Surrogate light chain expression during B lineage differentiation was examined by using indicator fluorochrome-filled liposomes in an enhanced immunofluorescence assay. Pro-B cells bearing surrogate light chain components were found in mice, but not in humans. A limited subpopulation of relatively large pre-B cells in both species expressed pre-B cell receptors. These cells had reduced expression of the recombinase activating genes, RAG-1 and RAG-2. Their receptor-negative pre-B cell progeny were relatively small, expressed RAG-1 and RAG-2, and exhibited selective down-regulation of VpreB and λ5expression. Comparative analysis of the 2 pre-B cell subpopulations indicated that loss of the pre-B cell receptors from surrogate light chain gene silencing was linked with exit from the cell cycle and light chain gene rearrangement to achieve B-cell differentiation.

Since the discovery that B cells belong to a discrete lymphoid lineage,1 much has been learned about this pathway of cellular differentiation (reviewed in2-5). B lymphopoiesis occurs in hemopoietic tissues, primarily embryonic liver and bone marrow in mammals. In these sites, lymphoid progenitors lacking immunoglobulin (Ig) expression (pro-B cells) give rise to large B lymphocyte precursors (pre-B cells) containing μ heavy chains (HCs).6-9 The replicating pre-B cells later exit the cell cycle to generate small pre-B cells that in turn become IgM-bearing B cells.10-14 The Ig V(D)J gene rearrangement events underlying this progression follow an orderly sequence in which D-JH rearrangement is followed by V-DJH rearrangement in pro-B cells.15-17 Pre-B cells generated through a productive VDJH rearrangement undergo several rounds of cell division before exiting the cell cycle. V-JL rearrangement in the kappa or lambda light chain (LC) loci preferentially occurs in these small resting pre-B cells.18-20 A productive VJL rearrangement allows the assembly of B-cell receptors (BCRs), composed of μHC, LC, and the Igα/β heterodimeric signal-transducing elements and their expression on newly formed B cells.21 22 

Differential expression of a large cohort of genes is required for the progression of B lineage differentiation,5,17,23 and the nonrearranging VpreB and λ5/14.1genes24-27 are central figures in this differentiation process. Their expression begins in pro-B cells and is maintained into the pre-B cell stage.28-31 The VpreB and λ5 protein products are immediately assembled to form surrogate light chains (SLCs) having an overall structure resembling conventional LCs.32,33 This structural similarity allows the SLC to bind nascent μHC proteins, thereby liberating them from the BiP retention protein in the endoplasmic reticulum.29,34 The μHC homodimers then associate with Igα/β heterodimers to allow transit of the newly assembled pre-BCR through the Golgi apparatus en route to the pre-B cell surface.35,36 Pre-BCRs are concentrated within lipid rafts, where they are constitutively associated with syk, lyn, BLNK, and PI-3 kinase signaling elements.37 The importance of SLC during early B lineage differentiation is evidenced by a block in pro-B to pre-B cell differentiation in mice and humans with nonfunctionalλ538,39 or VpreBmutations.40 Deficiencies in any of the other pre-BCR components41-44 or in the essential downstream signaling elements (reviewed in 45) also lead to abortive pro-B cell differentiation. Pre-BCR and its signaling competency are thus essential for growth and survival of the pre-B cell population.

SLCs, the unique components of pre-BCR, are easily detected on pre-B cell lines of mouse and human origin46-49 and on murine pro-B cell lines,48 but their cell surface expression by primary B lineage cells has been remarkably difficult to demonstrate.49-57 This difficulty has clouded the issue of exactly when and where SLCs exert their function during B lineage differentiation. Even in studies in which pre-BCR expression has been demonstrated, only a few pre-B cells could be identified by cell surface staining with anti-SLC antibodies.49,50,54,56,58The most easily identifiable SLC-bearing cells in mice have in fact been pro-B cells,53 whereas in human B lineage cells the appearance of cell surface SLCs has not been detectable before the onset of μHC expression in most studies.49,56,59 With rare exceptions,59,60 human pro-B cell lines have also been reported to lack cell surface SLCs.49,55,56 58Species differences may thus exist regarding when cell surface SLC expression begins. Given the very low levels at which SLC-containing receptors are expressed on primary B lineage cells, technical differences and variability in the antibody reagents used for SLC detection could also account for some of the discordant results.

The paradox posed by our understanding of the importance of pre-BCR in B lineage differentiation versus the uncertainty about when pro-BCR and pre-BCR are expressed led us to use an amplified immunofluorescence assay that can detect as few as 50 to 100 molecules per cell61 and a panel of monoclonal anti-VpreB/λ5 antibodies to readdress the issue of SLC expression during B lineage differentiation. The comparative analysis of mice and humans clearly indicates species variability in pro-BCR expression and, more important, reveals a conserved mode of selective SLC down-regulation to permit pre-B cells to exit the cell cycle to undergo B-cell differentiation. Models for mouse and human B cell differentiation indicated by this analysis have interesting implications for normal and neoplastic B lymphopoiesis.

Cells

Human pro-B (Nalm16, RS4;11, REH, and JEA2), pre-B (697, Nalm6, and OB5), and B cell lines (Daudi and Ramos) were cultured in RPMI 1640 medium containing 100 U/mL penicillin, 100 μg/mL streptomycin, 10% heat-inactivated fetal calf serum, 50 μM 2-mercaptoethanol, and 2 mM L-glutamine (Life Technologies, Grand Island, NY). Murine pro-B cell lines (Raw8.1, SCID7, 38B9, 40E1, 63-12, and D1F9) and the 70Z/3 pre-B cell line were grown in supplemented Dulbecco modified Eagle medium. SCID7 cultures contained interleukin-7 (1 ng/mL). Human bone marrow cells were obtained from ribs resected from donors of renal transplants and from long bones of 13- to 19-week-old previable fetuses in accordance with policies established by the UAB Institutional Review Board. Mouse bone marrow was obtained from 4- to 12-week-old Balb/c, C57BL/6, RAG-1−/−, RAG-2−/−, and μMT mice. Mononuclear cells were isolated from human bone marrow by Ficoll-Hypaque gradient centrifugation and from murine bone marrow after erythrocyte lysis by treatment with 0.8% ammonium chloride.

Antibodies

The SA-DA4.4 anti-human μHC, anti-human VpreB8,56anti-human VpreB (HSL96, 4G7), anti-human λ5 (HSL11), and anti-human pre-BCR (HSL2),58,59 anti-mouse VpreB (R3 and R5),62 and anti-mouse λ5 (LM34)48monoclonal antibodies (mAbs) were digoxigenin-labeled following the manufacturer's recommendations (Boehringer Mannheim, Germany). Fab fragments of sheep anti-digoxigenin antibodies were obtained from Boehringer Mannheim; the FcR blocking reagent was from Miltenyi Biotec (Auburn, CA); phycoerythrin (PE)-conjugated anti-human CD19, CD79b, and allophycoerythrin (APC)-conjugated anti-CD34 antibodies were from Becton Dickinson (Mountain View, CA); streptavidin-APC and biotin-conjugated goat antibodies to human μHC, λLC, and κLC were from Southern Biotechnology Associates (Birmingham, AL); mouse B220, CD19, CD24, CD43, BP-1, CD25, CD2, κLC, λLC, CD79a, CD79b, and CD16/CD32 (Fc Block) monoclonal antibodies were from BD PharMingen (San Diego, CA); and goat antibodies to mouse μHC were from Southern Biotechnology Associates and Jackson Immunoresearch (West Grove, PA). Fluorescein isothiocyanate (FITC)–conjugated monoclonal antibodies to mouse κLC (187.1) and λLC (JC5.1) were gifts from Dr John Kearney (University of Alabama at Birmingham).

Immunofluorescence analysis and cell sorting

The enhanced indirect immunofluorescence system61used fluorochrome-filled liposomes conjugated to Fab fragments of sheep antidigoxigenin antibodies as a second-step reagent. Viable cells pre-incubated with 200 μg/mL IgG and 20 μg/mL anti-FcγIIR antibody in phosphate-buffered saline containing 0.5% bovine serum albumin for 10 minutes at 4°C were incubated with a digoxigenin-conjugated anti-VpreB, anti-λ5, or anti–pre-BCR antibodies for 15 minutes before washing and incubation with antidigoxigenin conjugated fluorescent liposomes for 1 hour on ice with agitation, followed by washing and analysis by flow cytometry. Staining specificity was assessed by pre-incubation with a 100- to 1000-fold excess of the unlabeled primary antibody. B lineage cells enriched by magnetic cell sorting (Miltenyi Biotec) of bone marrow using anti-human CD19 or anti-mouse B220 antibodies were sorted using FACStar (Becton Dickinson) or MoFlow (Cytomation, Fort Collins, CO) instruments. Cells fixed by incubation in 0.05% paraformaldehyde solution at 4°C for 1 hour were permeabilized by treatment with 0.2% Tween 20 or 0.1% saponin in phosphate-buffered saline at room temperature for 20 minutes and were blocked with serum for 10 minutes before antibodies were added for intracellular staining.

Cell cycle analysis

Cells purified by immunofluorescence cell sorting were fixed in 95% ice-cold ethanol for more than 30 minutes before treatment with RNase A (50 μg/mL) for 30 minutes at 37°C, followed by staining with FITC-conjugated anti–Ki-67 or control antibodies (BD PharMingen) for 30 minutes on ice. The cells were then washed before incubation, with 40 μg/mL propidium iodide for 15 minutes at room temperature, and flow immunocytometric analysis.

Reverse transcription–polymerase chain reaction assays

Subpopulations of pro-B and pre-B cells purified by 2 sequential fluorescence-activated cell sorting (FACS) sorts were lysed in TRIzol reagent (Gibco, Grand Island, NY) before preparation of total RNA, as recommended by the manufacturer (Gibco). First-strand cDNA synthesis was performed using the SuperScript preamplification system (Life Technologies) in parallel with a control synthesis reaction without reverse transcriptase (RT) to test for genomic DNA contamination. Protocols for polymerase chain reaction (PCR) of human gene products involved denaturing at 94°C for 3 minutes, amplification by 36 cycles of 94°C for 1 minute, 30 seconds for annealing at 55°C for RAG-1 and RAG-2, 60°C for TdT and B29, or 65°C forβ-actin and VDJ-Cμ, 72°C for 30 seconds, and extension at 72°C for 5 minutes. Primers for PCR amplification were as follows: TdT,5′-ACACGAATGCAGAAAGCAGGA-3′, 5′-AGGCAACCTGAGCTTTTCAAA-3′;RAG-1, 5′-ATGACAGCAGATGACCTCCTA-3′, 5′-TACCTCCAGAAGTTTATGAAT-3′; RAG-2,5′-TTCTTGGCATACCAGGAGACA-3′, 5′-CTATTTGCTTCTGCACTGAAA-3′;λ5/14.1, 5′-ACTGTCGGATCCTCGCAGAGCAGG-3′, 5′-CAGTCAAGCTTCTATGAACATTCT-3′; VpreB,5′-GTAGAGGCATGCCAGCCGGTGCTG-3′, 5′-CTTGAAGCTTTCGAGGGACACGTGT-3′;B29, 5′-GAATCTCTCGCCACCCTCACC-3′, 5′-CCTTGCTGTCATCCTTGTCCA-3′; VDJ-Cμ,5′-GGGTCGACACGGCCGTGTATTACTGT-3′, 5′-TGGTGGCAGCAAGTAGACATC-3′; andβ-actin, 5′-GCGGGAAATCGTGCGTGACAT-3′, 5′-GTGGACTTGGGAGAGGACTGG-3′. Primers used for Vλ-Jλ PCR amplification have been previously described.63 PCR protocols for mouse gene products involved 36 cycles of amplification with annealing at 58°C for β-actin, VDJ-Cμ, B29,and λ5; 60°C for RAG-2; and 65°C forTdT, RAG-1, and VpreB for 1 minute followed by extension at 72°C for 1 minute and 94°C denaturation for 1 minute. Primers for mouse PCR amplification were as follows: TdT,5′-GAAGATGGGAACAACTCGAAGAG-3′, 5′-CAGGTGCTGGAACATTCTGGGAG-3′;RAG-1, 5′-TGAAAAGGCACCCGAAGAAGC-3′, 5′-GGTGCCACTCCACGGTCACTT-3′; RAG-2,5′-CACATCCACAAGCAGGAAGTACAC-3′, 5′-GGTTCAGGGACATCTCCTACTAAG-3′;λ5, 5′-GTTGGGTCTAGTGGATGGTGT-3′, 5′-TTGGTCTGTTTGGAGGGTTGG-3′; VpreB,5′-GCCACCATCCGCCTCTCCTGT-3′, 5′-CCCCACGGCACAGTAATACAG-3′; B29,5′-TCAGAAGAGGGACGCATTGTG-3′, 5′-TTCAAGCCCTCATAGGTGTGA-3′;VDJ-Cμ, 5′-CGCGCGGCCGCTGCAGCAGCCTGGGGC TGAG-3′, 5′-GGAATGGGCACATGCAGATCTC-3′; Vk-Ck,5′-GGCTGCAGSTTCAGTGGCAGTGGRTCWGGRAC-3′, 5′-CTCATTCCTGTTGAAGCTCTTGACAATGGG-3′; and β-actin,5′-CGCAGCTCAGTAACAGTC-3′, 5′-TACGAGGGCTATGCTCTC-3′. Cellular cDNA was serially diluted for template use in semiquantitative RT-PCR assays of VpreB and λ5 transcripts.

Surrogate light chain expression on B lineage cell lines of human and mouse origin

Pro-B, pre-B, and B cell lines were examined with conventional and enhanced indirect immunofluorescence assays in a preliminary survey of SLC expression during human and mouse B lineage differentiation. In the enhanced immunofluorescence assay, cells were incubated first with anti-SLC antibodies conjugated with digoxigenin and then with fluorochrome-loaded liposomes bearing antidigoxigenin antibodies. This method yielded approximately a 2-log enhancement of the mean fluorescence intensity for anti-VpreB and anti-λ5 staining of human and mouse pre-B cells over that observed by conventional indirect immunofluorescence (Figure 1). Specificity was verified by the inhibition of staining with unconjugated anti-VpreB or anti-λ5 antibodies. SLC components were also detected on mouse pro-B cell lines, with similar enhancement seen with the fluorochrome-loaded liposomes. In contrast, SLC components could not be detected on human pro-B cell lines (Figure 1) despite their production of intracellular VpreB and λ5 SLC proteins.56 58 This species distinction held for all mouse (SCID7, 38B9, 40E1, 63-12, D1F9, Raw8.1) and human (Nalm16, RS4;11, JEA2, and REH) pro-B cell lines included in the analysis. Cell surface expression of SLC on mouse pro-B cell lines versus its absence on human pro-B cell lines was confirmed using the entire panel of monoclonal antibodies against VpreB (HSL96, 4G7, VP245) and λ5/14.1 epitopes (HSL11, LM34). When mouse and human pre-B cell lines were examined for cell surface reactivity with anti-μHC antibodies, they were universally found to express this pre-BCR component, whereas none of the pro-B cell lines produced μHC (data not shown).

Fig. 1.

Analysis of SLC expression on human and mouse cell lines.

Pro-B and pre-B cell lines were analyzed for cell surface binding of anti-VpreB and anti-λ5 antibodies by conventional indirect immunofluorescence (dashed line) or by an enhanced immunofluorescence method using fluorochrome-loaded liposomes as a second-step reagent (solid line). Test cells incubated with an excess of unlabeled primary antibodies served as staining controls (shaded histograms). Results illustrated in this and subsequent figures used the VpreB8 anti-human VpreB, R3 anti-mouse Vpre-B, HSL11 anti-human λ5, and LM34 anti-mouse λ5 monoclonal antibodies. Comparable results were obtained with the other anti-VpreB, anti-λ5, and anti-preBCR antibodies used in these studies (see “Materials and methods”).

Fig. 1.

Analysis of SLC expression on human and mouse cell lines.

Pro-B and pre-B cell lines were analyzed for cell surface binding of anti-VpreB and anti-λ5 antibodies by conventional indirect immunofluorescence (dashed line) or by an enhanced immunofluorescence method using fluorochrome-loaded liposomes as a second-step reagent (solid line). Test cells incubated with an excess of unlabeled primary antibodies served as staining controls (shaded histograms). Results illustrated in this and subsequent figures used the VpreB8 anti-human VpreB, R3 anti-mouse Vpre-B, HSL11 anti-human λ5, and LM34 anti-mouse λ5 monoclonal antibodies. Comparable results were obtained with the other anti-VpreB, anti-λ5, and anti-preBCR antibodies used in these studies (see “Materials and methods”).

Close modal

Analysis of surrogate light chain expression by primary B lineage cells

Normal B lineage cells identifiable by their expression of the human CD19 or mouse B220 markers were examined for SLC expression using conventional and enhanced immunofluorescence staining methods. Although the usual difficulty was encountered in detecting SLC expression on bone marrow cells by conventional immunofluorescence, SLC-bearing B lineage cells were easily detected by the enhanced immunofluorescence method in human and mouse bone marrow samples (Figure2). Specificity of this immunofluorescence staining was validated by the results of blocking experiments with unlabeled antibody. The remarkable contrast between the staining results achieved with both methods was emphasized by the fact that we were unable to unambiguously detect SLC expression on mouse bone marrow cells by conventional indirect immunofluorescence (Figure 2B, lower panels). The frequency of SLC+ B lineage cells among bone marrow mononuclear cells identified by the enhanced immunofluorescence method was relatively low (range, 0.6%-4.8%).

Fig. 2.

Enhanced and conventional indirect immunofluorescence analysis of cell surface SLC expression by B lineage cells.

(A) CD19+ cells in human bone marrow and (B) B220+ cells in mouse bone marrow were examined with digoxigenin-labeled anti-VpreB and anti-λ5 mAbs and antidigoxigenin-coated fluorochrome-loaded liposomes as a second-step reagent or with biotinylated anti-VpreB mAb and streptavidin-PE.

Fig. 2.

Enhanced and conventional indirect immunofluorescence analysis of cell surface SLC expression by B lineage cells.

(A) CD19+ cells in human bone marrow and (B) B220+ cells in mouse bone marrow were examined with digoxigenin-labeled anti-VpreB and anti-λ5 mAbs and antidigoxigenin-coated fluorochrome-loaded liposomes as a second-step reagent or with biotinylated anti-VpreB mAb and streptavidin-PE.

Close modal

Enhanced immunofluorescence was used for the detection of the VpreB and λ5/14.1 components, and it was used with conventional indirect immunofluorescence to detect other lineage markers to determine the stages during which primary B lineage cells express cell surface SLC. The SLC+ subpopulation of the CD19+ B lineage cells in human bone marrow varied in frequency from 18.1% ± 1.9% (mean ± SE, n = 4) in fetal samples to 4.4% ± 0.5% (n = 6) in adult samples (Figure 3). In the experiments illustrated here the less sensitive, conventional immunofluorescence method was used for μHC detection, but the modest upward deflection observed with anti-μHC staining, especially for cells with the highest VpreB expression levels, suggested coordinate μHC expression by the SLC+ cells. By comparison, a complete lack of staining of VpreB+ cells was observed with the anti-κ/λ light chain antibodies (Figure 3A). Coexpression of μHC and VpreB by pre-B cells was also indicated in parallel experiments in which equivalent percentages of μHC+ and VpreB+ cells (14.2%± 1.5% vs 14.9%± 1.6%, mean ± SE; n = 3) were identified among CD19+ κ/λLC fetal bone marrow cells using the enhanced liposome method. Although the SLC+ B lineage cells in human bone marrow were predominately CD34, a subpopulation of the pre-B cells expressed this early hematopoietic cell marker at relatively low levels. The existence of a minor CD34+SLC+ subpopulation was confirmed by analysis of bone marrow cells with other anti-VpreB (HSL96, 4G7), anti-λ5 (HSL11), and anti-pre-BCR (HSL2) antibodies (data not shown). Notably, the proportion of SLC+CD19+ cells that retained CD34 expression was much greater in fetal bone marrow than in adult bone marrow (approximately 40% vs less than 5%).

Fig. 3.

Phenotypic characterization of SLC-bearing B lineage cells in bone marrow.

(A) Human adult or fetal cells and (B) mouse cells from wild-type or Rag-2−/− juvenile mice were incubated with, respectively, anti-CD19 or anti-B220 antibodies, plus digoxigenin-labeled anti-VpreB and antidigoxigenin-coated fluorochrome-loaded liposomes, before counterstaining with fluorochrome-labeled antibodies against CD34, μHC, or κ/λLC (A) or CD19, CD43, BP-1, or κ/λLC (B). Viable CD19+ cells (A) or B220+ cells (B) were electronically gated for this analysis. Coexpression of μHC by VpreB+ cells, indicated by the upward shift observed with conventional immunofluorescence (A, middle panels), was confirmed in parallel experiments in which the liposome-enhanced assay was also used to detect cell surface μHC expression (see text).

Fig. 3.

Phenotypic characterization of SLC-bearing B lineage cells in bone marrow.

(A) Human adult or fetal cells and (B) mouse cells from wild-type or Rag-2−/− juvenile mice were incubated with, respectively, anti-CD19 or anti-B220 antibodies, plus digoxigenin-labeled anti-VpreB and antidigoxigenin-coated fluorochrome-loaded liposomes, before counterstaining with fluorochrome-labeled antibodies against CD34, μHC, or κ/λLC (A) or CD19, CD43, BP-1, or κ/λLC (B). Viable CD19+ cells (A) or B220+ cells (B) were electronically gated for this analysis. Coexpression of μHC by VpreB+ cells, indicated by the upward shift observed with conventional immunofluorescence (A, middle panels), was confirmed in parallel experiments in which the liposome-enhanced assay was also used to detect cell surface μHC expression (see text).

Close modal

In bone marrow samples from 6 juvenile mice, the SLC+subset comprised 6.3 ± 1.5% of the B220+ B lineage cells. As in humans, all SLC+ cells in mice were CD19+, and none expressed κ or λ LCs. The heterogeneous CD43 and BP-1 expression patterns observed for mouse SLC+cells (Figure 3B) suggested that primary pro-B and pre-B cells in mice can express SLC on their cell surface, since CD43 expression is restricted to the pro-B and early pre-B cell stages.17Given that the BP-1 antigen is selectively expressed by pre-B and immature B cells,64 the detection of cell surface SLC expression on BP-1 cells also suggested that primary pro-B cells may express SLC-containing receptors. Conversely, the SLC expression on CD43/BP-1+ cells is indicative of pre-B cell receptor expression. The composite results thus suggest that pre-B cells are the only SLC-bearing cells in human bone marrow, whereas in the mouse pro-B cells and pre-B cells may express cell surface SLC.

Mouse, but not human, pro-B cells express cell surface surrogate light chain

To test the implied species variability in SLC expression, SLC-bearing cells in human bone marrow were purified by cell sorting on the basis of cell surface CD19 and VpreB expression and the absence of κ or λ LC. When permeabilized before immunofluorescence analysis, the CD19+VpreB+LC cells were found to contain μHC (Figure 4A, top panel), thereby indicating that all SLC-bearing cells in human bone marrow samples are pre-B cells, whereas pro-B cells lack cell surface SLC.

Fig. 4.

Analysis of intracellular μHC expression by pre-BCR+ and pre-BCR subpopulations.

(A) Human VpreB+ and VpreBCD34subpopulations of CD19+κ/λLC B lineage cells and (B) mouse VpreB+ and VpreBsubpopulations of B-lineage cells from bone marrow samples were purified by immunofluorescence cell sorting before cell permeabilization and analysis of intracellular μHC expression. Dashed lines indicate intracellular μHC staining; solid lines indicate background fluorescence.

Fig. 4.

Analysis of intracellular μHC expression by pre-BCR+ and pre-BCR subpopulations.

(A) Human VpreB+ and VpreBCD34subpopulations of CD19+κ/λLC B lineage cells and (B) mouse VpreB+ and VpreBsubpopulations of B-lineage cells from bone marrow samples were purified by immunofluorescence cell sorting before cell permeabilization and analysis of intracellular μHC expression. Dashed lines indicate intracellular μHC staining; solid lines indicate background fluorescence.

Close modal

Most of the B220+VpreB+ LC cells in mouse bone marrow also contained μHC. However, a discrete subpopulation of these cells (approximately 25%) did not contain μHC (Figure 4B, top right panel). This finding is concordant with the cell line data and with earlier reports indicating that primary pro-B cells in mice can express plasma membrane SLC in association with surrogate HC proteins.48 To verify that primary B lineage cells in mice can express cell surface SLC in the absence of μHC, bone marrow samples from RAG-1−/− and RAG-2−/− mice were examined. In these experiments, cell surface SLC expression was demonstrable for approximately 55% of the CD19+ cells, all of which were also CD43+ (Figure 3B, right panels). These results confirm the expression of SLC on a subpopulation of mouse pro-B cells.

Pre-BCR expression is limited to a subpopulation of pre-B cells

Previous studies using conventional indirect immunofluorescence have identified pre-BCR components on few, if any, of the μHC+ pre-B cells.49-51,53 55-57 It seemed possible that this reflected the relative insensitivity of the methods used for detecting pre-BCR expression. To our surprise, even with the fluorochrome-filled liposomes, we were unable to detect SLC components on most pre-B cells in human and murine bone marrow samples. When the cell surface SLC subpopulation of CD19+CD34 κ/λLC cells was isolated from human bone marrow samples and examined for intracellular μHC, they were found to contain μHC as a clear indication of their pre-B cell status (Figure 4A, lower panel). Correspondingly, when the subpopulation of mouse B220+LC bone marrow cells lacking cell surface VpreB was isolated, μHC expression was found in most, but not all, of these cells (Figure 4B, lower panel). This subpopulation of B220+VpreBLCμHCcells represents pro-B and possibly non-B lineage cells that lack cell surface SLC. These composite results indicate that most of the pre-B cells in mice and humans (60%-80%) lack pre-BCR.

Characterization of the pre-BCR+ and pre-BCR subpopulations of pre-B cells

Human pre-B cell subpopulations, VpreB+CD19+κ/λLC and VpreBCD34CD19+κ/λLC, were purified by 2 rounds of cell sorting. Analysis of these subpopulations indicated that most of the pre-BCR+subpopulation (ie, VpreB+) were relatively large cells, whereas the pre-BCR subpopulation of pre-B cells was composed primarily of relatively small cells (Figure5). In accordance with the cell size difference, the analysis of Ki-67 expression and DNA content indicated that a greater proportion of the pre-BCR+ pre-B cells were in the G1/S/G2/M stages of the cell cycle. The same trends were observed for the pre-BCR+ and pre-BCR subpopulations of mouse pre-B cells, though the strategy for identifying these subpopulations in mouse bone marrow samples was necessarily different. The mouse pre-B cell population of B220+ cells was identified by expression of the BP-1 antigen in the absence of κ/λLC expression. This strategy was used because all BP-1+/LC cells were shown to express intracellular μHC.64 Thirty percent of the latter subpopulation expressed cell surface IgM after overnight culture, thereby confirming that these cells are immediate B cell precursors. Pre-BCR expression is therefore restricted to a subpopulation of relatively large cycling pre-B cells, whereas receptor-negative pre-B cells are predominantly small, resting pre-B cells.

Fig. 5.

Cell size and cell cycle analysis of pre-BCR+ and pre-BCR subpopulations.

(A) Human VpreB+ and VpreBCD34subpopulations of CD19+κ/λLC B lineage cells and (B) mouse CD19+BP-1+κ/λLC pre-B cells were sorted on the basis of positive or negative cell surface VpreB expression before evaluation of relative cell size by light scatter profile analysis (upper panels) and cell cycle status assessment by Ki-67 expression and DNA content (lower panels).

Fig. 5.

Cell size and cell cycle analysis of pre-BCR+ and pre-BCR subpopulations.

(A) Human VpreB+ and VpreBCD34subpopulations of CD19+κ/λLC B lineage cells and (B) mouse CD19+BP-1+κ/λLC pre-B cells were sorted on the basis of positive or negative cell surface VpreB expression before evaluation of relative cell size by light scatter profile analysis (upper panels) and cell cycle status assessment by Ki-67 expression and DNA content (lower panels).

Close modal

Gene profile analysis of the pro-B and pre-B cell subpopulations

Pro-B cells in human bone marrow were identified in this analysis as CD19+CD34+SLC cells, and the 2 pre-B cell subpopulations were isolated as in the previous experiments. The mouse pro-B cell subpopulations, SLC+ and SLC, were isolated from RAG-2−/−mice to ensure the absence of more mature B lineage cells. Mouse pre-B cells were isolated from wild-type bone marrow as CD19+BP-1+κ/λLC cells, and the VpreB+ and Vpre-B subpopulations were then separated. Two rounds of cell sorting were conducted to ensure purity (more than 99.5%) of the subpopulations.

Several notable changes in gene expression were evident in this analysis (Figure 6). As anticipated, full-length μHC transcripts were either absent or were present only in trace levels in pro-B cells, and Tdt expression was detected exclusively in the pro-B subpopulations. RAG-1 andRAG-2 transcripts were down-regulated in the pre-BCR+ subpopulation, though RAG-2 transcripts could still be demonstrated, whereas both RAG-1 andRAG-2 expression were clearly evident in the VpreB subpopulation of pre-B cells. Correspondingly, the level of full-length light chain transcripts was dramatically increased in the small Vpre-B subpopulation of pre-B cells.B29, VpreB, and λ5 transcripts were expressed during all the pro-B and pre-B cell stages. However, VpreBand λ5 transcript levels were selectively down-regulated in the pre-BCR subpopulation of pre-B cells in both species (Figure 6), a finding that was verified by semiquantitative analysis (data not shown). Moreover, when the receptor-negative subpopulation of pre-B cells was permeabilized to allow examination of intracellular pre-BCR components, these cells were found to be virtually devoid of VpreB and λ5 proteins, whereas intracellular stores of μHC, Igα, and Igβ were all maintained (Figures 4, 6C, and data not shown). Selective loss of SLC expression is therefore a defining characteristic of the subpopulation of relatively small pre-B cells that no longer express pre-BCR.

Fig. 6.

Expression profiles for B lineage genes and intracellular proteins in pro-B and pre-B subpopulations.

(A) Human CD34+CD19+VpreBpro-B, CD19+ κ/λLCVpreB+pre-B, and CD19+κ/λLCCD34VpreBpre-B cells were isolated from fetal bone marrow samples. (B) Mouse CD19+ VpreB+ and CD19+VpreB pro-B cells were isolated from RAG2−/− bone marrow, and VpreB+ and VpreB subpopulations of pre-B cells (CD19+BP-1+ κ/λLC) were isolated from bone marrow of wild-type juvenile mice. Subpopulations were purified by 2 rounds of immunofluorescence-based sorting, and the sorted cells were used as templates for RT-PCR assays. PCR products were visualized on agarose gels by ethidium bromide staining. (C) Analysis of cytoplasmic VpreB, Igβ, and conventional LC protein expression within pre-BCR pre-B cells. Dashed lines indicate staining of pre-BCR cells. Solid lines represent staining in control pre-BCR+ cells (i) and CD19+κ/λLC+ B cells (ii) and (iii). Background staining with isotype-matched antibodies of irrelevant specificity is indicated by shaded histograms.

Fig. 6.

Expression profiles for B lineage genes and intracellular proteins in pro-B and pre-B subpopulations.

(A) Human CD34+CD19+VpreBpro-B, CD19+ κ/λLCVpreB+pre-B, and CD19+κ/λLCCD34VpreBpre-B cells were isolated from fetal bone marrow samples. (B) Mouse CD19+ VpreB+ and CD19+VpreB pro-B cells were isolated from RAG2−/− bone marrow, and VpreB+ and VpreB subpopulations of pre-B cells (CD19+BP-1+ κ/λLC) were isolated from bone marrow of wild-type juvenile mice. Subpopulations were purified by 2 rounds of immunofluorescence-based sorting, and the sorted cells were used as templates for RT-PCR assays. PCR products were visualized on agarose gels by ethidium bromide staining. (C) Analysis of cytoplasmic VpreB, Igβ, and conventional LC protein expression within pre-BCR pre-B cells. Dashed lines indicate staining of pre-BCR cells. Solid lines represent staining in control pre-BCR+ cells (i) and CD19+κ/λLC+ B cells (ii) and (iii). Background staining with isotype-matched antibodies of irrelevant specificity is indicated by shaded histograms.

Close modal

Since discovery of the SLC components, VpreB and λ5/14.1, their expression as unique pre-BCR components has been shown to be essential for pre-B cell growth and survival. Nevertheless, their detection on primary B lineage cells has been extremely challenging, largely because of their low levels of expression. This has led to conflicting views about when and where pro-BCR and pre-BCR are expressed during B lineage differentiation in mice and humans.49,51,54-60 The current studies used a sensitive immunofluorescence assay capable of detecting fewer than 100 molecules per cell61 to confirm the existence of a species difference regarding when cell surface SLC expression begins. They also define a conserved pattern of extinguished SLC expression in pre-B cells that allows them to exit the cell cycle and undergo the light chain rearrangements necessary for B-cell differentiation.

SLC components could not be detected on human pro-B cells with monoclonal antibodies recognizing the VpreB and λ5 proteins even with the enhanced immunofluorescence method. Primary pro-B cells and pro-B cell lines failed to express detectable cell surface SLC components despite their presence within these cells. Previous reports suggesting that human pro-B cells can express cell surface SLC in the absence of μHC57,59,60 may reflect, in part, differences in the phenotypic characteristics used to identify pro-B cells. CD34-bearing B lineage cells are generally considered to represent the human pro-B cell fraction, but, as did LeBien,65 we observed that CD19+CD34+ cells may express cell surface SLC, especially early in ontogeny when B lymphopoiesis is most active. However, all these CD34+SLC+ cells were shown to express μHC, thereby indicating that CD34 may be transiently expressed after B lineage cells undergo productive VDJHrearrangement to become pre-B cells.

SLC expression for mouse pro-B cells was reaffirmed in these studies, though a physiological role of this type of pro-BCR remains enigmatic. Cell surface expression of SLC is restricted to a subpopulation of the murine pro-B cells, and the SLC+ phenotype is faithfully reproduced by pro-B cell lines in which the SLC may be associated with surrogate HC proteins.48,51 One of the surrogate HC proteins has been identified as BILL-cadherin,66 a transmembrane glycoprotein that apparently lacks signal transducing capability. Igα/Igβ heterodimers are also expressed in low levels on mouse pro-B cells, and their ligation can induce progression of cellular differentiation.67 However, these Igα/Igβ heterodimers are associated with calnexin and have no apparent linkage with SLC components. The normal development of pro-B cells in VpreB- and λ5-deficient mice40 68 further argues against a functional role for SLC on mouse pro-B cells. The onset of intracellular SLC production before μHC production during B lineage development may simply reflect a preparatory step for subsequent pre- BCR assembly.

The pattern of pre-BCR expression conserved in mice and humans was characterized by the restriction of receptor expression to a limited subpopulation of the pre-B cells. Although the entire pre-B population exhibited the hallmark features of intracellular μHC presence and LC absence, the pre-BCR+ and pre-BCRsubpopulations of pre-B cells defined by the enhanced immunofluorescence method exhibited distinctive genotypic and phenotypic profiles. Receptor-positive pre-B cells were found to be relatively large, and many of these had entered the cell cycle. Consistent with the results of previous studies using other combinations of cell identification markers,53,69,70 this population of relatively large pre-B cells exhibited down-regulation of the recombination activating genes, RAG1 andRAG2. In contrast, pre-B cells that had no detectable pre-BCR were predominantly small, resting lymphocytes. These receptor-negative pre-B cells expressed RAG1 andRAG2, correspondingly exhibited enhanced levels of full-length LC transcripts, and were further distinguished by the selective down-regulation of SLC genes, VpreB andλ5/14.1. The latter feature was reflected by absence of the VpreB and λ5 proteins, although intracellular stores of the μHC, Igα and Igβ proteins were maintained in these cells. This selective loss of SLC production, without which μHC and Igα/Igβ cannot be expressed on the cell surface, thus terminates the expression of pre-BCR. In this regard, Igα/Igβ heterodimers have not been shown to bind to μHC before their association with SLC.36 Hence, selective down-regulation of SLC expression during pre-B cell differentiation leads inexorably to the loss of pre-BCR expression, an event that coincides with pre-B cell exit from the cell cycle.

Bone marrow DNA labeling studies initially revealed that large dividing pre-B cells give rise to small postmitotic pre-B cells that become cell surface IgM+ B cells after a 1- to 2-day interval.10,13,14 V-JL rearrangements in the κ and λ LC loci then take place in the small, postmitotic pre-B cells.18,20,70 The V(D)J rearrangement process is quiescent during the earlier pre-B phase when receptor-positive pre-B cells undergo mitosis. In addition to the corresponding down-regulation of RAG1 and RAG2, both of which are required for V(D)J rearrangement,71 phosphorylation of RAG2 leads to its efficient degradation before the S phase.72 This combination of events ensures protection against the creation of double-stranded DNA breaks and inappropriate cell death of the replicating, receptor-positive pre-B cells.

This analysis of SLC expression supports a B-cell differentiation scheme that differs for mice and humans primarily in the earlier onset of cell surface SLC expression in mice (Figure7). The extinction of pre-BCR expression by selective down-regulation of SLC production appears to have a profound impact on the pre-B cell differentiation process. The most well-documented function of pre-BCR is the promotion of pre-B cell growth and survival in conjunction with costimulatory signals provided by stromal cells or interleukin-7.73 Although evidence for pre-BCR promotion of HC allelic exclusion and LC gene rearrangement has also been suggested,74-76 these events can proceed in the absence of pre-BCR.38 68 The implication that pre-BCR expression is not directly involved in these differentiation events is reinforced by our observation that the loss of pre-BCR expression and withdrawal from the cell cycle coincide with the reinstatement ofRAG-1 and RAG-2 expression and VJLrearrangement. The selective SLC down-regulation to extinguish pre-BCR expression is thus an important prerequisite to B-cell differentiation. This sequence of events also ensures that SLC and conventional κ/λLC are not expressed simultaneously during primary B-cell differentiation, as indicated by the finding that SLC-bearing cells and κ/λLC-bearing cells belong to nonoverlapping populations in mouse and human bone marrow samples, irrespective of donor age.

Fig. 7.

Comparative models of normal B cell differentiation in humans and mice.

, μ heavy chains;, Igα/β;, VpreB/λ5;, BILL-cadherin;, calnexin; Tdt, terminal deoxyribonucleotidyl transferase. Pro-B cells depicted in this scheme express cell surface CD19.

Fig. 7.

Comparative models of normal B cell differentiation in humans and mice.

, μ heavy chains;, Igα/β;, VpreB/λ5;, BILL-cadherin;, calnexin; Tdt, terminal deoxyribonucleotidyl transferase. Pro-B cells depicted in this scheme express cell surface CD19.

Close modal

The model of primary B-cell differentiation supported by this analysis of SLC expression begs the question of how the VpreB andλ5/14.1 genes are turned off in concert at a key point in the pre-B cell differentiation process. A complex locus control region for the VpreB/λ5 genes has been demonstrated,77 but its function during primary B lineage differentiation is not entirely clear. Although the VpreBand λ5 promoter regions contain binding sites for Ikaros, EBF, E2A, and PAX5,4 expression of these transcription factors is maintained throughout pre-B cell differentiation. A possible mechanism for SLC suppression is suggested by the Ikaros-mediated transcriptional silencing that has been demonstrated for theλ5 gene in B cells.78-80 

The current observations are also relevant to the RAG and SLC expression observed for B lineage cells in secondary lymphoid tissues.56,81-86 Initially considered to reflect a reactivation of the pre-B cell gene program in germinal center B cells, further analysis of this phenomenon instead suggests an efflux of bone marrow pre-B cells to the periphery in response to inflammatory stimuli.86 However, a subpopulation of peripheral B cells with a restricted V gene repertoire has been reported to express SLC and κ or λ LC.85 We have so far been unable to identify SLC-bearing cells in normal peripheral lymphoid tissues, but background staining problems encountered in the analysis of peripheral B cells (Y.-H.W. and R.P.S., unpublished observations, 2001) indicate further study may be needed for a definitive resolution of this issue. This technical difficulty could reflect the expression of a newly identified family of Fc receptor homologues that are differentially expressed by B cells in peripheral lymphoid tissues.87,88 Finally, the dual expression of SLC and κ/λLC on leukemic cell lines48,49 is another remarkable exception to the rule established for normal B lymphopoiesis in humans and mice. Although this dual SLC/LC expression was previously interpreted as a reflection of the normal differentiation process,49 the current analysis suggests that the leukemic transitional pre-B/B cells must have been diverted from the normal differentiation pathway. In addition, the current differentiation model predicts that failure to down-regulate SLC expression would promote excessive pre-B cell proliferation at the expense of B-cell differentiation, a hypothesis that is being tested.

We thank Dr Larry Gartland for help with flow cytometry; Drs Peter Burrows, Flavius Martin, and John Kearney for helpful discussions; and Marsha Flurry and Ann Brookshire for help in preparing the manuscript. We thank Dr Alan Fantel (University of Washington, Seattle) for providing fetal bone marrow tissue samples.

Supported by National Institutes of Health grant AI39816 (M.D.C.). M.D.C. is a Howard Hughes Medical Institute Investigator.

Y.-H.W. and R.P.S. contributed equally to this manuscript.

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 U.S.C. section 1734.

1
Cooper
MD
Peterson
RDA
Good
RA
Delineation of the thymic and bursal lymphoid system in the chicken.
Nature.
205
1965
143
146
2
Tonegawa
S
Somatic generation of antibody diversity.
Nature.
302
1983
575
581
3
Rajewsky
K
Clonal selection and learning in the antibody system.
Nature.
381
1996
751
758
4
Reya
T
Grosschedl
R
Transcriptional regulation of B-cell differentiation.
Curr Opin Immunol.
10
1998
158
165
5
Rolink
AG
Schaniel
C
Andersson
J
Melchers
F
Selection events operating at various stages in B cell development.
Curr Opin Immunol.
13
2001
202
207
6
Owen
JJ
Cooper
MD
Raff
MC
In vitro generation of B lymphocytes in mouse foetal liver, a mammalian “bursa equivalent.”
Nature.
249
1974
361
363
7
Osmond
DG
Nossal
GJ
Differentiation of lymphocytes in mouse bone marrow, II: kinetics of maturation and renewal of antiglobulin-binding cells studied by double labeling.
Cell Immunol.
13
1974
132
145
8
Raff
MC
Megson
M
Owen
JJ
Cooper
MD
Early production of intracellular IgM by B-lymphocyte precursors in mouse.
Nature.
259
1976
224
226
9
Burrows
P
LeJeune
M
Kearney
JF
Evidence that murine pre-B cells synthesize mu heavy chains but no light chains.
Nature.
280
1979
838
840
10
Owen
JJ
Wright
DE
Habu
S
Raff
MC
Cooper
MD
Studies on the generation of B lymphocytes in fetal liver and bone marrow.
J Immunol.
118
1977
2067
2072
11
Gathings
WE
Lawton
AR
Cooper
MD
Immunofluorescent studies of the development of pre-B cells, B lymphocytes and immunoglobulin isotype diversity in humans.
Eur J Immunol.
7
1977
804
810
12
Burrows
PD
Kearney
JF
Lawton
AR
Cooper
MD
Pre-B cells: bone marrow persistence in anti-mu–suppressed mice, conversion to B lymphocytes, and recovery after destruction by cyclophosphamide.
J Immunol.
120
1978
1526
1531
13
Landreth
KS
Rosse
C
Clagett
J
Myelogenous production and maturation of B lymphocytes in the mouse.
J Immunol.
127
1981
2027
2034
14
Osmond
DG
Owen
JJ
Pre-B cells in bone marrow: size distribution profile, proliferative capacity and peanut agglutinin binding of cytoplasmic mu chain-bearing cell populations in normal and regenerating bone marrow.
Immunology.
51
1984
333
342
15
Maki
R
Kearney
J
Paige
C
Tonegawa
S
Immunoglobulin gene rearrangement in immature B cells.
Science.
209
1980
1366
1369
16
Alt
FW
Blackwell
TK
DePinho
RA
Reth
MG
Yancopoulos
GD
Regulation of genome rearrangement events during lymphocyte differentiation.
Immunol Rev.
89
1986
5
30
17
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.
173
1991
1213
1225
18
Coffman
RL
Weissman
IL
Immunoglobulin gene rearrangement during pre-B cell differentiation.
J Mol Cell Immunol.
1
1983
31
41
19
Ehlich
A
Schaal
S
Gy
H
Kitamura
D
Muller
W
Rajewsky
K
Immunoglobulin heavy and light chain genes rearrange independently at early stages of B cell development.
Cell.
72
1993
695
704
20
Engel
H
Rolink
A
Weiss
S
B cells are programmed to activate kappa and lambda for rearrangement at consecutive developmental stages.
Eur J Immunol.
29
1999
2167
2176
21
Hombach
J
Tsubata
T
Leclercq
L
Stappert
H
Reth
M
Molecular components of the B-cell antigen receptor complex of the IgM class.
Nature.
343
1990
760
762
22
Reth
M
Antigen receptors on B lymphocytes.
Annu Rev Immunol.
10
1992
97
121
23
Henderson
A
Calame
K
Transcriptional regulation during B cell development.
Annu Rev Immunol.
16
1998
163
200
24
Chang
H
Dmitrovsky
E
Hieter
PA
et al
Identification of three new Ig lambda-like genes in man.
J Exp Med.
163
1986
425
435
25
Sakaguchi
N
Melchers
F
Lambda 5, a new light-chain-related locus selectively expressed in pre-B lymphocytes.
Nature.
324
1986
579
582
26
Kudo
A
Melchers
F
A second gene, VpreB in the lambda 5 locus of the mouse, which appears to be selectively expressed in pre-B lymphocytes.
EMBO J.
6
1987
2267
2272
27
Bauer
SR
Huebner
K
Budarf
M
et al
The human Vpre B gene is located on chromosome 22 near a cluster of V lambda gene segments.
Immunogenetics.
28
1988
328
333
28
Schiff
C
Milili
M
Bossy
D
Fougereau
M
Organization and expression of the pseudo-light chain genes in human B-cell ontogeny.
Int Rev Immunol.
8
1992
135
145
29
Lassoued
K
Illges
H
Benlagha
K
Cooper
MD
Fate of surrogate light chains in B lineage cells.
J Exp Med.
183
1996
421
429
30
Martensson
IL
Melchers
F
Winkler
TH
A transgenic marker for mouse B lymphoid precursors.
J Exp Med.
185
1997
653
661
31
Person
C
Martensson
A
Martensson
I-L
Identification of a tissue- and differentiation stage-specific enhancer of the VpreB1 gene.
Eur J Immunol.
28
1998
787
798
32
Minegishi
Y
Hendershot
LM
Conley
ME
Novel mechanisms control the folding and assembly of lambda5/14.1 and VpreB to produce an intact surrogate light chain.
Proc Natl Acad Sci U S A.
96
1999
3041
3046
33
Melchers
F
Fit for life in the immune system? Surrogate L chain tests H chains that test L chains.
Proc Natl Acad Sci U S A.
96
1999
2571
2573
34
Hendershot
LM
Kearney
JF
A role for human heavy chain binding protein in the developmental regulation of immunoglobin transport.
Mol Immunol.
25
1988
585
595
35
Brouns
GS
de Vries
E
Neefjes
JJ
Borst
J
Assembled pre-B cell receptor complexes are retained in the endoplasmic reticulum by a mechanism that is not selective for the pseudo-light chain.
J Biol Chem.
271
1996
19272
19278
36
Benlagha
K
Guglielmi
P
Cooper
MD
Lassoued
K
Modifications of Igα and Igβ expression as a function of B lineage differentiation.
J Biol Chem.
274
1999
19389
19396
37
Guo
B
Kato
RM
Garcia-Lloret
M
Wahl
MI
Rawlings
DJ
Engagement of the human pre-B cell receptor generates a lipid raft-dependent calcium-signaling complex.
Immunity.
13
2000
243
253
38
Kitamura
D
Kudo
A
Schaal
S
Muller
W
Melchers
F
Rajewsky
K
A critical role of lambda 5 protein in B cell development.
Cell.
69
1992
823
831
39
Minegishi
Y
Coustan-Smith
E
Wang
YH
Cooper
MD
Campana
D
Conley
ME
Mutations in the human lambda5/14.1 gene result in B cell deficiency and agammaglobulinemia.
J Exp Med.
187
1998
71
77
40
Mundt
C
Licence
S
Shimizu
T
Melchers
F
Martensson
IL
Loss of precursor B cell expansion but not allelic exclusion in VpreB1/VpreB2 double-deficient mice.
J Exp Med.
193
2001
435
445
41
Kitamura
D
Rajewsky
K
Targeted disruption of mu chain membrane exon causes loss of heavy-chain allelic exclusion.
Nature.
356
1992
154
156
42
Gong
S
Nussenzweig
MC
Regulation of an early developmental checkpoint in the B cell pathway by Ig beta.
Science.
272
1996
411
414
43
Yel
L
Minegishi
Y
Coustan-Smith
E
et al
Mutations in the mu heavy-chain gene in patients with agammaglobulinemia.
N Engl J Med.
335
1996
1486
1493
44
Minegishi
Y
Coustan-Smith
E
Rapalus
L
Ersoy
F
Campana
D
Conley
ME
Mutations in Igα (CD79a) result in a complete block in B-cell development.
J Clin Invest.
104
1999
1115
1121
45
Conley
ME
Rohrer
J
Rapalus
L
Boylin
EC
Minegishi
Y
Defects in early B-cell development: comparing the consequences of abnormalities in pre-BCR signaling in the human and the mouse.
Immunol Rev.
178
2000
75
90
46
Pillai
S
Baltimore
D
Formation of disulfide-linked mu 2 omega 2 tetramers in pre-B cells by the 18K omega-immunoglobulin light chain.
Nature.
329
1987
172
174
47
Kerr
WG
Cooper
MD
Feng
L
Burrows
PD
Hendershot
LM
Mu heavy chains can associate with a pseudo-light chain complex (psi L) in human pre-B cell lines.
Int Immunol.
4
1989
355
361
48
Karasuyama
H
Rolink
A
Melchers
F
A complex of glycoproteins is associated with VpreB/lambda 5 surrogate light chain on the surface of mu heavy chain-negative early precursor B cell lines.
J Exp Med.
178
1993
469
478
49
Lassoued
K
Nunez
CA
Billips
L
et al
Expression of surrogate light chain receptors is restricted to a late stage in pre-B cell differentiation.
Cell.
73
1993
73
86
50
Nishimoto
N
Kubagawa
H
Ohno
T
Gartland
GL
Stankovic
AK
Cooper
MD
Normal pre-B cells express a receptor complex of mu heavy chains and surrogate light-chain proteins.
Proc Natl Acad Sci U S A.
88
1991
6284
6288
51
Shinjo
F
Hardy
RR
Jongstra
J
Monoclonal anti-lambda 5 antibody FS1 identifies a 130 kDa protein associated with lambda 5 and Vpre-B on the surface of early pre-B cell lines.
Int Immunol.
6
1994
393
399
52
Guelpa-Fonlupt
V
Tonnelle
C
Blaise
D
Fougereau
M
Fumoux
F
Discrete early pro-B and pre-B stages in normal human bone marrow as defined by surface pseudo-light chain expression.
Eur J Immunol.
24
1994
257
264
53
Karasuyama
H
Rolink
A
Shinkai
Y
Young
F
Alt
FW
Melchers
F
The expression of Vpre-B/lambda 5 surrogate light chain in early bone marrow precursor B cells of normal and B cell-deficient mutant mice.
Cell.
77
1994
133
143
54
Winkler
TH
Rolink
A
Melchers
F
Karasuyama
H
Precursor B cells of mouse bone marrow express two different complexes with the surrogate light chain on the surface.
Eur J Immunol.
25
1995
446
450
55
Meffre
E
Fougereau
M
Argenson
JN
Aubaniac
JM
Schiff
C
Cell surface expression of surrogate light chain (psi L) in the absence of mu on human pro-B cell lines and normal pro-B cells.
Eur J Immunol.
26
1996
2172
2180
56
Wang
YH
Nomura
J
Faye-Petersen
OM
Cooper
MD
Surrogate light chain production during B cell differentiation: differential intracellular versus cell surface expression.
J Immunol.
161
1998
1132
1139
57
Fluckiger
AC
Sanz
E
Garcia-Lloret
M
et al
In vitro reconstitution of human B-cell ontogeny: from CD34(+) multipotent progenitors to Ig-secreting cells.
Blood.
92
1998
4509
4520
58
Tsuganezawa
K
Kiyokawa
N
Matsuo
Y
et al
Flow cytometric diagnosis of the cell lineage and developmental stage of acute lymphoblastic leukemia by novel monoclonal antibodies specific to human pre-B-cell receptor.
Blood.
92
1998
4317
4324
59
Lemmers
B
Gauthier
L
Guelpa-Fonlupt
V
Fougereau
M
Schiff
C
The human (PsiL+mu−) proB complex: cell surface expression and biochemical structure of a putative transducing receptor.
Blood.
93
1999
4336
4346
60
Sanz
E
de la Hera
A
A novel anti-Vpre-B antibody identifies immunoglobulin-surrogate receptors on the surface of human pro-B cells.
J Exp Med.
183
1996
2693
2698
61
Scheffold
A
Assenmacher
M
Reiners-Schramm
L
Lauster
R
Radbruch
A
High-sensitivity immunofluorescence for detection of the pro- and anti- inflammatory cytokines gamma interferon and interleukin-10 on the surface of cytokine-secreting cells.
Nat Med.
6
2000
107
110
62
Stephan
RP
Elgavish
E
Karasuyama
H
Kubagawa
H
Cooper
MD
Analysis of VpreB expression during B lineage differentiation in λ5-deficient mice.
J Immunol.
167
2001
3734
3739
63
Farner
NL
Dörner
T
Lipsky
PE
Molecular mechanisms and selection influence the generation of the human VλJλ repertoire.
J Immunol.
162
1999
2137
2145
64
Cooper
MD
Mulvaney
D
Coutinho
A
Cazenave
PA
A novel cell surface molecule on early B-lineage cells.
Nature.
321
1986
616
618
65
LeBien
TW
Fates of human B-cell precursors.
Blood.
96
2000
9
23
66
Ohnishi
K
Shimizu
T
Karasuyama
H
Melchers
F
The identification of a nonclassical cadherin expressed during B cell development and its interaction with surrogate light chain.
J Biol Chem.
275
2000
31134
31144
67
Nagata
K
Nakamura
T
Kitamura
F
et al
The Igα/Igβ heterodimer on μ-negative proB cells is competent for transducing signals to induce early B cell differentiation.
Immunity.
7
1997
559
570
68
Rolink
A
Karasuyama
H
Grawunder
U
Haasner
D
Kudo
A
Melchers
F
B cell development in mice with a defective lambda 5 gene.
Eur J Immunol.
23
1993
1284
1288
69
Grawunder
U
Leu
TM
Schatz
DG
et al
Down-regulation of RAG1 and RAG2 gene expression in preB cells after functional immunoglobulin heavy chain rearrangement.
Immunity.
3
1995
601
608
70
Ghia
P
ten Boekel
E
Sanz
E
de la Hera
A
Rolink
A
Melchers
F
Ordering of human bone marrow B lymphocyte precursors by single-cell polymerase chain reaction analyses of the rearrangement status of the immunoglobulin H and L chain gene loci.
J Exp Med.
184
1996
2217
2229
71
Oettinger
MA
Schatz
DG
Gorka
C
Baltimore
D
RAG-1 and RAG-2, adjacent genes that synergistically activate V(D)J recombination.
Science.
248
1990
1517
1523
72
Lin
WC
Desiderio
S
Cell cycle regulation of V(D)J recombination-activating protein RAG-2.
Proc Natl Acad Sci U S A.
91
1994
2733
2737
73
Hess
J
Werner
A
Wirth
T
Melchers
F
Jack
HM
Winkler
TH
Induction of pre-B cell proliferation after de novo synthesis of the pre-B cell receptor.
Proc Natl Acad Sci U S A.
98
2001
1745
1750
74
Iglesias
A
Kopf
M
Williams
GS
Buhler
B
Kohler
G
Molecular requirements for the mu-induced light chain gene rearrangement in pre-B cells.
EMBO J.
10
1991
2147
2155
75
Loffert
D
Ehlich
A
Muller
W
Rajewsky
K
Surrogate light chain expression is required to establish immunoglobulin heavy chain allelic exclusion during early B cell development.
Immunity.
4
1996
133
144
76
Maki
K
Nagata
K
Kitamura
F
Takemori
T
Karasuyama
H
Immunoglobulin beta signaling regulates locus accessibility for ordered immunoglobulin gene rearrangements.
J Exp Med.
191
2000
1333
1340
77
Sabbattini
P
Georgiou
A
Sinclair
C
Dillon
N
Analysis of mice with single and multiple copies of transgenes reveals a novel arrangement for the lambda5-VpreB1 locus control region.
Mol Cell Biol.
19
1999
671
679
78
Brown
KE
Guest
SS
Smale
ST
Hahm
K
Merkenschlager
M
Fisher
AG
Association of transcriptionally silent genes with Ikaros complexes at centromeric heterochromatin.
Cell.
91
1997
845
854
79
Sabbattini
P
Lundgren
M
Georgiou
A
Chow
C
Warnes
G
Dillon
N
Binding of Ikaros to the λ5 promoter silences transcription through a mechanism that does not require heterochromatin formation.
EMBO J.
20
2001
2812
2822
80
Cobb
BS
Morales-Alcelay
S
Kleiger
G
Brown
KE
Fisher
AG
Smale
ST
Targeting of Ikaros to pericentromeric heterochromatin by direct DNA binding.
Genes Dev.
14
2000
2146
2160
81
Hikida
M
Mori
M
Takai
T
Tomochika
K
Hamatani
K
Ohmori
H
Reexpression of RAG-1 and RAG-2 genes in activated mature mouse B cells.
Science.
274
1996
2092
2094
82
Han
S
Zheng
B
Schatz
DG
Spanopoulou
E
Kelsoe
G
Neoteny in lymphocytes: Rag1 and Rag2 expression in germinal center B cells.
Science.
274
1996
2094
2097
83
Meffre
E
Papavasiliou
F
Cohen
P
et al
Antigen receptor engagement turns off the V(D)J recombination machinery in human tonsil B cells.
J Exp Med.
188
1998
765
772
84
Yu
W
Nagaoka
H
Jankovic
M
et al
Continued RAG expression in late stages of B cell development and no apparent re-induction after immunization.
Nature.
400
1999
682
687
85
Meffre
E
Davis
E
Schiff
C
et al
Circulating human B cells that express surrogate light chains and edited receptors.
Nat Immunol.
1
2000
207
213
86
Gartner
F
Alt
FW
Monroe
RJ
Seidl
KJ
Antigen-independent appearance of recombination activating gene (RAG)-positive bone marrow B cells in the spleens of immunized mice.
J Exp Med.
192
2000
1745
1754
87
Hatzivassiliou
G
Miller
I
Takizawa
J
et al
IRTA1 and IRTA2, novel immunoglobulin superfamily receptors expressed in B cells and involved in chromosome 1q21 abnormalities in B cell malignancy.
Immunity.
14
2001
277
289
88
Davis
RS
Wang
YH
Kubagawa
H
Cooper
MD
Identification of a family of Fc receptor homologs with preferential B cell expression.
Proc Natl Acad Sci U S A.
98
2001
9772
9777

Author notes

Max D. Cooper, Division of Developmental and Clinical Immunology, University of Alabama at Birmingham, WTI 378, 1824 6th Ave S, Birmingham, AL 35294-3300; e-mail: max.cooper@ccc.uab.edu.

Sign in via your Institution