10.3899/jrheum.100214 [DOI] [PubMed] [Google Scholar]
- 29. Perez-Andres M, Grosserichter-Wagener C, Teodosio C, van Dongen JJ, Orfao A, van Zelm MC.. The nature of circulating CD27+CD43+ B cells. J Exp Med 2011, 208, 2565–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Descatoire M, Weill JC, Reynaud CA, Weller S.. A human equivalent of mouse B-1 cells?. J Exp Med 2011, 208, 2563–4. doi: 10.1084/jem.20112232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Suo C, Dann E, Goh I, Jardine L, Kleshchevnikov V, Park JE, et al. Mapping the developing human immune system across organs. Science 2022, 376, eabo0510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Scheuermann RH, Racila E.. CD19 antigen in leukemia and lymphoma diagnosis and immunotherapy. Leuk Lymphoma 1995, 18, 385–97. doi: 10.3109/10428199509059636 [DOI] [PubMed] [Google Scholar]
- 33. Mahevas M, Patin P, Huetz F, Descatoire M, Cagnard N, Bole-Feysot C, et al. B cell depletion in immune thrombocytopenia reveals splenic long-lived plasma cells. J Clin Invest 2013, 123, 432–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Sanz I, Wei C, Jenks SA, Cashman KS, Tipton C, Woodruff MC, et al. Challenges and opportunities for consistent classification of human B cell and plasma cell populations. Front Immunol 2019, 10, 2458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Suryani S, Fulcher DA, Santner-Nanan B, Nanan R, Wong M, Shaw PJ, et al. Differential expression of CD21 identifies developmentally and functionally distinct subsets of human transitional B cells. Blood 2010, 115, 519–29. doi: 10.1182/blood-2009-07-234799 [DOI] [PubMed] [Google Scholar]
- 36. Anolik JH, Friedberg JW, Zheng B, Barnard J, Owen T, Cushing E, et al. B cell reconstitution after rituximab treatment of lymphoma recapitulates B cell ontogeny. Clin Immunol 2007, 122, 139–45. [DOI] [PubMed] [Google Scholar]
- 37. Sims GP, Ettinger R, Shirota Y, Yarboro CH, Illei GG, Lipsky PE.. Identification and characterization of circulating human transitional B cells. Blood 2005, 105, 4390–8. doi: 10.1182/blood-2004-11-4284 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Dieudonne Y, Gies V, Guffroy A, Keime C, Bird AK, Liesveld J, et al. Transitional B cells in quiescent SLE: an early checkpoint imprinted by IFN. J Autoimmun 2019, 102, 150–8. doi: 10.1016/j.jaut.2019.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Vossenkamper A, Blair PA, Safinia N, Fraser LD, Das L, Sanders TJ, et al. A role for gut-associated lymphoid tissue in shaping the human B cell repertoire. J Exp Med 2013, 210, 1665–74. doi: 10.1084/jem.20122465 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Taher TE, Ong VH, Bystrom J, Hillion S, Simon Q, Denton CP, et al. Association of defective regulation of autoreactive interleukin-6-producing transitional B lymphocytes with disease in patients with systemic sclerosis. Arthritis Rheumatol 2018, 70, 450–61. doi: 10.1002/art.40390 [DOI] [PubMed] [Google Scholar]
- 41. Woodruff M, Ramonell R, Cashman K, Nguyen D, Ley A, Kyu S, et al. Critically ill SARS-CoV-2 patients display lupus-like hallmarks of extrafollicular B cell activation. medRxiv 2020, preprint: not peer reviewed. doi: 10.1101/2020.04.29.20083717 [DOI] [Google Scholar]
- 42. Sosa-Hernandez VA, Torres-Ruiz J, Cervantes-Diaz R, Romero-Ramirez S, Paez-Franco JC, Meza-Sanchez DE, et al. B cell subsets as severity-associated signatures in COVID-19 patients. Front Immunol 2020, 11, 611004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Wirths S, Lanzavecchia A.. ABCB1 transporter discriminates human resting naive B cells from cycling transitional and memory B cells. Eur J Immunol 2005, 35, 3433–41. doi: 10.1002/eji.200535364 [DOI] [PubMed] [Google Scholar]
- 44. Lee J, Kuchen S, Fischer R, Chang S, Lipsky PE.. Identification and characterization of a human CD5+ pre-naive B cell population. J Immunol 2009, 182, 4116–26. doi: 10.4049/jimmunol.0803391 [DOI] [PubMed] [Google Scholar]
- 45. Palanichamy A, Barnard J, Zheng B, Owen T, Quach T, Wei C, et al. Novel human transitional B cell populations revealed by B cell depletion therapy. J Immunol 2009, 182, 5982–93. doi: 10.4049/jimmunol.0801859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Descatoire M, Weller S, Irtan S, Sarnacki S, Feuillard J, Storck S, et al. Identification of a human splenic marginal zone B cell precursor with NOTCH2-dependent differentiation properties. J Exp Med 2014, 211, 987–1000. doi: 10.1084/jem.20132203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Quach TD, Manjarrez-Orduno N, Adlowitz DG, Silver L, Yang H, Wei C, et al. Anergic responses characterize a large fraction of human autoreactive naive B cells expressing low levels of surface IgM. J Immunol 2011, 186, 4640–8. doi: 10.4049/jimmunol.1001946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Odendahl M, Jacobi A, Hansen A, Feist E, Hiepe F, Burmester GR, et al. Disturbed peripheral B lymphocyte homeostasis in systemic lupus erythematosus. J Immunol 2000, 165, 5970–9. doi: 10.4049/jimmunol.165.10.5970 [DOI] [PubMed] [Google Scholar]
- 49. Simon D, Balogh P, Bognar A, Kellermayer Z, Engelmann P, Nemeth P, et al. Reduced non-switched memory B cell subsets cause imbalance in B cell repertoire in systemic sclerosis. Clin Exp Rheumatol 2016, 34, 30–6. [PubMed] [Google Scholar]
- 50. Bemark M, Friskopp L, Saghafian-Hedengren S, Koethe S, Fasth A, Abrahamsson J, et al. A glycosylation-dependent CD45RB epitope defines previously unacknowledged CD27(-)IgM(high) B cell subpopulations enriched in young children and after hematopoietic stem cell transplantation. Clin Immunol 2013, 149, 421–31. [DOI] [PubMed] [Google Scholar]
- 51. Wangriatisak K, Thanadetsuntorn C, Krittayapoositpot T, Leepiyasakulchai C, Suangtamai T, Ngamjanyaporn P, et al. The expansion of activated naive DNA autoreactive B cells and its association with disease activity in systemic lupus erythematosus patients. Arthritis Res Ther 2021, 23, 179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Weller S, Braun MC, Tan BK, Rosenwald A, Cordier C, Conley ME, et al. Human blood IgM “memory” B cells are circulating splenic marginal zone B cells harboring a prediversified immunoglobulin repertoire. Blood 2004, 104, 3647–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Feng R, Zhao J, Sun F, Miao M, Sun X, He J, et al. Comparison of the deep immune profiling of B cell subsets between healthy adults and Sjogren’s syndrome. Ann Med 2022, 54, 472–83. doi: 10.1080/07853890.2022.2031272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Zhu L, Yin Z, Ju B, Zhang J, Wang Y, Lv X, et al. Altered frequencies of memory B cells in new-onset systemic lupus erythematosus patients. Clin Rheumatol 2018, 37, 205–12. [DOI] [PubMed] [Google Scholar]
- 55. Zhang W, Wang YF, Hu FL, Lu FA, Wu T, Feng YL, et al. Dysfunction of CD27(+)IgD(+) B cells correlates with aggravated systemic lupus erythematosus. Clin Rheumatol 2022, 41, 1551–9. doi: 10.1007/s10067-022-06051-z [DOI] [PubMed] [Google Scholar]
- 56. Roberts ME, Kaminski D, Jenks SA, Maguire C, Ching K, Burbelo PD, et al. Primary Sjogren’s syndrome is characterized by distinct phenotypic and transcriptional profiles of IgD+ unswitched memory B cells. Arthritis Rheumatol 2014, 66, 2558–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Corsiero E, Sutcliffe N, Pitzalis C, Bombardieri M.. Accumulation of self-reactive naive and memory B cell reveals sequential defects in B cell tolerance checkpoints in Sjogren’s syndrome. PLoS One 2014, 9, e114575e114575. doi: 10.1371/journal.pone.0114575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Gernert M, Tony HP, Schwaneck EC, Gadeholt O, Frohlich M, Portegys J, et al. Lymphocyte subsets in the peripheral blood are disturbed in systemic sclerosis patients and can be changed by immunosuppressive medication. Rheumatol Int 2022, 42(8), 1373–81. doi: 10.1007/s00296-021-05034-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Hu F, Zhang W, Shi L, Liu X, Jia Y, Xu L, et al. Impaired CD27(+)IgD(+) B cells with altered gene signature in rheumatoid arthritis. Front Immunol 2018, 9, 626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Laing AG, Lorenc A, Del Molino Del Barrio I, Das A, Fish M, Monin L, et al. A dynamic COVID-19 immune signature includes associations with poor prognosis. Nat Med 2020, 26(10), 1623–35. doi: 10.1038/s41591-020-1038-6 [DOI] [PubMed] [Google Scholar]
- 61. Bagnara D, Squillario M, Kipling D, Mora T, Walczak AM, Da Silva L, et al. A reassessment of IgM memory subsets in humans. J Immunol 2015, 195, 3716–24. doi: 10.4049/jimmunol.1500753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Klein U, Kuppers R, Rajewsky K.. Evidence for a large compartment of IgM-expressing memory B cells in humans. Blood 1997, 89, 1288–98. [PubMed] [Google Scholar]
- 63. Klein U, Rajewsky K, Kuppers R.. Human immunoglobulin (Ig)M+IgD+ peripheral blood B cells expressing the CD27 cell surface antigen carry somatically mutated variable region genes: CD27 as a general marker for somatically mutated (memory) B cells. J Exp Med 1998, 188, 1679–89. doi: 10.1084/jem.188.9.1679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Berkowska MA, Driessen GJ, Bikos V, Grosserichter-Wagener C, Stamatopoulos K, Cerutti A, et al. Human memory B cells originate from three distinct germinal center-dependent and -independent maturation pathways. Blood 2011, 118, 2150–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Korganow AS, Knapp AM, Nehme-Schuster H, Soulas-Sprauel P, Poindron V, Pasquali JL, et al. Peripheral B cell abnormalities in patients with systemic lupus erythematosus in quiescent phase: decreased memory B cells and membrane CD19 expression. J Autoimmun 2010, 34, 426–34. doi: 10.1016/j.jaut.2009.11.002 [DOI] [PubMed] [Google Scholar]
- 66. Szabo K, Papp G, Szanto A, Tarr T, Zeher M.. A comprehensive investigation on the distribution of circulating follicular T helper cells and B cell subsets in primary Sjogren’s syndrome and systemic lupus erythematosus. Clin Exp Immunol 2016, 183, 76–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Souto-Carneiro MM, Mahadevan V, Takada K, Fritsch-Stork R, Nanki T, Brown M, et al. Alterations in peripheral blood memory B cells in patients with active rheumatoid arthritis are dependent on the action of tumour necrosis factor. Arthritis Res Ther 2009, 11, R84. doi: 10.1186/ar2718 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Sellam J, Rouanet S, Hendel-Chavez H, Abbed K, Sibilia J, Tebib J, et al. Blood memory B cells are disturbed and predict the response to rituximab in patients with rheumatoid arthritis. Arthritis Rheum 2011, 63, 3692–701. doi: 10.1002/art.30599 [DOI] [PubMed] [Google Scholar]
- 69. Elmer E, Smargianaki S, Pettersson A, Skattum L, Ohlsson S, Hellmark T, et al. Increased frequencies of switched memory B cells and plasmablasts in peripheral blood from patients with ANCA-associated vasculitis. J Immunol Res 2020, 2020, 8209737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Fecteau JF, Cote G, Neron S.. A new memory CD27-IgG+ B cell population in peripheral blood expressing VH genes with low frequency of somatic mutation. J Immunol 2006, 177, 3728–36. [DOI] [PubMed] [Google Scholar]
- 71. Wei C, Anolik J, Cappione A, Zheng B, Pugh-Bernard A, Brooks J, et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J Immunol 2007, 178, 6624–33. doi: 10.4049/jimmunol.178.10.6624 [DOI] [PubMed] [Google Scholar]
- 72. Woodruff MC, Ramonell RP, Nguyen DC, Cashman KS, Saini AS, Haddad NS, et al. Extrafollicular B cell responses correlate with neutralizing antibodies and morbidity in COVID-19. Nat Immunol 2020, 21, 1506–16. doi: 10.1038/s41590-020-00814-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Stewart A, Ng JC, Wallis G, Tsioligka V, Fraternali F, Dunn-Walters DK.. Single-cell transcriptomic analyses define distinct peripheral B cell subsets and discrete development pathways. Front Immunol 2021, 12, 602539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. You X, Zhang R, Shao M, He J, Chen J, Liu J, et al. Double negative B cell is associated with renal impairment in systemic lupus erythematosus and acts as a marker for nephritis remission. Front Med 2020, 7, 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Wang Y, Lloyd KA, Melas I, Zhou D, Thyagarajan R, Lindqvist J, et al. Rheumatoid arthritis patients display B-cell dysregulation already in the naive repertoire consistent with defects in B-cell tolerance. Sci Rep 2019, 9, 19995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Moura RA, Quaresma C, Vieira AR, Goncalves MJ, Polido-Pereira J, Romao VC, et al. B-cell phenotype and IgD-CD27- memory B cells are affected by TNF-inhibitors and tocilizumab treatment in rheumatoid arthritis. PLoS One 2017, 12, e0182927. doi: 10.1371/journal.pone.0182927 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Mahmood Z, Muhammad K, Schmalzing M, Roll P, Dorner T, Tony HP.. CD27-IgD-memory B cells are modulated by in vivo interleukin-6 receptor (IL-6R) blockade in rheumatoid arthritis. Arthritis Res Ther 2015, 17, 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Cervantes-Diaz R, Sosa-Hernandez VA, Torres-Ruiz J, Romero-Ramirez S, Canez-Hernandez M, Perez-Fragoso A, et al. Severity of SARS-CoV-2 infection is linked to double-negative (CD27(-) IgD(-)) B cell subset numbers. Inflamm Res 2022, 71, 131–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Oliviero B, Varchetta S, Mele D, Mantovani S, Cerino A, Perotti CG, et al. Expansion of atypical memory B cells is a prominent feature of COVID-19. Cell Mol Immunol 2020, 17, 1101–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Bodenmiller B, Zunder ER, Finck R, Chen TJ, Savig ES, Bruggner RV, et al. Multiplexed mass cytometry profiling of cellular states perturbed by small-molecule regulators. Nat Biotechnol 2012, 30, 858–67. doi: 10.1038/nbt.2317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol 2018. doi: 10.1038/nbt.4314 [DOI] [PubMed] [Google Scholar]
- 82. Amir el AD, Davis KL, Tadmor MD, Simonds EF, Levine JH, Bendall SC, et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol 2013, 31, 545–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Marie-Cardine A, Divay F, Dutot I, Green A, Perdrix A, Boyer O, et al. Transitional B cells in humans: characterization and insight from B lymphocyte reconstitution after hematopoietic stem cell transplantation. Clin Immunol 2008, 127, 14–25. [DOI] [PubMed] [Google Scholar]
- 84. Bemark M, Holmqvist J, Abrahamsson J, Mellgren K.. Translational Mini-Review Series on B cell subsets in disease. Reconstitution after haematopoietic stem cell transplantation - revelation of B cell developmental pathways and lineage phenotypes. Clin Exp Immunol 2012, 167, 15–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Mraz M, Zent CS, Church AK, Jelinek DF, Wu X, Pospisilova S, et al. Bone marrow stromal cells protect lymphoma B-cells from rituximab-induced apoptosis and targeting integrin alpha-4-beta-1 (VLA-4) with natalizumab can overcome this resistance. Br J Haematol 2011, 155, 53–64. doi: 10.1111/j.1365-2141.2011.08794.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Loder F, Mutschler B, Ray RJ, Paige CJ, Sideras P, Torres R, et al. B cell development in the spleen takes place in discrete steps and is determined by the quality of B cell receptor-derived signals. J Exp Med 1999, 190, 75–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Allman D, Lindsley RC, DeMuth W, Rudd K, Shinton SA, Hardy RR.. Resolution of three nonproliferative immature splenic B cell subsets reveals multiple selection points during peripheral B cell maturation. J Immunol 2001, 167, 6834–40. doi: 10.4049/jimmunol.167.12.6834 [DOI] [PubMed] [Google Scholar]
- 88. Srivastava B, Quinn WJ 3rd, Hazard K, Erikson J, Allman D.. Characterization of marginal zone B cell precursors. J Exp Med 2005, 202, 1225–34. doi: 10.1084/jem.20051038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Pillai S, Cariappa A, Moran ST.. Positive selection and lineage commitment during peripheral B-lymphocyte development. Immunol Rev 2004, 197, 206–18. doi: 10.1111/j.0105-2896.2003.097.x [DOI] [PubMed] [Google Scholar]
- 90. Evans JG, Chavez-Rueda KA, Eddaoudi A, Meyer-Bahlburg A, Rawlings DJ, Ehrenstein MR, et al. Novel suppressive function of transitional 2 B cells in experimental arthritis. J Immunol 2007, 178, 7868–78. doi: 10.4049/jimmunol.178.12.7868 [DOI] [PubMed] [Google Scholar]
- 91. Yurasov S, Wardemann H, Hammersen J, Tsuiji M, Meffre E, Pascual V, et al. Defective B cell tolerance checkpoints in systemic lupus erythematosus. J Exp Med 2005, 201, 703–11. doi: 10.1084/jem.20042251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Wang H, Liu C, Chen W, Ding G.. The skewed frequency of B-cell subpopulation CD19(+) CD24 (hi) CD38 (hi) cells in peripheral blood mononuclear cells is correlated with the elevated serum sCD40L in patients with active systemic lupus erythematosus. J Cell Biochem 2019. doi: 10.1002/jcb.28427 [DOI] [PubMed] [Google Scholar]
- 93. Smets I, Prezzemolo T, Imbrechts M, Mallants K, Mitera T, Humblet-Baron S, et al. Treatment-induced BAFF expression and B cell biology in multiple sclerosis. Front Immunol 2021, 12, 676619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Chang NH, Li TT, Kim JJ, Landolt-Marticorena C, Fortin PR, Gladman DD, et al. Interferon-alpha induces altered transitional B cell signaling and function in Systemic Lupus Erythematosus. J Autoimmun 2015, 58, 100–10. doi: 10.1016/j.jaut.2015.01.009 [DOI] [PubMed] [Google Scholar]
- 95. Klarquist J, Cantrell R, Lehn MA, Lampe K, Hennies CM, Hoebe K, et al. Type I IFN drives experimental systemic lupus erythematosus by distinct mechanisms in CD4 T cells and B cells. Immunohorizons 2020, 4, 140–52. doi: 10.4049/immunohorizons.2000005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S, Duurland CL, et al. CD19(+)CD24(hi)CD38(hi) B cells are expanded in juvenile dermatomyositis and exhibit a pro-inflammatory phenotype after activation through toll-like receptor 7 and interferon-alpha. Front Immunol 2018, 9, 1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Lin W, Jin L, Chen H, Wu Q, Fei Y, Zheng W, et al. B cell subsets and dysfunction of regulatory B cells in IgG4-related diseases and primary Sjogren’s syndrome: the similarities and differences. Arthritis Res Ther 2014, 16, R118. doi: 10.1186/ar4571 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Cherukuri A, Salama AD, Carter CR, Landsittel D, Arumugakani G, Clark B, et al. Reduced human transitional B cell T1/T2 ratio is associated with subsequent deterioration in renal allograft function. Kidney Int 2017, 91, 183–95. doi: 10.1016/j.kint.2016.08.028 [DOI] [PubMed] [Google Scholar]
- 99. Burton H, Dorling A.. Transitional B cell subsets-a convincing predictive biomarker for allograft loss?. Kidney Int 2017, 91, 18–20. doi: 10.1016/j.kint.2016.10.028 [DOI] [PubMed] [Google Scholar]
- 100. Blair PA, Norena LY, Flores-Borja F, Rawlings DJ, Isenberg DA, Ehrenstein MR, et al. CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. Immunity 2010, 32, 129–40. doi: 10.1016/j.immuni.2009.11.009 [DOI] [PubMed] [Google Scholar]
- 101. Mauri C, Menon M.. Human regulatory B cells in health and disease: therapeutic potential. J Clin Invest 2017, 127, 772–9. doi: 10.1172/jci85113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Menon M, Blair PA, Isenberg DA, Mauri C.. A regulatory feedback between plasmacytoid dendritic cells and regulatory B cells is aberrant in systemic lupus erythematosus. Immunity 2016, 44, 683–97. doi: 10.1016/j.immuni.2016.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Rosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, et al. Regulatory B cells are induced by gut microbiota-driven interleukin-1beta and interleukin-6 production. Nat Med 2014, 20, 1334–9. doi: 10.1038/nm.3680 [DOI] [PubMed] [Google Scholar]
- 104. Flores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA, et al. CD19+CD24hiCD38hi B cells maintain regulatory T cells while limiting TH1 and TH17 differentiation. Sci Transl Med 2013, 5, 173ra23. [DOI] [PubMed] [Google Scholar]
- 105. Banko Z, Pozsgay J, Gati T, Rojkovich B, Ujfalussy I, Sarmay G.. Regulatory B cells in rheumatoid arthritis: alterations in patients receiving anti-TNF therapy. Clin Immunol 2017, 184, 63–9. [DOI] [PubMed] [Google Scholar]
- 106. Lin X, Wang X, Xiao F, Ma K, Liu L, Wang X, et al. IL-10-producing regulatory B cells restrain the T follicular helper cell response in primary Sjogren’s syndrome. Cell Mol Immunol 2019, 16, 921–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Cencioni MT, Ali R, Nicholas R, Muraro PA.. Defective CD19+CD24(hi)CD38(hi) transitional B-cell function in patients with relapsing-remitting MS. Mult Scler 2021, 27, 1187–97. [DOI] [PubMed] [Google Scholar]
- 108. Matsushita T, Hamaguchi Y, Hasegawa M, Takehara K, Fujimoto M.. Decreased levels of regulatory B cells in patients with systemic sclerosis: association with autoantibody production and disease activity. Rheumatology 2016, 55, 263–7. [DOI] [PubMed] [Google Scholar]
- 109. Zhu HQ, Xu RC, Chen YY, Yuan HJ, Cao H, Zhao XQ, et al. Impaired function of CD19(+) CD24(hi) CD38(hi) regulatory B cells in patients with pemphigus. Br J Dermatol 2015, 172, 101–10. [DOI] [PubMed] [Google Scholar]
- 110. Sheng JR, Rezania K, Soliven B.. Impaired regulatory B cells in myasthenia gravis. J Neuroimmunol 2016, 297, 38–45. doi: 10.1016/j.jneuroim.2016.05.004 [DOI] [PubMed] [Google Scholar]
- 111. Lin Y, Chang T, Lin J, Sun C, Wei C, Zhao J, et al. Regulatory B cells are decreased and functionally impaired in myasthenia gravis patients. Front Neurol 2022, 13, 808322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Guerrier T, Youinou P, Pers JO, Jamin C.. TLR9 drives the development of transitional B cells towards the marginal zone pathway and promotes autoimmunity. J Autoimmun 2012, 39, 173–9. doi: 10.1016/j.jaut.2012.05.012 [DOI] [PubMed] [Google Scholar]
- 113. Bell SE, Goodnow CC.. A selective defect in IgM antigen receptor synthesis and transport causes loss of cell surface IgM expression on tolerant B lymphocytes. EMBO J 1994, 13, 816–26. doi: 10.1002/j.1460-2075.1994.tb06324.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Sabouri Z, Perotti S, Spierings E, Humburg P, Yabas M, Bergmann H, et al. IgD attenuates the IgM-induced anergy response in transitional and mature B cells. Nat Commun 2016, 7, 13381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Brink R, Goodnow CC, Crosbie J, Adams E, Eris J, Mason DY, et al. Immunoglobulin M and D antigen receptors are both capable of mediating B lymphocyte activation, deletion, or anergy after interaction with specific antigen. J Exp Med 1992, 176, 991–1005. doi: 10.1084/jem.176.4.991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Jenks SA, Cashman KS, Woodruff MC, Lee FE, Sanz I.. Extrafollicular responses in humans and SLE. Immunol Rev 2019, 288, 136–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Huang W, Quach TD, Dascalu C, Liu Z, Leung T, Byrne-Steele M, et al. Belimumab promotes negative selection of activated autoreactive B cells in systemic lupus erythematosus patients. JCI Insight 2018, 3(17), e122525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Ramskold D, Parodis I, Lakshmikanth T, Sippl N, Khademi M, Chen Y, et al. B cell alterations during BAFF inhibition with belimumab in SLE. EBioMedicine 2019, 40, 517–27. doi: 10.1016/j.ebiom.2018.12.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Yagi-Numata N, Matsushita T, Takehara K, Hamaguchi Y.. Increased expression levels of FcgammaRIIB on naive and double-negative memory B cells in patients with systemic sclerosis. Clin Exp Rheumatol 2019, 37, 23–31. [PubMed] [Google Scholar]
- 120. Sato S, Fujimoto M, Hasegawa M, Takehara K.. Altered blood B lymphocyte homeostasis in systemic sclerosis: expanded naive B cells and diminished but activated memory B cells. Arthritis Rheum 2004, 50, 1918–27. doi: 10.1002/art.20274 [DOI] [PubMed] [Google Scholar]
- 121. von Borstel A, Abdulahad WH, Sanders JS, Rip J, Neys SFH, Hendriks RW, et al. Evidence for enhanced Bruton’s tyrosine kinase activity in transitional and naive B cells of patients with granulomatosis with polyangiitis. Rheumatology 2019, 58, 2230–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Hansen A, Odendahl M, Reiter K, Jacobi AM, Feist E, Scholze J, et al. Diminished peripheral blood memory B cells and accumulation of memory B cells in the salivary glands of patients with Sjogren’s syndrome. Arthritis Rheum 2002, 46, 2160–71. doi: 10.1002/art.10445 [DOI] [PubMed] [Google Scholar]
- 123. Mebius RE, Kraal G.. Structure and function of the spleen. Nat Rev Immunol 2005, 5, 606–16. doi: 10.1038/nri1669 [DOI] [PubMed] [Google Scholar]
- 124. Weill JC, Weller S, Reynaud CA.. Human marginal zone B cells. Annu Rev Immunol 2009, 27, 267–85. doi: 10.1146/annurev.immunol.021908.132607 [DOI] [PubMed] [Google Scholar]
- 125. Weller S, Faili A, Garcia C, Braun MC, Le Deist FF, de Saint Basile GG, et al. CD40-CD40L independent Ig gene hypermutation suggests a second B cell diversification pathway in humans. Proc Natl Acad Sci U S A 2001, 98, 1166–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Seifert M, Kuppers R.. Molecular footprints of a germinal center derivation of human IgM+(IgD+)CD27+ B cells and the dynamics of memory B cell generation. J Exp Med 2009, 206, 2659–69. doi: 10.1084/jem.20091087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Budeus B, Schweigle de Reynoso S, Przekopowitz M, Hoffmann D, Seifert M, Kuppers R.. Complexity of the human memory B-cell compartment is determined by the versatility of clonal diversification in germinal centers. Proc Natl Acad Sci U S A 2015, 112, E5281–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Seifert M, Kuppers R.. Human memory B cells. Leukemia 2016, 30, 2283–92. doi: 10.1038/leu.2016.226 [DOI] [PubMed] [Google Scholar]
- 129. Cerutti A, Cols M, Puga I.. Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes. Nat Rev Immunol 2013, 13, 118–32. doi: 10.1038/nri3383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Nagafuchi Y, Shoda H, Sumitomo S, Nakachi S, Kato R, Tsuchida Y, et al. Enhanced gut homing receptor expression of unswitched memory B cells in rheumatoid arthritis. Clin Exp Rheumatol 2017, 35, 354–5. [PubMed] [Google Scholar]
- 131. Newell KL, Clemmer DC, Cox JB, Kayode YI, Zoccoli-Rodriguez V, Taylor HE, et al. Switched and unswitched memory B cells detected during SARS-CoV-2 convalescence correlate with limited symptom duration. PLoS One 2021, 16, e0244855. doi: 10.1371/journal.pone.0244855 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Karlsen M, Hansen T, Nordal HH, Brun JG, Jonsson R, Appel S.. Expression of Toll-like receptor -7 and -9 in B cell subsets from patients with primary Sjogren’s syndrome. PLoS One 2015, 10, e0120383. doi: 10.1371/journal.pone.0120383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Si R, Zhao P, Yu Z, Qu Z, Sun W, Li T, et al. Increased non-switched memory B cells are associated with plasmablasts, serum IL-6 levels and renal functional impairments in IgAN patients. Immunol Invest 2020, 49, 178–90. [DOI] [PubMed] [Google Scholar]
- 134. Simon Q, Pers JO, Cornec D, Le Pottier L, Mageed RA, Hillion S.. In-depth characterization of CD24(high)CD38(high) transitional human B cells reveals different regulatory profiles. J Allergy Clin Immunol 2016, 137, 1577–84.e10. doi: 10.1016/j.jaci.2015.09.014 [DOI] [PubMed] [Google Scholar]
- 135. Carter MJ, Fish M, Jennings A, Doores KJ, Wellman P, Seow J, et al. Peripheral immunophenotypes in children with multisystem inflammatory syndrome associated with SARS-CoV-2 infection. Nat Med 2020, 26, 1701–7. [DOI] [PubMed] [Google Scholar]
- 136. Seifert M, Przekopowitz M, Taudien S, Lollies A, Ronge V, Drees B, et al. Functional capacities of human IgM memory B cells in early inflammatory responses and secondary germinal center reactions. Proc Natl Acad Sci U S A 2015, 112, E546–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Wu YC, Kipling D, Leong HS, Martin V, Ademokun AA, Dunn-Walters DK.. High-throughput immunoglobulin repertoire analysis distinguishes between human IgM memory and switched memory B-cell populations. Blood 2010, 116, 1070–8. doi: 10.1182/blood-2010-03-275859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Trend S, Leffler J, Teige I, Frendeus B, Kermode AG, French MA, et al. FcgammaRIIb expression is decreased on naive and marginal zone-like B cells from females with multiple sclerosis. Front Immunol 2020, 11, 614492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Liu Y, Feng Y, Tang S, Zhang L, Huang Z, Shi X, et al. Aberrant expression of inhibitory receptors on B cells in patients with Graves’ disease. Hum Immunol 2022, 83, 144–52. doi: 10.1016/j.humimm.2021.12.001 [DOI] [PubMed] [Google Scholar]
- 140. Danza A, Ruiz-Irastorza G.. Infection risk in systemic lupus erythematosus patients: susceptibility factors and preventive strategies. Lupus 2013, 22, 1286–94. doi: 10.1177/0961203313493032 [DOI] [PubMed] [Google Scholar]
- 141. Doyon-Laliberte K, Aranguren M, Poudrier J, Roger M.. Marginal zone B-cell populations and their regulatory potential in the context of HIV and other chronic inflammatory conditions. Int J Mol Sci 2022, 23(6), 3372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Weisel F, Shlomchik M.. Memory B cells of mice and humans. Annu Rev Immunol 2017, 35, 255–84. doi: 10.1146/annurev-immunol-041015-055531 [DOI] [PubMed] [Google Scholar]
- 143. Maurer D, Holter W, Majdic O, Fischer GF, Knapp W.. CD27 expression by a distinct subpopulation of human B lymphocytes. Eur J Immunol 1990, 20, 2679–84. doi: 10.1002/eji.1830201223 [DOI] [PubMed] [Google Scholar]
- 144. Tangye SG, Liu YJ, Aversa G, Phillips JH, de Vries JE.. Identification of functional human splenic memory B cells by expression of CD148 and CD27. J Exp Med 1998, 188, 1691–703. doi: 10.1084/jem.188.9.1691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Tarlinton D, Good-Jacobson K.. Diversity among memory B cells: origin, consequences, and utility. Science 2013, 341, 1205–11. doi: 10.1126/science.1241146 [DOI] [PubMed] [Google Scholar]
- 146. Good-Jacobson KL, Shlomchik MJ.. Plasticity and heterogeneity in the generation of memory B cells and long-lived plasma cells: the influence of germinal center interactions and dynamics. J Immunol 2010, 185, 3117–25. doi: 10.4049/jimmunol.1001155 [DOI] [PubMed] [Google Scholar]
- 147. Lanzavecchia A, Sallusto F.. Progressive differentiation and selection of the fittest in the immune response. Nat Rev Immunol 2002, 2, 982–7. doi: 10.1038/nri959 [DOI] [PubMed] [Google Scholar]
- 148. Tangye SG, Avery DT, Deenick EK, Hodgkin PD.. Intrinsic differences in the proliferation of naive and memory human B cells as a mechanism for enhanced secondary immune responses. J Immunol 2003, 170, 686–94. doi: 10.4049/jimmunol.170.2.686 [DOI] [PubMed] [Google Scholar]
- 149. Good KL, Bryant VL, Tangye SG.. Kinetics of human B cell behavior and amplification of proliferative responses following stimulation with IL-21. J Immunol 2006, 177, 5236–47. doi: 10.4049/jimmunol.177.8.5236 [DOI] [PubMed] [Google Scholar]
- 150. Weill JC, Reynaud CA.. IgM memory B cells: specific effectors of innate-like and adaptive responses. Curr Opin Immunol 2019, 63, 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Lindner JM, Cornacchione V, Sathe A, Be C, Srinivas H, Riquet E, et al. Human memory B cells harbor diverse cross-neutralizing antibodies against BK and JC polyomaviruses. Immunity 2019, 50, 668–76.e5. doi: 10.1016/j.immuni.2019.02.003 [DOI] [PubMed] [Google Scholar]
- 152. Manea ME, Mueller RB, Dejica D, Sheriff A, Schett G, Herrmann M, et al. Increased expression of CD154 and FAS in SLE patients’ lymphocytes. Rheumatol Int 2009, 30, 181–5. doi: 10.1007/s00296-009-0933-4 [DOI] [PubMed] [Google Scholar]
- 153. Jacobi AM, Reiter K, Mackay M, Aranow C, Hiepe F, Radbruch A, et al. Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arthritis Rheum 2008, 58, 1762–73. doi: 10.1002/art.23498 [DOI] [PubMed] [Google Scholar]
- 154. Wang J, Shan Y, Jiang Z, Feng J, Li C, Ma L, et al. High frequencies of activated B cells and T follicular helper cells are correlated with disease activity in patients with new-onset rheumatoid arthritis. Clin Exp Immunol 2013, 174, 212–20. doi: 10.1111/cei.12162 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Pugh-Bernard AE, Silverman GJ, Cappione AJ, Villano ME, Ryan DH, Insel RA, et al. Regulation of inherently autoreactive VH4-34 B cells in the maintenance of human B cell tolerance. J Clin Invest 2001, 108, 1061–70. doi: 10.1172/jci200112462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Desai-Mehta A, Lu L, Ramsey-Goldman R, Datta SK.. Hyperexpression of CD40 ligand by B and T cells in human lupus and its role in pathogenic autoantibody production. J Clin Invest 1996, 97, 2063–73. doi: 10.1172/jci118643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Grammer AC, Slota R, Fischer R, Gur H, Girschick H, Yarboro C, et al. Abnormal germinal center reactions in systemic lupus erythematosus demonstrated by blockade of CD154-CD40 interactions. J Clin Invest 2003, 112, 1506–20. doi: 10.1172/jci200319301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Ramanujam M, Steffgen J, Visvanathan S, Mohan C, Fine JS, Putterman C.. Phoenix from the flames: rediscovering the role of the CD40-CD40L pathway in systemic lupus erythematosus and lupus nephritis. Autoimmun Rev 2020, 19, 102668102668. doi: 10.1016/j.autrev.2020.102668 [DOI] [PubMed] [Google Scholar]
- 159. Roll P, Mahmood Z, Muhammad K, Feuchtenberger M, Dorner T, Tony HP.. Long-term repopulation of peripheral B-cell subsets after single and repeated rituximab infusions in patients with rheumatoid arthritis. Clin Exp Rheumatol 2015, 33, 347–53. [PubMed] [Google Scholar]
- 160. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O’Shea MA, et al. Evidence for HIV-associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV-infected viremic individuals. J Exp Med 2008, 205, 1797–805. doi: 10.1084/jem.20072683 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Liechti T, Kadelka C, Braun DL, Kuster H, Boni J, Robbiani M, et al. Widespread B cell perturbations in HIV-1 infection afflict naive and marginal zone B cells. J Exp Med 2019, 216, 2071–90. doi: 10.1084/jem.20181124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Cancro MP. Age-associated B cells. Annu Rev Immunol 2020, 38, 315–40. doi: 10.1146/annurev-immunol-092419-031130 [DOI] [PubMed] [Google Scholar]
- 163. Wang S, Wang J, Kumar V, Karnell JL, Naiman B, Gross PS, et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11c(hi)T-bet(+) B cells in SLE. Nat Commun 2018, 9, 1758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Rubtsov AV, Rubtsova K, Fischer A, Meehan RT, Gillis JZ, Kappler JW, et al. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c(+) B-cell population is important for the development of autoimmunity. Blood 2011, 118, 1305–15. doi: 10.1182/blood-2011-01-331462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Saadoun D, Terrier B, Bannock J, Vazquez T, Massad C, Kang I, et al. Expansion of autoreactive unresponsive CD21-/low B cells in Sjogren’s syndrome-associated lymphoproliferation. Arthritis Rheum 2013, 65, 1085–96. doi: 10.1002/art.37828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Tony HP, Roll P, Mei HE, Blumner E, Straka A, Gnuegge L, et al. Combination of B cell biomarkers as independent predictors of response in patients with rheumatoid arthritis treated with rituximab. Clin Exp Rheumatol 2015, 33, 887–94. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Raw mass cytometry data can be requested from the corresponding author.