Quick viewing(Text Mode)

Pre-BCR Signals and the Control of Ig Gene Rearrangements Jamie K

Pre-BCR Signals and the Control of Ig Gene Rearrangements Jamie K

Seminars in Immunology 18 (2006) 31–39

Review Pre-BCR signals and the control of Ig rearrangements Jamie K. Geier, Mark S. Schlissel ∗ UC-Berkeley, Department of Molecular & Biology, Division of Immunology, 439 Life Sciences Addition, Berkeley, CA 94720-3200, USA

Abstract Progenitor B lymphocytes that successfully assemble a heavy chain gene encoding an immunoglobulin capable of pairing with surrogate light chain proteins trigger their own further differentiation by signaling via the pre-BCR complex. The pre-BCR signals several rounds of proliferation and, in this expanded population, directs a complex, -specific set of epigenetic changes resulting in allelic exclusion of the heavy chain locus and activation of the light chain loci for V(D)J recombination. © 2005 Elsevier Ltd. All rights reserved.

Keywords: B cell development; Pre-B cell receptor (pre-BCR); Allelic exclusion; V(D)J recombination; Transcriptional regulation

1. Introduction a membrane complex with the surrogate light chain components (SLC) VpreB and ␭5, and the transmembrane proteins Ig-␣ and B cell differentiation is a highly dynamic process that requires -␤. Signaling through this receptor is associated with its internal- the coordinated action of various receptors, signaling molecules, ization and degradation [1–3]. The cytoplasmic domains of Ig-␣ factors and the V(D)J recombinase. Completion of and -␤ contain immunoreceptor tyrosine-based activation motifs immunoglobulin heavy chain (␮HC) gene rearrangement leads (ITAMs) which, when clustered in the membrane, provide a to surface expression of the pre-BCR complex which triggers docking site for the Syk kinase, Src family kinases (Fyn, Lyn, and a cascade of signaling events that alters the developing cell’s Blk), the Tec family kinase, Btk, and the adaptor proteins Grb2 response to growth factors and effects transcription and DNA and BLNK. BLNK (also known as BASH and SLP-65) recruits rearrangement. ␮HC expression results in pre-B cell expansion Btk, which is then phosphorylated by Lyn, Syk, or both [4,5]. and inhibits further rearrangement at the ␮HC locus, a process Phosphorylated Btk then phosphorylates PLC␥2, which known as allelic exclusion. Pre-B cells subsequently exit the hydrolyzes PIP2 to IP3 and diacylglycerol (DAG) causing cal- cell cycle and initiate of a second set of gene rearrangements cium mobilization and resulting in activation of calcium depen- at the immunoglobulin light chain (IgLC) loci. The timing of dent enzymes (reviewed in [6]). Proliferation is induced via acti- DNA rearrangements, which involve the generation and repair vation of MAP kinase pathways, and allelic exclusion is imposed of DNA double stranded breaks interspersed with bursts of pro- by a presently undefined mechanism [7,8]. Upon pre-BCR sig- liferative expansion, must be tightly regulated to ensure proper naling, cells initially undergo a proliferative burst accompanied development and to prevent genomic instability and leukemic by down regulation of SLC components and RAGs, followed by transformation. This review will focus on the function of the exit from the cell cycle and transition from pro-B/large pre-B pre-BCR as the critical regulator of the pre-antigenic phase of I (Hardy fraction C) to small pre-B II (Hardy fraction C) [9]. B cell development. Signaling also induces transcription factors such as NF-␬B, Spi- B and IRF-4 as well as re-expression of RAGs, contributing to ␬ ␬ 2. Pre-BCR signalling—an overview activation of the enhancers and germline transcription and rearrangement [10,11]. Pre-BCR signaling is initiated upon expression of a clono- typic immunoglobulin ␮ heavy chain (␮HC) that can assemble 3. Pre-BCR signaling does not require exogenous ligand

Once the pre-BCR assembles on the cell surface, tyrosine ∗ Corresponding author. Tel.: +1 510 643 2462; fax: +1 510 642 6845. kinase-dependent signaling begins. Ig-␣/␤ heterodimer surface E-mail address: [email protected] (M.S. Schlissel). expression facilitated by the ␮HC and SLC seems to be sufficient

1044-5323/$ – see front matter © 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.smim.2005.11.001 32 J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 to initiate signaling. Truncation mutants that lack extracellular tion [25], it is widely agreed that pre-BCR signaling is required domains can function to aggregate Ig-␣/␤ and signal B cell dif- for the proliferative expansion of pre-B cells expressing a suc- ferentiation [12,13]. In fact, pro- to pre-B cell development can cessfully rearranged IgHC gene which can assemble with SLCs. be driven by the Ig-␣ and -␤ ITAMS (along with other conserved This proliferation signal induces two to five rounds of cell divi- cytoplasmic motifs) targeted to the inner leaflet of the plasma sion and eliminates the dependence of developing cells on IL-7 membrane by fusion with a Lck myristoylation/palmitoylation [15]. Pre-B cells are known to be cycling in vivo based on their sequence [14]. These observations, along with the fact that a rapid incorporation of the nucleotide analog BrdU [1,26,27]. single pre-B cell in the absence of any other stromal cells can Transgenic IgHC expression induces proliferation in pro-B cells proliferate in response to pre-BCR expression [15], calls into cultured on stromal cells [28]. Targeted deletion of the Ig␮ mem- question the relevance of various extracellular ligands proposed brane exon or disruption of the SLC results in the failure to interact with the pre-BCR. However, soluble recombinant of pre-B cell proliferation [25]. pre-BCR can bind stromal cells [16]. The Schiff group identi- The MAP kinase pathway is involved in pre-B cell expansion fied a human pre-BCR ligand, galectin-1, that may be involved and perhaps various aspects of differentiation as well. In mature in receptor aggregation and activation of tyrosine kinase activ- B cells, Ras signaling is induced upon BCR crosslinking [29,30]. ity localized at the pre-BCR [3]. Another study by the Jack¨ Mice expressing a constitutively activated Ras mutant transgene lab implicated stromal heparin sulfate as a ligand for murine on a RAG-null background bypass the pre-BCR checkpoint and pre-BCR [17] (see also contribution by Espeli et al. in this contain almost normal numbers of B cells in their lymph nodes issue). Recent data has revealed another potential mechanism and spleen [31]. These RAS-transgenic B cells are abnormal in for pre-BCR signaling involving the SLCs. The conserved non- that they continue to express ␭5 and RAG2, which are normally immunoglobulin portions of ␭5 and VpreB may mediate con- down-regulated upon pre-BCR signaling. Their ␬ germline tran- stitutive receptor aggregation, signaling and internalization via script levels are similar to those observed in wild-type pre-B homotypic ionic interactions [18]. Various truncated or unusu- cells, but the transgenic cells also express some surface mark- ally structured ␮HCs may inherently allow aggregation in an ers associated with more mature stages of B cell development. SLC-independent fashion providing an explanation for the trun- When the activated Ras transgene is bred onto a RAG sufficient cated ␮HC transgene results mentioned above. but JH background, ␬ rearrangement is induced despite a lack The absence of both Ig-␣ and -␤ causes a developmental of HC expression [32]. The survival phenotype of Ras trans- arrest at the pro-B cell stage despite intact V(D)J recombina- genic B cell precursors is similar to that of RAG-null B cells tion and ␮HC expression revealing the requirement for these expressing transgenic ␮HC and Bcl-2. Indeed, levels of Bcl-2 molecules in pre-BCR signaling [19,20]. One role of the ITAMs are high in the activated Ras transgenic B cell precursors, a trait is recruitment of BLNK to Ig-␣. Fusing BLNK to a mutant Ig- characteristic of mature B cells [31] perhaps allowing survival ␣ lacking these tyrosines reconstitutes downstream signaling in the setting of both proliferation and ongoing Ig␬ locus rear- events. Thus, simply the formation of the pre-BCR leading to rangement. the membrane localization of Ig-␣ and -␤ is sufficient to trigger Mice expressing a transgenic human dominant negative Ras, pre-BCR signaling, allow downregulation of RAGs and SLC H-rasN17, show a developmental arrest at a stage correspond- proteins, and upregulation of transcription factors that result in ing to Hardy fraction “A”. This was not attributable to increased IgLC gene rearrangements. apoptosis. Over-expression of a membrane targeted, and thus The pre-BCR may serve yet another function, that of a molec- constitutively active, partner of Ras, Raf-1(Raf-CAAX), on ular clock which eliminates B cell precursors that fail to generate the H-rasN17 background overcame the block confirming the a functional IgLC gene within a certain window of time or that involvement of this MAP kinase pathway in B cell development express a ␮HC that is incapable of associating with any IgLC [33]. Mimicking the pre-BCR signal with these transgenes does ([21,22]; see also contribution by C. Vettermann and H.-M. Jack¨ not, however, induce allelic exclusion since V-to-DJ rearrange- in this issue). Pre-BCR signaling is associated with internal- ment proceeds despite the premature proliferation signal. ization and degradation of the pre-BCR [1–3]. Since the genes Thus, the MAPK pathway is necessary for proper pre-BCR encoding the SLC components ␭5 and VpreB are inactivated signaling, and when activated can substitute for the pre-BCR upon pre-BCR signaling [23], progressively less pre-BCR com- causing proliferative expansion, survival and ␬ activation, but plex can form on the surface as cells divide, culminating in not IgHC allelic exclusion, or ␭5 and RAG down-regulation. reduced proliferative signals. If a pre-B cell fails to make an These experiments demonstrate the existence of two separable IgLC that associates with its ␮HC, then ␮HC expression on pre-BCR signaling pathways; one causing proliferation and the the surface will eventually be lost, presumably leading to the induction of some transcription factors involved in differentia- cell’s arrested development and death [24]. The existence of tion, and the other mediating allelic exclusion. this clock may help protect the genome from RAG-associated genomic instability. 5. Allelic exclusion

4. Pre-BCR signals leading to proliferation Allelic exclusion remains an enigmatic aspect of pre-BCR signaling. Preventing further ␮HC gene rearrangement in a cell While there is debate as to whether the complete pre-BCR is that already expresses a ␮HC is necessary to ensure that each required for IgHC allelic exclusion and pre-B cell differentia- B cell has an unique antigen specificity. It is imposed at the J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 33

VH-to-DJH step in recombination while D-to-JH rearrangement expression amongst individual ␮HC genes could account for is unaffected [34]. The ability of RAG proteins to selectively the curious observation that some ␮HC transgenes allelically act at different loci depending on the cell type or stage in devel- exclude very stringently while others allow continuation of opment has been attributed to regulated accessibility of each D-proximal VH gene segment rearrangement [50]. Transgene rearranging locus within structure [35]. RAG proteins expression levels may also play a role. cannot cleave a nucleosomal substrate in vitro, implicating the D␮, an endogenously generated truncated ␮HC protein necessity of some sort of chromatin remodeling to render the formed by translation of a partially rearranged (DH-to-JH) IgHC DNA accessible to the recombinase [36–38]. The loci within allele [51] is also capable of signaling allelic exclusion. D␮ sur- purified nuclei that are accessible to cleavage by RAGs in vitro face expression blocks further development of B cell precursors, depend on the source of nuclei [39]. Thus, the accessibility of a process known as D␮ selection [42,52–54]. This selection is rearranging loci is a cell-type and stage-specific property of chro- thought to occur when D␮ expression causes ␮HC gene allelic matin structure. The accessibility of the IgHC locus changes exclusion, thus preventing the formation of a complete IgHC across the pro-B to pre-B cell transition, and pre-BCR signal- gene [55,56]. Proliferation is impaired in the D␮ transgenic ing induces a permanent loss of IgH locus accessibility. The mice, again pointing to different requirements for allelic exclu- mechanism or signaling pathways employed for such chromatin sion and proliferation. D␮ can cause increased germline ␬ and remodeling have not been delineated. diminished ␭5 transcription, yet it cannot promote full differ- entiation to the pre-B cell stage [56]. Considering that Ig-␤, ␭5 5.1. Requirements and signals and Syk are necessary for D␮ to signal, D␮ apparently forms a pre-BCR-like complex, but due to the lack of VH structure, this Surface expression of a ␮HC induces allelic exclusion and pseudo-pre-BCR fails to mimic essential aspects of the pre-BCR mice carrying a mutation in the transmembrane region of the signal and such cells eventually die from an inability to differ- ␮HC gene are unable to turn off IgHC gene segment rearrange- entiate perhaps due to ineffective pairing with IgLC [56,57]. ments [40]. Expression of a ␮HC transgene in mice prevents Pairing of D␮ to the SLC could be qualitatively different, and endogenous VH-to-DJH rearrangements [41]. It is controversial, different requirements for the maturation and surface expression however, whether SLC expression is required for IgHC allelic of the two types of pre-BCRs have been demonstrated [58]. But exclusion. One group reported that targeted disruption of the precisely how lack of the V regions in this circumstance has gene encoding ␭5 resulted in a failure of early pre-B cell pro- such a dramatic effect on B cell development when truncation liferation and the loss of allelic exclusion at the level of ␮HC mutants suffice to replace full-length proteins remains a mystery gene rearrangements. Surprisingly, however, these ␭5-deficient [12,13]. mice still displayed allelic exclusion at the level of surface ␮HC As mentioned above, proliferative signals transmitted expression [42]. Similar results were observed in mice lacking through the MAPK pathway are not sufficient to induce allelic VpreB1, VpreB2 and ␭5 [43,44]. This second set of researchers exclusion. Nor is BLNK required for allelic exclusion [59]. But detected no difference in the frequency of pre-B cells with two Syk, the tyrosine kinase recruited and activated upon pre-BCR ␮HC V(D)J rearrangements in the presence or absence of SLCs signaling upstream of BLNK, is necessary for normal B cell suggesting allelic exclusion at the level of gene rearrangement development and allelic exclusion. Irradiated mice reconstituted does not require full pre-BCR assembly. One potential explana- with Syk−/− ␮HC transgenic cells displayed incomplete ␮HC tion for this observation was provided in a report showing that gene allelic exclusion, however ZAP-70, another Syk family precocious expression of IgLC could replace the need for SLCs kinase expressed in B cells, appears to play a partially redundant to promote B cell development [45]. However, more compelling role with Syk. Syk−/− ZAP-70−/− double mutants completely data shows that ␮HC can be detected on the surface of IgLC- arrest at the pro-B cell stage according to their cell surface negative SLC-deficient cells demonstrating that some ␮HC are marker phenotype and cannot allelically exclude in the presence capable of reaching the cell surface without SLC or LC [46–49]. of a ␮HC transgene [60]. The few cells that are able to express ␮HC on the surface despite BLNK is the adaptor protein downstream of Syk that is the absence of SLC would then be capable of signaling progres- involved in cessation of proliferation and the activation of sion as well as allelic exclusion. Although it is not yet clear how germline ␬ transcription and rearrangement (see contribution by a ␮HC can reach the surface in the absence of SLC, it is possible R.W.Hendriks and R. Kersseboom in this issue). However, while that ␮HC can pair with other chaperone-type proteins in pre-B D␮ selection is impaired in the BLNK knock out, IgHC allelic cells, transiting to the surface and signaling allelic exclusion and exclusion in that mutant remains intact [61]. This may be due to other changes in gene regulation, but not proliferation [46–49]. another partially redundant adapter, LAT, which can substitute Differences in the ability of ␮HC gene products to be to some extent for BLNK in pre-B cell development [62].LATis expressed on the cell surface may be responsible for the sur- able to link Syk to downstream PLC␥ signaling, just as BLNK prising observation that 4–8% of pre-B cells do express two does [63]. Coincidentally, mice heterozygous for PLC␥1 and endogenous rearranged ␮HCs, only one of which is expressed missing PLC␥2 on an IgHC transgenic background fail to induce on the cell surface [22]. This may be due to inefficient pairing allelic exclusion as measured using a PCR assay for endoge- and thus weak surface expression of the first HC, resulting in nous VH gene-segment rearrangements [64]. The mechanism a failure of allelic exclusion followed by productive rearrange- for PLC␥s’ involvement in allelic exclusion is not yet under- ment of a second IgHC allele. In this vein, differences in surface stood, but perhaps calcium flux induces activation of relevant 34 J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 transcription factors. It would be interesting to know whether SWI-SNF, involved in heterochromatin formation, or active in over-expression of PLC␥ signaling cascade components could acetylation-dependent transcriptional regulation [72]. relieve the absolute requirement for HC surface expression in allelic exclusion. 6. Turning on kappa

5.2. Chromosomal choreography ␮HC expression causes several rounds of cell division fol- lowed by exit from the cell cycle, silencing of the ␮HC locus, Recently, large chromosomal movements have been corre- and up-regulation of the RAGs (among other molecules involved lated with allelic exclusion at the ␮HC locus. Fluorescent in in differentiation), as discussed above. The final step in differ- situ hybridization (FISH) experiments revealed that the ␮HC entiation induced by ␮HC is Ig␬ locus activation and V-to-J␬ locus associates with the nuclear periphery in most T cells, recombination [34,73]. Signaling cascades emanating from the but is more centrally located in LPS-stimulated B cells [65]. pre-BCR induce transcription factors and perhaps chromatin This phenomenon was shown to be RAG independent but IL- remodeling activities that bind cis-regulatory elements within 7R␣-dependent as well as cell type specific, suggesting a role the Ig␬ locus resulting in germline transcription and rearrange- for such chromosomal relocation in the regulation of recombi- ment. The mechanism of these events is currently the focus of nation. Remarkably, two-color FISH using probes from either intense research efforts. end of the ␮HC locus revealed a higher level of compaction It is generally agreed that Ig␬ rearrangement does not abso- within the locus in B cells undergoing V-to-DJ rearrangement lutely require ␮HC or pre-BCR expression. For example, one [65,66]. This compaction was lost in B cell precursors lacking can detect low levels of Ig␬ rearrangement in JH-deleted pro-B the B cell-restricted transcription factor Pax5 correlating with cells [74] or in IL-7-dependent ␮HC− pro-B cell clones upon a loss of rearrangement to distal V genes [67]. In transgenic T acute withdrawal of IL-7 [75]. This has led to the suggestion that cells expressing Pax5, central nuclear relocation of IgHC genes Ig␬ locus activation may be part of a developmental program that occurred, but the further level of compaction did not. This Pax5 is independent of pre-BCR expression. It has been proposed that induced nuclear relocation stimulated D-to-JH rearrangement in increases in Ig␬ rearrangement that are associated with pre-BCR the T cells and allowed proximal VH gene rearrangement that expression may be a consequence of the proliferative expan- normally is not found in T cells [68,69]. Thus, nuclear relocal- sion of these cells rather than a pre-BCR signal transmitted to ization and chromatin compaction may contribute to regulation the Ig␬ locus [25]. Strong data exists which contradicts this of the recombination machinery. Allelic exclusion requires that hypothesis, however. Pre-B cells positive for cytoplasmic ␮HC this accessibility be reversed and indeed, FISH studies have now contain far higher levels of J␬ locus dsDNA breaks associated demonstrated that co-localization to gamma satellite sequences with active V-to-J␬ rearrangement than do pro-B cells on a per of one ␮HC allele coincides with allelic exclusion and is then nanogram of DNA basis [73] and expression of a ␮HC transgene relaxed but reestablished during LC rearrangement [68]. in RAG-deficient mice results in increases in germline transcrip- IL-7 signaling is associated with histone acetylation and tion and recombinase accessibility in sorted populations of pre- germline transcription of the distal V gene segments, and is nec- as compared to pro-B cells [34,39]. Also, there is data link- essary for the large chromosomal movements of the IgH locus ing the pre-BCR-mediated activation of the Ras-MAP kinase [65]. Loss of IL-7 responsiveness upon pre-BCR signaling is pathway to ␬ locus activation [76]. Additional experimenta- accompanied by deacetylation of the distal V gene segments, tion will be required to determine whether the pathway from although subsequent exposure to IL-7 causes the locus to become pre-BCR expression to ␬ locus activation is direct or indirect, acetylated once again [70]. Treatment of activated splenic B cells however. with IL-7 greatly diminished gamma satellite association of the The two IgLC loci, ␬ and ␭, are expressed at a ratio of approx- IgH locus further implicating the shut-off of IL-7 receptor signal- imately 20:1 in mice [77]. The more frequently rearranged, and ing in allelic exclusion [68]. Thus, adoption of new proliferation therefore more frequently studied ␬ locus consists of approx- signals and loss of IL-7 responsiveness upon pre-BCR signaling imately 140 V␬ gene segments, 5 J␬ gene segments (four of may be necessary to establish allelic exclusion. which are functional), two germline transcript promoters, and Correlating with the initial activation of the IgHC locus are two enhancers (see Fig. 1). These two enhancers play partially antisense transcripts originating in intergenic regions between redundant roles in activating the ␬LC locus as deletion of either VH gene segments [71]. These transcripts are turned off by one results in a decrease in the ratio of ␬-to-␭ expression (more so the time B cell precursors reach the late pro-B stage (Hardy’s in the E␬3 deletion), but only when both enhancers are deleted fraction C, CD19+CD43+BP-1+). Whether these antisense tran- is the phenotype dramatic. Deletion of both completely abol- scripts play a role in opening up the locus or in inducing chro- ishes ␬ recombination, leaving only ␭ positive B cells [78]. ␬ matin compaction via an RNAi-dependent pathway has yet to germline transcripts are activated in pre-B cells and correlate be determined. with ␬ rearrangements [79]. The profiles driving these developmentally regulated changes in ␮HC locus structure remain to be dis- 6.1. Transcription factors and κ locus activation covered. A microarray study designed to elucidate changes in transcription associated with B cell differentiation identified four Many studies of Ig␬ locus regulation have utilized Abel- genes encoding proteins each of which is either associated with son murine leukemia virus (AMuLV)-transformed pro-B cell J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 35

AMuLV transformation arrests developing B cells in what appears to be an early pre-B cell-like stage by an unknown mechanism. The viral oncogene v-Abl immortalizes these cells rendering them IL-7 independent and apoptosis resistant, but blocks their further differentiation. Upon treatment of AMuLV- transformed cells with the abl kinase inhibitor STI-571 (com- monly called Gleevec) these pre-B cells activate RAG and germline ␬ transcription and rearrange their ␬ and ␭ loci at very high rates [11]. By analyzing DNA microarray data derived from such cells prior to and after treatment with STI-571, IRF-4 and Spi-B were identified as key factors in activating the ␬LC locus [11]. Co-transfection of cDNA expression vectors encoding the two proteins was sufficient to induce ␬ germline transcripts in an AMuLV-transformed pre-B cell line [11]. PU.1−/−Spi- B−/− primary pro-B cell cultures have lower levels of ␬ tran- scription than wild-type B cell lines, however IL-7 withdrawal still induced ␬ but not ␭ activation, suggesting that ␬ rear- rangement might be unaffected by the absence of these factors Fig. 1. Pre-BCR signaling activates pre-B cell proliferation, enforces allelic [83]. exclusion and activates Ig␬ locus rearrangement. The upper part diagrams PU.1 and IRF4 interact with the E␬3 enhancer in chromatin the signaling cascades initiated by homotypic surrogate light chain (SLC)- immunoprecipitation experiments [84,85]. Increased binding of dependent interactions. Although stroma cell-associated pre-BCR ligands have these proteins was associated with the enhancer’s increased been identified, it is unclear whether this kind of interaction is critical for cells passing through the pre-BCR checkpoint. The lower part is a schematic of the accessibility to restriction endonucleases as well as increased germline ␬ locus including the V␬ and J␬ gene segments and the constant region association with acetylated histones in B cell lines mimicking exon (C␬) in light grey and the intronic (E␬i) and 3 (E␬3) enhancers in dark grey. various stages of B cell development, leading to the suggestion Transcription factor binding sites are shown beneath their respective enhancers. that accessibility has a greater affect on enhancer occupancy ␬ Dark arrows represent germline transcript promoters. than protein levels [84]. IRF4 binds E␬3 but in the absence of IRF4, PU.1 binding is disrupted, suggesting coordinated bind- lines. While recapitulating certain aspects of pre-B cell biol- ing of the two molecules [82,85]. There are no binding sites ogy, these transformed cells are an imperfect model regarding in the ␬ intronic enhancer for IRF4/PU.1, thus the resulting gene regulation during the pro-to-pre-B cell transition. Treat- complete absence of ␬ rearrangements is intriguing. Interest- ment of cells with LPS or growth of temperature-sensitive ingly, IRF4,8−/− B cell precursors fail to exit the cell cycle, AMuLV-transformed pro-B cells at the non-permissive temper- cannot down-regulate SLC components, and express low lev- ature stimulates ␬ germline transcription and rearrangements els of RAG proteins, all of which could contribute to the lack [80,81]. This activation is dependent on the activation of NF-␬B, of ␬ activation [85]. Because of the similarity between this a transcription factor composed of the five members of the Rel- phenotype and that of the BLNK null mouse and the fact family that can form various homo- and heterodimers. Indeed, in that BLNK levels are unaffected by IRF4,8 inactivation, the vivo footprinting of AMuLV-transformed cells revealed an LPS- authors suggest that IRF4,8 might be downstream of BLNK dependent NF-␬B footprint in the ␬ intronic enhancer [10].A in a signaling pathway. BLNK might promote exit from the direct comparison of NF-␬B site occupancy in primary pro- and cell cycle and differentiation as well as E␬3 factor binding pre-B cells showed that the NF-␬B site is bound with protein in causing ␬ activation. It would be interesting to know whether both pro- and pre-B cells. Gel shift analysis of pro- and pre-B cell these IRF4,8−/− B cells are able to allelically exclude IgHC nuclei revealed different levels of the distinct NF-␬B complexes, rearrangement. but footprinting cannot distinguish the nature of the interacting Another well-studied transcription factor, E2A, can activate protein. Ig␬ germline transcription and recombinase accessibility in non- Occupancy of binding sites within E␬3 changes across the lymphoid cell lines [86]. There are E2A binding sites (known as pro-B to pre-B cell transition [10]. A Pax5 site is occupied in E-boxes) in both Ig␬ enhancers, and a recent study showed that pro-B cells but not in pre-B cells. Likewise, pre-B cells showed two such sites in E␬i were critical for V-to-J␬ rearrangement hypersensitivity at binding sites that were not present in pro-B [87]. While a direct role for E2A has not yet been demonstrated, cells, namely those for Ets family factors, PU.1 or Spi-B, and involvement of this factor has attracted attention because of CRE, a cyclic AMP response element. The binding sites for the abundance of E2A binding sites within Ig loci and their PU.1/IRF-4 [82], CRE, and Pax5 are in very close proximity to ability to recruit chromatin modifying factors [88]. Specific each other and the authors depict a model where Pax5 binding chromatin modifications associated with V(D)J recombination interferes with binding of other factors until pre-BCR signal- have yet to be identified, although regions of the IgHC locus ing somehow reduces Pax5 DNA binding allowing CRE and poised for recombination associate with hyper-acetylated his- PU.1/IRF-4 to bind the 3 enhancer and activate the ␬LC locus tones H3 and H4, and hypo-methylated histone H3 Lysine 9 [10]. [89–91]. 36 J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39

6.2. κ locus activation and IgLC allelic exclusion 7. Conclusion

Recent attention has focused on the mechanism of IgLC The transition of B cell precursors through the pre-BCR allelic exclusion. While productive IgLC gene rearrangement checkpoint (i.e. from the pro-B to pre-B cell stage) requires leads to the formation of a BCR and inactivation of the recom- tightly coordinated signals emanating from surface pre-BCR binase, mechanisms must exist to prevent the near simultane- that directs complex changes in the pattern of gene expres- ous rearrangement and expression of both ␬ alleles in a pre-B sion and even movement of entire chromosomes within the cell. Liang et al. [92] recently reported that high-level acti- nucleus. Alterations in nuclear transcription then redirect the vation of the ␬ locus in pre-B cells occurs in only a small V(D)J recombinase to different rearranging Ig loci creating fraction of pre-B cells at any given time. Using a - molecules that then replace the originating receptor with a new less GFP cDNA reporter knocked into the coding region of molecule, the BCR. While the earlier steps in signaling cascades J␬1, these investigators measured the percentage of cells turn- are quickly being teased apart despite the complexity of redun- ing on a ␬ allele at each stage in development using flow dant pathways, and enhancers responsible for IgHC and ␬ locus cytometry. They found that pro-B cells from heterozygous activation are known, pathways linking signaling to retargeting mice did not express the reporter, while only 5% of small of the V(D)J recombinase remain vague at best. While over- pre-B cells did. Further experiments showed that ␬ activation expression of particular signaling molecules or transcription was mono-allelic and thus may contribute to allelic exclu- factors can mimic certain aspects of development, the nature of sion of IgLC rearrangement. The authors suggest that some signaling pathways makes over-expression and knock-out stud- threshold level of a combination of particular enhancer-binding ies difficult to interpret. This is particularly true in light of a transcription factors might allow for the rare and probabilis- stochastic model of ␬ activation, which requires the cumulative tic activation and rearrangement of a single ␬ allele at a action of multiple factors. A better understanding of chromatin time. structure’s precise role in accessibility and the coordinated activ- In contrast, Bergman and co-workers found biallelic germline ity of chromatin remodeling enzymes and transcription factors transcription in single-cell RT-PCR assays performed on pre- will help identify key components involved in transmitting sig- B cells [93]. They corroborated this finding with RNA-FISH nals from the pre-BCR to the ␮HC and ␬LC loci, and may also performed on AMuLV-transformed pre-B cells stimulated with determine causality of these correlated events. LPS, which revealed that 91% of cells expressed ␬LC from both alleles. These results conflict with those from Liang et al. dis- Acknowledgements cussed above who suggest monoallelic germline transcription precedes rearrangement. Differences between these two sets of Work in the authors’ lab has been generously supported results may have to do with the amounts of germline transcripts by grants from the National Institutes of Health (HL48702, detected in two different assays (GFP expression versus high AI40221, and AI57487). We wish to thank other members of cycle-number PCR). the Schlissel lab for stimulating discussions. DNA cytosine methylation can inhibit recombinase activ- ity [94] and monoallelic DNA de-methylation of the Ig␬ locus References correlates with its activation [95,96]. DNA methylation sta-  tus is not affected by loss of E␬3 but surprisingly both alle- [1] Winkler TH, Rolink A, Melchers F, Karasuyama H. Precursor B cells of les are demethylated in E␬i−/− B cells. In the absence of mouse bone marrow express two different complexes with the surrogate both enhancers, however, alleles remain fully methylated [78]. light chain on the surface. Eur J Immunol 1995;25:446–50. [2] Brouns GS, de Vries E, Borst J. Assembly and intracellular transport of Results from the Schlissel group corroborate the correlation the human B cell antigen receptor complex. Int Immunol 1995;7:359–68. of demethylation with activation, as the modified ␬-GFP allele [3] Gauthier L, Rossi B, Roux F, Termine E, Schiff C. Galectin-1 is a stro- was unmethylated in GFP+ pre-B cells, but was highly methy- mal cell ligand of the pre-B cell receptor (BCR) implicated in synapse lated in GFP− cells. Surprisingly, unrearranged ␬ alleles in formation between pre-B and stromal cells and in pre-BCR triggering. mature B cells remain CpG methylated but are nonetheless Proc Natl Acad Sci USA 2002;99:13014–9. [4] Fu C, Turck CW, Kurosaki T, Chan AC. BLNK: a central linker protein highly transcribed casting doubt on causality in the relationship in B cell activation. Immunity 1998;9:93–103. between DNA methylation and Ig␬ locus activation in this later [5] DeFranco AL. The complexity of signaling pathways activated by the stage. BCR. Curr Opin Immunol 1997;9:296–308. The timing of replication during S phase is associated with [6] Rao A, Luo C, Hogan PG. Transcription factors of the NFAT family: gene expression [97]. At the Ig loci, where alleles appear to regulation and function. Annu Rev Immunol 1997;15:707–47. [7] Schlessinger J. SH2/SH3 signaling proteins. Curr Opin Genet Dev be activated one at a time, replication is asynchronous [98]. 1994;4:25–30. This observation has led to the hypothesis that early repli- [8] Corcoran AE. Immunoglobulin locus silencing and allelic exclusion. cation could be associated with the allelic choice for recom- Semin Immunol 2005;17:141–54. bination. Asynchronous replication begins in early embryo- [9] Hardy RR, Carmack CE, Shinton SA, Kemp JD, Hayakawa K. Resolu- genesis, thus pre-BCR signaling does not influence this phe- tion and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J Exp Med 1991;173:1213–25. nomenon. Each of these epigenetic events is correlated with [10] Shaffer AL, Peng A, Schlissel MS. In vivo occupancy of the kappa light ␬LC rearrangement but the order of events needs to be chain enhancers in primary pro- and pre-B cells: a model for kappa locus established. activation. Immunity 1997;6:131–43. J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 37

[11] Muljo SA, Schlissel MS. A small molecule Abl kinase inhibitor [32] Shaw AC, Swat W, Davidson L, Alt FW. Induction of Ig light chain induces differentiation of Abelson virus-transformed pre-B cell lines. gene rearrangement in heavy chain-deficient B cells by activated Ras. Nat Immunol 2003;4:31–7. Proc Natl Acad Sci USA 1999;96:2239–43. [12] Shaffer AL, Schlissel MS. A truncated heavy chain protein relieves the [33] Iritani BM, Forbush KA, Farrar MA, Perlmutter RM. Control of requirement for surrogate light chains in early B cell development. J B cell development by Ras-mediated activation of Raf. EMBO J Immunol 1997;159:1265–75. 1997;16:7019–31. [13] Corcos D, Dunda O, Butor C, Cesbron JY, Lores P, Bucchini D, et al. [34] Schlissel MS, Morrow T. Ig heavy chain protein controls B cell devel- Pre-B-cell development in the absence of lambda 5 in transgenic mice opment by regulating germ-line transcription and retargeting V(D)J expressing a heavy-chain disease protein. Curr Biol 1995;5:1140–8. recombination. J Immunol 1994;153:1645–57. [14] Bannish G, Fuentes-Panana EM, Cambier JC, Pear WS, Monroe JG. [35] Yancopoulos GD, DePinho RA, Zimmerman KA, Lutzker SG, Rosen- Ligand-independent signaling functions for the B lymphocyte antigen berg N, Alt FW. Secondary genomic rearrangement events in pre-B receptor and their role in positive selection during B lymphopoiesis. J cells: VHDJH replacement by a LINE-1 sequence and directed class Exp Med 2001;194:1583–96. switching. EMBO J 1986;5:3259–66. [15] Rolink AG, Winkler T, Melchers F, Andersson J. Precursor B cell [36] Golding A, Chandler S, Ballestar E, Wolffe AP, Schlissel MS. Nucle- receptor-dependent B cell proliferation and differentiation does not osome structure completely inhibits in vitro cleavage by the V(D)J require the bone marrow or fetal liver environment. J Exp Med recombinase. EMBO J 1999;18:3712–23. 2000;191:23–32. [37] McBlane F, Boyes J. Stimulation of V(D)J recombination by histone [16] Bradl H, Jack HM. Surrogate light chain-mediated interaction of acetylation. Curr Biol 2000;10:483–6. a soluble pre-B cell receptor with adherent cell lines. J Immunol [38] Kwon J, Imbalzano AN, Matthews A, Oettinger MA. Accessibility of 2001;167:6403–11. nucleosomal DNA to V(D)J cleavage is modulated by RSS positioning [17] Bradl H, Wittmann J, Milius D, Vettermann C, Jack HM. Interaction and HMG1. Mol Cell 1998;2:829–39. of murine precursor B cell receptor with stroma cells is controlled by [39] Stanhope-Baker P, Hudson KM, Shaffer AL, Constantinescu A, Schlissel the unique tail of lambda 5 and stroma cell-associated heparan sulfate. MS. Cell type-specific chromatin structure determines the targeting of J Immunol 2003;171:2338–48. V(D)J recombinase activity in vitro. Cell 1996;85:887–97. [18] Ohnishi K, Melchers F. The nonimmunoglobulin portion of lambda5 [40] Kitamura D, Rajewsky K. Targeted disruption of mu chain mem- mediates cell-autonomous pre-B cell receptor signaling. Nat Immunol brane exon causes loss of heavy-chain allelic exclusion. Nature 2003;4:849–56. 1992;356:154–6. [19] Pelanda R, Braun U, Hobeika E, Nussenzweig MC, Reth M. B cell [41] Nussenzweig MC, Shaw AC, Sinn E, Campos-Torres J, Leder P. Allelic progenitors are arrested in maturation but have intact VDJ recombination exclusion in transgenic mice carrying mutant human IgM genes. J Exp in the absence of Ig-alpha and Ig-beta. J Immunol 2002;169:865–72. Med 1988;167:1969–74. [20] Reichlin A, Hu Y, Meffre E, Nagaoka H, Gong S, Kraus M, et al. B cell [42] Loffert D, Ehlich A, Muller W, Rajewsky K. Surrogate light chain development is arrested at the immature B cell stage in mice carrying expression is required to establish immunoglobulin heavy chain a mutation in the cytoplasmic domain of immunoglobulin beta. J Exp allelic exclusion during early B cell development. Immunity 1996;4: Med 2001;193:13–23. 133–44. [21] Kline GH, Hartwell L, Beck-Engeser GB, Keyna U, Zaharevitz S, [43] Mundt C, Licence S, Shimizu T, Melchers F, Martensson IL. Loss of Klinman NR, et al. Pre-B cell receptor-mediated selection of pre-B precursor B cell expansion but not allelic exclusion in VpreB1/VpreB2 cells synthesizing functional mu heavy chains. J Immunol 1998;161: double-deficient mice. J Exp Med 2001;193:435–45. 1608–18. [44] Shimizu T, Mundt C, Licence S, Melchers F, Martensson IL. [22] ten Boekel E, Melchers F, Rolink AG. Precursor B cells showing H VpreB1/VpreB2/lambda 5 triple-deficient mice show impaired B cell chain allelic inclusion display allelic exclusion at the level of pre-B cell development but functional allelic exclusion of the IgH locus. J Immunol receptor surface expression. Immunity 1998;8:199–207. 2002;168:6286–93. [23] Burrows PD, Stephan RP, Wang YH, Lassoued K, Zhang Z, Cooper [45] Novobrantseva TI, Martin VM, Pelanda R, Muller W, Rajewsky K, MD. The transient expression of pre-B cell receptors governs B cell Ehlich A. Rearrangement and expression of immunoglobulin light chain development. Semin Immunol 2002;14:343–9. genes can precede heavy chain expression during normal B cell devel- [24] Lam KP, Kuhn R, Rajewsky K. In vivo ablation of surface immunoglob- opment in mice. J Exp Med 1999;189:75–88. ulin on mature B cells by inducible gene targeting results in rapid cell [46] Galler GR, Mundt C, Parker M, Pelanda R, Martensson IL, Winkler death. Cell 1997;90:1073–83. TH. Surface mu heavy chain signals down-regulation of the V(D)J- [25] Melchers F. The pre-B-cell receptor: selector of fitting immunoglobulin recombinase machinery in the absence of surrogate light chain compo- heavy chains for the B-cell repertoire. Nat Rev Immunol 2005;5:578–84. nents. J Exp Med 2004;199:1523–32. [26] Karasuyama H, Rolink A, Shinkai Y, Young F, Alt FW, Melchers F. [47] Su YW, Flemming A, Wossning T, Hobeika E, Reth M, Jumaa H. The expression of Vpre-B/lambda 5 surrogate light chain in early bone Identification of a pre-BCR lacking surrogate light chain. J Exp Med marrow precursor B cells of normal and B cell-deficient mutant mice. 2003;198:1699–706. Cell 1994;77:133–43. [48] Schuh W, Meister S, Roth E, Jack HM. Cutting edge: signaling and [27] Rolink A, Grawunder U, Winkler TH, Karasuyama H, Melchers F. IL-2 cell surface expression of a mu H chain in the absence of lambda 5: a receptor alpha chain (CD25, TAC) expression defines a crucial stage in paradigm revisited. J Immunol 2003;171:3343–7. pre-B cell development. Int Immunol 1994;6:1257–64. [49] Minegishi Y, Conley ME. Negative selection at the pre-BCR check- [28] Hess J, Werner A, Wirth T, Melchers F, Jack HM, Winkler TH. Induction point elicited by human mu heavy chains with unusual CDR3 regions. of pre-B cell proliferation after de novo synthesis of the pre-B cell Immunity 2001;14:631–41. receptor. Proc Natl Acad Sci USA 2001;98:1745–50. [50] Costa TE, Suh H, Nussenzweig MC. Chromosomal position of rearrang- [29] Lazarus AH, Kawauchi K, Rapoport MJ, Delovitch TL. Antigen-induced ing gene segments influences allelic exclusion in transgenic mice. Proc B lymphocyte activation involves the p21ras and ras.GAP signaling path- Natl Acad Sci USA 1992;89:2205–8. way. J Exp Med 1993;178:1765–9. [51] Reth MG, Alt FW. Novel immunoglobulin heavy chains are produced [30] Harwood AE, Cambier JC. B cell antigen receptor cross-linking triggers from DJH gene segment rearrangements in lymphoid cells. Nature rapid protein kinase C independent activation of p21ras1. J Immunol 1984;312:418–23. 1993;151:4513–22. [52] Gu H, Kitamura D, Rajewsky K. B cell development regulated by [31] Shaw AC, Swat W, Ferrini R, Davidson L, Alt FW. Activated Ras sig- gene rearrangement: arrest of maturation by membrane-bound D mu nals developmental progression of recombinase-activating gene (RAG)- protein and selection of DH element reading frames. Cell 1991;65: deficient pro-B lymphocytes. J Exp Med 1999;189:123–9. 47–54. 38 J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39

[53] Ehlich A, Martin V, Muller W, Rajewsky K. Analysis of the B- [74] Chen J, Trounstine M, Alt FW, Young F, Kurahara C, Loring JF, cell progenitor compartment at the level of single cells. Curr Biol et al. Immunoglobulin gene rearrangement in B cell deficient mice 1994;4:573–83. generated by targeted deletion of the JH locus. Int Immunol 1993;5: [54] Haasner D, Rolink A, Melchers F. Influence of surrogate L chain on 647–56. DHJH-reading frame 2 suppression in mouse precursor B cells. Int [75] Grawunder U, Haasner D, Melchers F, Rolink A. Rearrangement and Immunol 1994;6:21–30. expression of kappa light chain genes can occur without mu heavy [55] Tornberg UC, Bergqvist I, Haury M, Holmberg D. Regulation of B chain expression during differentiation of pre-B cells. Int Immunol lymphocyte development by the truncated immunoglobulin heavy chain 1993;5:1609–18. protein Dmu. J Exp Med 1998;187:703–9. [76] Iritani BM, Alberola-Ila J, Forbush KA, Perimutter RM. Distinct sig- [56] Malynn BA, Shaw AC, Young F, Stewart V, Alt FW. Truncated nals mediate maturation and allelic exclusion in lymphocyte progenitors. immunoglobulin Dmu causes incomplete developmental progression of Immunity 1999;10:713–22. RAG-deficient pro-B cells. Mol Immunol 2002;38:547–56. [77] McGuire KL, Vitetta ES. Kappa/lambda shifts do not occur during mat- [57] Horne MC, Roth PE, DeFranco AL. Assembly of the truncated uration of murine B cells. J Immunol 1981;127:1670–3. immunoglobulin heavy chain D mu into antigen receptor-like complexes [78] Inlay M, Alt FW, Baltimore D, Xu Y. Essential roles of the kappa light in pre-B cells but not in B cells. Immunity 1996;4:145–58. chain intronic enhancer and 3 enhancer in kappa rearrangement and [58] Fang T, Smith BP, Roman CA. Conventional and surrogate light chains demethylation. Nat Immunol 2002;3:463–8. differentially regulate Ig mu and Dmu heavy chain maturation and sur- [79] Schlissel MS. Regulation of activation and recombination of the murine face expression. J Immunol 2001;167:3846–57. Igkappa locus. Immunol Rev 2004;200:215–23. [59] Xu S, Wong SC, Lam KP. Cutting edge: B cell linker protein is [80] Schlissel MS, Baltimore D. Activation of immunoglobulin kappa gene dispensable for the allelic exclusion of immunoglobulin heavy chain rearrangement correlates with induction of germline kappa gene tran- locus but required for the persistence of CD5+ B cells. J Immunol scription. Cell 1989;58:1001–7. 2000;165:4153–7. [81] Chen YY, Wang LC, Huang MS, Rosenberg N. An active v-abl protein [60] Schweighoffer E, Vanes L, Mathiot A, Nakamura T, Tybulewicz VL. tyrosine kinase blocks immunoglobulin light-chain gene rearrangement. Unexpected requirement for ZAP-70 in pre-B cell development and Genes Dev 1994;8:688–97. allelic exclusion. Immunity 2003;18:523–33. [82] Eisenbeis CF, Singh H, Storb U. Pip, a novel IRF family member, is [61] Hayashi K, Yamamoto M, Nojima T, Goitsuka R, Kitamura D. Distinct a lymphoid-specific, PU. 1. Dependent transcriptional activator. Genes signaling requirements for Dmu selection, IgH allelic exclusion, pre-B Dev 1995;9:1377–87. cell transition, and tumor suppression in B cell progenitors. Immunity [83] Schweitzer BL, DeKoter RP. Analysis of gene expression and Ig tran- 2003;18:825–36. scription in PU.1/Spi-B-deficient progenitor B cell lines. J Immunol [62] Su YW, Herzog S, Lotz M, Feldhahn N, Muschen M, Jumaa H. The 2004;172:144–54. molecular requirements for LAT-mediated differentiation and the role [84] McDevit DC, Perkins L, Atchison ML, Nikolajczyk BS. The Ig kappa 3 of LAT in limiting pre-B cell expansion. Eur J Immunol 2004;34: enhancer is activated by gradients of chromatin accessibility and protein 3614–22. association. J Immunol 2005;174:2834–42. [63] Su YW, Jumaa H. LAT links the pre-BCR to calcium signaling. Immu- [85] Lu R, Medina KL, Lancki DW, Singh H. IRF-4,8 orchestrate the nity 2003;19:295–305. pre-B-to-B transition in lymphocyte development. Genes Dev 2003;17: [64] Wen R, Chen Y, Schuman J, Fu G, Yang S, Zhang W, et al. An important 1703–8. role of phospholipase C gamma 1 in pre-B-cell development and allelic [86] Romanow WJ, Langerak AW, Goebel P, Wolvers-Tettero IL, van Dongen exclusion. EMBO J 2004;23:4007–17. JJ, Feeney AJ, et al. E2A and EBF act in synergy with the V(D)J recom- [65] Kosak ST, Skok JA, Medina KL, Riblet R, Le Beau MM, binase to generate a diverse immunoglobulin repertoire in nonlymphoid Fisher AG, et al. Subnuclear compartmentalization of immunoglob- cells. Mol Cell 2000;5:343–53. ulin loci during lymphocyte development. Science 2002;296:158– [87] Inlay MA, Tian H, Lin T, Xu Y. Important roles for E protein bind- 62. ing sites within the immunoglobulin kappa chain intronic enhancer [66] Sayegh C, Jhunjhunwala S, Riblet R, Murre C. Visualization of looping in activating Vkappa Jkappa rearrangement. J Exp Med 2004;200: involving the immunoglobulin heavy-chain locus in developing B cells. 1205–11. Genes Dev 2005;19:322–7. [88] Bradney C, Hjelmeland M, Komatsu Y, Yoshida M, Yao TP, Zhuang Y. [67] Roldan E, Fuxa M, Chong W, Martinez D, Novatchkova M, Busslinger Regulation of E2A activities by histone acetyltransferases in B lympho- M, et al. Locus ‘decontraction’ and centromeric recruitment contribute to cyte development. J Biol Chem 2003;278:2370–6. allelic exclusion of the immunoglobulin heavy-chain gene. Nat Immunol [89] Johnson K, Pflugh DL, Yu D, Hesslein DG, Lin KI, Bothwell AL, et al. 2005;6:31–41. B cell-specific loss of histone 3 lysine 9 methylation in the V(H) locus [68] Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Bus- depends on Pax5. Nat Immunol 2004;5:853–61. slinger M. Pax5 induces V-to-DJ rearrangements and locus contrac- [90] Morshead KB, Ciccone DN, Taverna SD, Allis CD, Oettinger tion of the immunoglobulin heavy-chain gene. Genes Dev 2004;18: MA. Antigen receptor loci poised for V(D)J rearrangement are 411–22. broadly associated with BRG1 and flanked by peaks of histone [69] Hsu LY, Liang HE, Johnson K, Kang C, Schlissel MS. Pax5 activates H3 dimethylated at lysine 4. Proc Natl Acad Sci USA 2003;100: immunoglobulin heavy chain V to DJ rearrangement in transgenic thy- 11577–82. mocytes. J Exp Med 2004;199:825–30. [91] Chowdhury D, Sen R. Stepwise activation of the immunoglobulin mu [70] Chowdhury D, Sen R. Transient IL-7/IL-7R signaling provides a mech- heavy chain gene locus. EMBO J 2001;20:6394–403. anism for feedback inhibition of immunoglobulin heavy chain gene [92] Liang HE, Hsu LY, Cado D, Schlissel MS. Variegated transcriptional rearrangements. Immunity 2003;18:229–41. activation of the immunoglobulin kappa locus in pre-b cells con- [71] Bolland DJ, Wood AL, Johnston CM, Bunting SF, Morgan G, Chakalova tributes to the allelic exclusion of light-chain expression. Cell 2004;118: L, et al. Antisense intergenic transcription in V(D)J recombination. Nat 19–29. Immunol 2004;5:630–7. [93] Singh N, Bergman Y, Cedar H, Chess A. Biallelic germline tran- [72] Hoffmann R, Seidl T, Neeb M, Rolink A, Melchers F. Changes in scription at the kappa immunoglobulin locus. J Exp Med 2003;197: gene expression profiles in developing B cells of murine bone marrow. 743–50. Genome Res 2002;12:98–111. [94] Whitehurst CE, Schlissel MS, Chen J. Deletion of germline promoter [73] Constantinescu A, Schlissel MS. Changes in locus-specific V(D)J recom- PD beta 1 from the TCR beta locus causes hypermethylation that binase activity induced by immunoglobulin gene products during B cell impairs D beta 1 recombination by multiple mechanisms. Immunity development. J Exp Med 1997;185:609–20. 2000;13:703–14. J.K. Geier, M.S. Schlissel / Seminars in Immunology 18 (2006) 31–39 39

[95] Mostoslavsky R, Singh N, Kirillov A, Pelanda R, Cedar H, [97] Schubeler D, Scalzo D, Kooperberg C, van Steensel B, Delrow J, Chess A, et al. Kappa chain monoallelic demethylation and Groudine M. Genome-wide DNA replication profile for Drosophila the establishment of allelic exclusion. Genes Dev 1998;12:1801– melanogaster: a link between transcription and replication timing. Nat 11. Genet 2002;32:438–42. [96] Goldmit M, Schlissel M, Cedar H, Bergman Y. Differential accessibility [98] Mostoslavsky R, Singh N, Tenzen T, Goldmit M, Gabay C, Elizur S, et at the kappa chain locus plays a role in allelic exclusion. EMBO J al. Asynchronous replication and allelic exclusion in the immune system. 2002;21:5255–61. Nature 2001;414:221–5.