• NF-κB family members RelB and cRel are coordinately activated by BAFF and provide distinct survival signals.

  • In vivo and in vitro B-cell developmental defects are observed when both RelB and cRel are deleted.

Targeted deletion of BAFF causes severe deficiency of splenic B cells. BAFF-R is commonly thought to signal to nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB)–inducing kinase dependent noncanonical NF-κB RelB. However, RelB-deficient mice have normal B-cell numbers. Recent studies showed that BAFF also signals to the canonical NF-κB pathway, and we found that both RelB and cRel are persistently activated, suggesting BAFF signaling coordinates both pathways to ensure robust B-cell development. Indeed, we report now that combined loss of these 2 NF-κB family members leads to impaired BAFF-mediated survival and development in vitro. Although single deletion of RelB and cRel was dispensable for normal B-cell development, double knockout mice displayed an early B-cell developmental blockade and decreased mature B cells. Despite disorganized splenic architecture in Relb−/−cRel−/− mice, generation of mixed-mouse chimeras established the developmental phenotype to be B-cell intrinsic. Together, our results indicate that BAFF signals coordinate both RelB and cRel activities to ensure survival during peripheral B-cell maturation.

B-cell development originates in the bone marrow, where hematopoietic stem cell precursors commit to the B-cell lineage and immunoglobulin heavy-chain gene rearrangements occur.1,2  If rearrangement is successful, differentiation into the transitional B-cell compartment occurs. Cells that generate functional B-cell antigen receptors eventually leave the bone marrow and migrate to the spleen to complete their maturation process.3,4  The first B cells to arrive are referred to as transitional 1 (T1) B cells.5,6  T1 B cells are still subject to negative selection, where strong antigenic signals lead to apoptosis. In later transitional stages, some of the transitional B cells (transitional 2 [T2]) are allowed to develop into either mature follicular (FO) B cells, which can recirculate in the periphery, or marginal zone (MZ) B cells, which remain largely sessile.7,8 

The B-cell activation factor receptor belonging to the tumor necrosis factor (TNF) superfamily (BAFF-R, BR3) provides critical survival signals to all splenic B-cell subsets. Targeted deletion of BAFF ligand or BAFF-R results in a partial block at the T1 to T2 transition, resulting in severe deficiency of mature B cells.9,10  BAFF initiates the noncanonical nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) pathway via TRAF3, resulting in the stabilization of NF-κB–inducing kinase (NIK) and activation of a NF-κB essential modulator (NEMO)-independent IKK1 kinase complex. This mediates p100 processing, and nuclear translocation of RelB:p52 dimers.11  Recent human studies have shown that patients with germ-line mutations in NFKB2 have immunodeficiency. In some of the patients, there is a loss of B cells.12-14  It is likely that some of these B-cell developmental defects in the patients result from impaired BAFF-R signaling because of their nonprocessable p100. BAFF has also been reported to activate the canonical NF-κB pathway.15,16  Gene-targeted deletion of NFkB1 (p50), the primary binding partner of RelA and cRel, results in defective survival of B cells in response to BAFF.17  Although neither cRel−/− nor Rela−/− mice show a phenotype in B-cell numbers, doubly deficient B-cell precursors fail to develop the full mature subsets.18  This raises the question of whether the noncanonical NF-κB pathway and RelB play any role at all in safeguarding B-cell development. However, we note that RelA/cRel-deficiency is also known to diminish RelB expression and noncanonical signaling.19-22  The same considerations apply to interpreting other severe knockouts of the canonical pathway such as B-cell-specific NEMO or IKK2 knockouts.23,24  The fact that the Nfkb1−/−Nfkb2−/− mouse shows a phenotype similar to BAFF/BAFF-R–deficient mice (unlike either single mutant) suggests that both pathways may be redundant.11  However, studies of a compound knockout of the 2 transcriptional activators that mediate canonical and noncanonical pathways, respectively, have not been reported.

Here, we show that only cRel and RelB show persistent activation in response to BAFF, and we therefore examine the physiological consequence of their deletion singly or in combination. We find that both provide survival signals, albeit via distinct gene expression programs, and that these complement each other, such that only the doubly deficient mouse shows severe B-cell developmental deficiencies. Deficiencies in mature B-cell subsets are based not solely on survival defects but also a block in differentiation block at the transitional T1 stage that is cell autonomous and can be observed in an ex vivo differentiation assay.

Cell isolation and culture

Spleens were harvested from C57BL/6 wild-type, cRel−/−, Relb−/−, Relb−/−cRel−/−, or IκBα−/−IκBε−/−cRel−/−TNF−/− mice. B-cell isolation performed by anti-CD43 (Ly-48) microbeads and separated on LS column (Miltenyi Biotec) as in previous studies.25-28  Purity was confirmed to be between 92% and 95% (data not shown). Marginal Zone and Follicular B Cell Isolation Kit (Miltenyi Biotec) was used for separation of follicular B cells from whole splenocytes, with a purity of 83% to 87% CD23+ follicular B cells (supplemental Figure 1A, available on the Blood Web site). Complete media consisted of RPMI-1640, 10 mM N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid, 1 mM sodium pyruvate, 1 mM nonessential amino acids, 0.055 mM β-mercaptoethanol, 100 U penicillin/streptomycin, and 0.3 mg/mL glutamine.

Fluorescence-activated cell sorter (FACS) analysis of survival and B-cell development

Purified B cells were cultured in complete media with or without 50 ng/mL recombinant mouse BAFF/BLyS/TNFSF13B (R&D systems 2106BF). At indicated time points, B cells were collected and stained with 7-aminoactinomycin D (7AAD) (Invitrogen A1310). Cells were analyzed for survival using a C6 Accuri flow cytometer (BD Biosciences). B-cell development was obtained from single-cell suspensions of spleens incubated with fluorescently labeled antibodies for 30 minutes at 4°C in staining buffer (phosphate-buffered saline [PBS] with 0.5% bovine serum albumin or 2.5% fetal calf serum). Data were collected on a FACSCalibur or LSR II flow cytometer (BD Biosciences). FACS analysis was performed using FlowJo software (Tree Star Inc.). Antibodies used in this study from eBioscience (unless specified) included reagents specific for the following: CD21 (7G6), B220 (RA3-6B2), CD23 (B3B4), Ly5.1 (A20), Ly5.2 (RA3-6B2), CD93 (AA4.1), immunoglobulin (Ig) D (11-26), and IgM (115-096-020; Jackson Immunoresearch).

Immunohistochemistry

Spleens from wild-type or Relb−/−cRel−/− mice were frozen in optimum cutting temperature compound above liquid nitrogen and kept at −80°C. Five-micron sections were cut from frozen spleens, fixed in cold 4% paraformaldehyde, and allowed to dry. Sections were then hematoxylin and eosin stained or subjected to immunofluorescent labeling. Sections were blocked with 5% fetal bovine serum in PBS and incubated with anti-Moma1, anti-CD3 fluorescein isothiocyanate, and anti-B220 allophycocyanin (eBioscience) for 3 hours or overnight. Washed sections were mounted with Fluoromount-G (eBioscience), and images were captured on a CRI Nuance Multispectral Imaging (Caliper, Hopkinton, MA) system attached to a Nikon E800 fluorescent microscope.

Bone marrow chimeras

Donor bone marrow cells were isolated from femurs of wild-type, cRel−/−, Relb−/−, or Relb−/−cRel−/− mice, mixed in a 50:50 ratio, and injected (∼5 × 106 cells intravenously per mouse) into 2- to 3-month-old C57BL/6 or C57BL/6 x B6.SJL-Ptprca F1 recipients. Before injection, recipient mice were lethally irradiated (1000 rads). Chimeric mice were analyzed 8 weeks after bone marrow reconstitution. Origin and composition of lymphoid cells was determined by Ly5.1 and Ly5.2 markers.

In vitro B-cell differentiation

In vitro differentiation was performed as previously described.29  Briefly, bone marrow cells were cultured in complete Iscove modified Dulbecco medium with 20 ng/mL interleukin (IL) 7 for 3 to 4 days to enrich for IgM+ transitional B cells. Cells were then washed twice with PBS to remove IL-7 and plated with 50 ng/mL recombinant murine BAFF. On subsequent days, cells were stained with anti-B220, IgM, and IgD antibodies to determine maturation.

Biochemical analysis

Electrophoretic mobility shift assays (EMSAs) were conducted as described previously22,30  and modified to include antibodies specific for RelA (Santa Cruz SC372), RelB (SC226), cRel (SC71), or a combination thereof. For EMSAs focusing on cRel-DNA binding activity, nuclear extracts were preincubated with RelA and RelB antibodies for 15 minutes on ice prior to addition of radiolabeled probe to ablate their specific DNA binding activities. Similarly, nuclear extracts were preincubated with RelA and cRel antibodies or RelB and cRel antibodies when RelB- or RelA-DNA binding activity was the focus, respectively (see Figure 1B). Other antibodies used for immunoblotting were Akt (Cell Signaling Technology, CST#4691), phospho-Akt (Ser473) (CST#4060), nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor (IκB) α (SC371), IκBε (SC7155), α-tubulin (SC26), NFkB1/p50 and NFkB2/p52 (Dr Nancy Rice), and Bclxl (Abcam, #ab256).

Figure 1

BAFF-R stimulation triggers persistent RelB and cRel activity to control in vitro survival functions. (A) BAFF, which binds TACI and BAFF-R, is a key regulator of peripheral B-cell maintenance. BAFF-R signaling functions occur primarily in an NIK-dependent, noncanonical NF-κB manner, mediated through RelB activity, whereas TACI triggers NEMO-dependent canonical NF-κB. (B) Identification of activated NF-κB species in B cells stimulated with BAFF for 24 hours by supershift analysis. (C) RelA, RelB, and cRel EMSA (see “Materials and methods”) of B cells are used to reveal their respective activity kinetics following BAFF-R perturbation. (D) Quantification of RelA, RelB, and cRel activities of BAFF-stimulated primary B cells derived from EMSA. Naive is resting, unstimulated B cells; early and late activity correspond to activities following 1 to 5 hours and 24+ hours of BAFF stimulation, respectively. Representative of ≥4 experiments. *P < .05; **P < .01. (E) Summary table of known NF-κB–deficient mouse models and the effect on B-cell populations. Gray boxes denote a B-cell developmental defect. +++, cell numbers equate to typical wild-type level; ++++, greater than wild-type; ++, intermediate level; +, low; -, not present. EMSA for panels B-C representative of at least 3 experiments.

Figure 1

BAFF-R stimulation triggers persistent RelB and cRel activity to control in vitro survival functions. (A) BAFF, which binds TACI and BAFF-R, is a key regulator of peripheral B-cell maintenance. BAFF-R signaling functions occur primarily in an NIK-dependent, noncanonical NF-κB manner, mediated through RelB activity, whereas TACI triggers NEMO-dependent canonical NF-κB. (B) Identification of activated NF-κB species in B cells stimulated with BAFF for 24 hours by supershift analysis. (C) RelA, RelB, and cRel EMSA (see “Materials and methods”) of B cells are used to reveal their respective activity kinetics following BAFF-R perturbation. (D) Quantification of RelA, RelB, and cRel activities of BAFF-stimulated primary B cells derived from EMSA. Naive is resting, unstimulated B cells; early and late activity correspond to activities following 1 to 5 hours and 24+ hours of BAFF stimulation, respectively. Representative of ≥4 experiments. *P < .05; **P < .01. (E) Summary table of known NF-κB–deficient mouse models and the effect on B-cell populations. Gray boxes denote a B-cell developmental defect. +++, cell numbers equate to typical wild-type level; ++++, greater than wild-type; ++, intermediate level; +, low; -, not present. EMSA for panels B-C representative of at least 3 experiments.

Close modal

Transcriptome analysis

Total RNA was extracted from 50 ng/mL BAFF-stimulated wild-type B cells. Messenger RNA was isolated from 2 μg total RNA using oligo (dT) magnetic beads and fragmented at high temperature using divalent cations. Complementary DNA libraries were generated using the Illumina Truseq kits, and quantitation was performed using the Roche Light Cycler 480. Sequencing was performed on Illumina's HiSequation 2000, according to the manufacturer’s recommendations and prepared for RNA-sequencing (RNA-seq) analysis by the Broad Stem Cell Research Center core facility at the University of California, Los Angeles. Reads were aligned to the mouse mm10 genome and RefSeq genes31,32  with Tophat.33  Cufflinks CummRbund34  was used to ascertain differential expression of genes. Gene differential FPKMs were obtained from the cuffdiff program in the Tuxedo RNA-seq analysis suite. RNA-seq data are deposited in the National Center for Biotechnology Information’s Gene Expression Omnibus database (accession number GSE62559).

BAFF induces persistent NF-κB RelB and cRel activities that have complementary roles in mediating B-cell survival

BAFF is known to engage 3 BLyS family receptors, namely, B-cell maturation antigen (BCMA), transmembrane activator and calcium-modulating cyclophilin ligand-interactor protein (TACI), and BAFF-R (also known as BR3 or BLYS receptor),35  and to activate several signaling pathways including the canonical and noncanonical NF-κB pathways, whose transcriptional effectors are RelA/cRel and RelB, respectively (Figure 1A).

We examined which NF-κB transcription factors are activated by BAFF in murine splenic B cells. We used a modified EMSA procedure in which antibodies are used to ablate 2 of 3 activation-domain-containing NF-κB family members (Figure 1B). Stimulation with BAFF ligand resulted in activation of RelA-, cRel-, and RelB-containing dimers, but the time courses reveal that only cRel and RelB show persistent activation: RelA activity peaks at 1 hour and returns to basal levels within 8 hours. In contrast, cRel and RelB activities are not short lived, peaking at ∼3 hours and persisting beyond 24 hours (Figure 1C). Repeated experimental analysis showed that these conclusions were statistically significant (Figure 1D).

Numerous studies have investigated which signaling pathways mediate BAFF’s physiological functions, and these genetic knockout studies are summarized in Figure 1E. Of the 3 known BAFF-binding receptors, expression of BCMA is not observed in naive B-cell populations, and TACI deletion actually results in enhanced B-cell numbers and splenomegaly, whereas BAFF-R deletion phenocopies BAFF deletion in resulting in a near loss of peripheral B cells.36  BAFF activates both canonical and noncanonical signaling branches of the NF-κB system, yet deficiencies in only canonical components (eg, NEMO, RelA/cRel) have been reported to result in defective B-cell development (Figure 1E).37-48  However, the canonical pathway, and RelA in particular, is known to enable noncanonical signaling via NFkB2 and Relb expression20,21  (Figure 1A), suggesting that phenotypes observed in severe canonical deficiencies might be mediated by defects in noncanonical signaling.

As both RelB and cRel dimers are activated persistently by BAFF, we focused on examining their roles in mediating its key function, survival. Whole splenic B cells were isolated from Relb−/−, cRel−/−, and Relb−/−cRel−/− mice and stimulated with BAFF for up to 4 days. In the in vitro survival assay, RelB-deficient B cells respond poorly to 70 hours of BAFF stimulation, and cRel deletion also displays a minor defect in the BAFF response; however, compound deletion of both cRel and RelB resulted in a statistically more severe phenotype (Figure 2A) than single deletion compared with wild-type. We observe a similarly severe survival defect in a homogeneous population of FO (CD23+) B cells isolated from the spleen of wild-type and Relb−/−cRel−/− mice (Figure 2B). These data suggest that BAFF signaling coordinates the functions of both canonical and noncanonical NF-κB transcription factors to promote survival.

Figure 2

RelB and cRel coordinate together to provide proper BAFF-mediated survival signals in vitro. (A) FACS plots of in vitro survival assay of whole splenic wild-type, Relb−/−, cRel−/−, and Relb−/−cRel−/− B cells stimulated with BAFF ligand for 70 hours. Numbers represent the percentage of live cells (7AAD) found in culture. Graphical representation of the FACS plots (right), n = 3. (B) FACS plots of in vitro survival assay of FO (CD23+) wild-type and Relb−/−cRel−/− B cells stimulated with BAFF for 40 hours. (C) RNA-seq analysis from BAFF-stimulated CD23+wild-type, cRel−/−, and Relb−/−cRel−/− B cells at the indicated time points. Genes (517) were upregulated in BAFF-stimulated follicular B cells; 289 of these showed substantial expression defect in B cells lacking both cRel and RelB (middle and bottom panels). Of these, 127 showed expression defects even in the single cRel knockout (middle panel); 162 showed expression defects only in the Relb−/−cRel−/− double knockout (bottom panel). (D) cRel-dependent genes protect cells against cell death. Gene ontology analysis identifies distinct process terms for cRel-dependent vs RelB/cRel-dependent gene clusters. Whereas RelB/cRel-dependent clusters are significantly associated with terms describing metabolic processes, the cRel-dependent cluster shows overrepresentation of negative regulation of cell death/apoptosis. *P < .05; **P < .005; ***P < .001. Also see supplemental Figure 1. (E) List of representative cRel-dependent and RelB/cRel-dependent genes identified as “negative regulators of cell death” and “metabolic process,” respectively, by gene ontology analysis.

Figure 2

RelB and cRel coordinate together to provide proper BAFF-mediated survival signals in vitro. (A) FACS plots of in vitro survival assay of whole splenic wild-type, Relb−/−, cRel−/−, and Relb−/−cRel−/− B cells stimulated with BAFF ligand for 70 hours. Numbers represent the percentage of live cells (7AAD) found in culture. Graphical representation of the FACS plots (right), n = 3. (B) FACS plots of in vitro survival assay of FO (CD23+) wild-type and Relb−/−cRel−/− B cells stimulated with BAFF for 40 hours. (C) RNA-seq analysis from BAFF-stimulated CD23+wild-type, cRel−/−, and Relb−/−cRel−/− B cells at the indicated time points. Genes (517) were upregulated in BAFF-stimulated follicular B cells; 289 of these showed substantial expression defect in B cells lacking both cRel and RelB (middle and bottom panels). Of these, 127 showed expression defects even in the single cRel knockout (middle panel); 162 showed expression defects only in the Relb−/−cRel−/− double knockout (bottom panel). (D) cRel-dependent genes protect cells against cell death. Gene ontology analysis identifies distinct process terms for cRel-dependent vs RelB/cRel-dependent gene clusters. Whereas RelB/cRel-dependent clusters are significantly associated with terms describing metabolic processes, the cRel-dependent cluster shows overrepresentation of negative regulation of cell death/apoptosis. *P < .05; **P < .005; ***P < .001. Also see supplemental Figure 1. (E) List of representative cRel-dependent and RelB/cRel-dependent genes identified as “negative regulators of cell death” and “metabolic process,” respectively, by gene ontology analysis.

Close modal

Next we asked whether the cRel and RelB mediated their complementary functions by inducing the same or different sets of survival genes in B cells. We undertook RNA-seq of BAFF-stimulated FO (CD23+) wild-type, cRel−/−, and Relb−/−cRel−/− B cells. Here, we identified 517 genes upregulated in BAFF-stimulated wild-type B cells, 289 genes of which were substantially reduced in Relb−/−cRel−/− B cells (Figure 2C). Of these, 127 genes were largely cRel dependent, as they showed lower BAFF-induced expression in both cRel−/− or Relb−/−cRel−/− B cells, and 162 were identified as RelB dependent in the context of cRel deficiency and may thus be described as potential RelB target genes. Interestingly, gene ontology analysis focusing on biological process terms identified a large number of receptor-associated and mitochondrial antiapoptotic regulators within the cRel-dependent cluster, whereas metabolic regulators were primarily identified in the RelB/cRel-dependent cluster (Figure 2D-E). These data suggest that coordination of cRel and RelB activities by BAFF serves the expression of complementary sets of genes that ensure B-cell survival. Confirmatory reverse transcription–quantitative polymerase chain reaction showed that Bcl2 family members A1 and Bcl-xL required only cRel but not RelB for their expression, and the reduction is messenger RNA expression also resulted in a reduction in Bcl-xL protein (supplemental Figure 2).

Compound deletion of RelB and cRel results in severe peripheral B-cell developmental defects

As RelB and cRel are critical for BAFF-mediated survival functions in vitro, we examined B-cell development in vivo in mice lacking RelB, cRel, or both. Using an established FACS strategy,49  we confirmed that there is no defect in peripheral B-cell populations in cRel−/− mice and only a modest decrease in Relb−/− mice. However, we found a significant loss of mature B cells and an increase in the transitional compartment in double knockout mice (Figure 3A), whereas T-cell numbers were normal (supplemental Figure 3). Distinguishing mature B-cell subsets, we confirmed previous reports that Relb−/− spleens were deficient in the MZ B-cell population38  but now found that the double knockout mice generate significantly fewer FO B cells (Figure 3B; Table 1).

Figure 3

Compound deletion of RelB and cRel results in developmental block at the T1 stage. (A-C) FACS analysis of peripheral B-cell development in whole splenic extracts from wild-type, Relb−/−, cRel−/−, and Relb−/−cRel−/− mice. Identification of mature (B220+AA4.1) and transitional B cells (B220+AA4.1+). Mature B cells (B220+AA4.1) are further classified into FO B cells (CD21+IgM+) and mature MZ B cells (CD21highIgM+). Transitional B cells (B220+AA4.1+) are subdivided into T1 B cells (CD23IgM+) and T2 B cells (CD23+IgM+). Scatter plots are graphical representation of FACS plots: wild-type (●), Relb−/− (▪), cRel−/− (▲), and Relb−/−cRel−/− (▼). (D) Histologic analysis of splenic sections taken from wild-type and Relb−/−cRel−/− mouse using hematoxylin and eosin stain (H&E). (E) In vivo analysis of mature B-cell development in double knockout mouse using immunofluorescence of frozen splenic sections stained with anti-CD3 fluorescein isothiocyanate, anti-B220 allophycocyanin, and anti-MOMA-1 Alexa Fluor 405. *P < .05; **P < .005; ***P < .001. Also see supplemental Figure 2.

Figure 3

Compound deletion of RelB and cRel results in developmental block at the T1 stage. (A-C) FACS analysis of peripheral B-cell development in whole splenic extracts from wild-type, Relb−/−, cRel−/−, and Relb−/−cRel−/− mice. Identification of mature (B220+AA4.1) and transitional B cells (B220+AA4.1+). Mature B cells (B220+AA4.1) are further classified into FO B cells (CD21+IgM+) and mature MZ B cells (CD21highIgM+). Transitional B cells (B220+AA4.1+) are subdivided into T1 B cells (CD23IgM+) and T2 B cells (CD23+IgM+). Scatter plots are graphical representation of FACS plots: wild-type (●), Relb−/− (▪), cRel−/− (▲), and Relb−/−cRel−/− (▼). (D) Histologic analysis of splenic sections taken from wild-type and Relb−/−cRel−/− mouse using hematoxylin and eosin stain (H&E). (E) In vivo analysis of mature B-cell development in double knockout mouse using immunofluorescence of frozen splenic sections stained with anti-CD3 fluorescein isothiocyanate, anti-B220 allophycocyanin, and anti-MOMA-1 Alexa Fluor 405. *P < .05; **P < .005; ***P < .001. Also see supplemental Figure 2.

Close modal
Table 1

Summary of peripheral B-cell development in wild-type, Relb−/−, cRel−/−, and Relb−/−cRel−/− mice

PhenotypeTotal B cells (×106)
wild-typeRelb−/−cRel−/−Relb−/−cRel−/−
Total cells 75.4 ± 17 131 ± 15** 76.1 ± 8.3 104 ± 25* 
Total B cells 51.3 ± 6.9 39.8 ± 8.6* 41.6 ± 4.9 29.1 ± 7.8*** 
Mature 32.8 ± 6.7 23.3 ± 7.1* 30.1 ± 4.4 14.3 ± 4.2* 
Transitional 9.28 ± 1.9 7.85 ± 1.7 9.86 ± 2.5 12.7 ± 2.7* 
T1 3.00 ± 1.2 3.17 ± 1.2 3.58 ± 0.94 6.30 ± 2.0** 
T2 5.42 ± 0.64 5.21 ± 2.0 5.54 ± 1.3 6.48 ± 2.5 
MZ 3.1 ± 1.1 0.067 ± 0.052**** 3.2 ± 0.071 0.31 ± 0.34**** 
Follicular 25.3 ± 6.3 22.4 ± 5.1 24.1 ± 4.4 12.5 ± 3.6*** 
PhenotypeTotal B cells (×106)
wild-typeRelb−/−cRel−/−Relb−/−cRel−/−
Total cells 75.4 ± 17 131 ± 15** 76.1 ± 8.3 104 ± 25* 
Total B cells 51.3 ± 6.9 39.8 ± 8.6* 41.6 ± 4.9 29.1 ± 7.8*** 
Mature 32.8 ± 6.7 23.3 ± 7.1* 30.1 ± 4.4 14.3 ± 4.2* 
Transitional 9.28 ± 1.9 7.85 ± 1.7 9.86 ± 2.5 12.7 ± 2.7* 
T1 3.00 ± 1.2 3.17 ± 1.2 3.58 ± 0.94 6.30 ± 2.0** 
T2 5.42 ± 0.64 5.21 ± 2.0 5.54 ± 1.3 6.48 ± 2.5 
MZ 3.1 ± 1.1 0.067 ± 0.052**** 3.2 ± 0.071 0.31 ± 0.34**** 
Follicular 25.3 ± 6.3 22.4 ± 5.1 24.1 ± 4.4 12.5 ± 3.6*** 

****P < .0001, ***P < .001, **P < .01, *P < .05.

Dissection of transitional B-cell populations revealed an increased proportion of transitional T1 but not T2 cells (Figure 3C). T1 B cells are the initial class of B cells that give rise to the mature populations.50  In addition, we examined the B-cell maturation in the bone marrow of the double knockout and found no discernible defect compared with wild-type controls (supplemental Figure 4). The increase in T1 B cells found in the double knockout mouse suggested to us that RelB and cRel are required not only for the survival of developing B cells but also for peripheral B-cell development per se. Although the Relb−/−cRel−/− developmental phenotype is not as severe as that caused by BAFF deficiency, the block at the T1 stage is reminiscent.37  Further, histologic analysis corroborated these findings: in the Relb−/−cRel−/− spleen, the white pulp, although present, was not as dense (Figure 3D). The reduction in white pulp may be caused by reduced numbers of B cells in the spleen, as our FACS analysis suggested, or it may be a consequence of improper homing of the lymphocytes causing them to be dispersed throughout the spleen. To address this, we investigated the organization of B cells using immunofluorescence. Frozen sections of splenic tissue were stained with anti-CD3 and anti-B220 to identify the T-cell zone and B-cell zone, respectively. MOMA-1 (which stains MZ metallophilic macrophages) outlined the follicular rim surrounding the B-cell–rich zone. In agreement with our FACS data, the presence of B cells was greatly diminished in the Relb−/−cRel−/− spleen, as shown by the staining of B220 (red; Figure 3E). We also observed that the characteristic ringlike structure of MOMA-1 staining (blue) is consistently less organized in the double knockout mouse, suggestive of a defective splenic architecture, which may51,52  or may not contribute to the B-cell development phenotype in the mutant mice.

B-cell intrinsic developmental defect in vivo

We studied whether the B-cell developmental phenotype is in fact B-cell intrinsic by generating mixed bone marrow chimeras, where bone marrow–derived hematopoietic stem cells from both wild-type and mutant donor mice were injected into a lethally irradiated wild-type or mutant recipient mice. To distinguish cells derived from either donor, we used the CD45 congenic marker system, in which donor cells expressed the CD45.1 or CD45.2 variant and recipient mice were CD45.1.2 (supplemental Figure 5A-B).

Within this experimental system, we found that Relb−/−cRel−/− (CD45.2) hematopoietic stem cells were compromised in their development even when wild-type recipients provided a normal milieu of architecture and soluble factors (Figure 4A-B) akin to observations made in cRel- and RelB-deficient animals (Figure 3A-C). Again, the transitional B-cell populations are higher than the wild-type controls, whereas FO populations were severely decreased and MZ B cells were undetectable (Figure 4A-B). As a reference, we also generated chimeras with RelB and cRel single knockout bone marrow cells, with no observable phenotype (supplemental Figure 5C-D). When wild-type and Relb−/−cRel−/− donor bone marrow stem cells were injected into lethally irradiated Relb−/−cRel−/− recipient mice, wild-type donor stem cells were able to mature normally from T1 to T2, FO, and MZ B cells, whereas Relb−/−cRel−/− donor stem cells matured poorly (similar to when they were in wild-type host) (Figure 4C-D). These data provide strong evidence that the B-cell developmental phenotype of Relb−/−cRel−/− mice is in fact cell intrinsic, and not caused by improper splenic architecture or cytokine milieu.

Figure 4

B-cell developmental defect displayed in Relb−/−cRel−/− mouse is B-cell intrinsic. (A and C) Schematic for the generation of CD45.1 wild-type and CD45.2 Relb−/−cRel−/− mixed bone marrow chimeras. Bone marrow stem cells from CD45.1 wild-type and CD45.2 Relb−/−cRel−/− are mixed in a 50:50 ratio and then injected into a lethally irradiated CD45.1.2 wild-type or Relb−/−cRel−/− mouse. (B and D) Graphical plots of percentages of specific subsets relative to total B cells in indicated mixed bone marrow chimeras, as described previously. The gating strategy used in the analysis is depicted in Figure 3. Representative FACS plots see supplemental Figure 3. For panel B, n = 5; for panel D, n = 2. *P < .05; **P < .005; ***P < .001.

Figure 4

B-cell developmental defect displayed in Relb−/−cRel−/− mouse is B-cell intrinsic. (A and C) Schematic for the generation of CD45.1 wild-type and CD45.2 Relb−/−cRel−/− mixed bone marrow chimeras. Bone marrow stem cells from CD45.1 wild-type and CD45.2 Relb−/−cRel−/− are mixed in a 50:50 ratio and then injected into a lethally irradiated CD45.1.2 wild-type or Relb−/−cRel−/− mouse. (B and D) Graphical plots of percentages of specific subsets relative to total B cells in indicated mixed bone marrow chimeras, as described previously. The gating strategy used in the analysis is depicted in Figure 3. Representative FACS plots see supplemental Figure 3. For panel B, n = 5; for panel D, n = 2. *P < .05; **P < .005; ***P < .001.

Close modal

Relb−/−cRel−/− progenitors are defective in BAFF-mediated B-cell development in vitro

A true cell intrinsic phenotype is expected to be recapitulated in an in vitro differentiation assay in which all nonintrinsic components are held equal. We employed an in vitro B-cell development assay system described previously.11,53  In this system, transitional B cells are generated from bone marrow progenitors in the presence of IL-7. The cytokine is washed away, then recultured with BAFF ligand (Figure 5A). Expression of B-cell markers CD23 and CD21 has been reported to be regulated by BAFF.54  Further, FACS analysis suggested that RelB plays a role in CD21 expression (Figure 3B); thus we used IgD and B220 as our primary developmental markers in these analyses. Exposure of wild-type bone marrow cultures to BAFF for 4 days increased the fraction of IgD+ cells relative to untreated controls, demonstrating BAFF’s capacity to enhance B-cell development in vitro. In contrast, BAFF stimulation failed to increase the percentage of IgD+ cells present in Relb−/−cRel−/− bone marrow cultures (Figure 5B-C). Even in the absence of BAFF, the numbers of IgD+ cells were consistently lower in mutant cultures compared with wild-type. In fact, the reduction in B-cell numbers in this assay may be dominated by a BAFF-dependent survival defect (Figure 5D). Together, these experimental results further illustrate the necessity of RelB and cRel in BAFF-mediated B-cell maturation.

Figure 5

Relb−/−cRel−/− B-cell progenitors fail to respond to BAFF-mediated developmental signal in vitro. (A) Schematic of in vitro B-cell differentiation system. (B) Representative FACS plots of B220 and IgD expression in transitional B cells at the beginning (day 0) and end (day 4) of culture with or without BAFF. (C) Mean frequency of B220+IgD+ (T2-like) B cells following 4 days of culture as described in panel B. Numbers represent frequencies of live B220+ B cells in indicated gates. (D) Survival of in vitro cultured B cells. Live cells were identified by gating out 7AADHi population. Fold survival calculated from initial B-cell population (day 0). For FACS plots, n = 4. wt, wild-type.

Figure 5

Relb−/−cRel−/− B-cell progenitors fail to respond to BAFF-mediated developmental signal in vitro. (A) Schematic of in vitro B-cell differentiation system. (B) Representative FACS plots of B220 and IgD expression in transitional B cells at the beginning (day 0) and end (day 4) of culture with or without BAFF. (C) Mean frequency of B220+IgD+ (T2-like) B cells following 4 days of culture as described in panel B. Numbers represent frequencies of live B220+ B cells in indicated gates. (D) Survival of in vitro cultured B cells. Live cells were identified by gating out 7AADHi population. Fold survival calculated from initial B-cell population (day 0). For FACS plots, n = 4. wt, wild-type.

Close modal

Deregulating NF-κB accelerates splenic B-cell development

Given that NF-κB RelB/cRel activities are required for B-cell maturation past the T1 stage (Figure 3), we asked whether elevated NF-κB might be sufficient to accelerate B-cell maturation. Compound deficiency of IκBα and IκBε leads to elevation of both constitutive and stimulus-responsive NF-κB activity,55  inflammatory gene expression, and neonatal death.56  We restored viability by introducing compound cRel and TNF deficiencies, leaving RelA and RelB deregulated, and examined the B-cell compartment. Although the total splenocytes count is lower in IκBα−/−IκBε−/−cRel−/−TNF−/− mice than wild-type controls, the proportion of B220+ cells is relatively normal (∼40% to 50%). Interestingly, in contrast to hypo-NF-κB activity in Relb−/−cRel−/− mice, hyper-NF-κB activity in IκBα−/−IκBε−/−cRel−/−TNF−/− mice leads to reduced T1 cell numbers and a preponderance of MZ B cells (Figure 6). Although the reduced B-cell numbers also indicate restrictions in bone marrow developmental events, these observations are indicative of accelerated splenic B-cell development.

Figure 6

Deregulated NF-κB activity in IκBα−/−IκBε−/−cRel−/−TNF−/− leads to reduction in T1 cells and increased MZ B-cell generation. FACS analysis of peripheral B-cell development in whole splenic extracts from wild-type, cRel−/−, Relb−/−cRel−/−, and IκBα−/−IκBε-/cRel−/−TNF−/− mice. The gating strategy used in the analysis is depicted in Figure 3. Numbers represent percentage of denoted subsets as proportion of total B cells. Representative FACS results of 3 experiments shown.

Figure 6

Deregulated NF-κB activity in IκBα−/−IκBε−/−cRel−/−TNF−/− leads to reduction in T1 cells and increased MZ B-cell generation. FACS analysis of peripheral B-cell development in whole splenic extracts from wild-type, cRel−/−, Relb−/−cRel−/−, and IκBα−/−IκBε-/cRel−/−TNF−/− mice. The gating strategy used in the analysis is depicted in Figure 3. Numbers represent percentage of denoted subsets as proportion of total B cells. Representative FACS results of 3 experiments shown.

Close modal

For more than a decade, numerous reports have focused on the characterization of noncanonical NF-κB signaling in response to the BAFF signaling pathway,17,24,57  yet only mutants of the canonical pathway have BAFF−/−-like phenotypes. In this study, we report that the canonical NF-κB effectors cRel and RelA are activated following BAFF stimulation. Further, although RelA activation is transient, cRel activation is persistent and thus mimics the activation profile of RelB, the well-known effector of the noncanonical pathway.

Indeed, both cRel and RelB play a role in BAFF functions ex vivo, as either singly deficient B cell showed reduced BAFF-responsive survival. Remarkably, however, these survival functions appear to be mediated by different gene expression programs, with cRel being required for the expression of known prosurvival genes A1 and Bcl-xL,16,58  but Relb−/− B-cells showing no defects in their expression. Whereas unbiased transcriptome analysis of RelB-deficient B cells is compromised by the confounding secondary effects of systemic inflammation in the Relb−/− mouse,39 Relb−/−cRel−/− mice, which do not show an inflammatory phenotype, allowed us to identify the RelB-dependent gene expression program in the context of cRel deficiency. The complementary nature of the cRel- and RelB-dependent expression programs manifests itself in the fact that double deletion leads to a more severe B-cell survival defect not only ex vivo (Figure 2) but also in vivo (Figure 3).

Further, we found that double deletion of RelB and cRel results in a block in splenic B-cell development at the T1 to T2 stage. These mutant mice are not only deficient in mature cells but possess a twofold increase in the percentage of transitional B cells and almost fourfold enhancement in the population of T1 cells. This developmental blockade at the T1 to T2 stage mimics the phenotype of BAFF knockout mice, although the defect is not quite as severe, suggesting a role for RelA, or an NF-κB-independent mechanism. This developmental defect is B-cell intrinsic, as Relb−/−cRel−/− bone marrow stem cells failed to develop normally in mixed bone marrow chimeras and wild-type hematopoietic stem cells are capable of proper B-cell development in lethally irradiated Relb−/−cRel−/− mice.

How does BAFF coordinately activate cRel and RelB? Three TNF receptor superfamily members bind BAFF, namely, BCMA, TACI, and BAFF-R (also known as BR3 or BLYS receptor),51  with BAFF-R and TACI being expressed on peripheral B-cell subsets.59  TACI activates canonical NF-κB signaling,60  but TACI-deficient mice show no defect on B-cell development and B cells are capable of degrading IκBα in response to BAFF.15  Our results are consistent with BAFF-R triggering both canonical, IκBα-mediated NF-κB signaling and also noncanonical, IκBδ-mediated activation of RelA and cRel. As a key survival receptor, BAFF-R signaling maintains the B-cell pool. In contrast, TACI acts as a negative regulator of B-cell homeostasis, indirectly regulating peripheral B-cell numbers by competing for the amount of BAFF available for signaling through BAFF-R.51,61 

CD21 expression is reduced in the absence of RelB, and further decreased in the combined absence of RelB and cRel (Figure 3B). CD21 is known to be controlled by NF-κB-p52.62,63  Considering our data, CD21 is probably controlled by RelB:p52 complex during development, and cRel:p52 to a lesser extent (possibly providing compensation). Given that CD21 is essential in humoral immune response,64,65  it is likely RelB-deficient B cells will have impaired response to certain T-dependent antigens, especially complement-tagged antigens.66-68  Further functional studies are needed to address this.

Previous studies suggested that NF-κB regulates BAFF-R expression,69,70  although none of the NF-κB knockout studies cited in Figure 1E had examined BAFF-R expression. We examined BAFF-R expression in 2 ways: replicate RNA-seq did not show an expression defect in Relb−/−cRel−/− cells over wild-type controls (supplemental Figure 6A), thereby not corroborating transcriptional control by RelB/cRel. However, by FACS, BAFF-R staining was reduced (supplemental Figure 6B) and BAFF-responsive AKT phosphorylation (indicative of phosphatidylinositol 3-kinase activation) was also lower (supplemental Figure 6C). Together, this suggests that the phenotype of Relb−/−cRel−/− B cells may in part be reinforced by a reduction in BAFF-R surface expression. As BAFF-R is transcribed at the normal rate, this reduction is not a direct consequence of RelB/cRel deficiency, but probably rather a consequence of cell-biological deficiencies that hinder B-cell maturation from the T1 stage. Further, although the loss of cRel and RelB reduces the amount of p100, as observed elsewhere,71  BAFF-responsive noncanonical NFkB2/p100 degradation remained intact (supplemental Figure 6C).

The coordinate activation of cRel and RelB renders BAFF’s physiological responses relatively robust, as the single knockout shows little in vivo defect. At the same time, cell-context-specific control of the 2 branches of NF-κB signaling and activation of their respective gene expression programs may allow for fine tuning of BAFF’s broad physiological functions in controlling survival, B-cell maturation, cell growth, and even proliferation. Matching quantitative studies of signaling strength and dynamics to gene expression programs and cell biological level analyses may lead to a better understanding how BAFF functions in vivo and enable its pharmacologic manipulation in the clinical setting.

The data reported in this article have been deposited in the Gene Expression Omnibus database (accession number GSE62559).

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The authors thank L. Delacruz for irradiating donor mice and University of California, San Diego histology core at Moores Cancer Center for sectioning of frozen spleens for immunofluorescence. The authors also thank V. Shih and S. Basak for experimental advice and S. Hedrick, C. Murre, and R. Rickert for critical discussions.

This work was supported by grants from the National Institutes of Health, National Institute of Allergy and Infectious Diseases (R01 AI083453), National Cancer Institute (R01 CA141722), and National Institute of General Medical Sciences (R01 GM071573, P50 GM085764) (A.H.); and a Cell and Molecular Genetics training grant (J.V.A.).

Contribution: J.V.A., Y.C.L., A.H., M.A., M.D., and A.W.G. designed the research and interpreted data; J.V.A. and Y.C.L. performed all B-cell biochemistry; J.V.A., D.C.O., and M.D. performed immunofluorescence; J.V.A. and Y.C.L. performed in vitro B-cell development; J.V.A., H.B., and J.D.-T. carried out RNA-sequencing analysis; J.V.A., E.Y., M.A., and A.W.G. designed and made bone marrow chimeras; and J.V.A., Y.C.L., and A.H. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Alexander Hoffmann, University of California, Los Angeles, Los Angeles, CA 90095; e-mail: ahoffmann@ucla.edu.

1
Hardy
 
RR
Carmack
 
CE
Shinton
 
SA
Kemp
 
JD
Hayakawa
 
K
Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow.
J Exp Med
1991
, vol. 
173
 
5
(pg. 
1213
-
1225
)
2
Hardy
 
RR
Hayakawa
 
K
A developmental switch in B lymphopoiesis.
Proc Natl Acad Sci USA
1991
, vol. 
88
 
24
(pg. 
11550
-
11554
)
3
Allman
 
DM
Ferguson
 
SE
Cancro
 
MP
Peripheral B cell maturation. I. Immature peripheral B cells in adults are heat-stable antigenhi and exhibit unique signaling characteristics.
J Immunol
1992
, vol. 
149
 
8
(pg. 
2533
-
2540
)
4
Allman
 
DM
Ferguson
 
SE
Lentz
 
VM
Cancro
 
MP
Peripheral B cell maturation. II. Heat-stable antigen(hi) splenic B cells are an immature developmental intermediate in the production of long-lived marrow-derived B cells.
J Immunol
1993
, vol. 
151
 
9
(pg. 
4431
-
4444
)
5
Carsetti
 
R
Köhler
 
G
Lamers
 
MC
Transitional B cells are the target of negative selection in the B cell compartment.
J Exp Med
1995
, vol. 
181
 
6
(pg. 
2129
-
2140
)
6
Loder
 
F
Mutschler
 
B
Ray
 
RJ
, 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
, vol. 
190
 
1
(pg. 
75
-
90
)
7
Gerondakis
 
S
Siebenlist
 
U
Roles of the NF-kappaB pathway in lymphocyte development and function.
Cold Spring Harb Perspect Biol
2010
, vol. 
2
 
5
pg. 
a000182
 
8
Pillai
 
S
Cariappa
 
A
The follicular versus marginal zone B lymphocyte cell fate decision.
Nat Rev Immunol
2009
, vol. 
9
 
11
(pg. 
767
-
777
)
9
Harless Smith
 
S
Cancro
 
MP
BLyS: the pivotal determinant of peripheral B cell selection and lifespan.
Curr Pharm Des
2003
, vol. 
9
 
23
(pg. 
1833
-
1847
)
10
Schiemann
 
B
Gommerman
 
JL
Vora
 
K
, et al. 
An essential role for BAFF in the normal development of B cells through a BCMA-independent pathway.
Science
2001
, vol. 
293
 
5537
(pg. 
2111
-
2114
)
11
Claudio
 
E
Brown
 
K
Park
 
S
Wang
 
H
Siebenlist
 
U
BAFF-induced NEMO-independent processing of NF-kappa B2 in maturing B cells.
Nat Immunol
2002
, vol. 
3
 
10
(pg. 
958
-
965
)
12
Chen
 
K
Coonrod
 
EM
Kumánovics
 
A
, et al. 
Germline mutations in NFKB2 implicate the noncanonical NF-κB pathway in the pathogenesis of common variable immunodeficiency.
Am J Hum Genet
2013
, vol. 
93
 
5
(pg. 
812
-
824
)
13
Lindsley
 
AW
Qian
 
Y
Valencia
 
CA
Shah
 
K
Zhang
 
K
Assa’ad
 
A
Combined immune deficiency in a patient with a novel NFKB2 mutation.
J Clin Immunol
2014
, vol. 
34
 
8
(pg. 
910
-
915
)
14
Lee
 
CE
Fulcher
 
DA
Whittle
 
B
, et al. 
Autosomal-dominant B-cell deficiency with alopecia due to a mutation in NFKB2 that results in nonprocessable p100.
Blood
2014
, vol. 
124
 
19
(pg. 
2964
-
2972
)
15
Shinners
 
NP
Carlesso
 
G
Castro
 
I
, et al. 
Bruton’s tyrosine kinase mediates NF-kappa B activation and B cell survival by B cell-activating factor receptor of the TNF-R family [published correction appears in J Immunol. 2007;179(9):6369].
J Immunol
2007
, vol. 
179
 
6
(pg. 
3872
-
3880
)
16
Stadanlick
 
JE
Kaileh
 
M
Karnell
 
FG
, et al. 
Tonic B cell antigen receptor signals supply an NF-kappaB substrate for prosurvival BLyS signaling.
Nat Immunol
2008
, vol. 
9
 
12
(pg. 
1379
-
1387
)
17
Hatada
 
EN
Do
 
RK
Orlofsky
 
A
, et al. 
NF-kappa B1 p50 is required for BLyS attenuation of apoptosis but dispensable for processing of NF-kappa B2 p100 to p52 in quiescent mature B cells.
J Immunol
2003
, vol. 
171
 
2
(pg. 
761
-
768
)
18
Grossmann
 
M
O’Reilly
 
LA
Gugasyan
 
R
Strasser
 
A
Adams
 
JM
Gerondakis
 
S
The anti-apoptotic activities of Rel and RelA required during B-cell maturation involve the regulation of Bcl-2 expression.
EMBO J
2000
, vol. 
19
 
23
(pg. 
6351
-
6360
)
19
Basak
 
S
Shih
 
VF
Hoffmann
 
A
Generation and activation of multiple dimeric transcription factors within the NF-kappaB signaling system.
Mol Cell Biol
2008
, vol. 
28
 
10
(pg. 
3139
-
3150
)
20
Bren
 
GD
Solan
 
NJ
Miyoshi
 
H
Pennington
 
KN
Pobst
 
LJ
Paya
 
CV
Transcription of the RelB gene is regulated by NF-kappaB.
Oncogene
2001
, vol. 
20
 
53
(pg. 
7722
-
7733
)
21
Lombardi
 
L
Ciana
 
P
Cappellini
 
C
, et al. 
Structural and functional characterization of the promoter regions of the NFKB2 gene.
Nucleic Acids Res
1995
, vol. 
23
 
12
(pg. 
2328
-
2336
)
22
Shih
 
VF
Davis-Turak
 
J
Macal
 
M
, et al. 
Control of RelB during dendritic cell activation integrates canonical and noncanonical NF-κB pathways.
Nat Immunol
2012
, vol. 
13
 
12
(pg. 
1162
-
1170
)
23
Pasparakis
 
M
Schmidt-Supprian
 
M
Rajewsky
 
K
IkappaB kinase signaling is essential for maintenance of mature B cells.
J Exp Med
2002
, vol. 
196
 
6
(pg. 
743
-
752
)
24
Sasaki
 
Y
Derudder
 
E
Hobeika
 
E
, et al. 
Canonical NF-kappaB activity, dispensable for B cell development, replaces BAFF-receptor signals and promotes B cell proliferation upon activation.
Immunity
2006
, vol. 
24
 
6
(pg. 
729
-
739
)
25
Almaden
 
JV
Tsui
 
R
Liu
 
YC
, et al. 
A pathway switch directs BAFF signaling to distinct NFκB transcription factors in maturing and proliferating B cells.
Cell Reports
2014
, vol. 
9
 
6
(pg. 
2098
-
2111
)
26
Shokhirev
 
MN
Almaden
 
J
Davis-Turak
 
J
, et al. 
A multi-scale approach reveals that NF-κB cRel enforces a B-cell decision to divide.
Mol Syst Biol
2015
, vol. 
11
 
2
pg. 
783
 
27
Alves
 
BN
Tsui
 
R
Almaden
 
J
, et al. 
IκBε is a key regulator of B cell expansion by providing negative feedback on cRel and RelA in a stimulus-specific manner.
J Immunol
2014
, vol. 
192
 
7
(pg. 
3121
-
3132
)
28
Shokhirev
 
MN
Hoffmann
 
A
FlowMax: a computational tool for maximum likelihood deconvolution of CFSE time courses.
PLoS One
2013
, vol. 
8
 
6
pg. 
e67620
 
29
Rowland
 
SL
Leahy
 
KF
Halverson
 
R
Torres
 
RM
Pelanda
 
R
BAFF receptor signaling aids the differentiation of immature B cells into transitional B cells following tonic BCR signaling.
J Immunol
2010
, vol. 
185
 
8
(pg. 
4570
-
4581
)
30
Werner
 
SL
Barken
 
D
Hoffmann
 
A
Stimulus specificity of gene expression programs determined by temporal control of IKK activity.
Science
2005
, vol. 
309
 
5742
(pg. 
1857
-
1861
)
31
Kent
 
WJ
Sugnet
 
CW
Furey
 
TS
, et al. 
The human genome browser at UCSC.
Genome Res
2002
, vol. 
12
 
6
(pg. 
996
-
1006
)
32
Waterston
 
RH
Lindblad-Toh
 
K
Birney
 
E
, et al. 
Mouse Genome Sequencing Consortium
Initial sequencing and comparative analysis of the mouse genome.
Nature
2002
, vol. 
420
 
6915
(pg. 
520
-
562
)
33
Trapnell
 
C
Pachter
 
L
Salzberg
 
SL
TopHat: discovering splice junctions with RNA-Seq.
Bioinformatics
2009
, vol. 
25
 
9
(pg. 
1105
-
1111
)
34
Trapnell
 
C
Roberts
 
A
Goff
 
L
, et al. 
Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks.
Nat Protoc
2012
, vol. 
7
 
3
(pg. 
562
-
578
)
35
Day
 
ES
Cachero
 
TG
Qian
 
F
, et al. 
Selectivity of BAFF/BLyS and APRIL for binding to the TNF family receptors BAFFR/BR3 and BCMA.
Biochemistry
2005
, vol. 
44
 
6
(pg. 
1919
-
1931
)
36
Yan
 
M
Wang
 
H
Chan
 
B
, et al. 
Activation and accumulation of B cells in TACI-deficient mice.
Nat Immunol
2001
, vol. 
2
 
7
(pg. 
638
-
643
)
37
Harless
 
SM
Lentz
 
VM
Sah
 
AP
, et al. 
Competition for BLyS-mediated signaling through Bcmd/BR3 regulates peripheral B lymphocyte numbers.
Curr Biol
2001
, vol. 
11
 
24
(pg. 
1986
-
1989
)
38
Weih
 
DS
Yilmaz
 
ZB
Weih
 
F
Essential role of RelB in germinal center and marginal zone formation and proper expression of homing chemokines.
J Immunol
2001
, vol. 
167
 
4
(pg. 
1909
-
1919
)
39
Weih
 
F
Carrasco
 
D
Durham
 
SK
, et al. 
Multiorgan inflammation and hematopoietic abnormalities in mice with a targeted disruption of RelB, a member of the NF-kappa B/Rel family.
Cell
1995
, vol. 
80
 
2
(pg. 
331
-
340
)
40
Köntgen
 
F
Grumont
 
RJ
Strasser
 
A
, et al. 
Mice lacking the c-rel proto-oncogene exhibit defects in lymphocyte proliferation, humoral immunity, and interleukin-2 expression.
Genes Dev
1995
, vol. 
9
 
16
(pg. 
1965
-
1977
)
41
Alcamo
 
E
Mizgerd
 
JP
Horwitz
 
BH
, et al. 
Targeted mutation of TNF receptor I rescues the RelA-deficient mouse and reveals a critical role for NF-kappa B in leukocyte recruitment.
J Immunol
2001
, vol. 
167
 
3
(pg. 
1592
-
1600
)
42
Sha
 
WC
Liou
 
HC
Tuomanen
 
EI
Baltimore
 
D
Targeted disruption of the p50 subunit of NF-kappa B leads to multifocal defects in immune responses.
Cell
1995
, vol. 
80
 
2
(pg. 
321
-
330
)
43
Caamaño
 
JH
Rizzo
 
CA
Durham
 
SK
, et al. 
Nuclear factor (NF)-kappa B2 (p100/p52) is required for normal splenic microarchitecture and B cell-mediated immune responses.
J Exp Med
1998
, vol. 
187
 
2
(pg. 
185
-
196
)
44
Franzoso
 
G
Carlson
 
L
Xing
 
L
, et al. 
Requirement for NF-kappaB in osteoclast and B-cell development.
Genes Dev
1997
, vol. 
11
 
24
(pg. 
3482
-
3496
)
45
Miosge
 
LA
Blasioli
 
J
Blery
 
M
Goodnow
 
CC
Analysis of an ethylnitrosourea-generated mouse mutation defines a cell intrinsic role of nuclear factor kappaB2 in regulating circulating B cell numbers.
J Exp Med
2002
, vol. 
196
 
8
(pg. 
1113
-
1119
)
46
Kaisho
 
T
Takeda
 
K
Tsujimura
 
T
, et al. 
IkappaB kinase alpha is essential for mature B cell development and function.
J Exp Med
2001
, vol. 
193
 
4
(pg. 
417
-
426
)
47
Senftleben
 
U
Cao
 
Y
Xiao
 
G
, et al. 
Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway.
Science
2001
, vol. 
293
 
5534
(pg. 
1495
-
1499
)
48
Jellusova
 
J
Miletic
 
AV
Cato
 
MH
, et al. 
Context-specific BAFF-R signaling by the NF-κB and PI3K pathways.
Cell Reports
2013
, vol. 
5
 
4
(pg. 
1022
-
1035
)
49
Allman
 
D
Pillai
 
S
Peripheral B cell subsets.
Curr Opin Immunol
2008
, vol. 
20
 
2
(pg. 
149
-
157
)
50
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
, vol. 
167
 
12
(pg. 
6834
-
6840
)
51
Mackay
 
F
Schneider
 
P
Cracking the BAFF code.
Nat Rev Immunol
2009
, vol. 
9
 
7
(pg. 
491
-
502
)
52
Rickert
 
RC
Jellusova
 
J
Miletic
 
AV
Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease.
Immunol Rev
2011
, vol. 
244
 
1
(pg. 
115
-
133
)
53
Rowland
 
SL
DePersis
 
CL
Torres
 
RM
Pelanda
 
R
Ras activation of Erk restores impaired tonic BCR signaling and rescues immature B cell differentiation.
J Exp Med
2010
, vol. 
207
 
3
(pg. 
607
-
621
)
54
Gorelik
 
L
Cutler
 
AH
Thill
 
G
, et al. 
Cutting edge: BAFF regulates CD21/35 and CD23 expression independent of its B cell survival function.
J Immunol
2004
, vol. 
172
 
2
(pg. 
762
-
766
)
55
Kearns
 
JD
Basak
 
S
Werner
 
SL
Huang
 
CS
Hoffmann
 
A
IkappaBepsilon provides negative feedback to control NF-kappaB oscillations, signaling dynamics, and inflammatory gene expression.
J Cell Biol
2006
, vol. 
173
 
5
(pg. 
659
-
664
)
56
Goudeau
 
B
Huetz
 
F
Samson
 
S
, et al. 
IkappaBalpha/IkappaBepsilon deficiency reveals that a critical NF-kappaB dosage is required for lymphocyte survival.
Proc Natl Acad Sci USA
2003
, vol. 
100
 
26
(pg. 
15800
-
15805
)
57
Enzler
 
T
Bonizzi
 
G
Silverman
 
GJ
, et al. 
Alternative and classical NF-kappa B signaling retain autoreactive B cells in the splenic marginal zone and result in lupus-like disease.
Immunity
2006
, vol. 
25
 
3
(pg. 
403
-
415
)
58
Castro
 
I
Wright
 
JA
Damdinsuren
 
B
, et al. 
B cell receptor-mediated sustained c-Rel activation facilitates late transitional B cell survival through control of B cell activating factor receptor and NF-kappaB2.
J Immunol
2009
, vol. 
182
 
12
(pg. 
7729
-
7737
)
59
Stadanlick
 
JE
Cancro
 
MP
BAFF and the plasticity of peripheral B cell tolerance.
Curr Opin Immunol
2008
, vol. 
20
 
2
(pg. 
158
-
161
)
60
Mackay
 
F
Schneider
 
P
TACI, an enigmatic BAFF/APRIL receptor, with new unappreciated biochemical and biological properties.
Cytokine Growth Factor Rev
2008
, vol. 
19
 
3-4
(pg. 
263
-
276
)
61
Bossen
 
C
Cachero
 
TG
Tardivel
 
A
, et al. 
TACI, unlike BAFF-R, is solely activated by oligomeric BAFF and APRIL to support survival of activated B cells and plasmablasts.
Blood
2008
, vol. 
111
 
3
(pg. 
1004
-
1012
)
62
Debnath
 
I
Roundy
 
KM
Weis
 
JJ
Weis
 
JH
Analysis of the regulatory role of BAFF in controlling the expression of CD21 and CD23.
Mol Immunol
2007
, vol. 
44
 
9
(pg. 
2388
-
2399
)
63
Debnath
 
I
Roundy
 
KM
Weis
 
JJ
Weis
 
JH
Defining in vivo transcription factor complexes of the murine CD21 and CD23 genes.
J Immunol
2007
, vol. 
178
 
11
(pg. 
7139
-
7150
)
64
Molina
 
H
Holers
 
VM
Li
 
B
, et al. 
Markedly impaired humoral immune response in mice deficient in complement receptors 1 and 2.
Proc Natl Acad Sci USA
1996
, vol. 
93
 
8
(pg. 
3357
-
3361
)
65
Thiel
 
J
Kimmig
 
L
Salzer
 
U
, et al. 
Genetic CD21 deficiency is associated with hypogammaglobulinemia.
J Allergy Clin Immunol
2012
, vol. 
129
 
3
(pg. 
801
-
810
)
66
Haas
 
KM
Hasegawa
 
M
Steeber
 
DA
, et al. 
Complement receptors CD21/35 link innate and protective immunity during Streptococcus pneumoniae infection by regulating IgG3 antibody responses.
Immunity
2002
, vol. 
17
 
6
(pg. 
713
-
723
)
67
Cherukuri
 
A
Cheng
 
PC
Sohn
 
HW
Pierce
 
SK
The CD19/CD21 complex functions to prolong B cell antigen receptor signaling from lipid rafts.
Immunity
2001
, vol. 
14
 
2
(pg. 
169
-
179
)
68
Cherukuri
 
A
Cheng
 
PC
Pierce
 
SK
The role of the CD19/CD21 complex in B cell processing and presentation of complement-tagged antigens.
J Immunol
2001
, vol. 
167
 
1
(pg. 
163
-
172
)
69
Yuan
 
H
Wang
 
Y
Wu
 
X
, et al. 
Characterization of the 5′-flanking region and regulation of transcription of human BAFF-R gene.
DNA Cell Biol
2010
, vol. 
29
 
3
(pg. 
133
-
139
)
70
Woo
 
YJ
Yoon
 
BY
Jhun
 
JY
, et al. 
Regulation of B cell activating factor (BAFF) receptor expression by NF-ΚB signaling in rheumatoid arthritis B cells.
Exp Mol Med
2011
, vol. 
43
 
6
(pg. 
350
-
357
)
71
Fusco
 
AJ
Savinova
 
OV
Talwar
 
R
Kearns
 
JD
Hoffmann
 
A
Ghosh
 
G
Stabilization of RelB requires multidomain interactions with p100/p52.
J Biol Chem
2008
, vol. 
283
 
18
(pg. 
12324
-
12332
)

Author notes

*

J.V.A. and Y.C.L. contributed equally to this study.

Sign in via your Institution