Visual Abstract

Upon Ag exposure, naive B cells expressing BCR able to bind Ag can undergo robust proliferation and differentiation that can result in the production of Ab-secreting and memory B cells. The factors determining whether an individual naive B cell will proliferate following Ag encounter remains unclear. In this study, we found that polyclonal naive murine B cell populations specific for a variety of foreign Ags express high levels of the orphan nuclear receptor Nur77, which is known to be upregulated downstream of BCR signaling as a result of cross-reactivity with self-antigens in vivo. Similarly, a fraction of naive human B cells specific for clinically-relevant Ags derived from respiratory syncytial virus and HIV-1 also exhibited an IgMLOW IgD+ phenotype, which is associated with self-antigen cross-reactivity. Functionally, naive B cells expressing moderate levels of Nur77 are most likely to proliferate in vivo following Ag injection. Together, our data indicate that BCR cross-reactivity with self-antigen is a common feature of populations of naive B cells specific for foreign Ags and a moderate level of cross-reactivity primes individual cells for optimal proliferative responses following Ag exposure.

During hematopoietic development, B cell progenitors undergo VDJ recombination to generate Ig expressed on the cell surface as a BCR (1). The random nature of this process gives way to the broad spectrum of receptor specificities needed to recognize diverse pathogens but can also yield cells expressing receptors that bind self-antigens. To prevent Ab-mediated autoimmunity, self-reactive B cells are subject to tolerance mechanisms including receptor editing, clonal deletion, and the restriction of functional responsiveness, termed anergy (2). The clones that survive these checkpoints comprise the repertoire of naive B cells that may respond to vaccination or infection (3). There is growing evidence suggesting that Ab responses are hindered by peripheral tolerance mechanisms due to cross-reactivity of Ag-specific BCR with host-derived self-antigens. Studies of individual Ab lineages have identified isolated cases where such cross-reactivity may influence activation of particular clones (410). Additionally, several studies have found increased protective Ab responses in animals and patients prone to autoimmunity (1113). However, it is not clear from these examples how universal this feature may be for naive B cell populations targeting different foreign Ags.

Such studies pose a number of technical difficulties, as they require analysis of rare Ag-specific B cells prior to somatic hypermutation. Previous work has shown that as many as 20% of the BCRs in the naive human repertoire exhibit detectable reactivity with self-antigens (14). In this work however, the specificity of these B cells was not fully defined. More recently, Zikherman and colleagues (1518) used a reporter mouse in which BCR signaling in response to self-antigen binding was assessed by the expression of eGFP as a surrogate for the expression of the orphan nuclear receptor Nur77. These studies revealed that mature follicular B cells expressed a range of Nur77, and high levels were associated with self-antigen reactivity and diminished function (15). Importantly, whereas other factors such as CD40 and TLR signaling could result in modest Nur77eGFP upregulation in vitro, Nur77eGFP expression can be detected in naive B cells at steady-state in the absence of these signals (16). These studies also revealed that Nur77eGFP expression was low in naive MD4 B cells, which express a transgenic BCR specific for the foreign Ag hen egg lysozyme (15, 16). These data may indicate that naive B cells specific for foreign Ags exhibit low cross-reactivity with self-antigens. However, this conclusion is drawn upon the examination of a single BCR, while there are potentially hundreds of thousands of unique BCRs able to bind a given Ag. Furthermore, the MD4 BCR was originally isolated from an animal postimmunization following germinal center selection and extensive somatic hypermutation, which may have reduced cross-reactivity from the unmutated BCR from which the MD4 Ab was derived. A more recent study analyzed B cells with a fixed VH186.2 transgenic H chain specific for the foreign Ag 4-hydroxy-3-nitrophenyl (NP) paired to different nontransgenic L chains. In this context, positive selection was more evident in cells expressing BCR that exhibited higher self-antigen reactivity (18). These data examining oligoclonal populations of B cells indicates that mild self-reactivity is a key part of B cell development and suggests that nontransgenic polyclonal populations of foreign Ag-specific naive B cells would have similar features.

A recent study examining the reactivity of foreign and self-reactivity of Abs produced by cultures of thousands of single the transitional and mature human B cells may have shed light on this question (19). These data revealed that on average, half of the B cells binding both foreign and self-antigens were deleted between the transitional and mature stage (19). Importantly, self-antigen cross-reactivity was never completely removed from the foreign Ag-specific populations, and the extent of deletion appeared to vary from Ag to Ag (19).

In this study, we used Ag-specific enrichment to analyze populations of naive murine B cells specific for both model Ags as well as pathogen-derived Ags and found that Nur77eGFP was expressed similarly among most populations. Unexpectedly, these levels were indistinguishable from the larger population of naive B cells whose specificities were unknown. Similarly, naive human B cells specific for influenza virus, respiratory syncytial virus (RSV), or HIV-1 proteins contained B cells with the IgD+ IgMLOW phenotype that has been previously reported to contain human self-antigen-reactive B cells (20, 21). Importantly, the level of self-antigen cross-reactivity appears to influence the likelihood that a naive B cell will respond to Ag immunization. Naive Ag-specific B cells with high Nur77eGFP levels were ∼2-fold less likely to proliferate in response to Ag injection compared with their counterparts that expressed moderate levels of Nur77eGFP. However the cells with low Nur77eGFP levels were also less likely to proliferate compared with the population expressing moderate levels of Nur77eGFP, indicating that some level of cross-reactivity with self-antigen may support a cell state optimally poised to proliferate during an immune response. Together, our data suggest that all polyclonal populations of B cells specific for foreign Ags contain BCRs with a range of self-antigen reactivities that tunes cells for optimal function.

Six- to fourteen-week-old male and female mice were used for experiments. CD45.2 (C57BL/6), CD45.1 congenic (B6.SJL-Ptprca Pep3b/BoyJ), IgHa congenic (B6.Cg-Gpi1a Thy1a Igha/J), Nur77eGFP (C57BL/6-Tg(Nr4a1-EGFP/cre)820Khog/J) (22), MD4 (C57BL/6-Tg(IghelMD4)4Ccg/J) (4) and Rag1−/− (B6.129S7-Rag1tm1Mom/J) (23) mice were purchased from the Jackson Laboratory (Bar Harbor, ME). IgHa and Nur77eGFP were backcrossed to generate IgHa/a Nur77eGFP mice before use. MD4 and Rag1−/− were backcrossed to generate MD4 Rag1−/−. Animals were maintained in a specific pathogen-free facility in accordance with Fred Hutchinson Cancer Research Center Institutional Animal Care and Use Committee approval and National Institutes of Health guidelines.

Purified R-PE and allophycocyanin were purchased from ProZyme, and OVA was purchased from MilliporeSigma. Biotinylated peptides (GGGEQKLISEEDLGGG) conjugated to NP (NP-biotin) or DNP (DNP-biotin) were purchased from GenScript.

NP650 was created by first conjugating streptavidin–allophycocyanin (PJ27S; ProZyme) to DyLight 650 NHS ester following the manufacturer’s instructions (Thermo Fisher Scientific). The molar concentration of streptavidin–allophycocyanin–DyLight 650 was calculated by measuring the absorbance of streptavidin–allophycocyanin at 280 nm using the extinction coefficient of 0.268 μM−1cm−1. The resulting streptavidin–allophycocyanin–DyLight 650 was mixed at a molar ratio of 1:6 with NP-biotin and incubated for 30 min on ice. The resulting conjugate was purified from excess NP-biotin using a 100-kDa Amicon Ultra size exclusion column (MilliporeSigma), and the concentration was determined as described for streptavidin–allophycocyanin–DyLight 650.

RSV prefusion and postfusion F Ags, HIV-1 envelope (Env) 426CTM4ΔV1–3, and influenza hemagglutinin (HA) stem were produced as described previously (2426).

Purified Ags were biotinylated using an EZ-Link Sulfo-NHS-LC-Biotinylation kit (Thermo Fisher Scientific) using a 1:1.3 M ratio of biotin to Ag. Unconjugated biotin was removed by centrifugation using an Amicon Ultra size exclusion column (MilliporeSigma) with a smaller m.w. cutoff than the Ag. To determine the average number of biotin molecules bound to each Ag, streptavidin–PE (PJRS25; ProZyme) was mixed with a fixed amount of biotinylated Ag in increasing concentrations and incubated at room temperature for 30 min. Samples were run on an SDS-PAGE gel (Bio-Rad Laboratories) and transferred to nitrocellulose prior to incubation with streptavidin–Alexa Fluor 680 (Thermo Fisher Scientific) diluted 1:10,000 in 1× DPBS containing 0.2% Tween 20 (MilliporeSigma) to determine the ratio at which there was excess biotinylated Ag available for streptavidin–Alexa Fluor 680 to bind. To make tetramers, biotinylated Ags were mixed with streptavidin–PE or streptavidin–allophycocyanin (PJ27S; ProZyme) at the ratio determined above or at a 5 to 1 ratio using the biotin concentration provided by the manufacturer. Following a 30-min incubation on ice, unconjugated biotinylated Ag was often removed by several rounds of dilution and concentration using a 100 kDa Amicon Ultra (MilliporeSigma) or 300 kDa Nanosep centrifugal devices (Pall). Tetramers were stored at 1 μM in 1× DPBS at 4°C or 1× DPBS containing 50% glycerol at −20°C prior to use. Control PE594, PE650, and allophycocyanin–DyLight 755 (Allophycocyanin755) tetramers were created by mixing a biotinylated control Ag with streptavidin–PE preconjugated with DyLight 594 (PE594) or DyLight 650 (PE650), and streptavidin–Allophycocyanin755 following the manufacturer’s instructions for DyLight conjugation (Thermo Fisher Scientific). On average, PE594/PE650 and Allophycocyanin755 contained four to eight DyLight molecules per PE or allophycocyanin. The concentration of each tetramer was calculated by measuring the absorbance of PE at 565 nm combined with an extinction coefficient of 1.96 μM−1cm−1 or allophycocyanin at 650 nm using an extinction coefficient of 0.7 μM−1cm−1.

The spleen and inguinal, axillary, brachial, cervical, mesenteric, and periaortic lymph nodes from individual mice were pooled and dissociated with forceps prior to incubation with 0.8 mg/ml Dispase (Thermo Fisher Scientific), 0.2 mg/ml Collagenase P (Roche), and 0.1 mg/ml of DNase I (Roche) in RPMI 1640 (Thermo Fisher Scientific) for 20 min at 37°C. Tissue fragments were next forced through a 100-micron mesh to create single-cell suspensions, which were filtered using another 100-μm mesh to remove tissue debris and centrifuged at 300 × g for 5 min at 4°C.

Venipuncture was used to obtain blood from healthy, HIV-seronegative adult volunteers enrolled in the General Quality Control study in Seattle, WA. This protocol was approved by the Fred Hutch institutional review board, and informed consent was obtained from all study participants before enrollment into the parent protocols. PBMCs were isolated from whole blood using Accuspin System Histopaque-1077 (MilliporeSigma), and the resulting cell fraction was washed in 1× DPBS and centrifuged at 300 × g for 5 min at 4°C. The supernatant was aspirated and the cell pellet resuspended in 10% DMSO in heat-inactivated FBS and cryopreserved in liquid nitrogen before use.

Ag-specific B cells were enriched using previously described protocols (2730). For labeling with fluorescent model Ags, 0.2-ml samples containing 1–2 × 108 cells were incubated with 1 pmole of allophycocyanin, NP650, or PE for 25 min on ice in FACS buffer (1× DPBS containing 1% heat-inactivated newborn calf serum) containing 2 μg of anti-Fc receptor Ab 2.4G2 (Bio X Cell) for murine cells or 2.5% heat-inactivated mouse serum (Thermo Fisher Scientific) and 2.5% heat-inactivated rat serum (Thermo Fisher Scientific) for human cells. For Ag tetramer labeling, cells were first incubated with 1–3 pmole of tetramer containing a control Ag conjugated to PE650, PE594, and/or Allophycocyanin755 for 10 min on ice prior to incubation with 1 pmole of allophycocyanin- or PE-conjugated Ag tetramer for 25 min on ice.

After the incubation, 15 ml of FACS buffer was added and the sample was centrifuged at 300 × g for 5 min at 4°C. The supernatant was discarded and the pellet was resuspended prior to the addition of 25 μl of anti-allophycocyanin and/or 25 μl of anti-PE microbeads (Miltenyi Biotec). Following a 15–30 min incubation on ice, 5 ml of FACS buffer was added, and the sample was passed over a magnetized LS column (Miltenyi Biotec). The tube and column were washed once with FACS buffer and then removed from the magnetic field. Five milliliters of FACS buffer was pushed through the column with a plunger twice to elute column-bound cells.

Cells from the column-bound fraction and 1/40th of the column flow through fractions were incubated in 50 μl of FACS buffer containing a mixture of Abs and a 0.05 μl/ml fixable viability dye (FVD) eFluor 506 (eBioscience), FVD eFluor 780 (eBioscience), or Ghost Dye Violet 510 (Tonbo Biosciences) for 30 min on ice. Abs for murine experiments included various combinations of 2 μg/ml GL7 Fitc (GL7; BD Biosciences), 2 μg/ml anti-IgD PerCP-Cy5.5 (11-26c.2a; BD Biosciences), 4 μg/ml anti-IgMb BV650 (AF6-78; BD Biosciences), 4 μg/ml anti-Igλ BV421 or BUV395 (R26-46; BD Biosciences), 2 μg/ml anti-Igκ BV605 (187.1; BD Biosciences), 2 μg/ml anti-CD9 PE (eBioKMC8; eBioscience), 2 μg/ml anti-CD19 BUV395 or BUV737 (1D3; BD Biosciences), 2 μg/ml anti-CD21 PE-Cy7 (eBio8D9; eBioscience), 2 μg/ml anti-CD23 BV786 (B3B4; BD Biosciences), 2 μg/ml anti-CD38 Pacific Blue (90; BioLegend), 2 μg/ml anti-B220 BV711 or BV786 (RA3-6B2; BD Biosciences), 2 μg/ml anti-CD45.1 PE-eFluor 610 (A20; eBioscience), 2 μg/ml anti-CD45.2 PerCP-Cy5.5 (104; eBioscience), 2 μg/ml anti-CD45.2 PE-Cy7 (104; BioLegend), 4 μg/ml anti-CD79b BUV661 (HM79b; BD Biosciences), 2 μg/ml anti-CD93 BV421 or BV650 (AA4.1; BD Biosciences), 2 μg/ml anti-CD3 BV510 (145-2C11; BD Biosciences), 2 μg/ml anti-F4/80 BV510 (BM8; BioLegend), and 2 μg/ml anti-Gr-1 BV510 (RB6-8C5; BD Biosciences). Following surface staining, samples were centrifuged for 5 min at 300 × g and supernatant was discarded. For intracellular Ig assessment, samples were resuspended and incubated with 250 μl of Cytofix/Cytoperm (BD Biosciences) for 30 min on ice. Four milliliters of permeabilization buffer (BD Biosciences) was added, samples were centrifuged for 5 min at 400 × g, and supernatant was discarded. Samples were resuspended with 40 μg/ml F(ab′)2 goat anti-IgG (H+L) Alexa Fluor 350 and incubated for 30 min on ice. Four milliliters of permeabilization buffer (BD Biosciences) was added, samples were centrifuged for 5 min at 400 × g, and supernatant was discarded.

For human PBMC experiments, cells were labeled with various combinations of 6.24 μg/ml anti-IgM FITC (G20-127; BD Biosciences), 0.3 μg/ml anti-IgD PerCP-Cy5.5 (IA6-2; BD Biosciences), 25 μg/ml anti-Igλ Pacific Blue (MHL-38; BioLegend), 2.5 μg/ml anti-Igκ BV786 (G20-193; BD Biosciences), 50 μl/ml of anti-CD10 BUV395 (HI10a; BD Biosciences), 20 μl/ml of anti-CD19 PE-Cy7 (HIB19; BD Biosciences), 100 μl/ml of anti-CD19 BUV496 (SJ25C1; BD Biosciences), 20 μg/ml anti-CD20 eFluor 450 (2H7; eBioscience), 25 μl/ml anti-CD20 BUV395 (2H7; BD Biosciences), 50 μl/ml of anti-CD27 BV650 or BV480 (L128; BD Biosciences), 20 μg/ml anti-CD79b allophycocyanin/Fire750 (MHL-38; BioLegend), 60 μl/ml of anti-CD3 BV711 (UCT1; BD Biosciences), 60 μl/ml of anti-CD14 BV711 (MφP9; BD Biosciences), and 2.8 μg/ml of anti-CD16 BV711 (3G8; BioLegend). Following surface staining, samples were centrifuged for 5 min at 300 × g and supernatant was discarded. Human samples were resuspended with 250 μl of Cytofix/Cytoperm (BD Biosciences) or 2% paraformaldehyde and incubated for 15 min on ice prior to the addition of 4 ml of FACS buffer. Human samples were centrifuged for 5 min at 400 × g, and supernatant was discarded.

Prior to flow cytometry, both human and murine samples were resuspended in FACS buffer containing 20,000 Fluorescent AccuCheck counting beads (Thermo Fisher Scientific) to calculate cell numbers. Flow cytometry was performed on a five-laser (355, 405, 488, 561, 640 nm) LSR II, LSRFortessa, or FACSymphony (BD Biosciences) and analyzed with FlowJo software (Tree Star, Ashland, OR).

Naive follicular B cells were enriched from pooled spleen and lymph nodes from 18–20 mice using a B cell negative selection kit (Miltenyi Biotec, Auburn, CA) supplemented with Biotin-conjugated anti-CD93 (BD Biosciences) to eliminate transitional B cells (31). The B cells were washed in warm RPMI medium, adjusted to a final concentration of 2 × 107 cells/ml in prewarmed RPMI medium, and incubated with 5 μM CellTrace Violet (CTV) (Thermo Fisher Scientific) for 12 min at 37°C prior to the addition of media containing 10% newborn calf serum. Following washing, cell surface marker staining was performed as described above. B cells expressing different levels of Nur77eGFP were sorted by a FACSAria II cell sorter (BD Biosciences) using side scatter height/side scatter width–based duplet discrimination, followed by gating on CD9 CD3 F4/80 Gr-1 FVD eFluor 780. CD9 was used to exclude marginal zone cells (32).

Purified CTV-labeled B cells were adjusted to a concentration of 2 × 106 cells/ml in RPMI (Thermo Fisher Scientific, Logan, UT) containing 10% FBS (Thermo Fisher Scientific), 100 U/ml penicillin (Thermo Fisher Scientific), 100 μg/ml streptomycin (Thermo Fisher Scientific), 2 mM l-glutamine (Thermo Fisher Scientific), and 27.5 μM 2-ME (MilliporeSigma). Five milliliters of cells was added per well of a six-well flat bottomed plate and cultured in the presence or absence of 1–10 μg/ml F(ab′)2 goat anti-mouse IgG (H+L) (Jackson ImmunoResearch) for 72 h at 37°C. Following incubation, cell surface marker staining was performed as described above prior to analysis of CTV dilution in gated CD19+ CD3 Gr-1 F4/80 FVD eFluor 780 cells using flow cytometry.

A total of 1–3 × 106 of FACS-purified B cells in 0.1 ml of 1× DPBS was injected retro-orbitally into CD45.1+ recipient mice. The following day, animals were injected s.c. in the base of the tail with 50 μl of CFA (MilliporeSigma) mixed 1 to 1 with 1× DPBS with or without 62.5 pmoles of NP650. Seven days following Ag injection, NP650+ cells were enriched and CD45.2+ CD45.1 donor B cells were assessed using flow cytometry as described above.

Single human B cells were isolated using a FACSAria II cell sorter (BD Biosciences) following tetramer enrichment and cell surface marker staining using human PBMCs as described above. Specifically, Env-specific B cells were sorted using side scatter height/side scatter width–based duplet discrimination, followed by gating on CD19+ CD20+ CD3 CD14 CD16 FVD B cells that bound HIV-1 Env–PE (or allophycocyanin) tetramers but not PE650 (or PE594 or Allophycocyanin755) tetramers containing control Ags. Single cells were sorted into individual wells of 96-well PCR plates (Eppendorf) containing 10 μl/well of ice-cold lysis buffer containing 0.25 μl 12.5 U RNase out (Thermo Fisher Scientific), 2.5 μl 5× SuperScript IV First Strand Buffer (Thermo Fisher Scientific), 0.625 μl 0.1 M DTT (Thermo Fisher Scientific), 0.3125 μl 10% Igepal detergent (MilliporeSigma), and 6.625 μl DEPC-treated water. Plates were sealed with adhesive PCR plate seals (Thermo Fisher Scientific), centrifuged briefly, and immediately frozen on dry ice before storage at −80°C.

Reverse transcription was performed using SuperScript IV (Thermo Fisher Scientific) as previously described (33, 34). Briefly, 3 μl of reverse transcription reaction mix consisting of 1.5 μl of 50 μM random hexamers (Thermo Fisher Scientific), 0.4 μl 25 mM dNTPs (Thermo Fisher Scientific), 0.5 μl 10 U SuperScript IV RT, and 0.6 μl water was added to each well containing a single-sorted B cell in 10 μl lysis buffer and incubated at 50°C for 1 h. Following reverse transcription, 2 μl of cDNA was added to 19 μl of PCR mix containing 0.2 μl 0.5 U HotStarTaq Polymerase (Qiagen), 0.075 μl 50 μM 3′ reverse primers, 0.115 μl 50 μM 5′ forward primers, 0.24 μl 25 mM dNTPs, 1.9 μl 10× buffer (Qiagen), and 16.5 μl water. The PCR program for IgM/IgG and Igκ was 50 cycles of 94°C for 30 s, 57°C for 30 s, and 72°C for 55 s, followed by 72°C for 10 min. The PCR program for Igλ was 50 cycles of 94°C for 30 s, 60°C for 30 s, and 72°C for 55 s, followed by 72°C for 10 min. After the first round of PCR, 2 μl of the PCR product was added to 19 μl of the second round PCR so that the final reaction contained 0.2 μl 0.5 U HotStarTaq Polymerase, 0.075 μl 50 μM 3′ reverse primers, 0.075 μl 50 μM 5′ forward primers, 0.24 μl 25 mM dNTPs, 1.9 μl 10× Buffer, and 16.5 μl water. PCR programs were the same as the first round of PCR. Five microliters of the PCR product was run on an agarose gel to confirm the presence of an ∼500-bp H chain band or 450-bp L chain band. Five microliters from PCR reactions showing the presence of H chain or L chain amplicons was mixed with 2 μl of ExoSAP-IT (Thermo Fisher Scientific) and incubated at 37°C 15 min, followed by 80°C for 15 min to hydrolyze excess primers and nucleotides. Hydrolyzed second-round PCR products were sequenced by Genewiz with the respective reverse primer, and sequences were analyzed using IMGT/V-Quest to identify V, D, and J gene segments. Sequences with more than five mutations or <85% V gene similarity were excluded from Supplemental Table I.

Paired H chain VDJ and L chain VJ sequences that lacked signs of somatic hypermutation from Env-specific or control B cells of unknown specificity were cloned into pTT3-derived expression vectors containing the human IgG1, Igκ, or Igλ constant regions (25) using In-Fusion cloning (Clontech), and sequences were confirmed using Sanger sequencing (Genewiz). Expression vectors were transfected into 293F cells and Abs purified from culture supernatant using protein A columns (Thermo Fisher Scientific) followed by media exchange into 1× DPBS in house or by the Fred Hutch Ab Development Shared Resource.

Bio-layer interferometry (BLI) using the Octet Red instrument (Forte Bio) was performed at room temperature with shaking at 1000 RPM following the manufacturer’s instructions. Briefly, 100 μg/ml of cloned Abs were loaded on anti-human IgG Fc capture biosensors (Forte Bio) for 240 s, followed by a wash step in kinetics buffer (1× DPBS, 0.01% BSA, 0.02% Tween 20, and 0.005% NaN3 [pH 7.4]) for 60 s. After washing, 1 μM HIV-1 Env was associated for 300 s, followed by dissociation into kinetics buffer for an additional 300 s. The HIV-1 Env–specific Ab VRC01 was used as a positive control in all experiments, and the RSV-specific Ab palivizumab was used as a negative control.

The ZEUS IFA ANA HEp-2 Test System was used following the protocol recommended by the manufacturer (Zeus Scientific) with minor changes. Briefly, HEp-2–coated slides from the kit were incubated with 25 μl of Ab at 0.1 mg/ml for 30 min at room temperature. Slides were then washed three times in 1× DPBS, followed by incubation of 25 μl of 0.01 mg/ml goat anti-human IgG Alexa Fluor 594 (Thermo Fisher Scientific) in 1× DPBS for 30 min at room temperature in the dark. After washing three times with 1× DPBS, slides were coated in 50% glycerol, and coverslips were applied. The EVOS Cell Imaging System (Thermo Fisher Scientific) was used to acquire images, which were analyzed using ImageJ to determine the average Alexa Fluor 594 fluorescence per HEp-2 cell.

Prism software (GraphPad) was used to calculate p values using an unpaired two-tailed t test, a ratio paired two-tailed t test, or one-way ANOVA followed by Dunnett multiple comparison test.

To assess the degree to which cross-reactivity with self-antigen could exist within polyclonal naive B cells specific for foreign Ags, we measured Nur77eGFP expression in rare naive follicular Ag-specific B cells using previously described Ag-specific enrichment approaches (2730). In our initial experiments, we analyzed B cells specific for a complex model Ag we called NP650, which contains four NP molecules bound to streptavidin–allophycocyanin and ∼6 molecules of DyLight 650. In Nur77eGFP transgenic mice, a relatively large population, ∼0.4%, of naive follicular B cells bound NP650, which was increased to ∼18% following enrichment using anti-allophycocyanin microbeads (Fig. 1A, 1B). The binding of NP650 was confirmed to be BCR mediated, because MD4 Rag1−/− B cells expressing a transgenic BCR specific for hen egg lysozyme did not bind this Ag above the limit of detection even with enrichment (Fig. 1B). Additionally, we confirmed that immunization with NP650 in CFA resulted in activation and differentiation of B cells specific for NP, allophycocyanin and DL650 when these populations were assessed individually (Supplemental Fig. 1).

FIGURE 1.

Expression of Nur77eGFP in naive follicular B cells specific for a model Ag. (A) Representative flow cytometric analysis of CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells. (B) Representative flow cytometric analysis of NP650 binding to follicular B cells from pooled spleen and lymph node samples from MD4 Rag1−/− and Nur77eGFP transgenic animals with or without enrichment of NP650-binding cells with allophycocyanin-specific microbeads prior to analysis. The fraction “depleted” of NP650-binding cells with anti-allophycocyanin microbeads is also shown. (C) Nur77eGFP expression by NP650+ and NP650NEGATIVE follicular B cells from Nur77eGFP and GFPNEGATIVE control. (D) Combined data from seven experiments displaying the gMFI of Nur77eGFP within NP650+ follicular B cells normalized to the NP650NEGATIVE follicular B cells from individual mice using enrichment (n = 18) or following incubation with 5 nM of NP650 at 37°C (n = 6) for 3 h. (E) Nur77eGFP gMFI of NP650+ follicular B cells from individual samples (n = 5) in which one fraction was enriched for NP650 as described in (B), and a second fraction was labeled with NP650 immediately before analysis. The line connects matched samples from two combined experiments. (F) Representative gating of B cells binding low, mid, and high levels of NP650. (G) Combined data from four experiments displaying the Nur77eGFP gMFI of follicular B cells binding low, mid, and high levels of NP650 normalized to the NP650NEGATIVE B cells from individual mice using the enrichment protocol on ice or following incubation at 37°C (n = 6) for 3 h. The bars in (D) and (G) represent the means, and p values (**p < 0.002, ***p < 0.0001) were determined using an unpaired two-tailed t test.

FIGURE 1.

Expression of Nur77eGFP in naive follicular B cells specific for a model Ag. (A) Representative flow cytometric analysis of CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells. (B) Representative flow cytometric analysis of NP650 binding to follicular B cells from pooled spleen and lymph node samples from MD4 Rag1−/− and Nur77eGFP transgenic animals with or without enrichment of NP650-binding cells with allophycocyanin-specific microbeads prior to analysis. The fraction “depleted” of NP650-binding cells with anti-allophycocyanin microbeads is also shown. (C) Nur77eGFP expression by NP650+ and NP650NEGATIVE follicular B cells from Nur77eGFP and GFPNEGATIVE control. (D) Combined data from seven experiments displaying the gMFI of Nur77eGFP within NP650+ follicular B cells normalized to the NP650NEGATIVE follicular B cells from individual mice using enrichment (n = 18) or following incubation with 5 nM of NP650 at 37°C (n = 6) for 3 h. (E) Nur77eGFP gMFI of NP650+ follicular B cells from individual samples (n = 5) in which one fraction was enriched for NP650 as described in (B), and a second fraction was labeled with NP650 immediately before analysis. The line connects matched samples from two combined experiments. (F) Representative gating of B cells binding low, mid, and high levels of NP650. (G) Combined data from four experiments displaying the Nur77eGFP gMFI of follicular B cells binding low, mid, and high levels of NP650 normalized to the NP650NEGATIVE B cells from individual mice using the enrichment protocol on ice or following incubation at 37°C (n = 6) for 3 h. The bars in (D) and (G) represent the means, and p values (**p < 0.002, ***p < 0.0001) were determined using an unpaired two-tailed t test.

Close modal

We next compared the level of Nur77eGFP in naive follicular B cells that bound NP650 compared with cells that were NP650NEGATIVE (Fig. 1C). This analysis revealed a roughly 1:1 ratio of Nur77eGFP geometric mean fluorescence intensities (gMFI) between the two populations (Fig. 1D). These results raised the concern that the enrichment methodology resulted in an upregulation of Nur77eGFP expression. For the enrichment protocol, fluorescent Ags are bound to BCRs expressed by Ag-specific B cells, followed by magnetic enrichment using microbeads specific for the fluorochrome. Although the labeling and enrichment procedure is carried out largely on ice, it is possible that BCR signaling and subsequent Nur77eGFP upregulation could occur during short portions of this 2–3 h procedure when the cells are not on ice. Against this notion, Nur77eGFP was not upregulated in enriched NP650-specific B cells compared with a portion of the sample that was analyzed immediately following NP650 labeling without enrichment (Fig. 1E). In contrast, Nur77eGFP upregulation was present in NP650-specific B cells if the samples were incubated with NP650 at 37°C for 3 h (Fig. 1D). Nur77eGFP expression was correlated with the level of NP650 bound by the cells when the samples were incubated at 37°C for 3 h, but not when samples were kept on ice (Fig. 1F, 1G). Together, these data indicate that Nur77eGFP expression is not altered by Ag-specific enrichment and instead is reflective of in vivo BCR signaling.

We next assessed whether B cells specific for individual components of NP650 expressed either high or low levels of Nur77eGFP. This did not appear to be the case because the NP-specific and allophycocyanin-specific naive follicular B cells expressed indistinguishable Nur77eGFP gMFI and robust coefficient of variation (rCV) compared with their counterparts that did not bind these Ags (Fig. 2A–D). These results appear to conflict with the results of a recent study that found low Nur77eGFP levels in NP-specific B cells using a transgenic H chain paired to endogenously produced λ (Igλ) L chains (18). In agreement with this report, we found that Nur77eGFP levels were slightly lower in NP-specific naive follicular B cells using Igλ (Fig. 2E, 2F). This did not appear to be unique to NP-specific cells because Nur77eGFP expression was slightly lower in all naive follicular B cells using Igλ, including those specific for the model Ag DNP (Fig. 2E, 2F). Lower Nur77eGFP expression in B cells using Igλ likely resulted from slightly lower surface BCR expression compared with their counterparts using Igκ, which was detected when surface levels of CD79b (Igβ) were assessed (Fig. 2G, 2H). Similarly, human Igλ+ naive follicular B cells expressed slightly lower CD79b levels compared with their Igκ+ counterparts (Supplemental Fig. 2). Differences in Nur77eGFP expression between B cells expressing Igλ and Igκ were eliminated when normalized to the level of surface BCR (Fig. 2I, 2J).

FIGURE 2.

Expression of Nur77eGFP in naive follicular B cells specific for model Ags. Representative flow cytometric analysis of (A) allophycocyanin-specific and PE-specific B cells, and (B) DNP and NP-specific B cells within CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells from pooled spleen and lymph node samples from Nur77eGFP transgenic animals enriched with allophycocyanin-specific and PE-specific microbeads prior to analysis. The control PE594 and Allophycocyanin755 tetramers are included in (B) to gate out B cells specific for streptavidin, allophycocyanin, and PE (28). The percentage of B cells in the enriched fraction that are within each gate are shown on each plot, and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (C and D) Combined data from 14 experiments displaying the (C) gMFI and (D) rCV of Nur77eGFP within the listed Ag-specific follicular B cells normalized to AgNEGATIVE follicular B cells from individual IgHb or congenic IgHa mice (n = 8–18) within the enriched fraction. (E) Representative flow cytometric analysis of Igκ and Igλ expression by NP-specific, DNP-specific, and AgNEGATIVE follicular B cells. (F) Combined data from two experiments displaying the gMFI of Nur77eGFP within the listed population of Igκ+ or Igλ+ follicular B cell population from individual mice (n = 8) normalized to AgNEGATIVE follicular B cells. (G) Representative flow cytometric analysis and (H) gMFI of surface CD79b expression on Igκ+ or Igλ+ B cells. Lines in (H) connect data points from individual mice (n = 10) from two experiments. (I) Representative flow cytometric analysis and (J) gMFI of Nur77eGFP by Igκ+ or Igλ+ B cells when normalized to the level of CD79b expressed by each cell. Lines in (J) connect data points from individual mice (n = 8) from two experiments. The p values (*p < 0.05, **p < 0.01, ***p < 0.001) in (C) and (F) were determined using an unpaired two-tailed t test and (H) using a ratio paired two-tailed t test.

FIGURE 2.

Expression of Nur77eGFP in naive follicular B cells specific for model Ags. Representative flow cytometric analysis of (A) allophycocyanin-specific and PE-specific B cells, and (B) DNP and NP-specific B cells within CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells from pooled spleen and lymph node samples from Nur77eGFP transgenic animals enriched with allophycocyanin-specific and PE-specific microbeads prior to analysis. The control PE594 and Allophycocyanin755 tetramers are included in (B) to gate out B cells specific for streptavidin, allophycocyanin, and PE (28). The percentage of B cells in the enriched fraction that are within each gate are shown on each plot, and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (C and D) Combined data from 14 experiments displaying the (C) gMFI and (D) rCV of Nur77eGFP within the listed Ag-specific follicular B cells normalized to AgNEGATIVE follicular B cells from individual IgHb or congenic IgHa mice (n = 8–18) within the enriched fraction. (E) Representative flow cytometric analysis of Igκ and Igλ expression by NP-specific, DNP-specific, and AgNEGATIVE follicular B cells. (F) Combined data from two experiments displaying the gMFI of Nur77eGFP within the listed population of Igκ+ or Igλ+ follicular B cell population from individual mice (n = 8) normalized to AgNEGATIVE follicular B cells. (G) Representative flow cytometric analysis and (H) gMFI of surface CD79b expression on Igκ+ or Igλ+ B cells. Lines in (H) connect data points from individual mice (n = 10) from two experiments. (I) Representative flow cytometric analysis and (J) gMFI of Nur77eGFP by Igκ+ or Igλ+ B cells when normalized to the level of CD79b expressed by each cell. Lines in (J) connect data points from individual mice (n = 8) from two experiments. The p values (*p < 0.05, **p < 0.01, ***p < 0.001) in (C) and (F) were determined using an unpaired two-tailed t test and (H) using a ratio paired two-tailed t test.

Close modal

Overall, the expression of Nur77eGFP in DNP-specific naive follicular B cells was also similar to the DNPNEGATIVE population (Fig. 2B–D). Unexpectedly, increased expression of Nur77eGFP was detected in naive follicular B cells specific for the model Ag R-PE (Fig. 2A, 2C). Previous work has demonstrated that the PE-specific population is large and dominated by a large fraction of cells using VH1-81 in IgHb mouse strains such as C57BL/6 (35). The increased expression of Nur77eGFP appeared linked to the usage of VH1-81 because the PE-specific population from IgHa mice expressed similar levels of Nur77eGFP compared with the allophycocyanin-specific and AgNEGATIVE populations (Fig. 2C). Together, these data suggest that a range of cross-reactivity with self-antigens is common within populations of B cells specific for foreign Ags.

We next considered whether these results were applicable to Ags derived from human pathogens. For this, we examined the populations of B cells specific for an influenza HA stem Ag (26), an HIV-1 Env Ag called 426CTM4ΔV1–V3 (25), and Ags representing the prefusion and postfusion forms of the RSV F protein (24, 36) using tetramer enrichment. We found that the follicular B cells specific for influenza HA stem, HIV-1 Env, RSV prefusion F, and postfusion F expressed a similar level of Nur77eGFP (Fig. 3). These data suggested that a range of self-antigen cross-reactivity is a common feature among populations of naive B cells specific for pathogen-derived Ags.

FIGURE 3.

Expression of Nur77eGFP in naive follicular B cells specific for pathogen-derived Ags. Representative flow cytometric analysis of (A) influenza HA stem-specific and HIV-1 Env–specific B cells and (B) RSV prefusion F-specific and postfusion F-specific B cells within CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells from pooled spleen and lymph node samples from Nur77eGFP transgenic animals enriched with allophycocyanin-specific and PE-specific microbeads prior to analysis. The control PE594 and Allophycocyanin755 tetramers are included to gate out B cells specific for purification tags, trimerization domains, streptavidin, allophycocyanin, and PE (28). The percentage of B cells in the enriched fraction that are within each gate are shown on each plot, and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (C and D) Combined data from four experiments displaying the (C) gMFI and (D) rCV of Nur77eGFP within the listed Ag-specific follicular B cells normalized to the AgNEGATIVE follicular B cells from individual mice (n = 8) within the enriched fraction.

FIGURE 3.

Expression of Nur77eGFP in naive follicular B cells specific for pathogen-derived Ags. Representative flow cytometric analysis of (A) influenza HA stem-specific and HIV-1 Env–specific B cells and (B) RSV prefusion F-specific and postfusion F-specific B cells within CD19+ B220+ CD23+ CD21/35MID CD3 Gr-1 F4/80 FVD follicular B cells from pooled spleen and lymph node samples from Nur77eGFP transgenic animals enriched with allophycocyanin-specific and PE-specific microbeads prior to analysis. The control PE594 and Allophycocyanin755 tetramers are included to gate out B cells specific for purification tags, trimerization domains, streptavidin, allophycocyanin, and PE (28). The percentage of B cells in the enriched fraction that are within each gate are shown on each plot, and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (C and D) Combined data from four experiments displaying the (C) gMFI and (D) rCV of Nur77eGFP within the listed Ag-specific follicular B cells normalized to the AgNEGATIVE follicular B cells from individual mice (n = 8) within the enriched fraction.

Close modal

B cells expressing BCRs able to bind self-antigens often exhibit reduced responsiveness to BCR stimulation (410). Previous work demonstrated that calcium signaling in response to BCR stimulation was reduced in B cells expressing high levels of Nur77eGFP compared with cells expressing low levels (15). In this study, we assessed whether this reduced calcium flux was mirrored by reduced proliferation of FACS-purified mature follicular B cells expressing high levels of Nur77eGFP following BCR stimulation with anti-Ig in vitro. For this, we incubated FACS-purified naive B cells expressing high, mid, and low levels of Nur77eGFP with anti-Ig for 3 d in vitro and measured CTV dilution. In these experiments, we found that 39% of cells expressing high levels of Nur77eGFP had diluted CTV following stimulation with 2 μg/ml anti-Ig for 3 d in vitro (Fig. 4A–C). In contrast, 61% of B cells expressing low levels of Nur77eGFP had low CTV levels in response to the same amount of anti-Ig (Fig. 4B, 4C). In contrast, 44% of B cells expressing moderate levels of Nur77eGFP had low CTV levels in response to the same amount of anti-Ig (Fig. 4B, 4C). These differences in CTV dilution were diminished as the concentration of anti-Ig was increased (Fig. 4C), indicating that strong BCR stimulation could overcome proliferative defects. Similar differences in proliferation were detected when B cells binding NP650 were examined within populations expressing high or low levels of Nur77eGFP (Fig. 4D). These data suggest that Nur77eGFP expression is a surrogate for reduced function in B cells specific for foreign Ags.

FIGURE 4.

In vitro proliferation of naive follicular B cells expressing different levels of Nur77eGFP. (A) Representative flow cytometric analysis of FACS-purified Nur77eGFP-low, FACS-purified Nur77eGFP-mid, and Nur77eGFP-high naive follicular B cells and the unsorted fraction of B cells. (B) B cells expressing different levels of Nur77eGFP were FACS-purified and labeled with CTV and cultured in vitro for 72 h in the presence or absence of 2 μg/ml of anti-Ig prior to flow cytometric analysis of CTV dilution within gated CD19+ CD3 Gr-1 F4/80 FVD B cells. (C) Combined data from three experiments displaying the average frequency ± SD of CTVLOW cells following in vitro culture with 2, 5, 10, or 25 μg/ml of anti-Ig for 72 h. (D) Displays the average frequency ± SD of CTVLOW cells within NP650+ B cells from the experiments described in (C). The p values (*p < 0.05, **p < 0.01, ***p < 0.001) were determined using an unpaired two-tailed t test.

FIGURE 4.

In vitro proliferation of naive follicular B cells expressing different levels of Nur77eGFP. (A) Representative flow cytometric analysis of FACS-purified Nur77eGFP-low, FACS-purified Nur77eGFP-mid, and Nur77eGFP-high naive follicular B cells and the unsorted fraction of B cells. (B) B cells expressing different levels of Nur77eGFP were FACS-purified and labeled with CTV and cultured in vitro for 72 h in the presence or absence of 2 μg/ml of anti-Ig prior to flow cytometric analysis of CTV dilution within gated CD19+ CD3 Gr-1 F4/80 FVD B cells. (C) Combined data from three experiments displaying the average frequency ± SD of CTVLOW cells following in vitro culture with 2, 5, 10, or 25 μg/ml of anti-Ig for 72 h. (D) Displays the average frequency ± SD of CTVLOW cells within NP650+ B cells from the experiments described in (C). The p values (*p < 0.05, **p < 0.01, ***p < 0.001) were determined using an unpaired two-tailed t test.

Close modal

The modest in vitro response of B cells expressing high levels of Nur77eGFP suggested that these cells may respond poorly to Ag immunization in vivo. To test this, 1–3 × 106 FACS-purified mature CD45.2+ follicular B cells that expressed high, moderate, or low levels of Nur77eGFP were adoptively transferred into CD45.1+ recipients 1 d prior to injection of NP650 in CFA or CFA alone. We used NP650 for these experiments because the frequency of B cells able to bind this combination Ag is large enough to reliably detect hundreds of these cells within the CD45.2+ donor population after transfer (Fig. 5A). Seven days after the injection of CFA alone, the differences in Nur77eGFP expression were maintained within the three groups of NP650-specific cells (Fig. 5B). Following the injection of NP650 in CFA, the majority of donor NP650-specific B cells in all three groups had fully diluted levels of CTV compared with their counterparts in control mice injected with CFA alone (Fig. 5C, 5D), indicating proliferation. However, significantly fewer, 66%, NP650-specific cells that expressed high levels of Nur77eGFP had low CTV compared with the 79% of donor cells that expressed low levels of Nur77eGFP (Fig. 5C, 5D). Unexpectedly, NP650-specific B cells that expressed moderate levels of Nur77eGFP contained the highest frequency of CTVLOW cells, 88% (Fig. 5D). These data suggest that the expression of moderate levels of Nur77eGFP corresponds with optimal function.

FIGURE 5.

In vivo proliferation of naive B cells expressing different levels of Nur77eGFP. Naive follicular B cells expressing low, mid, or high levels of Nur77eGFP were FACS purified, CTV labeled, and adoptively transferred into CD45.1+ recipient mice 1 d prior to s.c. injection of 62.5 pmoles NP650 in CFA or CFA alone. Seven days later, pooled spleen and lymph nodes from individual mice were analyzed following simultaneous NP650 and CD45.2-based enrichment. (A) Representative flow cytometric analysis of donor CD45.2+ CD45.1+ NP650+ B cells from enriched fractions. (B) Nur77eGFP gMFI in donor NP650+ B cells in mice 7 d after the injection of CFA. Representative of two similar experiments. (C) Representative CTV dilution 7 d after the injection of NP650 in CFA or CFA alone within donor NP650+ B cells expressing different levels of Nur77eGFP at the time of transfer (D) Combined data from four experiments showing the percentages of CTVLOW donor NP650+ B cells in each group. (E) Combined data from four experiments showing the percentages of donor B cells that were CTVHIGH NP650+ following injection of NP650 in CFA or CFA alone. (F) Same data as (E) expressed as a percentage of cells that remained CTVHIGH NP650+ in animals injected with NP650 in CFA compared CFA alone. (G) The percentage of cells that remained CTVHIGH NP650+ is plotted versus the percentage of CTVLOW cells. The bar graphs in (D)–(F) represent the mean ± SD (n = 9–12), and p values (*p < 0.05, **p < 0.01, ***p < 0.001) in (B) and (D)–(F) were determined using an unpaired two-tailed t test.

FIGURE 5.

In vivo proliferation of naive B cells expressing different levels of Nur77eGFP. Naive follicular B cells expressing low, mid, or high levels of Nur77eGFP were FACS purified, CTV labeled, and adoptively transferred into CD45.1+ recipient mice 1 d prior to s.c. injection of 62.5 pmoles NP650 in CFA or CFA alone. Seven days later, pooled spleen and lymph nodes from individual mice were analyzed following simultaneous NP650 and CD45.2-based enrichment. (A) Representative flow cytometric analysis of donor CD45.2+ CD45.1+ NP650+ B cells from enriched fractions. (B) Nur77eGFP gMFI in donor NP650+ B cells in mice 7 d after the injection of CFA. Representative of two similar experiments. (C) Representative CTV dilution 7 d after the injection of NP650 in CFA or CFA alone within donor NP650+ B cells expressing different levels of Nur77eGFP at the time of transfer (D) Combined data from four experiments showing the percentages of CTVLOW donor NP650+ B cells in each group. (E) Combined data from four experiments showing the percentages of donor B cells that were CTVHIGH NP650+ following injection of NP650 in CFA or CFA alone. (F) Same data as (E) expressed as a percentage of cells that remained CTVHIGH NP650+ in animals injected with NP650 in CFA compared CFA alone. (G) The percentage of cells that remained CTVHIGH NP650+ is plotted versus the percentage of CTVLOW cells. The bar graphs in (D)–(F) represent the mean ± SD (n = 9–12), and p values (*p < 0.05, **p < 0.01, ***p < 0.001) in (B) and (D)–(F) were determined using an unpaired two-tailed t test.

Close modal

An increase in the percentage of CTVLOW cells could be a reflection of a higher number of divisions made by the naive cells that were activated in response to Ag. In these experiments, the responding cells in each group were essentially fully CTV diluted (Fig. 5C), indicating seven or more cell divisions. These data could reflect differences in migration of activated cells out of the spleen and lymph nodes, but the differences in calcium signaling reported previously (15) led us to hypothesize that differences in the frequency of CTVLOW cells resulted from differences in the number of naive B cells that participated in the response and divided at least once. The frequency of naive B cells that failed to divide can be determined by comparing the frequency of Ag-specific donor B cells that are CTVHIGH in mice injected with and without Ag (30). Notably, naive CTVHIGH B cells binding low levels of NP650 were excluded from these analyses because previous experiments revealed that the frequency of these cells was unaltered by NP650 injection (Supplemental Fig. 3). Within recipient animals receiving Nur77eGFP-high cells, ∼0.19% of donor B cells remained undivided and CTVHIGH in animals injected with NP650 in CFA, compared with ∼0.26% in animals injected with CFA alone (Fig. 5E). Although this decrease was not significant, it suggested that ∼three-fourths (19/26%) of Nur77eGFP-high NP650-specific naive B cells fail to proliferate in response to immunization (Fig. 5F). In contrast, only 45% of NP650-specific naive B cells that expressed moderate levels of Nur77eGFP failed to proliferate (Fig. 5E, 5F). An intermediate frequency, 61%, of NP650-specific naive B cells that expressed low levels of Nur77eGFP failed to proliferate (Fig. 5E, 5F). Importantly, the average frequencies of naive B cells that failed to proliferate inversely correlated with the average frequencies of CTV diluted cells (Fig. 5G). Together, these data indicate that naive foreign Ag-specific cells expressing moderate levels of Nur77eGFP are more than twice as likely to respond to Ag immunization compared with their counterparts expressing high levels of Nur77eGFP.

Having observed functional impairment in vaccine-relevant populations in mice, we sought to gauge the level of self-antigen cross-reactivity within analogous populations of naive human B cells specific for pathogen-derived Ags. Unlike Nur77eGFP expression in mice, high levels of intracellular Nur77 in human B cells do not appear to correlate with self-antigen reactivity (37). Instead, human naive B cells expressing low levels of IgM have been shown to be enriched for self-antigen–reactive B cells (20, 21), a phenotype commonly exhibited by transgenic B cells specific for self-antigens (3842). Additionally, both human and murine B cells expressing low levels of IgM have reduced calcium signaling in response to BCR stimulation similar to cells expressing high levels of Nur77eGFP (15, 20, 21, 43). Overall, naive follicular murine B cells specific for NP650 or HIV-1 Env expressed indistinguishable levels of IgM compared with cells that did not bind these Ags (Fig. 6A, 6B). In agreement with published reports (15), naive B cells of unknown specificity that expressed higher levels of Nur77eGFP expressed reduced levels of surface IgM (Fig. 6C–E). Similar results were found within murine B cells specific for NP650 or HIV-1 Env (Fig. 6E). Downregulated IgM levels in cells expressing high levels of Nur77eGFP was mirrored by an overall decrease in surface BCR levels when assessed based upon CD79b staining (Fig. 6F, 6G). Together, these data suggest that self-antigen binding results in Nur77eGFP upregulation and a corresponding downregulation of IgM BCRs.

FIGURE 6.

Expression of IgM on naive follicular B cells specific for model Ags. Representative flow cytometry analysis and quantitation of (A, B, D, and E) IgM and (F and G) CD79b expression by naive follicular murine B cells specific for NP650 or HIV-1 Env compared with cells that did not bind these Ags. Fluorescence minus one (FMO) controls are displayed for comparison. (C) Representative flow cytometric gating of Nur77eGFP-Low (red), Nur77eGFP-Mid (gray), and Nur77eGFP-high (blue) naive follicular murine B cells for the analyses conducted in (D)–(G). Data points are from individual mice [(B), n = 10; (E and G), n = 8] combined from two experiments, and p values (*p < 0.05, ***p < 0.001) were determined using an unpaired two-tailed t test.

FIGURE 6.

Expression of IgM on naive follicular B cells specific for model Ags. Representative flow cytometry analysis and quantitation of (A, B, D, and E) IgM and (F and G) CD79b expression by naive follicular murine B cells specific for NP650 or HIV-1 Env compared with cells that did not bind these Ags. Fluorescence minus one (FMO) controls are displayed for comparison. (C) Representative flow cytometric gating of Nur77eGFP-Low (red), Nur77eGFP-Mid (gray), and Nur77eGFP-high (blue) naive follicular murine B cells for the analyses conducted in (D)–(G). Data points are from individual mice [(B), n = 10; (E and G), n = 8] combined from two experiments, and p values (*p < 0.05, ***p < 0.001) were determined using an unpaired two-tailed t test.

Close modal

Using Ag tetramer-based enrichment, we examined IgM expression in IgD+ naive human B cells specific for HIV-1 Env and RSV F prefusion or postfusion F proteins (Fig. 7A–C). Because most people are naturally infected with RSV early in their lives, we excluded memory B cells and focused upon naive B cells by gating upon cells that expressed IgD but not CD27 (Fig. 7A). Cells expressing CD10 were also excluded because transitional B cells circulating in the blood express this protein (44). Using this gating strategy, we found that each Ag-specific population expressed a similar level of IgM compared with their counterparts that did not bind Ag (Fig. 7D, 7E). These features suggest that populations of B cells specific for foreign Ags exhibit cross-reactivity with self-antigens.

FIGURE 7.

Expression of IgM on naive human B cells specific for pathogen-derived Ags. (A) Representative flow cytometric analysis of CD19+ CD20+ IgD+ CD10 CD27 CD3 CD14 CD16 FVD naive B cells specific for (B) HIV-1 Env, (C) RSV prefusion F, or postfusion F from the fractions enriched using anti-PE and/or anti-allophycocyanin microbeads from 100 million PBMC. The percentage of B cells in the enriched fraction that are within each gate are shown on each plot and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (D) Representative IgM expression by HIV-1 Env tetramer-binding naive B cells compared with tetramerNEGATIVE naive B cells. Fluorescence minus one controls for IgM are also displayed for both populations. (E) Combined data from six experiments displaying the average IgM gMFI within the listed Ag-specific B cell population normalized to the tetramerNEGATIVE B cells. Each data point is the average of two to four independent assessments of samples from individual donors (n = 3–7).

FIGURE 7.

Expression of IgM on naive human B cells specific for pathogen-derived Ags. (A) Representative flow cytometric analysis of CD19+ CD20+ IgD+ CD10 CD27 CD3 CD14 CD16 FVD naive B cells specific for (B) HIV-1 Env, (C) RSV prefusion F, or postfusion F from the fractions enriched using anti-PE and/or anti-allophycocyanin microbeads from 100 million PBMC. The percentage of B cells in the enriched fraction that are within each gate are shown on each plot and the bold percentages represent the frequency of B cells binding the tetramer within the entire unfractionated sample. (D) Representative IgM expression by HIV-1 Env tetramer-binding naive B cells compared with tetramerNEGATIVE naive B cells. Fluorescence minus one controls for IgM are also displayed for both populations. (E) Combined data from six experiments displaying the average IgM gMFI within the listed Ag-specific B cell population normalized to the tetramerNEGATIVE B cells. Each data point is the average of two to four independent assessments of samples from individual donors (n = 3–7).

Close modal

To directly analyze BCR cross-reactivity with self-antigens, we next cloned 27 BCRs from HIV-1 Env–specific B cells and assessed binding to the HEp-2 cell line, a common test of self-antigen reactivity (45). The cloned BCRs were selected from a data set of 103 HIV-1 tetramer-binding B cells from human PBMCs in which paired H and L chain sequences were recovered using single-cell RT-PCR (Supplemental Table I). The 27 cloned Abs exhibited no signs of somatic hypermutation, and 24 bound HIV Env above the limit of detection when assessed by BLI (Fig. 8A, 8B). We next assessed self-antigen reactivity of the 24 Env-specific cloned Abs compared with a set of 18 Abs cloned from naive B cells of unknown specificities (Supplemental Table I) along with an Env-specific positive control, 4E10 (46), and the RSV-specific negative control, palivizumab (47). Using this approach, 8.3% of the Env-specific Abs bound to HEp-2 cells above background, which was similar to the 11% of Abs derived from B cells of unknown specificity that also bound (Fig. 8D, 8E). Together, these data indicate that self-antigen cross-reactivity is a common feature among B cells and is composed of a range of affinity within a given population.

FIGURE 8.

Self-antigen cross-reactivity of HIV-1 Env–specific naive human B cells. (A) BLI measurements of the binding of six Abs cloned from HIV-1 Env–tetramer+ naive B cells to purified Env. (B) Frequency of the 27 cloned Abs that bound to Env above the BLI limit of detection. (C) Representative immunofluorescence images (scale bar, 100 μm) displaying the binding of two Env-specific Abs derived from naive B cells, E81G01 and E82A03, to human HEp-2 cells compared with a positive control Ab, 4E10, and a negative control Ab, palivizumab. (D) Mean (±SD) Alexa Fluor 594 fluorescence per HEp-2 cell for 24 Env-specific Abs and 18 Abs cloned from naive B cells of unknown specificity compared with 4E10 and palivizumab. Data were obtained from five independent experiments for each Ab, and * denotes Abs with significantly (*p < 0.01, **p < 0.0001) higher fluorescence compared with palivizumab using one-way ANOVA followed by Dunnett multiple comparison test. (E) Percentage of Env-specific and control Abs from (D) with significant (p < 0.05) binding to HEp-2 cells compared with palivizumab.

FIGURE 8.

Self-antigen cross-reactivity of HIV-1 Env–specific naive human B cells. (A) BLI measurements of the binding of six Abs cloned from HIV-1 Env–tetramer+ naive B cells to purified Env. (B) Frequency of the 27 cloned Abs that bound to Env above the BLI limit of detection. (C) Representative immunofluorescence images (scale bar, 100 μm) displaying the binding of two Env-specific Abs derived from naive B cells, E81G01 and E82A03, to human HEp-2 cells compared with a positive control Ab, 4E10, and a negative control Ab, palivizumab. (D) Mean (±SD) Alexa Fluor 594 fluorescence per HEp-2 cell for 24 Env-specific Abs and 18 Abs cloned from naive B cells of unknown specificity compared with 4E10 and palivizumab. Data were obtained from five independent experiments for each Ab, and * denotes Abs with significantly (*p < 0.01, **p < 0.0001) higher fluorescence compared with palivizumab using one-way ANOVA followed by Dunnett multiple comparison test. (E) Percentage of Env-specific and control Abs from (D) with significant (p < 0.05) binding to HEp-2 cells compared with palivizumab.

Close modal

Overall, our results demonstrate that B cells specific for foreign Ag display a breadth of self-reactivity and diminished function that may limit vaccine responses. We initially hypothesized that different populations of B cells would have different levels of cross-reactivity with self-antigens based upon the unique properties of the epitopes displayed by each Ag. Although this could still be true, our data suggest that cross-reactivity with self-antigen is generally present among polyclonal BCRs specific for foreign Ags including small haptens, large complex Ags, and even rationally designed immunogens. This suggests that cross-reactivity with self-antigens may have more to do with intrinsic biochemical features of the BCRs themselves rather than similarity between the self- and foreign Ags. There are a number of ways this could occur, for example Abs using VH4–34 are known to be more self-reactive compared with other alleles (4850), whereas the unique CDRH3 regions expressed by BCRs using these alleles can mediate binding to diverse sets of foreign Ags. Ab cross-reactivity to structurally distinct self- and foreign Ags could also occur if different amino acids within the H and L chain CDR3s mediated binding to the two Ags.

Uniquely high cross-reactivity with self-antigen within the PE-specific B cell population was surprising. Cross-reactivity with self-antigens was previously reported within the PE-specific memory B cell population, but this was attributed to somatic hypermutation in the germinal center (51). Our data suggest that this self-reactivity was present in the naive population and either not selected against or increased in the germinal center. Interestingly, increased levels of Nur77eGFP were not detected in PE-specific B cells in IgHa mice, which lack the VH1–81 allele used by the majority of the large PE-specific B cell population in IgHb C57/BL6 mice (35). This could suggest that VH1–81 exhibits a mild level of unrecognized self-reactivity with self-antigens that maintains a higher frequency of PE-specific B cells through increased positive selection similar to results obtained from studies of cells expressing a transgenic H chain specific for NP (18).

In our analyses we found slightly lower expression of Nur77eGFP on B cells using Igλ compared with their Igκ counterparts. Although we initially hypothesized that this was the result of Igλ having inherently lower levels of self-antigen reactivity because overall, B cells that express low levels of Nur77eGFP expressed higher levels of CD79b and IgM. This is likely because binding to self-antigens results in Nur77eGFP upregulation and subsequent downregulation of surface BCR. However, our data instead suggests that the slightly lower Nur77eGFP levels in B cells using Igλ are instead a result of slightly lower BCR expression rather than slightly lower levels of self-antigen reactivity.

Our results contrast with studies of CD4+ and CD8+ T cells showing that increased cross-reactivity with self-peptide bound to MHC class I was associated with higher CD5 expression and enhanced responsiveness (52, 53). Instead, we find that there is a “sweet spot” in that B cells with too little or too much cross-reactivity with self-antigen function poorly. Moderate signaling in response to self-antigen may provide the level of tonic BCR signaling known to be essential for B cell survival (54). We hypothesize that the differences between B and T cells are the result of differences in positive and negative selection. If T cells with higher affinity for self-antigens are more effectively deleted from the repertoire, the remaining CD5HIGH T cells may represent the sweet spot of reactivity necessary for optimal function. In contrast, mild reactivity with self-antigens appears to increase positive selection of developing B cells (18). The ability of B cells to undergo somatic hypermutation may reduce the need for stringent deletion of cells expressing BCRs with higher affinity for self-antigen. Indeed, naive B cells with high affinity for both foreign and self-antigens are likely the source of “redeemed” Abs that appear to lose self-reactivity as a consequence of somatic hypermutation and affinity in the germinal center (55, 56). Together these data suggest that the benefit of redeeming potentially pathogenic Abs to become protective may outweigh the risks of maintaining B cells cross-reactive with self-antigens. Because TCRs cannot be somatically mutated and redeemed, the benefits of keeping T cells with cross-reactivity to self-peptides/MHC is lower.

During the preparation of this article, it was reported that a population of Nur77eGFP-high CD4+ exhibits reduced function (57) similar to what we and others have observed for Nur77eGFP-high B cells. These Nur77eGFP-high T cells appear to compose only a portion of the CD5HIGH CD4+ T cell populations analyzed previously (52), which may have concealed their reduced function. In light of these new data, B and T cells expressing moderate levels of Nur77eGFP may represent the sweet spot for responsiveness to foreign Ags.

One limitation to our analyses of in vivo responsiveness is that we only measured this for B cells specific for our combination Ag NP650. The reason we did not assess multiple Ag-specific populations is because frequency of individual populations is typically below 0.01% of the total B cell population. Below this level we could not obtain enough donor cells sorted upon Nur77eGFP expression level to reliably detect naive Ag-specific B cells that failed to respond to immunization. For this reason, we chose to conjugate NP, streptavidin, allophycocyanin, and Dylight 650 to form NP650. Using this combination Ag, we essentially simultaneously probed the response of multiple naive populations together. Therefore, our data showing that superior response of the naive B cells expressing moderate levels of Nur77eGFP is not confided to a single Ag-specific population but reflective of the larger response targeting all of these Ags. Although it is possible that one Ag-specific population is dominating this response and skewing our results, we think it is more likely that an unusual response from any one population of Ag-specific B cells in this combination would be masked by the response of the other populations.

In our experiments, we also found that naive B cells binding lower levels of foreign Ag were less likely to proliferate in response to immunization. In fact, we could find no evidence that the third of naive B cells binding lowest levels of Ag proliferated using these immunization conditions. These results were not surprising and do not mean that these B cells bind Ag with an affinity below the threshold for activation and are therefore irrelevant. However, it is possible that with increased T cell help, increased Ag availability, Ag multimerization, or different adjuvants could induce these lower binding B cells to participate in a relevant immune response. It will also be interesting to determine whether more low-affinity B cells participate in the response to Ags provided by infection rather than Ag immunization. In fact, some infections are able to stimulate the activation of B cells expressing BCRs with affinities below the limit of detection for most assays (58).

Our results indicate that two key affinities help control which cells respond to Ag exposure. First, the BCR affinity for the injected Ag must be high enough to allow activation. However, overlaid on top of this is the BCR affinity for self-antigens, in which a moderate amount primes cells for optimal responsiveness. Future work is focused upon the elucidation of other factors that tune the functional potential of naive B cells.

We thank P. Culver, R. Putnam, L. Yates, and R. Eberts for administrative support, D. Alwan, E. Naibert, A. MacCamy, and E. Hayes for technical expertise, M. D. Grey, A. T. McGuire, and L. Stamatatos for providing HIV-1 Env, B. Graham for plasmids encoding RSV prefusion and post-fusion F Ags, P. Kwong for plasmids encoding influenza HA stem, and M. J. McElrath for PBMCs from Seattle Area Control cohort.

This work was supported by the National Institutes of Health under Awards R01AI122912 (to J.J.T.) and T32AI118690 (to J.B.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

Allophycocyanin755

allophycocyanin–DyLight 755

BLI

bio-layer interferometry

CTV

CellTrace Violet

Env

envelope

FVD

fixable viability dye

gMFI

geometric mean fluorescence intensity

HA

hemagglutinin

NP

4-hydroxy-3-nitrophenyl

rCV

robust coefficient of variation

RSV

respiratory syncytial virus.

1
Reth
,
M. G.
,
F. W.
Alt
.
1984
.
Novel immunoglobulin heavy chains are produced from DJH gene segment rearrangements in lymphoid cells.
Nature
312
:
418
423
.
2
Cambier
,
J. C.
,
S. B.
Gauld
,
K. T.
Merrell
,
B. J.
Vilen
.
2007
.
B-cell anergy: from transgenic models to naturally occurring anergic B cells?
Nat. Rev. Immunol.
7
:
633
643
.
3
Havenar-Daughton
,
C.
,
R. K.
Abbott
,
W. R.
Schief
,
S.
Crotty
.
2018
.
When designing vaccines, consider the starting material: the human B cell repertoire.
Curr. Opin. Immunol.
53
:
209
216
.
4
Goodnow
,
C. C.
,
J.
Crosbie
,
S.
Adelstein
,
T. B.
Lavoie
,
S. J.
Smith-Gill
,
R. A.
Brink
,
H.
Pritchard-Briscoe
,
J. S.
Wotherspoon
,
R. H.
Loblay
,
K.
Raphael
, et al
.
1988
.
Altered immunoglobulin expression and functional silencing of self-reactive B lymphocytes in transgenic mice.
Nature
334
:
676
682
.
5
Klinman
,
N. R.
1996
.
The “clonal selection hypothesis” and current concepts of B cell tolerance.
Immunity
5
:
189
195
.
6
Fulcher
,
D. A.
,
A.
Basten
.
1994
.
Reduced life span of anergic self-reactive B cells in a double-transgenic model.
J. Exp. Med.
179
:
125
134
.
7
Cyster
,
J. G.
,
S. B.
Hartley
,
C. C.
Goodnow
.
1994
.
Competition for follicular niches excludes self-reactive cells from the recirculating B-cell repertoire.
Nature
371
:
389
395
.
8
Goodnow
,
C. C.
,
C. G.
Vinuesa
,
K. L.
Randall
,
F.
Mackay
,
R.
Brink
.
2010
.
Control systems and decision making for antibody production.
Nat. Immunol.
11
:
681
688
.
9
Nemazee
,
D.
2006
.
Receptor editing in lymphocyte development and central tolerance.
Nat. Rev. Immunol.
6
:
728
740
.
10
Nemazee
,
D.
2017
.
Mechanisms of central tolerance for B cells.
Nat. Rev. Immunol.
17
:
281
294
.
11
Bonsignori
,
M.
,
K.
Wiehe
,
S. K.
Grimm
,
R.
Lynch
,
G.
Yang
,
D. M.
Kozink
,
F.
Perrin
,
A. J.
Cooper
,
K. K.
Hwang
,
X.
Chen
, et al
.
2014
.
An autoreactive antibody from an SLE/HIV-1 individual broadly neutralizes HIV-1.
J. Clin. Invest.
124
:
1835
1843
.
12
Moody
,
M. A.
,
I.
Pedroza-Pacheco
,
N. A.
Vandergrift
,
C.
Chui
,
K. E.
Lloyd
,
R.
Parks
,
K. A.
Soderberg
,
A. T.
Ogbe
,
M. S.
Cohen
,
H. X.
Liao
, et al
.
2016
.
Immune perturbations in HIV-1-infected individuals who make broadly neutralizing antibodies.
Sci. Immunol.
1
: aag0851.
13
Schroeder
,
K. M. S.
,
A.
Agazio
,
P. J.
Strauch
,
S. T.
Jones
,
S. B.
Thompson
,
M. S.
Harper
,
R.
Pelanda
,
M. L.
Santiago
,
R. M.
Torres
.
2017
.
Breaching peripheral tolerance promotes the production of HIV-1-neutralizing antibodies.
J. Exp. Med.
214
:
2283
2302
.
14
Wardemann
,
H.
,
S.
Yurasov
,
A.
Schaefer
,
J. W.
Young
,
E.
Meffre
,
M. C.
Nussenzweig
.
2003
.
Predominant autoantibody production by early human B cell precursors.
Science
301
:
1374
1377
.
15
Zikherman
,
J.
,
R.
Parameswaran
,
A.
Weiss
.
2012
.
Endogenous antigen tunes the responsiveness of naive B cells but not T cells.
Nature
489
:
160
164
.
16
Noviski
,
M.
,
J. L.
Mueller
,
A.
Satterthwaite
,
L. A.
Garrett-Sinha
,
F.
Brombacher
,
J.
Zikherman
.
2018
.
IgM and IgD B cell receptors differentially respond to endogenous antigens and control B cell fate.
eLife
7
: e35074.
17
Tan
,
C.
,
J. L.
Mueller
,
M.
Noviski
,
J.
Huizar
,
D.
Lau
,
A.
Dubinin
,
A.
Molofsky
,
P. C.
Wilson
,
J.
Zikherman
.
2019
.
Nur77 links chronic antigen stimulation to B cell tolerance by restricting the survival of self-reactive B cells in the periphery.
J. Immunol.
202
:
2907
2923
.
18
Noviski
,
M.
,
C.
Tan
,
J.
Huizar
,
V.
Vykunta
,
J. L.
Mueller
,
J.
Zikherman
.
2019
.
Optimal development of mature B cells requires recognition of endogenous antigens.
J. Immunol.
203
:
418
428
.
19
Watanabe
,
A.
,
K. Y.
Su
,
M.
Kuraoka
,
G.
Yang
,
A. E.
Reynolds
,
A. G.
Schmidt
,
S. C.
Harrison
,
B. F.
Haynes
,
E. W.
St Clair
,
G.
Kelsoe
.
2019
.
Self-tolerance curtails the B cell repertoire to microbial epitopes.
JCI Insight
4
: 122551.
20
Duty
,
J. A.
,
P.
Szodoray
,
N. Y.
Zheng
,
K. A.
Koelsch
,
Q.
Zhang
,
M.
Swiatkowski
,
M.
Mathias
,
L.
Garman
,
C.
Helms
,
B.
Nakken
, et al
.
2009
.
Functional anergy in a subpopulation of naive B cells from healthy humans that express autoreactive immunoglobulin receptors.
J. Exp. Med.
206
:
139
151
.
21
Quách
,
T. D.
,
N.
Manjarrez-Orduño
,
D. G.
Adlowitz
,
L.
Silver
,
H.
Yang
,
C.
Wei
,
E. C.
Milner
,
I.
Sanz
.
2011
.
Anergic responses characterize a large fraction of human autoreactive naive B cells expressing low levels of surface IgM.
J. Immunol.
186
:
4640
4648
.
22
Moran
,
A. E.
,
K. L.
Holzapfel
,
Y.
Xing
,
N. R.
Cunningham
,
J. S.
Maltzman
,
J.
Punt
,
K. A.
Hogquist
.
2011
.
T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse.
J. Exp. Med.
208
:
1279
1289
.
23
Mombaerts
,
P.
,
J.
Iacomini
,
R. S.
Johnson
,
K.
Herrup
,
S.
Tonegawa
,
V. E.
Papaioannou
.
1992
.
RAG-1-deficient mice have no mature B and T lymphocytes.
Cell
68
:
869
877
.
24
McLellan
,
J. S.
,
M.
Chen
,
M. G.
Joyce
,
M.
Sastry
,
G. B.
Stewart-Jones
,
Y.
Yang
,
B.
Zhang
,
L.
Chen
,
S.
Srivatsan
,
A.
Zheng
, et al
.
2013
.
Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus.
Science
342
:
592
598
.
25
McGuire
,
A. T.
,
M. D.
Gray
,
P.
Dosenovic
,
A. D.
Gitlin
,
N. T.
Freund
,
J.
Petersen
,
C.
Correnti
,
W.
Johnsen
,
R.
Kegel
,
A. B.
Stuart
, et al
.
2016
.
Specifically modified Env immunogens activate B-cell precursors of broadly neutralizing HIV-1 antibodies in transgenic mice.
Nat. Commun.
7
:
10618
.
26
Yassine
,
H. M.
,
J. C.
Boyington
,
P. M.
McTamney
,
C. J.
Wei
,
M.
Kanekiyo
,
W. P.
Kong
,
J. R.
Gallagher
,
L.
Wang
,
Y.
Zhang
,
M. G.
Joyce
, et al
.
2015
.
Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection.
Nat. Med.
21
:
1065
1070
.
27
Pape
,
K. A.
,
J. J.
Taylor
,
R. W.
Maul
,
P. J.
Gearhart
,
M. K.
Jenkins
.
2011
.
Different B cell populations mediate early and late memory during an endogenous immune response.
Science
331
:
1203
1207
.
28
Taylor
,
J. J.
,
R. J.
Martinez
,
P. J.
Titcombe
,
L. O.
Barsness
,
S. R.
Thomas
,
N.
Zhang
,
S. D.
Katzman
,
M. K.
Jenkins
,
D. L.
Mueller
.
2012
.
Deletion and anergy of polyclonal B cells specific for ubiquitous membrane-bound self-antigen.
J. Exp. Med.
209
:
2065
2077
.
29
Taylor
,
J. J.
,
K. A.
Pape
,
M. K.
Jenkins
.
2012
.
A germinal center-independent pathway generates unswitched memory B cells early in the primary response.
J. Exp. Med.
209
:
597
606
.
30
Taylor
,
J. J.
,
K. A.
Pape
,
H. R.
Steach
,
M. K.
Jenkins
.
2015
.
Humoral immunity. Apoptosis and antigen affinity limit effector cell differentiation of a single naïve B cell.
Science
347
:
784
787
.
31
Allman
,
D.
,
R. C.
Lindsley
,
W.
DeMuth
,
K.
Rudd
,
S. A.
Shinton
,
R. R.
Hardy
.
2001
.
Resolution of three nonproliferative immature splenic B cell subsets reveals multiple selection points during peripheral B cell maturation.
J. Immunol.
167
:
6834
6840
.
32
Won
,
W. J.
,
J. F.
Kearney
.
2002
.
CD9 is a unique marker for marginal zone B cells, B1 cells, and plasma cells in mice.
J. Immunol.
168
:
5605
5611
.
33
Tiller
,
T.
,
C. E.
Busse
,
H.
Wardemann
.
2009
.
Cloning and expression of murine Ig genes from single B cells.
J. Immunol. Methods
350
:
183
193
.
34
Wu
,
X.
,
Z. Y.
Yang
,
Y.
Li
,
C. M.
Hogerkorp
,
W. R.
Schief
,
M. S.
Seaman
,
T.
Zhou
,
S. D.
Schmidt
,
L.
Wu
,
L.
Xu
, et al
.
2010
.
Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1.
Science
329
:
856
861
.
35
Pape
,
K. A.
,
R. W.
Maul
,
T.
Dileepan
,
A. S.
Paustian
,
P. J.
Gearhart
,
M. K.
Jenkins
.
2018
.
Naive B cells with high-avidity germline-encoded antigen receptors produce persistent IgM+ and transient IgG+ memory B cells.
Immunity
48
:
1135
1143.e4
.
36
McLellan
,
J. S.
,
Y.
Yang
,
B. S.
Graham
,
P. D.
Kwong
.
2011
.
Structure of respiratory syncytial virus fusion glycoprotein in the postfusion conformation reveals preservation of neutralizing epitopes.
J. Virol.
85
:
7788
7796
.
37
Ashouri
,
J. F.
,
A.
Weiss
.
2017
.
Endogenous Nur77 is a specific indicator of antigen receptor signaling in human T and B cells.
J. Immunol.
198
:
657
668
.
38
Goodnow
,
C. C.
,
J.
Crosbie
,
H.
Jorgensen
,
R. A.
Brink
,
A.
Basten
.
1989
.
Induction of self-tolerance in mature peripheral B lymphocytes.
Nature
342
:
385
391
.
39
Pewzner-Jung
,
Y.
,
D.
Friedmann
,
E.
Sonoda
,
S.
Jung
,
K.
Rajewsky
,
D.
Eilat
.
1998
.
B cell deletion, anergy, and receptor editing in “knock in” mice targeted with a germline-encoded or somatically mutated anti-DNA heavy chain.
J. Immunol.
161
:
4634
4645
.
40
Roark
,
J. H.
,
A.
Bui
,
K. A.
Nguyen
,
L.
Mandik
,
J.
Erikson
.
1997
.
Persistence of functionally compromised anti-double-stranded DNA B cells in the periphery of non-autoimmune mice.
Int. Immunol.
9
:
1615
1626
.
41
Benschop
,
R. J.
,
K.
Aviszus
,
X.
Zhang
,
T.
Manser
,
J. C.
Cambier
,
L. J.
Wysocki
.
2001
.
Activation and anergy in bone marrow B cells of a novel immunoglobulin transgenic mouse that is both hapten specific and autoreactive.
Immunity
14
:
33
43
.
42
Bancroft
,
T.
,
B. L.
DeBuysscher
,
C.
Weidle
,
A.
Schwartz
,
A.
Wall
,
M. D.
Gray
,
J.
Feng
,
H. R.
Steach
,
K. S.
Fitzpatrick
,
M. M.
Gewe
, et al
.
2019
.
Detection and activation of HIV broadly neutralizing antibody precursor B cells using anti-idiotypes.
J. Exp. Med.
216
:
2331
2347
.
43
Kirchenbaum
,
G. A.
,
J. B.
St Clair
,
T.
Detanico
,
K.
Aviszus
,
L. J.
Wysocki
.
2014
.
Functionally responsive self-reactive B cells of low affinity express reduced levels of surface IgM.
Eur. J. Immunol.
44
:
970
982
.
44
Sims
,
G. P.
,
R.
Ettinger
,
Y.
Shirota
,
C. H.
Yarboro
,
G. G.
Illei
,
P. E.
Lipsky
.
2005
.
Identification and characterization of circulating human transitional B cells.
Blood
105
:
4390
4398
.
45
Meroni
,
P. L.
,
P. H.
Schur
.
2010
.
ANA screening: an old test with new recommendations.
Ann. Rheum. Dis.
69
:
1420
1422
.
46
Haynes
,
B. F.
,
J.
Fleming
,
E. W.
St Clair
,
H.
Katinger
,
G.
Stiegler
,
R.
Kunert
,
J.
Robinson
,
R. M.
Scearce
,
K.
Plonk
,
H. F.
Staats
, et al
.
2005
.
Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies.
Science
308
:
1906
1908
.
47
Liu
,
M.
,
G.
Yang
,
K.
Wiehe
,
N. I.
Nicely
,
N. A.
Vandergrift
,
W.
Rountree
,
M.
Bonsignori
,
S. M.
Alam
,
J.
Gao
,
B. F.
Haynes
,
G.
Kelsoe
.
2015
.
Polyreactivity and autoreactivity among HIV-1 antibodies.
J. Virol.
89
:
784
798
.
48
Pugh-Bernard
,
A. E.
,
G. J.
Silverman
,
A. J.
Cappione
,
M. E.
Villano
,
D. H.
Ryan
,
R. A.
Insel
,
I.
Sanz
.
2001
.
Regulation of inherently autoreactive VH4-34 B cells in the maintenance of human B cell tolerance.
J. Clin. Invest.
108
:
1061
1070
.
49
Jenks
,
S. A.
,
E. M.
Palmer
,
E. Y.
Marin
,
L.
Hartson
,
A. S.
Chida
,
C.
Richardson
,
I.
Sanz
.
2013
.
9G4+ autoantibodies are an important source of apoptotic cell reactivity associated with high levels of disease activity in systemic lupus erythematosus.
Arthritis Rheum.
65
:
3165
3175
.
50
Richardson
,
C.
,
A. S.
Chida
,
D.
Adlowitz
,
L.
Silver
,
E.
Fox
,
S. A.
Jenks
,
E.
Palmer
,
Y.
Wang
,
J.
Heimburg-Molinaro
,
Q. Z.
Li
, et al
.
2013
.
Molecular basis of 9G4 B cell autoreactivity in human systemic lupus erythematosus.
J. Immunol.
191
:
4926
4939
.
51
Gitlin
,
A. D.
,
L.
von Boehmer
,
A.
Gazumyan
,
Z.
Shulman
,
T. Y.
Oliveira
,
M. C.
Nussenzweig
.
2016
.
Independent roles of switching and hypermutation in the development and persistence of B lymphocyte memory.
Immunity
44
:
769
781
.
52
Mandl
,
J. N.
,
J. P.
Monteiro
,
N.
Vrisekoop
,
R. N.
Germain
.
2013
.
T cell-positive selection uses self-ligand binding strength to optimize repertoire recognition of foreign antigens.
Immunity
38
:
263
274
.
53
Fulton
,
R. B.
,
S. E.
Hamilton
,
Y.
Xing
,
J. A.
Best
,
A. W.
Goldrath
,
K. A.
Hogquist
,
S. C.
Jameson
.
2015
.
The TCR’s sensitivity to self peptide-MHC dictates the ability of naive CD8(+) T cells to respond to foreign antigens.
Nat. Immunol.
16
:
107
117
.
54
Lam
,
K. P.
,
R.
Kühn
,
K.
Rajewsky
.
1997
.
In vivo ablation of surface immunoglobulin on mature B cells by inducible gene targeting results in rapid cell death.
Cell
90
:
1073
1083
.
55
Reed
,
J. H.
,
J.
Jackson
,
D.
Christ
,
C. C.
Goodnow
.
2016
.
Clonal redemption of autoantibodies by somatic hypermutation away from self-reactivity during human immunization.
J. Exp. Med.
213
:
1255
1265
.
56
Sabouri
,
Z.
,
P.
Schofield
,
K.
Horikawa
,
E.
Spierings
,
D.
Kipling
,
K. L.
Randall
,
D.
Langley
,
B.
Roome
,
R.
Vazquez-Lombardi
,
R.
Rouet
, et al
.
2014
.
Redemption of autoantibodies on anergic B cells by variable-region glycosylation and mutation away from self-reactivity.
Proc. Natl. Acad. Sci. USA
111
:
E2567
E2575
.
57
Zinzow-Kramer
,
W. M.
,
A.
Weiss
,
B. B.
Au-Yeung
.
2019
.
Adaptation by naïve CD4+ T cells to self-antigen-dependent TCR signaling induces functional heterogeneity and tolerance.
Proc. Natl. Acad. Sci. USA
116
:
15160
15169
.
58
Di Niro
,
R.
,
S. J.
Lee
,
J. A.
Vander Heiden
,
R. A.
Elsner
,
N.
Trivedi
,
J. M.
Bannock
,
N. T.
Gupta
,
S. H.
Kleinstein
,
F.
Vigneault
,
T. J.
Gilbert
, et al
.
2015
.
Salmonella infection drives promiscuous B cell activation followed by extrafollicular affinity maturation.
Immunity
43
:
120
131
.

The authors have no financial conflicts of interest.

Supplementary data