Fuxa 2007

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Transcriptional regulation in early B cell development

Martin Fuxa1 and Jane A Skok1,2

Transcription factors and signalling molecules are important for Lineage commitment in early B-cell
both lineage commitment and lineage-specific regulation. The development
B cell specification factor Pax5 plays a dual role in B lineage An early step in hematopoiesis is the commitment of
commitment. Simultaneously, it potentiates and limits multipotent progenitors to common myeloid progenitors
lineage choice by activating genes that are required for the B [1] or lymphoid-primed multipotent progenitors [2]. The
cell program while repressing lineage-inappropriate genes; latter subsequently differentiate to the earliest lymphocyte
more than 100 of the latter have now been identified. In this progenitors, which upregulate the lymphoid-specific
context, repression of the tyrosine kinase Flt3 has been shown recombinase genes Rag1 and Rag2 and give rise to common
to be essential for B lineage commitment. Regulation of antigen lymphoid progenitors (CLPs) that have their characteristic
receptor recombination constitutes another level at which B, T and natural killer cell developmental potential [3].
lineage specificity is determined, and the identification of two
factors, E47 and FOXP1, which regulate the activity of the The generation of lymphoid progenitors and B lympho-
recombinase enzymes in B lineage cells, provides insight into cytes depends on signalling through the c-Kit, Flt3 and
the mechanisms that determine this. New information IL-7 receptors; in their absence, early B cell progenitors in
regarding the control of ordered recombination and allelic the bone marrow are severely affected [4,5]. In addition,
exclusion comes from studies of cis-acting elements within the developmental control of early B lymphopoiesis is
Ig loci. exerted by a regulatory network of key transcription
Addresses factors that include PU.1 (an ets-family member), Ikaros,
1
The Department of Immunology and Molecular Pathology, Division of Bcl11a (a zinc finger transcription factor), E2A (a helix-
Infection and Immunity, University College London, London W1T 4JF, loop-helix protein), EBF (early B cell factor) and Pax5
United Kingdom
2
Department of Pathology, New York University School of Medicine, 550
(Figure 1) [3,6]. Whereas PU.1 appears to be crucial for
1st Avenue, MSB 531, New York, NY 10016, USA both myeloid and lymphoid progenitors [7], Ikaros con-
trols the development of lymphoid progenitors [8]. In the
Corresponding author: Skok, Jane A (jane.skok@med.nyu.edu) absence of Bcl11a, early B lymphopoiesis is blocked at
the CLP stage before the pre-pro-B stage [9]. E2A and
Current Opinion in Immunology 2007, 19:129–136 EBF are considered primary B-cell fate determinants and
co-ordinately activate the expression of B cell specific
This review comes from a themed issue on
Lymphocyte development genes. However, the mere activation of the B lymphocyte
Edited by James Hagman and Dietmar Kappes transcription program is not sufficient to commit B cell
progenitors to the B lymphoid lineage in the absence of
Available online 9th February 2007 the paired domain protein Pax5. Targeted deletion of
0952-7915/$ – see front matter Pax5 arrests B-cell development at the pro-B cell stage,
# 2006 Elsevier Ltd. All rights reserved. and these Pax5/ cells have the capacity to self-renew
extensively and retain a broad developmental potential
DOI 10.1016/j.coi.2007.02.002
similar to that of multipotent progenitors [10,11].

Role of Pax5 in potentiating and limiting


Introduction lineage choice
Hematopoietic development is a highly regulated multi- Pax5 fulfils a dual role in B lineage commitment by
step process in which pluripotent hematopoietic stem activating B cell specific genes while simultaneously
cells (HSCs) differentiate through intermediate progeni- repressing lineage-inappropriate genes [11]. Target genes
tors to produce all mature cell types of the blood. Limita- that are activated by Pax5 code for essential components
tion of lineage choice during development is largely of the pre-B- and B-cell receptor (pre-BCR and BCR,
regulated by a combination of transcription factors and respectively) signalling pathways [3]. These include Ebf1
signalling pathways. B lymphopoiesis is initiated by the (see Update), the receptor signalling chain Iga (encoded
entry of progenitors into the B-cell lineage transcription by mb-1) [10], the stimulatory co-receptors CD19 [12] and
programme and the concomitant sequential rearrange- CD21 [13], and the central adaptor protein BLNK [14].
ment of the immunoglobulin genes through V(D)J recom- Lineage choices are limited by Pax5 acting as a transcrip-
bination. In this review, we will discuss various aspects of tional repressor. In this capacity, the targets include
early B-cell development and will focus on some of Notch1 and c-fms, which encodes M-CSFR (macrophage
the most recent advances in transcriptional regulation colony stimulating factor receptor), which are important
of B-cell specification and differentiation. for the differentiation of progenitors into myeloid

www.sciencedirect.com Current Opinion in Immunology 2007, 19:129–136


130 Lymphocyte development

Figure 1

Early B-cell development in the bone marrow. A schematic diagram of early B lymphopoiesis, showing the successive differentiation stages and
the rearrangement status of IgH and IgL genes. DH–JH rearrangements are initiated in the earliest lymphocyte progenitors (ELPs) at a low level
and are completed as the cell progresses to the CLP and pre-pro-B cell (also referred to as CLP2) stage. VH–DJH recombination takes place in
pro-B cells, and successful rearrangement leads to expression of the Igm protein as part of the pre-BCR in large pre-B cells. Subsequent
rearrangement of the IgL locus in small pre-B cells results in the expression of the BCR (consisting of m heavy and k or l light chains) on
immature B cells (Imm. B). The approximate points of the developmental arrest in mice that have defective transcription factors PU.1, Ikaros,
Bcl11a, E2A, EBF, Pax5 and Foxp1 (black) or signalling components involved in signalling through c-Kit, Flt3 or IL-7R (grey) are indicated above.
E2A- and EBF-deficient progenitors in the bone marrow resemble pre-pro-B cells but lack all Ig rearrangements. Abbreviation: HSC,
haematopoietic stem cell.

lineages and T cells, respectively [11,15] (Figure 2). A that repression of Flt3 by Pax5 is an essential step in this
recent microarray screen that investigated the repressor process. Pax5-mediated gene repression was furthermore
function of Pax5 identified more than 100 target genes shown to be essential for normal homeostasis of the
[16]. Grouped according to category, these included hematopoietic system, because ectopic expression of
secreted proteins, cell adhesion molecules, signal trans- the Pax5-repressed chemokine gene Ccl3 in B cells results
ducers and nuclear proteins that are expressed in ery- in increased osteoclast formation and bone loss [16].
throid, myeloid and/or T lymphoid cells. Continuous
repression by Pax5 is known to be required, because Immunoglobulin gene rearrangement
reactivation of these repressed targets is observed follow- Although the initial stages of B lineage commitment
ing conditional deletion of Pax5 in pro-B and mature B depend upon the correct functioning of signalling path-
cells. Surprisingly, even at the transition to the plasma cell ways and transcription factors, ongoing differentiation is
stage, when Pax5 is physiologically lost during terminal additionally dependent on successful rearrangement of
differentiation, repressed genes are reactivated and con- immunoglobulin (Ig) gene loci, and stage-specific assembly
tribute to the plasma cell transcriptional program [16]. of immunoglobulin heavy chain (IgH) and light (IgL)
chain genes defines each phase of B-cell development.
Self-renewal capacity and developmental plasticity are In essence, recombination involves pairing of compatible
shared characteristics of Pax5/ pro-B cells and early conserved recombination signal sequences that flank the
haematopoietic progenitors. Hence, a key function of V, D and J gene segments of the Ig and T-cell receptor
Pax5 in B-lineage commitment is repression of genes (Tcr) gene segments [18–20]. Formation of a synaptic
that are required for maintaining the undifferentiated complex and subsequent introduction of double-stranded
state. Evidence in favour of this hypothesis comes from DNA breaks requires the recombinase enzymes RAG1
the recent observation that Pax5 binds to the promoter and RAG2. These proteins are expressed from an early
region of Flt3 and represses transcription [17]. This stage in lymphoid development and are present in lym-
tyrosine kinase receptor is required for efficient formation phoid progenitor cells as well as in B and T cells [18–20].
of the CLP. In contrast to wild-type pro-B cells, Pax5-
deficient progenitors were shown to express Flt3 and to The function of antigen receptor rearrangement is to
proliferate in a dose-dependent manner in response to generate sufficient diversity to enable recognition of
Flt3L. Furthermore, enforced expression of Flt3 in hae- any antigen encountered while at the same time ensuring
matopoietic cells impairs B lymphopoiesis. These data that each lymphocyte expresses a receptor that has a
contribute to our understanding of the process by which single specificity. As a common machinery is required
cells commit to the B lymphoid lineage by demonstrating for recombination of variable (V), diversity (D) and joining

Current Opinion in Immunology 2007, 19:129–136 www.sciencedirect.com


Transcriptional regulation in early B cell development Fuxa and Skok 131

Figure 2

Dual role of Pax5 at B lineage commitment. Pax5 activates B cell specific genes (e.g. CD19, CD22 and mb-1) and simultaneously represses
lineage-inappropriate genes at B cell commitment. The latter are essential for diverse cellular functions such as cell–cell communication,
adhesion and migration in T lymphoid, myeloid and erythroid cells as well as in multipotent progenitors.

(J) gene segments in all immune receptor loci, devel- lineage cells, it might play a different role at later stages of
opmentally ordered changes in accessibility are crucial for B-cell development. It is now known that the nuclear
maintaining the integrity of this process [21]. Regulation periphery can consist of more than one subcompartment,
of accessibility is exerted at several levels to ensure favouring opposing outcomes for transcriptional regula-
lineage specificity, sequential rearrangement of IgH tion. In yeast, nuclear pore association has been shown to
and IgL genes and individual gene segments, and allelic optimize transcription of an inducible gene [26,27];
exclusion. location of genes in these regions could have a similar
function in mammalian cells and might explain the find-
Lineage specificity ing that Ig loci are relocated to this subcompartment in
Enrichment of IgH and Igk at the nuclear periphery has mature B lineage cells in which these loci are being
been shown to be important in limiting Ig recombination to actively transcribed [28] (JAS, unpublished).
the appropriate lineage [22,23]. In non-B lineage cells, IgH
and Igk are anchored with their 50 ends positioned closest to Ordered rearrangement
the periphery and their 30 ends located more centrally Recombination begins in the B-cell lineage at the pro-B
[24,25]. At the IgH locus, a region of 1 Mb at the 50 cell stage with rearrangement of the IgH locus, whereas
end has been shown to be required for colocalization with recombination of IgL takes place at the later pre-B cell
the nuclear lamina. Silencing of genes at the nuclear stage. Ordered recombination is not limited to the loci but
periphery might result from an exclusion of transcription extends to the gene segments so that DH segments are
factors, enrichment of transcriptional repressors or a com- first joined to JH segments of the IgH loci, which is
bination of both. Although expression of B lineage tran- followed by VH–DJH recombination.
scription factors has been implicated in movement towards
the centre of the nucleus [23,24], the factors involved in The transcription factors E2A and EBF control the initial
maintaining the default position of these loci at the per- DH–JH rearrangement step by activating expression of the
iphery in their silent state have not been identified to date. Rag genes and promoting accessibility of the DH–JH
region within the IgH locus [29,30]. PAX5, by contrast,
It should be noted that, although peripheral location induces large-scale contraction of the IgH locus; this is
is clearly important for limiting accessibility in non-B essential for the second stage of VH–DJH recombination,

www.sciencedirect.com Current Opinion in Immunology 2007, 19:129–136


132 Lymphocyte development

enabling synapse formation of distant gene segments that Allelic exclusion


facilitates rearrangement [24]. RAG-mediated recombi- Allelic exclusion ensures monoallelic expression of anti-
nation is primarily determined by localized changes in gen receptor loci, which is essential for single specificity
chromatin structure, which are regulated at multiple antigen recognition. Accumulating evidence supports a
levels including onset of sense and antisense germline ‘feedback inhibition’ model for establishing allelic
transcription [21,31], alterations in histone modifications exclusion [38]. Functional rearrangement leads to
[32,33] and nucleosome remodelling [34]. expression of Igm as part of the pre-BCR; this acts as
an important checkpoint preventing ongoing rearrange-
Transcriptional control of the Rag genes themselves ment of the second DJH rearranged allele at the pre-B
constitutes another aspect of regulation. The previously cell stage of development when IgL rearrangement takes
identified enhancer of RAG transcription (Erag) regulates place. Following a functional rearrangement at the IgH
RAG expression specifically in B cells but not in T locus, RAG protein expression is transiently downregu-
lymphocytes, and its targeted deletion leads to a partial lated upon pre-BCR signalling. Signalling through the
block at the pro-B cell stage [35]. More insight into IL-7R is attenuated at this stage and contributes to
transcriptional regulation of RAG expression comes from allelic exclusion by decreasing accessibility at the IgH
two recent studies that demonstrate B lineage specific locus [39]. Decontraction occurs at the IgH locus follow-
control [36,37]. ing recombination and is instrumental in establishing
allelic exclusion as this process physically separates
Borghesi et al. [36] show that loss of the transcription distant gene segments, preventing further interaction.
factor E47, an E2A gene product, results in diminished In addition, following successful recombination of one
CLP and pre-pro-B subsets as well as a complete absence IgH allele, the second allele is repositioned at centro-
of pro-B cells. Developmental progression to other meric heterochromatin at the pre-B cell stage of devel-
lineages is reduced but remains intact. In addition, this opment while IgL recombination takes place [25].
study clearly demonstrates that E47 is required for RAG1 Recruitment of the second allele to a repressive com-
expression and recombinase activity as early as the CLP partment of the nucleus is thought to act as a back-up
stage. Interestingly, reduced recombinase expression is mechanism for establishing allelic exclusion by prevent-
apparent in B cells, occurs to a lesser extent in dendritic ing accessibility to recombinase enzymes during IgL
and natural killer cells, but is absent in T cells. recombination when the recombinase enzymes are once
again upregulated (Figure 3).
In agreement with the B lineage specific requirements for
V(D)J recombination activity, Hu et al. [37] demonstrate In contrast to the IgH locus, repositioning of the Igk locus
a significant role for the forkhead protein FOXP1 in this to centromeric clusters occurs at the pre-B cell stage of
context. Targeted deletion of Foxp1 results in embryonic development, limiting accessibility to a single allele
lethality, but reconstitution experiments using Foxp1/ during recombination [25,40]. Although recruitment
fetal liver cells in Rag2-deficient recipients leads to a of the IgH locus to centromeric clusters and deacetylation
dramatic reduction in mature B cell numbers and a differ- have been correlated with a decrease in IL-7R signalling,
entiation defect in the transition from pro-B to pre-B cells. little is known about the mechanisms that govern regu-
No obvious equivalent impairment in thymocyte devel- lation of reversible locus contraction and repositioning of
opment was detected. Molecular characterization of these loci to heterochromatic regions. Recently, however, a cis-
Foxp1/ pro-B cells demonstrated reduced expression of acting silencing element has been identified that has a
the B-cell regulator genes Tcfe2a (which encodes E2A) and role in directing the Igk locus to centromeric heterochro-
Ebf in addition to reduced expression of B cell specific matin and association with the transcription factor Ikaros
genes regulated downstream from the E2A and EBF path- [41]. This silencer, known as Sis (silencer in the inter-
ways. These include Pax5, components of the pre-BCR, vening sequence), is located between the most 30 Vk gene
and Rag1 and Rag2. Further analysis demonstrated that and the Jk gene segments and is the first cis-acting
Foxp1 mutant pro-B cells have a substantial reduction in element to be identified that is involved in centromeric
the frequency of VH–DJH recombination involving both recruitment of the Ig loci.
proximal and distal VH gene segments, whereas DH–JH
recombination is less severely affected. This phenotype is Monoallelic silencing [40] and a low level of monoallelic
reminiscent of the V(D)J recombination defect seen in activation [42] have been implicated in allelic exclusion at
Erag/ mice [35]. The fact that both E2A and FOXP1 the Igk locus and, in this context, enhancer elements
bind the Erag enhancer in vivo and exhibit similar loss-of- might be important. Both IgH and Igk loci contain an
function phenotypes suggests that these two transcription intronic enhancer located between the J gene segments
factors and this cis-acting element in the RAG locus are and the constant region. A recent study demonstrated that
essential for regulating V(D)J recombinase activity during deletion of the Igk locus intronic enhancer/matrix attach-
B lymphopoiesis. The way in which they co-operate ment region (MiEk/) severely reduces accessibility and
remains to be determined. rearrangement of this locus in pre-B cells [43]. This has

Current Opinion in Immunology 2007, 19:129–136 www.sciencedirect.com


Transcriptional regulation in early B cell development Fuxa and Skok 133

Figure 3

Conformational changes and subnuclear relocation of immunoglobulin loci during B-cell development. (a) Germline organization of the murine Ig loci.
The mouse IgH and Igk loci together with the relative position of V gene segment families (yellow rectangles), indicating the most distal and proximal
family. In addition, D (orange vertical lines) and J segments (green vertical lines) as well as the constant regions (C, light blue rectangles) are shown.
Previously characterized enhancers and promotors are represented as red ovals and grey rectangles, respectively. The 1 Mb region that encompasses
the majority of the most distal VH gene family (J558) and appears to be important for adherence of the IgH locus to the nuclear periphery is depicted as
a dark grey bar. The location of the cis-acting element, Sis, which is required for targeting the Igk locus to centromeric heterochromatin, is shown as a
dark blue oval. The asterisk indicates the nonfunctional Jk pseudogene segment. (b) Subnuclear relocation and locus contraction of the IgH and Igk
loci. A diagram of the nucleus illustrates the location of the IgH and Igk loci and the rearrangement status at different stages of B-cell development. The
expression of Pax5, the pre-BCR and the BCR at the corresponding differentiation stages is indicated. The distal (50 ) and proximal (30 ) domains (dark
and light coloured circles, respectively) are depicted together with their location relative to the repressive compartments at the nuclear periphery (grey
shaded area) and centromeric heterochromatin (dark grey). The locus contraction/decontraction and monoallelic, centromeric recruitment states are
schematically shown for progenitors, pro-B, pre-B and activated B cells.

been attributed to reduced accessibility and germline the effect, if any, of enhancer replacement on IgH re-
transcription. Replacing the endogenous intronic enhan- arrangement remains unclear.
cer of the Igk locus (MiEk) with the intronic heavy chain
enhancer (Em) promotes changes in accessibility and Conclusions
greatly increases premature recombination of the Igk In this review, we have discussed recent advances in
locus at the pro-B cell stage of development. In addition, transcriptional regulation of early B-cell development.
Igk rearrangement is reduced at the pre-B cell stage. In Detailed molecular analyses have provided important
these EmR replacement loci, Igk rearrangement mimics insight into the gene repression function of Pax5 during
the normal stage-specific order of IgH recombination, B-cell development. The transcriptional regulation of this
indicating a role for the Em enhancer in directing ordered lineage commitment factor is determined by its inter-
rearrangement. Further analysis will be required to deter- action with distinct partner proteins. For instance, Pax5
mine whether these EmR replacement loci undergo pre- cooperates with PU.1 to recruit co-repressors of the
mature changes in nuclear organization [25]. In addition, Groucho protein family in a context-depending manner

www.sciencedirect.com Current Opinion in Immunology 2007, 19:129–136


134 Lymphocyte development

to repressed target genes [3]. By contrast, cooperative 2. Adolfsson J, Mansson R, Buza-Vidas N, Hultquist A, Liuba K,
Jensen CT, Bryder D, Yang L, Borge OJ, Thoren LA et al.:
binding of Pax5 and Ets proteins to the mb-1 promoter Identification of Flt3+ lympho-myeloid stem cells lacking
results in transcriptional activation [44]. Further studies erythro-megakaryocytic potential a revised road map for adult
blood lineage commitment. Cell 2005, 121:295-306.
are necessary to identify other Pax5 interaction partners
3. Busslinger M: Transcriptional control of early B cell
that are required for the downregulation of diverse line- development. Annu Rev Immunol 2004, 22:55-79.
age-inappropriate cellular functions and the activation of
4. Waskow C, Paul S, Haller C, Gassmann M, Rodewald HR: Viable
the B-cell program during B-cell development. c-KitW/W mutants reveal pivotal role for c-kit in the
maintenance of lymphopoiesis. Immunity 2002, 17:277-288.
Identification of E2A and FOXP1 as two key factors that 5. Vosshenrich CA, Cumano A, Muller W, Di Santo JP, Vieira P:
regulate recombinase activity specifically in B cells pro- Thymic stromal-derived lymphopoietin distinguishes fetal
from adult B cell development. Nat Immunol 2003, 4:773-779.
vides insight into the mechanisms that control lineage-
specific recombination. Although these proteins bind to a 6. Medina KL, Singh H: Genetic networks that regulate B
lymphopoiesis. Curr Opin Hematol 2005, 12:203-209.
previously characterized enhancer region in the Rag locus,
additional regulatory elements appear to be necessary for 7. Dakic A, Metcalf D, Di Rago L, Mifsud S, Wu L, Nutt SL: PU.1
regulates the commitment of adult hematopoietic progenitors
lineage specificity. Further investigations will determine and restricts granulopoiesis. J Exp Med 2005, 201:1487-1502.
the mechanism of transcription factor cooperation at these 8. Allman D, Miller JP: Common lymphoid progenitors, early
sites. B-lineage precursors, and IL-7: characterizing the trophic and
instructive signals underlying early B cell development.
Immunol Res 2003, 27:131-140.
Transcriptional regulators that influence changes in
9. Liu P, Keller JR, Ortiz M, Tessarollo L, Rachel RA, Nakamura T,
nuclear location of Ig loci during B-cell development Jenkins NA, Copeland NG: Bcl11a is essential for normal
remain to be identified. The molecular mechanisms that lymphoid development. Nat Immunol 2003, 4:525-532.
underlie locus contraction are currently unknown and we 10. Nutt SL, Urbanek P, Rolink A, Busslinger M: Essential functions
have little insight into the factors that regulate centro- of Pax5 (BSAP) in pro-B cell development: difference between
fetal and adult B lymphopoiesis and reduced V-to-DJ
meric recruitment. Future investigations should aid in recombination at the IgH locus. Genes Dev 1997, 11:476-491.
characterizing the transcription factor composition of
11. Nutt SL, Heavey B, Rolink AG, Busslinger M: Commitment to the
distinct nuclear subcompartments, which will further B-lymphoid lineage depends on the transcription factor Pax5.
our understanding of these events. Nature 1999, 401:556-562.
12. Nutt SL, Morrison AM, Dorfler P, Rolink A, Busslinger M:
Update Identification of BSAP (Pax-5) target genes in early B-cell
development by loss- and gain-of-function experiments.
A recent study [45] that has analyzed differential regu- EMBO J 1998, 17:2319-2333.
lation of two promoters within the Ebf1 gene adds yet 13. Horcher M, Souabni A, Busslinger M: Pax5/BSAP maintains the
another layer of complexity to the control of transcrip- identity of B cells in late B lymphopoiesis. Immunity 2001,
tional regulation during B lymphopoiesis. Activation of 14:779-790.
the distal Ebf1 promoter is induced by E2A and EBF1 in 14. Schebesta M, Pfeffer PL, Busslinger M: Control of pre-BCR
addition to IL-7 signaling, whereas the stronger proximal signaling by Pax5-dependent activation of the BLNK gene.
Immunity 2002, 17:473-485.
promoter is activated by the combination of PAX5, ETS1
15. Souabni A, Cobaleda C, Schebesta M, Busslinger M: Pax5
and PU.1. Absence of PAX5 impairs the function of the promotes B lymphopoiesis and blocks T cell development by
proximal Ebf1 promoter and accumulation of EBF1 repressing Notch1. Immunity 2002, 17:781-793.
protein. In concert with this, the replication timing and 16. Delogu A, Schebesta A, Sun Q, Aschenbrenner K, Perlot T,
subnuclear localization of the Ebf1 locus are altered. This  Busslinger M: Gene repression by Pax5 in B cells is essential for
blood cell homeostasis and is reversed in plasma cells.
indicates that the presence of an additional promoter can Immunity 2006, 24:269-281.
further refine control of the expression of a locus and has Identification of 110 Pax5-repressed genes provides important insight
into the repression function of Pax5 in B lineage commitment. These
wider implications for feedback loops within the regulat- genes fulfil diverse functions in different hematopoietic cell types and
ory network that orchestrates B cell development. encode proteins involved in receptor signalling, cell adhesion, migration,
transcriptional control and cellular metabolism. Expression analyses of
hematopoietic progenitors revealed that Pax5/ pro-B cells and com-
Acknowledgements mon lymphoid progenitors display lymphoid and myeloid promiscuity of
We thank Meinrad Busslinger and Adrian Erlebacher for critical reading of gene expression. Conditional deletion of Pax5 in committed pro-B cells
the manuscript. This work was supported by a Wellcome trust project grant and mature B cells demonstrates that Pax5 activity is continuously
and New York University School of Medicine start-up laboratory funding. required for the repression of these lineage-inappropriate genes.
Pax5-repressed genes are also reactivated in plasma cells, indicating
that the physiological loss of Pax5 during terminal differentiation con-
References and recommended reading tributes to the plasma cell transcription program.
Papers of particular interest, published within the period of review,
have been highlighted as: 17. Holmes ML, Carotta S, Corcoran LM, Nutt SL: Repression of Flt3
 by Pax5 is crucial for B-cell lineage commitment.
 of special interest Genes Dev 2006, 20:933-938.
 of outstanding interest The role of Pax5 in lineage commitment is extended by the finding that
Pax5 represses a crucial growth factor receptor, Flt3, which is required for
efficient formation of early lymphoid progenitors. Pax5-deficient pro-B
1. Akashi K, Traver D, Miyamoto T, Weissman IL: A clonogenic cells express abundant Flt3 that is rapidly silenced upon the introduction
common myeloid progenitor that gives rise to all myeloid of Pax5, which binds to the Flt3 promoter in vivo. Enforced expression of
lineages. Nature 2000, 404:193-197. Flt3 in wild-type progenitors significantly impairs B-cell development and

Current Opinion in Immunology 2007, 19:129–136 www.sciencedirect.com


Transcriptional regulation in early B cell development Fuxa and Skok 135

indicates that the repression of Flt3 by Pax5 is essential for normal 34. Maes J, O’Neill LP, Cavelier P, Turner BM, Rougeon F,
B-lymphopoiesis. Goodhardt M: Chromatin remodeling at the Ig loci prior to V(D)J
recombination. J Immunol 2001, 167:866-874.
18. Hesslein DG, Schatz DG: Factors and forces controlling V(D)J
recombination. Adv Immunol 2001, 78:169-232. 35. Hsu LY, Lauring J, Liang HE, Greenbaum S, Cado D, Zhuang Y,
Schlissel MS: A conserved transcriptional enhancer regulates
19. Bassing CH, Swat W, Alt FW: The mechanism and regulation RAG gene expression in developing B cells. Immunity 2003,
of chromosomal V(D)J recombination. Cell 2002, 19:105-117.
109:S45-S55.
36. Borghesi L, Aites J, Nelson S, Lefterov P, James P,
20. Schlissel MS: Regulating antigen-receptor gene assembly.  Gerstein R: E47 is required for V(D)J recombinase activity
Nat Rev Immunol 2003, 3:890-899. in common lymphoid progenitors. J Exp Med 2005,
21. Yancopoulos GD, Alt FW: Developmentally controlled and 202:1669-1677.
tissue-specific expression of unrearranged VH gene E47, an alternative splice product of the transcription factor E2A, is shown
segments. Cell 1985, 40:271-281. to be required for expression of both Rag1 and V(D)J recombinase activity
in multipotent hematopoietic and early B-cell progenitors. The loss of E47
22. Kosak ST, Skok JA, Medina KL, Riblet R, Le Beau MM, Fisher AG, results in a ten-fold reduction of the CLP subset that lacks D–JH rear-
Singh H: Subnuclear compartmentalization of immunoglobulin rangements and a profound arrest before the pro-B cell stage. Other
loci during lymphocyte development. Science 2002, downstream lymphoid progeny of CLPs are, however, still intact in these
296:158-162. mice, albeit at reduced numbers. Although recombinase activity is inhib-
ited in early B lineage precursors in E47-deficient animals, loss of either
23. Sato H, Saito-Ohara F, Inazawa J, Kudo A: Pax-5 is essential for k E47 or its cis-acting target Erag has little effect on recombinase activity in
sterile transcription during Ig k chain gene rearrangement. thymic T lineage precursors. This report defines a role for E47 in lineage
J Immunol 2004, 172:4858-4865. progression at the CLP stage and in regulating B cell specific recombi-
nase activity.
24. Fuxa M, Skok J, Souabni A, Salvagiotto G, Roldan E, Busslinger M:
Pax5 induces V-to-DJ rearrangements and locus contraction 37. Hu H, Wang B, Borde M, Nardone J, Maika S, Allred L, Tucker PW,
of the immunoglobulin heavy-chain gene. Genes Dev 2004,  Rao A: Foxp1 is an essential transcriptional regulator of B cell
18:411-422. development. Nat Immunol 2006, 7:819-826.
The forkhead transcription factor Foxp1 is essential for early B-cell
25. Roldan E, Fuxa M, Chong W, Martinez D, Novatchkova M, development and leads to a differentiation defect in the transition from
 Busslinger M, Skok JA: Locus ‘decontraction’ and centromeric the pro-B cell to the pre-B cell stage. Foxp1 deficiency is associated with
recruitment contribute to allelic exclusion of the decreased expression of all B lineage genes and of Rag1 and Rag2 in
immunoglobulin heavy-chain gene. Nat Immunol 2005, early B-cell progenitors in the fetal liver. Foxp1 binds to the Erag enhancer
6:31-41. and its loss leads to impaired DH–JH and VH–DJH rearrangement in a B-
This study uses three-dimensional DNA fluorescence in situ hybridiza- cell lineage specific way. The loss-of-function phenotype of Foxp1
tion analysis to demonstrate that distal V gene segments are brought resembles that of E2A+/ EBF+/ compound heterozygous mice. This
into contact with the proximal domains of Ig loci by DNA looping. report identifies Foxp1 as an essential participant in the transcriptional
Reversible locus contraction facilitates synapse formation and recom- regulatory network of B lymphopoiesis.
bination of gene segments separated by a large distance in cells
that are undergoing recombination. Pre-BCR signalling induces 38. Jung D, Giallourakis C, Mostoslavsky R, Alt FW: Mechanism and
decontraction, which prevents ongoing recombination at the second control of V(D)J recombination at the immunoglobulin heavy
DJH rearranged IgH allele, and centromeric recruitment, which reposi- chain locus. Annu Rev Immunol 2006, 24:541-570.
tions the non-functional allele within a repressive subcompartment
of the nucleus during IgL rearrangement. Thus, both decontraction 39. Chowdhury D, Sen R: Transient IL-7/IL-7R signaling provides a
and centromeric recruitment are important for establishing allelic mechanism for feedback inhibition of immunoglobulin heavy
exclusion. chain gene rearrangements. Immunity 2003, 18:229-241.
26. Taddei A, Gasser SM: Multiple pathways for telomere tethering: 40. Goldmit M, Ji Y, Skok J, Roldan E, Jung S, Cedar H, Bergman Y:
functional implications of subnuclear position for  Epigenetic ontogeny of the Igk locus during B cell
heterochromatin formation. Biochim Biophys Acta 2004, development. Nat Immunol 2005, 6:198-203.
1677:120-128. This study analyzes the epigenetic changes that occur at the Igk locus
during B-cell development. The authors report that, in pre-B cells, one
27. Schmid M, Arib G, Laemmli C, Nishikawa J, Durussel T, allele is preferentially packaged into active chromatin whereas the other
Laemmli UK: Nup-PI: the nucleopore-promoter interaction of allele is repositioned at centromereric heterochromatin and is associated
genes in yeast. Mol Cell 2006, 21:379-391. with HP1 and Ikaros.
28. Yang Q, Riblet R, Schildkraut CL: Sites that direct nuclear 41. Liu Z, Widlak P, Zou Y, Xiao F, Oh M, Li S, Chang MY, Shay JW,
compartmentalization are near the 50 end of the mouse  Garrard WT: A recombination silencer that specifies
immunoglobulin heavy-chain locus. Mol Cell Biol 2005, heterochromatin positioning and Ikaros association in the
25:6021-6030. immunoglobulin k locus. Immunity 2006, 24:405-415.
Using yeast artificial chromosome-based single-copy isotransgenic
29. Romanow WJ, Langerak AW, Goebel P, Wolvers-Tettero IL, mice, these authors identified a cis-acting element residing in the V–J
van Dongen JJ, Feeney AJ, Murre C: E2A and EBF act in synergy intervening sequence. This element, termed Sis, negatively regulates
with the V(D)J recombinase to generate a diverse rearrangement in this locus and appears necessary for targeting germ-
immunoglobulin repertoire in nonlymphoid cells. Mol Cell 2000, line Igk transgenes to centromeric heterochromatin in pre-B and B cells.
5:343-353. Sis is furthermore shown to associate with Ikaros — a repressor protein
30. Goebel P, Janney N, Valenzuela JR, Romanow WJ, Murre C, that also colocalizes with centromeric heterochromatin. The data sug-
Feeney AJ: Localized gene-specific induction of gest that Sis participates in the monoallelic silencing aspect of allelic
accessibility to V(D)J recombination induced by E2A and exclusion regulation through targeting to centromeric heterochromatin
early B cell factor in nonlymphoid cells. J Exp Med 2001, domains.
194:645-656.
42. Liang HE, Hsu LY, Cado D, Schlissel MS: Variegated
31. Bolland DJ, Wood AL, Johnston CM, Bunting SF, Morgan G, transcriptional activation of the immunoglobulin k locus in
Chakalova L, Fraser PJ, Corcoran AE: Antisense intergenic pre-B cells contributes to the allelic exclusion of light-chain
transcription in V(D)J recombination. Nat Immunol 2004, expression. Cell 2004, 118:19-29.
5:630-637.
43. Inlay MA, Lin T, Gao HH, Xu Y: Critical roles of the
32. McBlane F, Boyes J: Stimulation of V(D)J recombination by  immunoglobulin intronic enhancers in maintaining the
histone acetylation. Curr Biol 2000, 10:483-486. sequential rearrangement of IgH and Igk loci. J Exp Med 2006,
203:1721-1732.
33. Chowdhury D, Sen R: Stepwise activation of the To identify cis-acting elements that regulate ordered rearrangement, the
immunoglobulin m heavy chain gene locus. EMBO J 2001, authors of this study replaced the endogenous matrix attachment
20:6394-6403. region/Igk intronic enhancer (MiEk) with its heavy chain counterpart

www.sciencedirect.com Current Opinion in Immunology 2007, 19:129–136


136 Lymphocyte development

(Em). Replacement substantially increases accessibility of both Vk and cooperates with Runx1 and mediates epigenetic changes
Jk segments to the recombinase enzymes in pro-B cells and induces Igk associated with mb-1 transcription. Nat Immunol 2004,
rearrangement in these cells. Replacement loci, however, do not sup- 5:1069-1077.
port Igk rearrangement in pre-B cells. This suggests an important role for
Em and MiEk in stage-specific ordered rearrangement. 45. Roessler S, Györy I, Imhof S, Spivakov M, Williams RR, Busslinger
M, Fisher AG, Grosschedl R: Distinct promoters mediate the
44. Maier H, Ostraat R, Gao H, Fields S, Shinton SA, Medina KL, regulation of Ebf1 gene expression by IL-7 and Pax5.
Ikawa T, Murre C, Singh H, Hardy RR et al.: Early B cell factor Mol Cell Biol 2006, in press.

Endeavour
The quarterly magazine for the history and
philosophy of science.

You can access Endeavour online on


ScienceDirect, where you’ll find book reviews,
editorial comment and a collection of beautifully
illustrated articles on the history of science.

Featuring:

Information revolution: William Chambers, the publishing pioneer by A. Fyfe


Does history count? by K. Anderson
Waking up to shell shock: psychiatry in the US military during World War II by H. Pols
Deserts on the sea floor: Edward Forbes and his azoic hypothesis for a lifeless deep ocean by T.R. Anderson and T. Rice
‘Higher, always higher’: technology, the military and aviation medicine during the age of the two world wars by C. Kehrt
Bully for Apatosaurus by P. Brinkman

Coming soon:

Environmentalism out of the Industrial Revolution by C. Macleod


Pandemic in print: the spread of influenza in the Fin de Siècle by J. Mussell
Earthquake theories in the early modern period by F. Willmoth
Science in fiction - attempts to make a science out of literary criticism by J. Adams
The birth of botanical Drosophila by S. Leonelli

And much, much more. . .

Endeavour is available on ScienceDirect, www.sciencedirect.com

Current Opinion in Immunology 2007, 19:129–136 www.sciencedirect.com

You might also like