You are on page 1of 12

Cell Stem Cell

Article

A Distinctive DNA Damage Response in Human


Hematopoietic Stem Cells Reveals an
Apoptosis-Independent Role for p53 in Self-Renewal
Michael Milyavsky,1,2,7 Olga I. Gan,1,2,7 Magan Trottier,2,5,7 Martin Komosa,5 Ofer Tabach,6 Faiyaz Notta,1,2
Eric Lechman,1,2 Karin G. Hermans,1,2 Kolja Eppert,1,2 Zhanna Konovalova,1,2 Olga Ornatsky,3 Eytan Domany,6
M. Stephen Meyn,2,4,5 and John E. Dick1,2,*
1Department of Stem Cell and Developmental Biology, Campbell Family Cancer Research Institute, Ontario Cancer Institute, Toronto General

Research Institute, University Health Network


2Department of Molecular Genetics
3Institute for Biomaterials and Biomedical Engineering
4Department of Pediatrics

University of Toronto, Toronto, ON M5G 1L7, Canada


5Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
6Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot 76100, Israel
7These authors contributed equally to this work

*Correspondence: jdick@uhnres.utoronto.ca
DOI 10.1016/j.stem.2010.05.016

SUMMARY outcomes: repair of DNA damage and genomic restoration or,


if damaged DNA cannot be sufficiently repaired, execution of
Highly regenerative tissues such as blood must cell death or senescence programs (Vousden and Lane, 2007).
possess effective DNA damage responses (DDR) In highly regenerative tissues like blood, short-lived but highly
that balance long-term regeneration with protection proliferative committed progenitors (colony-forming cells, CFCs)
from leukemogenesis. Hematopoietic stem cells generate large numbers of differentiated cells. In turn, CFCs are
(HSCs) sustain life-long blood production, yet their derived from more primitive multipotential progenitors (MPPs)
with limited self-renewal potential (Majeti et al., 2007). The entire
response to DNA damage remains largely unex-
hematopoietic system is ultimately generated from HSCs that
plored. We report that human HSCs exhibit delayed
have both the greatest longevity and the highest potential for
DNA double-strand break rejoining, persistent self-renewal (Benveniste et al., 2003; Rossi et al., 2008). A single
gH2AX foci, and enhanced p53- and ASPP1-depen- HSC can sustain hematopoiesis for the lifetime of a mouse,
dent apoptosis after g-radiation compared to pro- pointing to its potency (Osawa et al., 1996). Because of geno-
genitors. p53 inactivation or Bcl-2 overexpression toxic stresses, the bone marrow (BM) is likely to harbor progen-
reduced radiation-induced apoptosis and preserved itor cells with acquired mutations (Ji et al., 1995). However, their
in vivo repopulating HSC function. Despite similar short lifespan precludes the accumulation of multiple mutations
protection from irradiation-induced apoptosis, only required for malignant transformation in a single progenitor cell
Bcl-2-overexpressing HSCs showed higher self- clone. By contrast, the long lifespan of HSCs provides time to
renewal capacity, establishing that intact p53 posi- potentially accumulate enough DNA damage-induced mutations
to initiate a leukemogenic process (Dick, 2008). Despite a high
tively regulates self-renewal independently from
rate of hematopoietic regeneration, leukemia is a relatively rare
apoptosis. The reduced self-renewal of HSCs with disease; it is likely that that strong evolutionary pressure exists
inactivated p53 was associated with increased for HSCs to develop distinct DDR strategies that emphasize
spontaneous gH2AX foci in secondary transplants genetic integrity over survival in the face of genotoxicity. Indeed,
of HSCs. Our data reveal distinct physiological roles of all tissues, the hematopoietic system is the most radiosensi-
of p53 that together ensure optimal HSC function: tive, and it is DNA damage to HSCs, not progenitors, that ulti-
apoptosis regulation and prevention of gH2AX foci mately limits the regeneration of hematopoiesis (Dainiak, 2002;
accumulation upon HSC self-renewal. Mettler and Voelz, 2002; Nijnik et al., 2007; Rossi et al., 2007).
Patients with inherited DDR defects and mice that model these
diseases frequently exhibit marked genomic instability, progres-
INTRODUCTION sive BM failure resulting from impairment of HSC function, and
a high incidence of hematopoietic malignancies (Niedernhofer,
Corruption of genetic information by endogenous and exoge- 2008). Collectively, these data argue that DDR is essential for
nous sources of DNA damage threatens the health of an maintaining normal HSC function and for preventing leukemo-
organism. A complex network of DNA damage responses genesis.
(DDR) has evolved to sense and respond to DNA damage. In In humans, where genotoxic stress is widely employed clini-
multicellular organisms, activation of DDR results in two primary cally in chemo- and radiotherapy, BM injury is thought to be

186 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

caused primarily by induction of apoptosis (Gudkov and Komar- obtained in rodents to humans (Rangarajan et al., 2004).
ova, 2003). Both p53 and Bcl-2 proteins are well-characterized Together, little is known about regulators of DNA damage and
regulators of apoptosis. The p53 tumor suppressor is a widely apoptosis within human HSCs and the effects of p53 and Bcl-2
expressed transcription factor that, in addition to apoptosis alterations on long-term HSC function.
induction, plays critical roles in several DDR pathways, including The development of tools to purify and assay human HSCs
activation of cell cycle checkpoints, suppression of homologous (Majeti et al., 2007; McKenzie et al., 2006), combined with the
recombination, and others (Bertrand et al., 2004; Vousden and ability to genetically manipulate HSCs (Mazurier et al., 2004),
Lane, 2007). As with other DDR genes, germline mutations in provides the basis for a genetic approach to gain mechanistic
p53 increase the incidence of hematological malignancy (Nie- insight into the responses of human HSCs to DNA damage.
dernhofer, 2008), and somatic aberrations of p53 and Bcl-2 are Our experiments establish that HSCs delay DSB rejoining,
frequent in human leukemias and lymphomas (Prokocimer and exhibit persistent DDR activation, and are more sensitive to the
Rotter, 1994; Yip and Reed, 2008). Neutralization of Bcl-2 by cytotoxic effects of g-radiation compared to committed progen-
the direct transcriptional target of p53, such as Puma, is respon- itors. Irradiated human HSCs and progenitors undergo apo-
sible for apoptosis in many cell types (Michalak et al., 2008) and ptosis via p53/ASPP1 and Bcl-2. Upon serial transplantation,
overexpression of Bcl-2 alone counteracts many apoptotic HSCs with disabled p53 accumulated DNA damage and
stimuli (Domen et al., 1998; Domen and Weissman, 2003; exhibited decreased self-renewal capacity compared to Bcl-2-
Strasser et al., 1994). The recent identification of additional overexpressing cells, pointing to distinct functions of p53 in
components of the p53 pathway may help explain the mecha- apoptosis and maintenance of self-renewal.
nism that underlies cell-specific variations in sensitivity to p53-
mediated apoptosis (Vousden and Lane, 2007). In vitro experi- RESULTS
ments suggest that p53-interacting proteins, such as
apoptosis-stimulating proteins of p53 (ASPPs) family and others, Delayed Double-Strand Break Rejoining and Sustained
fine tune cellular fate toward apoptosis or survival by altering the DDR Foci in Primitive Hematopoietic Cells
transcriptional activity of p53, although their physiological signif- The human hematopoietic system is hierarchically organized
icance in cells that comprise the hematopoietic hierarchy is where cells at various stages of differentiation can be distin-
unknown (Samuels-Lev et al., 2001). guished by cell surface phenotype: HSCs are highly enriched
The high susceptibility of hematopoietic cells to genotoxic in mature lineage-negative (Lin!) CD34+CD38! fractions, and
stress, such as radiation, prompted numerous studies in murine even more so in the Lin!CD34+CD38!CD90+CD45RA! fraction,
models that established that BM progenitors vary substantially in whereas Lin!CD34+CD38!CD90!CD45RA! population con-
their radiosensitivity and functional recovery after irradiation tains fewer HSCs and many MPPs. More mature progenitor/
(Down et al., 1995; Meng et al., 2003; van Bekkum, 1991; Wage- precursor cells, including CFCs, are Lin!CD34+CD38+ (Majeti
maker, 1995; Wang et al., 2006). However, modern advances in et al., 2007). We designate these fractions as ‘‘HSCs’’,
flow cytometric analysis and functional assays of early hemato- ‘‘MPPs’’, and ‘‘Progenitors’’, respectively.
poietic development limit the impact of these pioneering studies. To investigate the response of these phenotypic subpopula-
In some cases (Down et al., 1995; van Bekkum, 1991; Wage- tions to genotoxic stress, purified fractions of cord blood (CB)
maker, 1995), the precise cell surface phenotype of population cells were irradiated ex vivo. The most biologically significant
subsets was not assessed, whereas in others putative stem DNA lesion induced by ionizing radiation (IR) is a double-strand
cell populations are now known to comprise both progenitors break (DSB). To investigate DSB repair, we used the neutral
and HSCs (Meng et al., 2003; Wang et al., 2006). comet assay to detect DSB rejoining in cells exposed to 15 Gy
It has also been difficult to establish the function of p53 in g-IR. We found that the Progenitor fraction rejoined 19.4% and
HSCs compared to other hematopoietic cells via mouse models. 36.4% of breaks by 30 and 60 min post IR, respectively (Fig-
BM progenitors from p53 knockout as well as Bcl-2 transgenic ure 1A), a rate that is slower than what we and others observe
mice exhibit increased resistance to radiation-induced apo- in human fibroblasts (Figure S1 available online; Stenerlow
ptosis in short-term survival assays (Lotem and Sachs, 1993; et al., 2003). The initial response of the HSC/MPP population
Michalak et al., 2008; Wu et al., 2005). However, the rapid trans- was even more striking: no detectable rejoining was seen in
formation of p53 null BM cells complicates analysis of p53 func- the first 30 min (<1%) and significantly less repair than in the
tion in normal nonmalignant long-term HSCs, also precluding Progenitor population at 1 hr post IR (29.9% versus 36.4%)
examination of self-renewal by retransplantation (Dumble et al., (Figure 1A).
2007; TeKippe et al., 2003). When attempts were made to The delay in initiating DSB rejoining suggested that the DDR of
examine the function of putatively nonmalignant HSCs from the quiescent HSC/MPP population differs from more differenti-
p53-deficient mice, conflicting results were obtained; HSC repo- ated hematopoietic cells and human fibroblasts. To investigate
pulation was either increased, similar, or even decreased (Chen this possibility, we examined another facet of the damage
et al., 2008; Liu et al., 2009; Marusyk et al., 2010). Moreover, with response: the formation of subnuclear foci of phosphorylated
increased p53 dosage, HSCs exhibit reduced competitive repo- H2AX (gH2AX) at and around DSB sites. One hour after 3 Gy
pulation potential (Dumble et al., 2007). IR, no detectable differences in the induction of gH2AX foci
Finally, species-specific differences in irradiation suscepti- were observed between HSC/MPP and Progenitor fractions;
bility (van Bekkum, 1991), oncogene function, cytokine require- both populations displayed approximately 26 induced gH2AX
ments, telomere/telomerase biology, and the inbred nature of foci/cell (Figures 1B and 1C). However, 12 hr after IR, signifi-
mouse strains complicate direct extrapolation of the knowledge cantly more gH2AX foci remained in HSC/MPP compared to

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 187
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 1. IR-Induced Cellular Response in Hematopoietic Fractions In Vitro


Lin-CB cells were sorted into CD34+CD38!/lowCD45RA! (HSC- and MPP-containing fraction) and CD34+CD38+ (Progenitors).
(A) DSB rejoining measured after 15 Gy IR via the neutral comet assay in HSC/MPP (blue bars) and Progenitor (red bars) fractions. Percent of initial olive tail
moment (OTM) was calculated by dividing OTM at each time point by OTM immediately after IR. 150–350 nuclei were scored for each time point (bars represent
means ± SE, *p < 0.005).
(B) Number of gH2AX foci per cell in HSC/MPP and Progenitor subpopulations 1 and 12 hr after damage. 80–120 nuclei were scored for each time point (bars
represent means ± SE, *p < 0.001).
(C) Representative immunofluorescence staining for gH2AX (red) and DNA (blue) in HSC/MPP- and Progenitor-enriched fractions 1 and 3 hr after 3 Gy IR.
(D) Assessment of cell death in sorted primitive HSC, MPP, and Progenitor fractions 18 hr post IR by double staining with Annexin and 7-AAD (n = 3).
(E) Survival of clonogenic progenitors from HSC, MPP, and progenitor fractions calculated by dividing number of colonies scored in the irradiated plate by the
number of colonies from the same fraction counted in the nonirradiated plate (n = 3, bars represent means ± standard deviation, *p < 0.05 between progenitors
and other fractions). See also Figure S1.

Progenitor fraction (7.1 versus 2.7 foci/nucleus, respectively), rately. Both populations showed a similar proportion of apo-
and the number of gH2AX foci/nucleus remained above the unir- ptotic cells (59% and 55%, respectively) that was significantly
radiated baseline in 82% of HSCs/MPPs compared to only 44% higher than irradiated Progenitor cells (35%). With loss of clono-
Progenitor cells (Figures 1B and 1C). Taken together, these data genic potential as a biological readout of cell death, irradiated
indicate that DDR foci persist longer in primitive cells compared HSC and MPP fractions exhibited decreased clonogenic survival
to progenitors. compared to the Progenitor fraction (Figure 1E), correlating
closely with their higher apoptotic rate.
Increased Apoptosis Induction in Primitive To examine the possible effect of the microenvironment on
Hematopoietic Cells IR-induced apoptosis in HSC/MPP and Progenitor fractions,
In some cells, the DDR machinery responds to IR-induced DNA we performed in vivo IR experiments. NOD/SCID mice were
damage by triggering apoptosis (Vousden and Lane, 2007). Both transplanted with Lin! human CB cells to obtain human hemato-
early (Annexin+, 7AAD!) and late (Annexin+, 7AAD+) stages of poietic engraftment. Upon IR (3 Gy) of the engrafted recip-
apoptosis were detected 18 hr after 3 Gy IR for all fractions ients (5–10 weeks posttransplantation), the apoptosis rate
(Figure 1D). HSC and MPP fractions were investigated sepa- of Lin!CD34+CD38! and Lin!CD34+CD38+ human cells was

188 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Table 1. Differential Induction of Apoptosis in Response to In apoptosis (Figure 3B; Figure S2A). These data provide evidence
Vivo Irradiation in Stem and Progenitor Cell Subpopulations that CFCs respond to genotoxic stress via a p53/Bcl-2-depen-
Annexin V + Cells (%) dent apoptotic mechanism, and this mechanism must be
partially responsible for the loss of their clonogenic potential.
Lin!CD34+CD38! Lin!CD34+ CD38+
Fraction Fraction
To determine whether the differences in DDR between HSCs/
(Mean ± SE) (Mean ± SE) MPPs and Progenitor cells could be due to the differential
Nonirradiated 6.9 ± 1.8 7.6 ± 4.2
expression of molecular regulators of p53-dependent apoptosis,
marrow (n = 3) we carried out global gene expression profiling of both cell frac-
tions, with and without IR, and with and without p53 inactivation
Irradiated 33.3 ± 9.7 17.9 ± 6.0
marrow (n = 3) (unpublished data). Because these studies required short-term
culture (4 days), we were not able to employ CD38 in the sorting
Fold increase in 4.8 2.3
apoptosis in
strategy because it becomes dissociated from HSC/MPP func-
response to radiation tion upon culture (Dorrell et al., 2000). As an alternative, we
Human hematopoietic cells were purified from NOD/SCID mice trans-
employed CD90 because it is a useful marker to distinguish
planted with CB cells 5–10 weeks before. The cohort of the recipients a population enriched for HSCs (CD34+CD90+), whereas CD34+
was irradiated with 3 Gy and sacrificed 1.5 hr later. In every experiment CD90! cells are Progenitor enriched, but devoid of HSCs (Danet
BM cells were obtained from 2–3 nonirradiated mice and from 4–6 irradi- et al., 2001). One molecular regulator of p53, expression of which
ated recipients. Lin! human cells were purified and incubated in was much higher in CD34+CD90+ was PPP1R13B mRNA,
cytokine-supplemented medium for the additional 18 hr followed by encoding the apoptosis-stimulating protein of p53 (ASPP1).
CD34, CD38, and Annexin staining for flow cytometry analysis. Mean Independent validation of ASPP1 expression by real-time PCR
percentage ± standard error (SE) of Annexin V+ SYTOX Blue! cells confirmed its higher expression in CD34+CD90+ cells and in
in each fraction is shown. n indicates the number of independent
freshly isolated Lin!CD34+CD38! HSC-enriched cells as com-
experiments.
pared with CD34+CD90! and Lin!CD34+CD38+ Progenitors,
respectively (Figure 3C; Figure S2B). To elucidate the functional
significance of ASPP1 after induction of apoptosis, we knocked
assessed (Table 1). IR-induced apoptosis of human hema-
down ASPP1 protein expression (Figure S2C). After transduction
topoietic cells in the BM microenvironment was 2-fold higher
of Lin-CB cells with shASPP1-encoding lentiviruses and IR,
in Lin!CD34+CD38! cells compared to Lin!CD34+CD38+
the proportion of IR-induced Annexin+ cells was significantly
Progenitors.
reduced only in the CD34+CD90+ primitive fraction and not in
Collectively, our in vitro and in vivo irradiation data indicated
the more mature CD34+CD90! cell population (Figures 3D and
that the HSC/MPP fraction possessed higher susceptibility to
3E), providing evidence that the HSC-specific expression of
IR-induced apoptosis than Progenitor cells.
ASPP1 is functionally important.
These results reveal that in response to IR, the HSC-enriched
Inactivation of p53 or Bcl-2 Overexpression Blocks fraction is more sensitive to p53-dependent apoptosis, whereas
IR-Induced Apoptosis more mature cells undergo lower apoptosis induction and they
The roles of p53 and Bcl-2 in IR-induced apoptosis are not are also less sensitive to p53 inactivation. Furthermore, the
well established in immature human hematopoietic cells. To high ASPP1 expression in HSC-enriched fraction is partially
determine their role, we constructed lentivirus vectors express- responsible for the inherent susceptibility of those cells to
ing either shRNA targeting p53 (shp53) or a dominant-negative IR-induced apoptosis.
form of p53 (GSE56) (Ossovskaya et al., 1996) and the human
Bcl-2 cDNA (Figure 2A). Transduced hematopoietic cells Abrogation of p53/Bcl-2-Mediated Apoptosis Allows
showed considerable downregulation of p53 protein, overex- Myelopoiesis Recovery after IR
pression of inactive p53, or overexpression of Bcl-2, respectively Because our results showed a clear distinction in apoptosis
(Figure 2B). After 3 Gy IR, the expression of several p53 tran- induction between HSCs/MPPs and Progenitor cells as well as
scriptional target genes, p21, PUMA, and Killer/DR5, was abro- only partial rescue of clonogenic activity when apoptosis was in-
gated in Lin-CB cells that expressed either the shp53 or GSE56 hibited by disabling p53 or overexpressing Bcl2, it was important
lentiviruses (Figure 2C), confirming the functional inactivation of to examine the biological consequences on primitive cells after
p53 with both vectors. Transduction of Lin-CB cells with the Bcl- abrogation of IR-induced apoptosis. Lin-CB cells were trans-
2 lentivirus did not affect the induction of p53 target genes estab- duced with control, GSE56, or Bcl-2 lentiviruses, irradiated
lishing the specificity of these genetic tools (Figure 2C). (3 Gy), and grown in cytokine-dependent cultures with equal
To determine the involvement of p53/Bcl-2 in cell death of input cell numbers. Continuous production of immature CD34+
human hematopoietic cells, we measured apoptosis induction cells, as well as functionally defined CFCs in this long-term
in Lin-CB cells expressing shp53, GSE56, or Bcl-2 at 18–24 hr culture assay, serves as a functional indicator for the mainte-
after IR. Inactivation of p53 (p53 knockdown [KD]) or overexpres- nance of primitive cells, including HSCs (Piacibello et al., 2002).
sion of Bcl-2 resulted in a significant decrease in the post-IR The myeloid differentiation potential in control and experi-
apoptotic frequency compared to controls (Figure 3A). Clono- mental cultures beyond 4 weeks of incubation was similar,
genic assays revealed that both genetic modifications increased although in the first 2 weeks, Bcl-2-overexpressing cells
the number of CFC after IR, but this partial functional rescue exhibited EPO-independent differentiation (Figure S3). Overall,
was much less effective than the protection from IR-induced unirradiated cells achieved greater than 1000-fold expansion

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 189
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 2. Inactivation of p53 and Overexpression of Bcl-2 in Lin-CB


(A) Diagrams of the lentiviral vectors used for inactivation of p53 (H1-shp53-GIP and MA-GSE56) and overexpression of Bcl-2. Control vectors are: H1-shRFP for
shRNA interference, MA for GSE56 and Bcl-2 overexpression.
(B) Left: western blot analysis of p53 protein levels in TEX cell line after infection with shp53 and IR with 3 Gy. Right: western blot analysis of p53 and Bcl-2 protein
levels in CB cells infected with either control (MA), GSE56-, or Bcl-2-expressing lentiviruses. Accumulation of inactive p53 protein is visible as a result of GSE56
overexpression.
(C) Analysis of p53 mRNA and p53 target genes 4 days after shp53 lentiviral transduction of Lin-CB cells. Relative expression levels of the indicated genes were
determined by Q-PCR. Profiling of p21, PUMA, and Killer/DR5 by Q-PCR in Lin-CB-expressing shRFP, shp53, MA, GSE56, and Bcl-2 (nonirradiated [NT], white
bars; 3 hr post IR [IR], black bars). Representative experiments are separated by vertical lines. Gene-specific expression levels were normalized to the levels of the
GAPDH housekeeping control for the same cDNA sample.

over the course of 10 weeks, with GSE56- and Bcl-2-infected cells that are essential for the recovery of long-term myeloid
cultures showing slightly higher cell numbers at later time points cell production after IR.
compared to control (Figure 4A). In all groups, nonirradiated cells
exhibited a slight expansion of CD34+ cells over 3 weeks (Fig- Prevention of IR-Induced p53-Mediated Apoptosis
ure 4B). By contrast, irradiated control cells failed to expand, Protects HSCs
whereas both inactivation of p53 and overexpression of Bcl-2 Human HSCs can be assayed by reconstitution of hematopoi-
permitted sustainable cell production. Upon IR, the control esis in vivo utilizing the NOD/SCID repopulation assay (Dick,
group showed a sharp decrease in the number of CD34+ cells 2008). Lin-CB cells were transduced with control, p53KD-, and
and CFCs (Figures 4B and 4C) in agreement with the absence Bcl-2-expressing viruses, sorted for EGFP+ cells, exposed to
of overall cell production. However, IR of cells with inactivated IR or not, and transplanted intrafemorally into NOD/SCID mice
p53 or overexpressed Bcl-2 resulted in a modest initial decrease (Figure 5A). Recipients of nonirradiated cells from all three exper-
in the CD34+ fraction followed by robust recovery up to input imental groups were highly engrafted in the injected bone with
levels (Figure 4B) and steady expansion of CFCs (Figure 4C). human cells (Figure 5B), demonstrating similar multilineage
However, p53 inactivation or Bcl-2 overexpression did not engraftment provided by transduced HSCs (Figure S4). The
completely restore the expansion potential of hematopoietic intrafemoral injection method provides a unique ability to study
cells after IR: as a result, the total and CFC cell production was one of the central functions of HSCs, namely their migration to
about 10-fold lower relative to the nonirradiated group (Figures other hematopoietic territories. Despite the similar level of the
4A–4C). Nevertheless, these long-term in vitro cultures reveal engraftment in the injected bone, engraftment in the noninjected
that blocking IR-induced apoptosis by p53 inactivation or Bcl- bones was significantly higher in recipients of cells overexpress-
2 overexpression effectively protects primitive hematopoietic ing Bcl-2 or p53KD cells (Figure 5B). The improved repopulation

190 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 3. p53, Bcl-2, and ASPP1 Regulates Apoptosis and Survival of Lin-CB Cells after IR
(A) Apoptosis of EGFP+ population was analyzed by annexin staining and flow cytometry 18–24 hr after 3 Gy IR. Results obtained separately for shRFP and MA
(designated Ctrl thereafter), as well as for shp53 and GSE56 (designated p53KD [knockdown]) showed similar trend and therefore were combined for mean calcu-
lations (n = 2–8).
(B) Clonogenic potential (CFC) of sorted Lin-CB Ctrl, p53KD-, or Bcl-2-overexpressing cells (n = 5–11).
(C) Relative expression of ASPP1 in CD34+CD90+ and CD34+CD90! cells isolated after 4 days of ex vivo culture.
(D and E) Lin! CB cells were transduced with the indicated shRNAs, sorted 4 days later into CD34+CD90+ and CD34+CD90! fractions followed by 3 Gy IR.
Apoptosis was analyzed by annexin staining and flow cytometry 18–24 hr post-IR.
Bars represent means ± standard deviation, *p < 0.05 versus Ctrl. See also Figure S2.

capacity of p53KD cells was probably due to a 1.7-fold increase genic progenitors, whereas p53 inactivation prior to IR resulted
in HSC frequency as determined by limiting dilution analysis in 96% and 80% inactivation of HSCs and CFCs, respectively.
(Table S1). Therefore, p53 inactivation conferred radioprotection of HSCs
Irradiated control cells failed to establish grafts in most of the by 5.7-fold, an effect that was much higher than the 1.7-fold
recipients (17 out of 21) with median engraftment levels <0.1% protection of CFCs (Figure 3B; Figure S2A and Table S2).
(Figure 5C). On the contrary, most recipients of irradiated Based on the long-term in vivo and in vitro results, we
p53KD- or Bcl-2-overexpressing cells were engrafted (25 out conclude that p53 inactivation or Bcl-2 overexpression offers
of 27 and 15 out of 21, respectively). Median levels of engraft- significant, but not absolute, radioprotection. We established
ment in these two groups (6.6% and 5.6%, respectively) were that p53/Bcl2-dependent apoptosis regulates human HSCs
significantly higher in comparison with the irradiated control. under homeostatic and genotoxic stress conditions and that
The differentiation of engrafted cells was unaffected by IR in HSCs are more dependent on this pathway than committed
the p53KD and Bcl-2 groups (Figure S4). Importantly, IR did progenitors.
not impair the ability of p53KD- or Bcl-2-overexpressing cells
to migrate (Figure 5C). HSC Self-Renewal Is Regulated by Bcl-2 and p53
Examination of engraftment levels does not provide the means Accumulation of DNA damage in several murine genetic models
to quantify the number of HSCs contributing to repopulation. of DNA repair results in severely diminished HSC function (Nijnik
However, applying Poisson statistics to the engraftment data et al., 2007; Rossi et al., 2007). Our initial observations revealed
enabled us to estimate that 3 Gy of IR reduced the HSC that freshly isolated human HSCs exhibited distinct patterns of
frequency in control and p53KD cells by 154- and 25-fold, DDR as compared to progenitors (Figure 1), so it was important
respectively. Interestingly in the control vector-transduced to determine whether p53 and Bcl-2 modulation would affect
group, 3 Gy of IR inactivated 99.3% of HSC and 90% of clono- HSC genome integrity after repopulation of NOD/SCID mice.

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 191
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 4. Effect of p53 Inactivation or Bcl-2 Overexpression on Long-Term Myelopoiesis In Vitro


Four days after lentiviral transduction, EGFP+ cells were sorted from control (MA), GSE56-, or Bcl-2-overexpressing groups, divided into two groups (NT and IR
with 3 Gy), and placed into long-term cultures (100,000 cells per culture).
(A) Cell expansion.
(B) CD34+ cell number per culture as analyzed by flow cytometry.
(C) CFC dynamics in cultures (n = 3).
See also Figure S3.

After transplantation, HSCs proliferate to initiate hematopoietic of apoptosis. The increase in gH2AX foci seen solely in p53KD
repopulation, and in addition, HSCs must self-renew to ensure cells is probably due to a loss of one or more p53 functions
long-term repopulation. Serial transplantation is an effective that must be independent of apoptosis, because abrogation of
assay to measure the capacity of HSCs to self-renew; however, apoptosis induction by Bcl-2 overexpression did not result in
retransplantation could generate significant replicative or other an elevation in foci numbers.
forms of stress. We purified human Lin! cells from the BM of Our findings that p53 helps to control the accumulation of
highly engrafted mice and quantified gH2AX foci. We found spontaneous DNA damage in human HSCs throughout times
that human Lin! cells from primary recipients of control (MA), of expansion, such as during serial transplantation, raises the
p53KD-, and Bcl-2-overexpressing cells harbored approxi- question of whether HSC self-renewal capacity is affected
mately the same mean number and similar distribution of spon- when p53 is disabled. Equally, the reduced numbers of gH2AX
taneous gH2AX foci as human fibroblasts (Table S3, Figure 6A; foci when Bcl-2 is overexpressed brings up a similar query.
Wilson et al., 2010). However, upon serial transplantation there To determine whether p53 or Bcl-2 modulation affected HSC
was a striking elevation in the number of gH2AX foci per cell self-renewal, we examined human cell engraftment of secondary
only in human Lin! cells with disabled p53 as compared to the recipients.
control or Bcl-2-overexpressing cells that maintain wild-type In nonirradiated HSCs, only Bcl-2 overexpression resulted in
p53 (Table S3). Examination of the distribution of foci showed significantly higher self-renewal ability with median engraftment
that human cells with disabled p53 had a high proportion of cells levels of 0.15%, 0.58%, and 3.6% for control, p53KD, and Bcl-2
exhibiting more than 5 gH2AX foci/nucleus and a complete groups, respectively (Figure 6B). The p53KD group showed
absence of foci-negative cells compared to the other experi- a trend to lower engraftment compared to the Bcl-2 group.
mental groups (Figure 6A). By contrast, the Bcl-2-overexpress- When irradiated cells were serially transplanted, we found
ing cells possessed lower numbers of gH2AX foci/nucleus a striking difference: HSCs with disabled p53 exhibited signifi-
even compared to control. These results indicate that the cantly lower self-renewal capacity in comparison with Bcl-2-
changes in gH2AX foci numbers are uncoupled from the process overexpressing cells. Only HSCs from the Bcl-2-overexpressing

192 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 5. Repopulating Potential of Genetically Modified HSCs after IR


(A) Study design. Lin-CB cells were transduced with Ctrl (shRFP or MA), p53KD (shp53 or GSE56), or Bcl-2 lentiviruses, cultured for 4 days, sorted for EGFP+ cells,
divided into two groups (NT and IR with 3 Gy), and injected intrafemorally into preconditioned immunodeficient mice. Seven to ten weeks later, the secondary
transplants were performed and mice were analyzed again 7–10 weeks later.
(B and C) Human engraftment of the nonirradiated (B) and irradiated (C) Lin-CB cells in the injected femurs (RF) and in the noninjected bones (BM) of primary
recipients. Each symbol stands for engraftment level in one recipient. Horizontal lines represent median for each group, *p < 0.05 in comparison with relevant
Ctrl group (n = 6).
See also Figure S4 and Tables S1 and S2.

group were able to consistently re-establish human engraftment HSC self-renewal capacity. By contrast, Bcl-2-overexpressing
(Figure 6C). This was surprising because p53 inactivation and HSCs with intact p53 were able to sustain HSC self-renewal,
Bcl-2 overexpression similarly protected HSCs from radiation establishing that the negative impact of disabled p53 on HSC
injury resulting in similar levels of engraftment in primary recipi- self-renewal must be due to impairment of apoptosis-independent
ents. Together, these data link the function of these two key p53 function. Taken together, these results indicate that p53 has
molecular regulators to HSC self-renewal under both homeo- discrete functions in human HSCs that are balanced to facilitate
static and stress conditions. genome stability and optimal self-renewal: one is apoptosis
dependent and serves to negatively regulate HSCs, especially
DISCUSSION after IR, whereas the other is apoptosis independent, positively
regulating HSCs especially during serial transplantation.
Our results establish that the DDR of human HSCs differs in The major pathway for DSB repair in quiescent human cells is
multiple ways from more mature hematopoietic populations. nonhomologous end joining (NHEJ), a rapid but error-prone
Primitive HSCs/MPPs exhibit delayed DSB rejoining and persis- method of DSB repair responsible for the majority of genomic
tent DDR foci and undergo higher levels of p53/ASPP1-depen- structural variants and chromosome translocations in human
dent apoptosis compared to Progenitor population in response cancers (Raphael et al., 2008). In mammalian cells, the protein
to IR. p53 inactivation or Bcl-2 overexpression effectively antag- components of NHEJ begin to assemble at DSB sites seconds
onized IR-induced apoptosis and provided profound rescue after IR (Mari et al., 2006), and NHEJ-mediated repair and rejoin-
of HSC repopulation potential in primary transplanted mice. ing of DSBs is detected within 10 min of DSB induction (Gulston
However, in serial transplanted mice, HSCs with disabled p53 et al., 2004). For quiescent human fibroblasts, "50% of DSBs
exhibited persistently high spontaneous gH2AX foci in the are rejoined within 30 min post-IR (Stenerlow et al., 2003).
engrafted cells and this correlated with markedly decreased However, no rejoining of DSBs occurred in the first 30 min

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 193
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

Figure 6. Effect of p53 Inactivation and Bcl-2 Overexpression on DNA Damage Accumulation and HSC Self-Renewal
(A) gH2AX foci distribution in Lin! human hematopoietic cells isolated from primary and secondary recipients of cells transduced with MA, GSE56, and Bcl-2
lentiviruses. Percent of cells with <1, 1–5, and >5 foci per nucleus is depicted.
(B and C) Comparison of human engraftment in the injected femurs between primary and secondary recipients. Nonirradiated (B) and irradiated (C) cells were
retransplanted to assay HSC self-renewal ability. Horizontal lines represent median for each group, *p < 0.05 in comparison with relevant experimental group.

post-IR in quiescent HSC/MPP populations, indicating that the HSC function after DNA damage (Wouters et al., 2007),
distinct DDR of HSCs/MPPs begins immediately post-IR with the HSC/MPP subset still showed increased susceptibility to
the initial ‘‘decision’’ to delay NHEJ repair of DSBs. The resolu- IR-induced apoptosis after in vivo IR. Thus, our studies offer
tion of gH2AX foci was also delayed in the HSC/MPP fraction. a first step in the exploration of extrinsic and intrinsic factors
The persistence of these DDR protein foci could be the result that might dictate the relative apoptotic sensitivity of different
of increased numbers of residual DSBs in the HSC/MPP fraction cells in a tissue.
compared to Progenitor cells. However, at 6 hr, the differences in Our findings that p53 and Bcl-2 are important mediators of
DSB rejoining between the two populations are minor, suggest- IR-induced apoptosis in primitive human hematopoietic cells
ing instead that the persistence of gH2AX foci after rejoining of extends prior observations made in mice (Domen et al., 1998;
their associated DSBs represent another aspect of the altered Lotem and Sachs, 1993; Wu et al., 2005). Interestingly, despite
DDR of HSCs. These data also provide a note of caution regard- effective abrogation of apoptosis by p53KD and Bcl-2 overex-
ing extrapolating DDR results from one cell type to another (e.g., pression measured at early time points, immediate clonogenic
fibroblasts and HSCs) and point to the importance of investi- survival still decreased by 80%, indicating that some CFCs either
gating the DDR in cell types that make up hierarchically orga- died or became senescent at a later time through p53- and Bcl-
nized tissues. 2-independent mechanisms. By contrast, in two independent
Although the detailed mechanisms that account for the assays for primitive cells (long-term in vitro cultures and serial
cell type-specific activation of DDR are still elusive, our results repopulation of NOD/SCID mice), irradiated cells with inactive
identify ASPP1 as one endogenous factor that accounts for the p53 or overexpression of Bcl-2 regenerated myelopoiesis or
high susceptibility of HSCs/MPPs to p53-dependent apoptosis. repopulated mice. These results demonstrate that not only do
ASPP1, a p53-interacting protein stimulating p53 apoptotic HSCs and progenitors differ in apoptosis induction, they also
potential (Samuels-Lev et al., 2001), was expressed at higher differ in the extent to which the p53/Bcl-2 pathway is involved.
levels in HSC/MPP fraction, and upon its silencing, only HSC/ After p53 inactivation and IR, the higher degree of relative HSC
MPP, but not Progenitor cells, were rescued from IR-induced protection compared to more mature CFC (5.7- and 1.7-fold,
apoptosis. Although the BM microenvironment may impact respectively), combined with their ability to repopulate primary

194 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

recipients, argues that HSCs utilize this pathway to a greater oped hematological malignancies (irrespective of IR exposure)
extent than progenitors. during 14–18 weeks of observation. Collectively, these results
Importantly, neither p53 disruption nor Bcl-2 overexpression underscore the differences in tumor suppressor mechanisms
rescued long-term HSC function to the nonirradiated level, sug- between species. Moreover, studies that focus on modulation
gesting that additional DDR alterations may contribute to the of p53 and Bcl-2 in primary human cells may be more directly
increased radiosensitivity of HSCs. Survival after IR exposure relevant for potential therapeutic manipulation to enhance HSC
is the integrated result of multiple p53/Bcl-2-dependent and expansion and transplantation potential without promoting
-independent factors, including the extent and quality of DNA transformation.
repair, the initiation and release of cell cycle checkpoints, and
the threshold for triggering apoptosis. The delays in DSB rejoin- EXPERIMENTAL PROCEDURES
ing and persistent gH2AX foci we observe in human HSCs/
MMPs may be among those p53/Bcl-2-independent DDR alter- Sample Collection and Purification
Lineage depletion and CD34+ enrichment of cord blood (CB) samples were
ations that help determine HSC radiosensitivity. Such alterations
achieved by negative selection with the StemSep system according to the
may be part of an overall HSC strategy that emphasizes accu- manufacturer’s protocol (Stem Cell Technologies, Canada). Sorting experi-
racy of DNA repair over efficiency, with HSCs harboring unre- ments and antibodies are described in detail in Supplemental Experimental
paired damage eliminated by p53-mediated apoptosis. Procedures.
One implication of the distinct effects that p53 and Bcl-2
exhibit in serially transplanted mice, despite providing similar Immunofluorescence and Neutral Comet Assay
protection from apoptosis, is that p53 has both a positive and Cells were labeled with anti-gH2AX (1:3000, Upstate, MA) and secondary
negative roles in the regulation of HSC function under both antibodies.
The number of gH2AX foci as well as ‘‘Olive Tail Moments’’ were estimated
homeostatic and stress conditions. When p53 is disabled, the
as detailed in Supplemental Experimental Procedures.
balance between these roles collectively results in a net negative
effect on HSC self-renewal. As seen in the primary transplanted Viral Constructs and Transduction
mice, p53 inactivation decreases apoptosis sensitivity and Full-length human Bcl-2 cDNA (kindly provided by Dr. L. Penn, Ontario Cancer
increases HSC frequency and migration—each is a positive Institute, Toronto, Canada) and dominant-negative p53 peptide GSE56 cDNA
component to HSC function and self-renewal. However, (Ossovskaya et al., 1996) were subcloned into bidirectional lentiviral construct
disabling p53 comes at a price for the HSC self-renewal. It is MA1.
p53 and ASPP1 shRNAs were designed with the Dharmacon algorithm
possible that increased gH2AX foci numbers in secondary grafts
(http://www.dharmacon.com). 19 nucleotide sense siRNA sequences are
of p53KD cells reflect the engagement of the complex network of detailed in Supplemental Experimental Procedures.
DDR proteins that reduce HSC self-renewal. Therefore, the net Viral particles pseudotyped with VSV-G were generated by transient trans-
consequence of these numerous (and sometimes antagonistic) fection and titrated on HeLa cells as described (Mazurier et al., 2004).
functions of intact p53 is reflected in self-renewal capacity. Thawed Lin! CB cells were incubated with lentivirus, at multiplicity of infec-
The genetic bypass of p53-dependent apoptosis by Bcl-2 over- tion of 50–100, in X-VIVO 10 (BioWhittaker, Waldersville, MD) medium supple-
mented with 1% BSA and SCF (100 ng/ml), FLT3L (100 ng/ml), TPO (15 ng/ml),
expression established that the effect of p53 on genome integ-
G-CSF (10 ng/ml), and IL-6 (10 ng/ml) for 16 hr. The cells were washed and
rity, and thus on HSC self-renewal, was apoptosis independent.
resuspended in the new transduction medium for additional 60–72 hr. At the
Our results may argue that p53 apoptosis-independent end of gene transduction procedure, cells were sorted for EGFP-expressing
mechanisms become activated to prevent damaged HSCs cells, irradiated (if required) on ice with the indicated dose, and used for
from self-renewing. experiments.
Because transplantation itself is a nonphysiological situation
for HSCs, our findings point to a role for the p53/Bcl-2 pathway Ex Vivo Experiments
in transplant-mediated cellular stress. Although the nature of Annexin V Apoptosis Detection Kit (BD PharMingen) and SYTOX Blue Dead
Cell Stain (Invitrogen) were used according to manufacturer’s protocols.
such stress is ill defined, the accumulation of spontaneous
For long-term cultures, Lin-CB cells sorted after transduction were seeded
gH2AX in hematopoietic progenitors provides an important in IMDM, 10% FCS (Sigma) supplemented with FLT3L (50 ng/ml), TPO (20 ng/
novel marker for the study of this type of stress. The opposing ml), SCF (50 ng/ml), and IL-6 (10 ng/ml) at the density of 1 3 105 cells/ml as
roles of p53 inactivation and Bcl-2 overexpression on the rate described in Piacibello et al. (2002). Every week cells were counted, washed,
of gH2AX foci accumulation, which became evident only after and resuspended at the density of 1 3 105 cells/ml in fresh medium and growth
a second round of transplantation, provide for the first time factors. Harvested cells were also used to assay clonogenic cell content in
methylcellulose (Guenechea et al., 2001) and for immunophenotype by flow
insight into dynamics, stability, and mechanisms that control
cytometry.
the human HSC genome upon BM transplantation, albeit in
a xenotransplant setting. NOD/SCID Mouse Repopulation
The findings presented here establish the importance of All animal experimental protocols were approved by the Animal Care
experimentation with primary human cells to extend studies on Committee of the University Health Network (Toronto, Canada). Cells were
p53-null mice. Although resistant to IR-induced apoptosis, transplanted intrafemorally (IF) as described previously (McKenzie et al.,
mouse p53-deficient hematopoietic cells become immortalized 2007) into 8- to 10-week-old NOD/LtSz-Prkdcscid (NOD-SCID) mice which
in vitro (Palacios et al., 1996) and undergo leukemic transforma- were irradiated (2.5 Gy) and injected intraperitoneally with 200 mg of anti-
CD122 antibodies 24 hr before transplantation.
tion beginning at 10 weeks posttransplantation (Marusyk et al.,
Supplemental Experimental Procedures contains cell doses injected,
2010; Yoshida et al., 2002). By contrast, we saw no evidence human engraftment flow cytometry analysis on FACScalibur or LSR II cytom-
of immortalization in long-term cultures and none of the 46 sec- eters (Becton Dickinson) and antibodies used, lineage depletion of human cells
ondary recipients transplanted with p53-inactivated cells devel- from engrafted mice, and secondary transplantation procedure.

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 195
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

For in vivo IR experiments, preconditioned NOD/SCID mice were trans- Bertrand, P., Saintigny, Y., and Lopez, B.S. (2004). p53’s double life: Transac-
planted with 50,000 Lin! CB cells. 5–10 weeks after transplantation, the cohort tivation-independent repression of homologous recombination. Trends Genet.
of the recipients was irradiated with 3 Gy and sacrificed in 1.5 hr. Lineage-posi- 20, 235–243.
tive cell depletion from human hematopoietic engraftment was performed and Chen, J., Ellison, F.M., Keyvanfar, K., Omokaro, S.O., Desierto, M.J., Eckhaus,
cells incubated in IMDM, 10% FCS (Sigma) supplemented with FLT3L (50 ng/ M.A., and Young, N.S. (2008). Enrichment of hematopoietic stem cells with
ml), TPO (20 ng/ml), SCF (50 ng/ml), and IL-6 (10 ng/ml) for additional 18 hr SLAM and LSK markers for the detection of hematopoietic stem cell function
followed by staining of CD34, CD38, Annexin, and CYTOX Blue for flow cytom- in normal and Trp53 null mice. Exp. Hematol. 36, 1236–1243.
etry analysis.
Dainiak, N. (2002). Hematologic consequences of exposure to ionizing radia-
tion. Exp. Hematol. 30, 513–528.
Quantitative RT-PCR and Microarrays
RNA was extracted with the Trizol reagent (Invitrogen) and reverse transcribed Danet, G.H., Lee, H.W., Luongo, J.L., Simon, M.C., and Bonnet, D.A. (2001).
with SuperScript III (Invitrogen). Real-time PCR reactions were prepared with Dissociation between stem cell phenotype and NOD/SCID repopulating
SYBR Green PCR Master Mix (Applied Biosystems) in triplicates and analyzed activity in human peripheral blood CD34(+) cells after ex vivo expansion.
on Applied Biosystems 7900HT instruments. Absolute gene expression was Exp. Hematol. 29, 1465–1473.
quantified with SDS software (Applied Biosystems) based on the standard Dick, J.E. (2008). Stem cell concepts renew cancer research. Blood 112,
curve method and presented after normalization for GAPDH. Supplemental 4793–4807.
Experimental Procedures contain list of primers. RNA from 10,000 or more Domen, J., and Weissman, I.L. (2003). Hematopoietic stem cells and other
sorted cells was used for microarray analysis by Human HT-12 Expression hematopoietic cells show broad resistance to chemotherapeutic agents
Beadchip (Illumina). Data were preprocessed and normalized with the Biocon- in vivo when overexpressing bcl-2. Exp. Hematol. 31, 631–639.
ductor package lumi, which included log2 transformation.
Domen, J., Gandy, K.L., and Weissman, I.L. (1998). Systemic overexpression
of BCL-2 in the hematopoietic system protects transgenic mice from the
Statistical Analysis consequences of lethal irradiation. Blood 91, 2272–2282.
The significance of differences among groups was determined by Student’s
Dorrell, C., Gan, O.I., Pereira, D.S., Hawley, R.G., and Dick, J.E. (2000). Expan-
t test and Mann-Whitney test (SigmaStat software; Jandel).
sion of human cord blood CD34(+)CD38(-) cells in ex vivo culture during retro-
viral transduction without a corresponding increase in SCID repopulating cell
ACCESSION NUMBERS (SRC) frequency: Dissociation of SRC phenotype and function. Blood 95,
102–110.
The microarray data are available in the Gene Expression Omnibus (GEO)
Down, J.D., Boudewijn, A., van Os, R., Thames, H.D., and Ploemacher, R.E.
database (http://www.ncbi.nlm.nih.gov/gds) under the accession number
(1995). Variations in radiation sensitivity and repair among different hematopoi-
GSE21830.
etic stem cell subsets following fractionated irradiation. Blood 86, 122–127.
Dumble, M., Moore, L., Chambers, S.M., Geiger, H., Van Zant, G., Goodell,
SUPPLEMENTAL INFORMATION
M.A., and Donehower, L.A. (2007). The impact of altered p53 dosage on hema-
topoietic stem cell dynamics during aging. Blood 109, 1736–1742.
Supplemental Information includes Supplemental Experimental Procedures,
four figures, and three tables and can be found with this article online at Gudkov, A.V., and Komarova, E.A. (2003). The role of p53 in determining sensi-
doi:10.1016/j.stem.2010.05.016. tivity to radiotherapy. Nat. Rev. Cancer 3, 117–129.
Guenechea, G., Gan, O.I., Dorrell, C., and Dick, J.E. (2001). Distinct classes of
ACKNOWLEDGMENTS human stem cells that differ in proliferative and self-renewal potential.
Nat. Immunol. 2, 75–82.
We thank S. Harding and Dr. R.G. Bristow (Princess Margaret Hospital, Gulston, M., de Lara, C., Jenner, T., Davis, E., and O’Neill, P. (2004). Process-
Toronto, Ontario) for helping with immunofluorescent imaging; P. Scheufler, ing of clustered DNA damage generates additional double-strand breaks in
P. Savage, and the obstetrics unit of Trillium Hospital (Mississauga, Ontario) mammalian cells post-irradiation. Nucleic Acids Res. 32, 1602–1609.
for providing cord blood samples and co-op students for processing cord
Ji, W., Qu, G.Z., Ye, P., Zhang, X.Y., Halabi, S., and Ehrlich, M. (1995). Frequent
blood samples; S. Zhao, L. Jamieson, and P.A. Penttilä (Hospital for Sick Chil-
detection of bcl-2/JH translocations in human blood and organ samples by
dren, Toronto, Ontario) for sorting; and the J.E.D. lab members for critical
a quantitative polymerase chain reaction assay. Cancer Res. 55, 2876–2882.
review of the manuscript. This work was supported by postdoctoral fellow-
ships from the European Molecular Biology Organization (EMBO) and Euro- Liu, Y., Elf, S.E., Miyata, Y., Sashida, G., Liu, Y., Huang, G., Di Giandomenico,
pean Hematology Association and Canadian Institutes for Health Research S., Lee, J.M., Deblasio, A., Menendez, S., et al. (2009). p53 regulates hemato-
(CIHR) (M.M.) and The Netherland Society for Scientific Research (NOW) poietic stem cell quiescence. Cell Stem Cell 4, 37–48.
(K.G.H.) and grants from The Stem Cell Network of National Centers of Excel- Lotem, J., and Sachs, L. (1993). Hematopoietic cells from mice deficient in
lence, Canadian Cancer Society Research Institute, Terry Fox Foundation, wild-type p53 are more resistant to induction of apoptosis by some agents.
Genome Canada through the Ontario Genomics Institute, Ontario Institute Blood 82, 1092–1096.
for Cancer Research, Premier’s Summit Award funded from the province of Majeti, R., Park, C.Y., and Weissman, I.L. (2007). Identification of a hierarchy of
Ontario, Leukemia and Lymphoma Society, and Canadian Institutes for Health multipotent hematopoietic progenitors in human cord blood. Cell Stem Cell 1,
Research, and a Canada Research Chair. This research was funded in part by 635–645.
the Ontario Ministry of Health and Long Term Care (OMOHLTC). The views ex-
Mari, P.O., Florea, B.I., Persengiev, S.P., Verkaik, N.S., Bruggenwirth, H.T.,
pressed do not necessarily reflect those of the OMOHLTC.
Modesti, M., Giglia-Mari, G., Bezstarosti, K., Demmers, J.A., Luider, T.M.,
et al. (2006). Dynamic assembly of end-joining complexes requires interaction
Received: November 18, 2009
between Ku70/80 and XRCC4. Proc. Natl. Acad. Sci. USA 103, 18597–18602.
Revised: March 24, 2010
Accepted: May 14, 2010 Marusyk, A., Porter, C.C., Zaberezhnyy, V., and DeGregori, J. (2010). Irradia-
Published online: July 8, 2010 tion selects for p53-deficient hematopoietic progenitors. PLoS Biol. 8,
e1000324.
REFERENCES Mazurier, F., Gan, O.I., McKenzie, J.L., Doedens, M., and Dick, J.E. (2004).
Lentivector-mediated clonal tracking reveals intrinsic heterogeneity in the
Benveniste, P., Cantin, C., Hyam, D., and Iscove, N.N. (2003). Hematopoietic human hematopoietic stem cell compartment and culture-induced stem cell
stem cells engraft in mice with absolute efficiency. Nat. Immunol. 4, 708–713. impairment. Blood 103, 545–552.

196 Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc.
Cell Stem Cell
DNA Damage Response in Hematopoietic Stem Cells

McKenzie, J.L., Gan, O.I., Doedens, M., Wang, J.C., and Dick, J.E. (2006). Indi- Rossi, D.J., Bryder, D., Seita, J., Nussenzweig, A., Hoeijmakers, J., and Weiss-
vidual stem cells with highly variable proliferation and self-renewal properties man, I.L. (2007). Deficiencies in DNA damage repair limit the function of
comprise the human hematopoietic stem cell compartment. Nat. Immunol. 7, haematopoietic stem cells with age. Nature 447, 725–729.
1225–1233.
Rossi, D.J., Jamieson, C.H., and Weissman, I.L. (2008). Stems cells and the
McKenzie, J.L., Gan, O.I., Doedens, M., and Dick, J.E. (2007). Reversible cell pathways to aging and cancer. Cell 132, 681–696.
surface expression of CD38 on CD34-positive human hematopoietic repopu-
Samuels-Lev, Y., O’Connor, D.J., Bergamaschi, D., Trigiante, G., Hsieh, J.K.,
lating cells. Exp. Hematol. 35, 1429–1436.
Zhong, S., Campargue, I., Naumovski, L., Crook, T., and Lu, X. (2001). ASPP
Meng, A., Wang, Y., Van Zant, G., and Zhou, D. (2003). Ionizing radiation and
proteins specifically stimulate the apoptotic function of p53. Mol. Cell 8,
busulfan induce premature senescence in murine bone marrow hematopoietic
781–794.
cells. Cancer Res. 63, 5414–5419.
Stenerlow, B., Karlsson, K.H., Cooper, B., and Rydberg, B. (2003). Measure-
Mettler, F.A., Jr., and Voelz, G.L. (2002). Major radiation exposure—what to
ment of prompt DNA double-strand breaks in mammalian cells without
expect and how to respond. N. Engl. J. Med. 346, 1554–1561.
including heat-labile sites: results for cells deficient in nonhomologous end
Michalak, E.M., Villunger, A., Adams, J.M., and Strasser, A. (2008). In several
joining. Radiat. Res. 159, 502–510.
cell types tumour suppressor p53 induces apoptosis largely via Puma but
Noxa can contribute. Cell Death Differ. 15, 1019–1029. Strasser, A., Harris, A.W., Jacks, T., and Cory, S. (1994). DNA damage can
induce apoptosis in proliferating lymphoid cells via p53-independent mecha-
Niedernhofer, L.J. (2008). DNA repair is crucial for maintaining hematopoietic
nisms inhibitable by Bcl-2. Cell 79, 329–339.
stem cell function. DNA Repair (Amst.) 7, 523–529.
Nijnik, A., Woodbine, L., Marchetti, C., Dawson, S., Lambe, T., Liu, C., TeKippe, M., Harrison, D.E., and Chen, J. (2003). Expansion of hematopoietic
Rodrigues, N.P., Crockford, T.L., Cabuy, E., Vindigni, A., et al. (2007). DNA stem cell phenotype and activity in Trp53-null mice. Exp. Hematol. 31,
repair is limiting for haematopoietic stem cells during ageing. Nature 447, 521–527.
686–690. van Bekkum, D.W. (1991). Radiation sensitivity of the hemopoietic stem cell.
Osawa, M., Hanada, K., Hamada, H., and Nakauchi, H. (1996). Long-term lym- Radiat. Res. 128, S4–S8.
phohematopoietic reconstitution by a single CD34-low/negative hematopoi-
Vousden, K.H., and Lane, D.P. (2007). p53 in health and disease. Nat. Rev.
etic stem cell. Science 273, 242–245.
Mol. Cell Biol. 8, 275–283.
Ossovskaya, V.S., Mazo, I.A., Chernov, M.V., Chernova, O.B., Strezoska, Z.,
Wagemaker, G. (1995). Heterogeneity of radiation sensitivity of hemopoietic
Kondratov, R., Stark, G.R., Chumakov, P.M., and Gudkov, A.V. (1996). Use
stem cell subsets. Stem Cells 13 (Suppl 1), 257–260.
of genetic suppressor elements to dissect distinct biological effects of sepa-
rate p53 domains. Proc. Natl. Acad. Sci. USA 93, 10309–10314. Wang, Y., Schulte, B.A., LaRue, A.C., Ogawa, M., and Zhou, D. (2006). Total
Palacios, R., Bucana, C., and Xie, X. (1996). Long-term culture of lymphohe- body irradiation selectively induces murine hematopoietic stem cell senes-
matopoietic stem cells. Proc. Natl. Acad. Sci. USA 93, 5247–5252. cence. Blood 107, 358–366.
Piacibello, W., Bruno, S., Sanavio, F., Droetto, S., Gunetti, M., Ailles, L., San- Wilson, P.F., Nham, P.B., Urbin, S.S., Hinz, J.M., Jones, I.M., and Thompson,
toni de Sio, F., Viale, A., Gammaitoni, L., Lombardo, A., et al. (2002). Lentiviral L.H. (2010). Inter-individual variation in DNA double-strand break repair in
gene transfer and ex vivo expansion of human primitive stem cells capable of human fibroblasts before and after exposure to low doses of ionizing radiation.
primary, secondary, and tertiary multilineage repopulation in NOD/SCID Mutat. Res. 683, 91–97.
mice. Nonobese diabetic/severe combined immunodeficient. Blood 100,
Wouters, A., Pauwels, B., Lardon, F., and Vermorken, J.B. (2007). Review:
4391–4400.
Implications of in vitro research on the effect of radiotherapy and chemo-
Prokocimer, M., and Rotter, V. (1994). Structure and function of p53 in normal therapy under hypoxic conditions. Oncologist 12, 690–712.
cells and their aberrations in cancer cells: Projection on the hematologic cell
Wu, W.S., Heinrichs, S., Xu, D., Garrison, S.P., Zambetti, G.P., Adams, J.M.,
lineages. Blood 84, 2391–2411.
and Look, A.T. (2005). Slug antagonizes p53-mediated apoptosis of hemato-
Rangarajan, A., Hong, S.J., Gifford, A., and Weinberg, R.A. (2004). Species-
poietic progenitors by repressing puma. Cell 123, 641–653.
and cell type-specific requirements for cellular transformation. Cancer Cell
6, 171–183. Yip, K.W., and Reed, J.C. (2008). Bcl-2 family proteins and cancer. Oncogene
27, 6398–6406.
Raphael, B.J., Volik, S., Yu, P., Wu, C., Huang, G., Linardopoulou, E.V., Trask,
B.J., Waldman, F., Costello, J., Pienta, K.J., et al. (2008). A sequence-based Yoshida, K., Aizawa, S., Watanabe, K., Hirabayashi, Y., and Inoue, T. (2002).
survey of the complex structural organization of tumor genomes. Genome Stem-cell leukemia: p53 deficiency mediated suppression of leukemic differ-
Biol. 9, R59. entiation in C3H/He myeloid leukemia. Leuk. Res. 26, 1085–1092.

Cell Stem Cell 7, 186–197, August 6, 2010 ª2010 Elsevier Inc. 197

You might also like