In this issue of Blood, Yi et al reveal an important role for the protein phosphatase Wip1 (PPM1D) in the regulation of B-cell homeostasis.1  Mice deficient in the Wip1 gene display increased apoptosis in the pre–B-cell compartment and a reduction in peripheral B-cell numbers, a phenotype exacerbated with age and upon serial transplantations of bone marrow (BM) cells.1  Even though Wip1 has the ability to modulate multiple signaling pathways in the cell, the restoration of B-cell numbers upon deletion of the p53 gene1  suggests that an autoregulatory loop between p53 and Wip1 is of importance to maintain normal production of B lymphocytes.

Stage-specific expansion in the early B-cell compartment. The figure displays a schematic drawing of early B-cell development indicating stage-specific recombination events of the immunoglobulin heavy-chain (IgHR) and light-chain (IgLR) genes and the expansion or deletion (apoptosis indicated by a cross) of early pre–B cells. The lower part of the figure indicates the specific stages of the cell cycle (G1 [gap I], S [synthesis], G2 [gap II], and M [mitosis]), as well as the cell-cycle block in G1 induced by p21 and the ability of Wip1 to modulate the activity of p53 in G2 phase.

Stage-specific expansion in the early B-cell compartment. The figure displays a schematic drawing of early B-cell development indicating stage-specific recombination events of the immunoglobulin heavy-chain (IgHR) and light-chain (IgLR) genes and the expansion or deletion (apoptosis indicated by a cross) of early pre–B cells. The lower part of the figure indicates the specific stages of the cell cycle (G1 [gap I], S [synthesis], G2 [gap II], and M [mitosis]), as well as the cell-cycle block in G1 induced by p21 and the ability of Wip1 to modulate the activity of p53 in G2 phase.

Close modal

The development of mature B lymphocytes from hematopoietic stem cells in the BM is a complex process in which proliferation and expansion need to be coordinated with DNA recombination events as well as the selection of functional progenitor cells (see figure). In order to better understand the molecular interplay underlying the homeostatic expansion of B-lymphoid cells, Yi and colleges explored the functional role of the p53-activated serine/threonine protein phosphatase Wip12  in the formation and expansion of B-lymphoid cells.1 Wip1 was originally identified as a p53 target gene,2  and subsequent experiments revealed that this protein acts in an autoregulatory loop to reduce the functional activity of p53.3  The current report reveals that in the absence of Wip1, the formation of the pre–B-cell compartment is impaired.1  The early pre–B-cell stage represents one of the major windows for cell proliferation in early B-cell development, as coordinated interleukin-7 and pre–B-cell receptor signaling drives the proliferation of cells carrying a functional immunoglobulin (Ig) heavy-chain gene,4  and it is therefore reasonable that targeting of this developmental stage impairs the formation of mature B-lineage cells. p53 can block cell-cycle progression by induction of p21 expression; however, deletion of p21 could not rescue B-cell development in Wip1-deficient mice, arguing against the idea that the loss of pre–B cells would be due to a disruption of cell-cycle progression.1  Analysis of the cell-cycle status of defined B-cell compartments supported this notion, because no significant differences in G1-S-G2 composition could be detected. Rather, the phenotype appeared to be related to an increased apoptosis in the pre–B-cell compartment, a phenotype that could be rescued by loss of p53 function, indicating a need for harnessing p53 to control apoptosis in early B-cell development.

p53 is activated by DNA damage, and it is tempting to speculate that Ig rearrangements, known to induce double-strand breaks and a DNA damage response, would result in increased p53 activity that needs to be modulated by Wip1. However, while a CDK/cyclin A–mediated degradation of RAG-2 restricts Ig recombination to the G1 phase of the cell cycle,5  Wip1 has been suggested to act mainly in G2 phase, where the protein is able to potently release a cell-cycle block.6  Furthermore, DNA damage generated during G1 phase would likely result in sustained p21 expression and a cell-cycle block at the G1-S transition. Hence, the p53 response modulated by Wip1 is unlikely to be related to the Ig recombination process per se but rather to other DNA-damaging events occurring during the replication process.

The reduction in pre–B-cell numbers in Wip1-deficient mice could not be compensated for by peripheral expansion of mature B cells because the reduction in cell numbers was consistent in blood, lymph nodes, and spleen,1  possibly indicating an additional need of modulated p53 activity in peripheral cells. Even though Wip1 deficiency results in impaired T-cell development7  and an expanded peripheral pool of neutrophils8  that could potentially impact the peripheral expansion of the mature B-cell compartments, Yi et al use BM chimera experiments to demonstrate that Wip1 deficiency impacts B-cell development in a cell-autonomous manner.1  The impact of reduced Wip1 activity was further exacerbated upon serial transplantation or aging, suggesting an involvement of immature long-lived progenitor compartments. In line with this notion, the exacerbated phenotype was associated with reduced numbers of pre-pro– and pro–B cells that was not observed to the same extent in young adult mice.1  In further support of a role for Wip1 in early progenitors, recently published data suggest that regulation of p53 activity by Wip1 impacts long-lived hematopoietic progenitors in the BM.9 

Although there exists an apparent need to harness p53 activity to achieve normal B-cell production, there is likely a need to balance rather than abolish the activity of this protein, because loss of p53 function is a common event in human malignancies. Of special interest in the context of this report is the finding that a mutated or deleted TP53 gene is one of the strongest independent predictors of inferior treatment outcome in childhood B-lineage acute lymphoblastic leukemia.10  In a situation where the functional dose of a transcription factor like p53 is of critical importance, the use of an autoregulatory loop such as that created by the induction of Wip1 transcription by p53 presents an elegant solution to preserve a high output of normal cells while still preventing uncontrolled malignant proliferation of progenitor cells. Therefore, the extended insight to Wip1 function provided by this report is an important contribution to our understanding of regulatory events in early B-lymphocyte development.

Conflict-of-interest disclosure: The author declares no competing financial interests.

1
Yi
 
W
Hu
 
X
Chen
 
Z
, et al. 
Phosphatase Wip1 controls antigen-independent B-cell development in a p53-dependent manner.
Blood
2015
, vol. 
126
 
5
(pg. 
620
-
628
)
2
Fiscella
 
M
Zhang
 
H
Fan
 
S
, et al. 
Wip1, a novel human protein phosphatase that is induced in response to ionizing radiation in a p53-dependent manner.
Proc Natl Acad Sci USA
1997
, vol. 
94
 
12
(pg. 
6048
-
6053
)
3
Takekawa
 
M
Adachi
 
M
Nakahata
 
A
, et al. 
p53-inducible wip1 phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation.
EMBO J
2000
, vol. 
19
 
23
(pg. 
6517
-
6526
)
4
Ochiai
 
K
Maienschein-Cline
 
M
Mandal
 
M
, et al. 
A self-reinforcing regulatory network triggered by limiting IL-7 activates pre-BCR signaling and differentiation.
Nat Immunol
2012
, vol. 
13
 
3
(pg. 
300
-
307
)
5
Lee
 
J
Desiderio
 
S
Cyclin A/CDK2 regulates V(D)J recombination by coordinating RAG-2 accumulation and DNA repair.
Immunity
1999
, vol. 
11
 
6
(pg. 
771
-
781
)
6
Shaltiel
 
IA
Aprelia
 
M
Saurin
 
AT
, et al. 
Distinct phosphatases antagonize the p53 response in different phases of the cell cycle.
Proc Natl Acad Sci USA
2014
, vol. 
111
 
20
(pg. 
7313
-
7318
)
7
Schito
 
ML
Demidov
 
ON
Saito
 
S
Ashwell
 
JD
Appella
 
E
Wip1 phosphatase-deficient mice exhibit defective T cell maturation due to sustained p53 activation.
J Immunol
2006
, vol. 
176
 
8
(pg. 
4818
-
4825
)
8
Liu
 
G
Hu
 
X
Sun
 
B
, et al. 
Phosphatase Wip1 negatively regulates neutrophil development through p38 MAPK-STAT1.
Blood
2013
, vol. 
121
 
3
(pg. 
519
-
529
)
9
Chen
 
Z
Yi
 
W
Morita
 
Y
, et al. 
Wip1 deficiency impairs haematopoietic stem cell function via p53 and mTORC1 pathways.
Nat Commun
2015
, vol. 
6
 pg. 
6808
 
10
Krentz
 
S
Hof
 
J
Mendioroz
 
A
, et al. 
Prognostic value of genetic alterations in children with first bone marrow relapse of childhood B-cell precursor acute lymphoblastic leukemia.
Leukemia
2013
, vol. 
27
 
2
(pg. 
295
-
304
)
Sign in via your Institution