13
Article Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and Prevents Malignant Transformation Graphical Abstract Highlights d Aspp1 is predominantly expressed in HSCs and restricts self- renewal capacity d Aspp1 induces apoptosis in damaged HSCs and inhibits persistent DNA damage foci d Aspp1 coordinates with p53 to limit HSC self-renewal and DNA damage accumulation d Double deletion of Aspp1 and p53 in HSCs leads to hematological malignancy in vivo Authors Masayuki Yamashita, Eriko Nitta, Toshio Suda Correspondence [email protected] (E.N.), [email protected] (T.S.) In Brief Suda and colleagues show that the hematopoietic stem cell (HSC) pool integrity is coordinately controlled by p53 and its proapoptotic activator Aspp1. Aspp1 has both p53-dependent and -independent functions in regulating HSC self-renewal and DNA damage tolerance, and concomitant deletion of Aspp1 and p53 in HSCs leads to hematological malignancies. Accession Numbers GSE69032 Yamashita et al., 2015, Cell Stem Cell 17, 23–34 July 2, 2015 ª2015 Elsevier Inc. http://dx.doi.org/10.1016/j.stem.2015.05.013

Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

Article

Aspp1 Preserves Hematop

oietic Stem Cell PoolIntegrity and Prevents Malignant Transformation

Graphical Abstract

Highlights

d Aspp1 is predominantly expressed in HSCs and restricts self-

renewal capacity

d Aspp1 induces apoptosis in damaged HSCs and inhibits

persistent DNA damage foci

d Aspp1 coordinates with p53 to limit HSC self-renewal and

DNA damage accumulation

d Double deletion of Aspp1 and p53 in HSCs leads to

hematological malignancy in vivo

Yamashita et al., 2015, Cell Stem Cell 17, 23–34July 2, 2015 ª2015 Elsevier Inc.http://dx.doi.org/10.1016/j.stem.2015.05.013

Authors

Masayuki Yamashita, Eriko Nitta,

Toshio Suda

[email protected] (E.N.),[email protected] (T.S.)

In Brief

Suda and colleagues show that the

hematopoietic stem cell (HSC) pool

integrity is coordinately controlled by p53

and its proapoptotic activator Aspp1.

Aspp1 has both p53-dependent and

-independent functions in regulating HSC

self-renewal and DNA damage tolerance,

and concomitant deletion of Aspp1 and

p53 in HSCs leads to hematological

malignancies.

Accession Numbers

GSE69032

Page 2: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

Cell Stem Cell

Article

Aspp1 Preserves Hematopoietic Stem Cell PoolIntegrity and Prevents Malignant TransformationMasayuki Yamashita,1 Eriko Nitta,1,2,* and Toshio Suda1,3,*1Department of Cell Differentiation, The Sakaguchi Laboratory of Developmental Biology, School of Medicine, Keio University,

35 Shinano-machi, Shinjuku-ku, Tokyo 160-8582, Japan2Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku,Chiba 260-8670, Japan3Cancer Science Institute, National University of Singapore, 14 Medical Drive MD6, Centre for Translational Medicine, 117599, Singapore

*Correspondence: [email protected] (E.N.), [email protected] (T.S.)

http://dx.doi.org/10.1016/j.stem.2015.05.013

SUMMARY

Quiescent hematopoietic stem cells (HSCs) areprone to mutagenesis, and accumulation of muta-tions can result in hematological malignancies. Themechanisms through which HSCs prevent suchdetrimental accumulation, however, are unclear.Here, we show that Aspp1 coordinates with p53 tomaintain the genomic integrity of the HSC pool.Aspp1 is preferentially expressed in HSCs and re-stricts HSC pool size by attenuating self-renewal un-der steady-state conditions. After genotoxic stress,Aspp1 promotes HSC cycling and induces p53-dependent apoptosis in cells with persistent DNAdamage foci. Beyond these p53-dependent func-tions, Aspp1 attenuates HSC self-renewal and accu-mulation of DNA damage in p53 null HSCs. Conse-quently, concomitant loss of Aspp1 and p53 leadsto the development of hematological malignancies,especially T cell leukemia and lymphoma. Together,these data highlight coordination between Aspp1and p53 in regulating HSC self-renewal and DNAdamage tolerance and suggest that HSCs possessspecific mechanisms that prevent accumulation ofmutations and malignant transformation.

INTRODUCTION

In adult mammals, a vast number of blood cells of all kinds are

derived from hematopoietic stem cells (HSCs), a rare population

in bone marrow (BM). HSCs are the only cells within the blood

system that have both self-renewal and multi-lineage potential,

giving rise to daughter stem cells and committed progenitor

cells, thus achieving lifelong hematopoiesis (Orkin and Zon,

2008). To avoid being lost by various stresses, HSCs are mostly

kept in a quiescent state (Arai et al., 2004; Wilson et al., 2008;

Yoshihara et al., 2007).

Growing evidence has suggested that HSCs have unique

characteristics in their DNA damage response owing to their

quiescence (de Laval et al., 2013; Mohrin et al., 2010). HSCs

tend to repair DNA double strand breaks (DSBs), the severest

form of DNA damage, by error-prone nonhomologous end

joining (NHEJ) rather than homologous recombination. Because

of the error-prone nature of NHEJ, surviving HSCs may retain

mutations (Mohrin et al., 2010). Once a mutation favorable for

cell survival is maintained, the mutation can be passed to

daughter stem cells and pooled in the HSC population and can

be transmitted to a variety of mature hematopoietic cells (Blan-

pain et al., 2011). Thus, HSCs can serve as a reservoir for muta-

tion accumulation, which may eventually lead to hematopoietic

failure or malignant transformation (Rossi et al., 2008). Recent

deep sequencing analyses suggest that clonal expansion of

HSCs with somatic mutation can precede the development of

leukemia and lymphoma, and these ‘‘pre-leukemic HSCs’’ can

survive after chemotherapy, contributing to relapse of the dis-

ease (Corces-Zimmerman et al., 2014; Quivoron et al., 2011;

Shlush et al., 2014).

Given that quiescent HSCs are vulnerable to mutagenesis due

to low-fidelity DNA repair, there should be some compensatory

mechanisms by which mutations are removed from the HSC

pool. One of the most plausible molecular pathways to guard

the HSC genome is through the well-known tumor suppressor

p53. Upon genotoxic stress, p53 can induce cell-cycle arrest,

senescence, or apoptosis to prevent mutation accumulation.

Greater p53 activation is observed in hematopoietic stem and

progenitor cells (HSPCs) than in myeloid progenitor cells (Mohrin

et al., 2010), and p53 also regulates HSC quiescence under

normal conditions (Liu et al., 2009).

The apoptosis-stimulating protein of p53 (Aspp) family pro-

teins Aspp1, Aspp2, and iAspp are co-factors of p53 that specif-

ically regulate its proapoptotic function (Bergamaschi et al.,

2003; Samuels-Lev et al., 2001). Aspp1 and Aspp2 enhance

the proapoptotic function of p53 by activating p53-mediated

transcription of proapoptotic genes, whereas iAspp plays the

opposite role. All three proteins contain ankyrin repeats, an

SH3 domain and a proline-rich region in their C termini, by which

they can associate with several proteins, including p53 (Trigiante

and Lu, 2006). Among the Aspp members, loss of ASPP1

expression due to aberrant methylation of its promoter has

been reported as an independent poor prognosis factor of hu-

man acute lymphoblastic leukemia (ALL) (Agirre et al., 2006).

Previous studies showed that Aspp1 was highly expressed in

murine hematopoietic cells as well as vascular endothelial cells

(Hirashima et al., 2004, 2008). In addition, a recent study identi-

fied Aspp1 as an apoptosis enhancer of human cord blood stem

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 23

Page 3: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

cells (Milyavsky et al., 2010). Together these findings suggest

that Aspp1 could be a potent regulator of hematopoiesis, espe-

cially of HSCs. However, the significance of Aspp1 in adult

hematopoiesis and the causal relationship between Aspp1

deficiency and hematological malignancy have not been

investigated.

Here we identify Aspp1 as a selective regulator of HSCs and

examine its function in protecting the integrity of the HSC pool

and the underlying mechanisms.

RESULTS

HSCs Are Predisposed to Activate p53-DependentProapoptotic Signaling after DNA DamageHSPCsactivate greater levels of p53 comparedwithmyeloidpro-

genitor cells after genotoxic stress (Mohrin et al., 2010). To further

assess the activity of p53 in HSCs, we treated mice with 2 Gy or

4 Gy ionizing radiation (IR), isolated myeloid progenitor-enriched

LKS� (lineage�Sca-1�c-Kit+), HSPC-enrichedLSK (lineage–Sca-

1+c-Kit+), and HSC-enriched long-term HSC (LT-HSC) (CD150+

CD41�CD48�LSK) populations, and evaluated p53 activation

and changes inmRNA levels of several p53 target genes (Figures

A

B

C D

E F G H

I J

Figure 1. Preferential Expression of Aspp1

in the HSC Population and Increased HSC

Pool Size in Aspp1–/– Mice

(A) Relative Aspp1 mRNA levels in purified BM

subpopulations, assessed by qRT-PCR (n = 4).

(B) Expression of LacZ in BM subpopulations of

Aspp1+/LacZ mice, measured by flow cytometry

after being stained with fluorescein di-b-D-gal-

actopyranoside (FDG).

(C) Quantification of FDG fluorescence shown in

(B). The fluorescence intensity was converted into

geometric mean, and the mean values were

normalized to Aspp1+/+ control (n = 3).

(D) Western blotting using lysates of lineage+,

LKS�, and LSK cells.

(E) BM cellularity of adult WT and Aspp1�/� mice

(n = 10).

(F–H) Absolute number of myeloid cells, B cells,

and T cells (F), common lymphoid progenitor

subpopulations (G), and myeloid progenitor sub-

populations (H) in the BM of adult WT and

Aspp1�/� mice (n = 10).

(I) Representative flow cytometry profiles of LSK

cells (left) and CD150+CD41�CD48�LSK cells (LT-

HSCs; middle) in adult WT and Aspp1�/� BM cells.

LT-HSCs are further classified based on CD150

expression levels (right).

(J) Absolute number of LSK and LT-HSC pop-

ulations in the BM of adult WT and Aspp1�/� mice

(n = 10; ��p < 0.01 [versus WT]).

Data shown are mean ± SD. *p < 0.05, **p < 0.01.

See also Figures S1 and S2.

S1A and S1C). LSK cells and LT-HSCs

consistently showed higher responsive-

ness to IR compared with LKS� cells, as

observed by strikingly enhanced phos-

phorylation of p53 and expression of p53

target genes after DNA damage (Figures

S1C and S1D). Of note, when treated with 4 Gy IR, LT-HSCs ex-

hibited stronger upregulation of proapoptotic genes Bax and

Noxa when compared with LKS� cells, whereas upregulation of

survival-contributing p21 and Mdm2 genes was comparable

between the two populations (Figure S1D). These observations

suggest a p53-dependent stem cell-specific machinery that

guides HSCs toward apoptosis rather than survival particularly

after severe DNA damage to protect the HSC pool against unre-

pairable DNA damage accumulation.

Aspp1 Is Highly Expressed in Adult HSCsThe driving force toward p53-dependent apoptosis in HSCs

encouraged us to investigate co-factors of p53, especially the

molecules that facilitate distinct responses between p53-depen-

dent apoptosis and survival. We therefore focused on Aspp1,

which is an apoptosis-specific co-activator of p53 (Trigiante

and Lu, 2006) that is preferentially expressed in the stem cell

population of human cord blood (Milyavsky et al., 2010). Aspp1

mRNA levels were evaluated by qRT-PCR in murine HSC, he-

matopoietic progenitor, and differentiated cell populations iso-

lated from BM of adult mice (Figures 1A, S1A, and S1B).

Aspp1 was highly expressed in LSK cells and LT-HSCs. To

24 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.

Page 4: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

confirm Aspp1 expression in the HSC-enriched population, we

performed flow cytometric analysis using fluorescein di-b-D-gal-

actopyranoside (FDG) (Nolan et al., 1988) and Aspp1-LacZ

reporter (Aspp1+/LacZ) mice, in which part of the Aspp1 gene is

replaced with an IRES-LacZ cassette (Hirashima et al., 2008).

Consistent with the qRT-PCR data, FDG fluorescence was pref-

erentially detected in LSK cells and LT-HSCs (Figures 1B and

1C). Aspp1 protein was specifically detected in LSK cells, but

not in lineage+ or LKS� cells (Figure 1D). Among the Aspp family

of genes, LT-HSCs showed the highest enrichment of Aspp1

(Figure S1E). Evaluation of Aspp family expression by reanalysis

of RNA-seq and mass spectrometry data (Cabezas-Wallscheid

et al., 2014) also revealed the highest expression of Aspp1

among Aspp family members (Figure S1F).

Aspp1 Negatively Regulates the HSC Pool Size In VivoThe preferential expression of Aspp1 in HSCs led us to speculate

that Aspp1 might be responsible for significant regulation of

stem cell maintenance. To assess the physiological effect on he-

matopoiesis by disruption of the Aspp1 gene, we examined pe-

ripheral blood (PB) and BM status of 10- to 12-week-old mice

lacking Aspp1 because of homozygous replacement by the

LacZ gene (hereafter referred to as Aspp1�/� mice). Aspp1�/�

mice grew up healthy without distinctive abnormality from WT

mice in aC57BL/6 background, although the number of offspring

was significantly reduced for an unknown reason (Table S1).

However, complete blood count tests revealed a significant in-

crease in the number of white blood cells (WBCs) in Aspp1�/�

mice compared with WT mice (Table S2), accompanied with

increased numbers of myeloid cells (Gr1+Mac1+), B cells

(B220+CD19+), and T cells (CD3+) (Figure S2A). The mean

corpuscular hemoglobin (MCH) and platelet counts of Aspp1�/�

mice were also slightly elevated (Table S2). The number of BM

mononuclear cells (MNCs) was slightly increased in Aspp1�/�

mice (Figure 1E). Flow cytometric analysis revealed that the in-

crease in BM cellularity was derived from an increase in myeloid

cell population (Figure 1F). In addition, whereas the number

of lymphoid cells and common lymphoid progenitor cells

(CLPs; IL-7Ra+Flt3+lineage�Sca-1lowc-Kitlow) was comparable

between WT and Aspp1�/� mice (Figures 1F and 1G), the gran-

ulocyte-macrophage progenitor cell (GMP; CD34+FcgRhigh

lineage�Sca-1�c-Kit+) population was significantly increased in

Aspp1�/� mice BM (Figures 1H and S2B). Most significantly,

we observed an approximate 1.5- and 2-fold increase in the ab-

solute number of LSK cells and LT-HSCs in Aspp1�/� mice,

respectively (Figures 1I, 1J, and S2C). Of note, the increased

HSC pool size was accompanied with increased numbers of

CD150high LT-HSCs (Figure 1J), which are myeloid biased and

have a more robust self-renewal potential (Beerman et al.,

2010; Morita et al., 2010). Taken together, these data indicate

that Aspp1 negatively controls HSC pool size.

Sustained Self-Renewal Capacity of Aspp1–/– HSCs inSerial BM TransplantationThe increase in HSC population of Aspp1�/� mice can theoreti-

cally be caused by changes in cell-intrinsic factors (e.g., self-

renewal, differentiation, cell-cycle regulation, and apoptosis)

and/or cell-extrinsic factors (such as niche factors). To evaluate

long-term self-renewal capacity of bona fide HSCs, we per-

formed serial competitive BM transplantation using Aspp1-defi-

cient HSCs.Aspp1�/� LSK cells displayedmoderately enhanced

reconstitution capacity in the first round of transplantation, as

measured by the percentage of CD45.2+ cells monthly in PB

MNCs (Figure S3A) and 4 months after transplantation in BM

MNCs, LSK cells, and CD150+ LSK cells (Figure S3B). Further-

more, the reconstitution capacity of Aspp1�/� LSK cells was

significantly sustained, while WT LSK cells displayed diminished

reconstitution capacity in the second round of transplantation

(Figures S3C and S3D). Serial competitive transplantation further

clarified that the reconstitution capacity of Aspp1�/� LT-HSCs

was significantly sustained especially after tertiary transplanta-

tion (Figure 2A), suggesting sustained self-renewal capacity in

Aspp1-deficient HSCs. These results indicate that Aspp1 nega-

tively regulates the self-renewal capacity of HSCs in a cell-

intrinsic manner, especially under cumulative stress after serial

BM transplantation.

Aspp1 Negatively Regulates HSC Quiescence uponExposure to StressesNext we analyzed the mechanisms through which Aspp1 pro-

motes the decreased self-renewal capacity of HSCs in serial

BM transplantation. We examined the gene expression profile

of WT and Aspp1�/� LSK cells before and after BM transplanta-

tion using a multiplex qRT-PCR system. Consistent with the

sustained self-renewal capacity of Aspp1�/� LSK cells, gene

expression analysis revealed that multiple genes important for

the maintenance of HSCs were significantly upregulated in

Aspp1�/� LSK cells (Figure 2B). Among them, several quies-

cence-related genes including Tek, Ndn, and Cdkn1c were

significantly upregulated in Aspp1�/� LT-HSCs isolated after

transplantation (Figure S3E).

Quiescence is an important mechanism for the resistance to

stress stimuli and therefore for the maintenance of self-renewal

capacity of HSCs (Arai et al., 2004). Thus, we assessed the

cell-cycle status of Aspp1�/� HSCs. BrdU uptake was signifi-

cantly decreased in Aspp1�/� LT-HSCs in vivo (Figure 2C).

Furthermore, when we analyzed the quiescence of donor-

derived LSK cells in primary and secondary recipients using

the Ki67 proliferation marker and Hoechst33342 to stain DNA

content, we found that serial transplantation caused a progres-

sive decrease in Ki67-negative cells in control donor-derived

LSK cells, whereas Aspp1�/� LSK cells retained more Ki67-

negative cells (Figure 2D). These results indicate that Aspp1�/�

HSPCs were more quiescent and resistant to the loss of

quiescence after serial transplantation.

To further investigate the resistance to stress stimuli, we

administered a single dose of 5-FU, a chemotherapeutic agent

that selectively kills proliferating hematopoietic cells (Lerner

and Harrison, 1990; Van Zant, 1984), into Aspp1�/� mice and

control mice and monitored the recovery of PB and BM status.

Aspp1�/� mice displayed an enhanced recovery of WBCs, he-

moglobin, and platelets (Figure S3F). Consistent with this, we

observed rapid recovery in the number of BM CD150+

CD41�CD48�lineage�Sca-1+ cells prior to the recovery of PB

in Aspp1�/� mice (Figure S3G). Furthermore, Aspp1�/� LT-

HSCs remained quiescent upon 5-FU treatment, whereas the

majority of WT LT-HSCs entered the cell cycle (Figure S3H). In

contrast, no significant changes were observed in the apoptosis

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 25

Page 5: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

ratio in Aspp1�/� WBCs or LT-HSCs (Figure S3I). These findings

support the hypothesis that Aspp1�/� HSCs may remain quies-

cent upon exposure to stress stimuli and are more resistant to

myelotoxic agents.

Aspp1Promotes Apoptosis andControls Persistent DNADamage in HSCsAspp1 has been shown to induce apoptosis in response to DNA

damage in cancer cell lines and human cord blood stem cells

(Milyavsky et al., 2010; Samuels-Lev et al., 2001). Therefore,

we evaluated the apoptosis of donor-derived LSK cells induced

by primary and secondary transplantation using the apoptosis

marker Annexin V. The percentage of Annexin V-positive cells

in donor-derived LSK cells was comparable between both

groups of primary recipients, but a marked reduction was

observed in donor-derived LSK cells in secondary recipients

that received Aspp1�/� LSK cells as donors (Figures 3A and

3B). Similar to the findings under the transplantation stresses,

the percentages of early apoptotic cells were significantly

decreased in Aspp1�/� LT-HSCs and LSK cells upon 4 Gy IR

in vivo (Figures 3C, S4A, and S4B). LKS� cells, which contain

myeloid progenitor cells but no HSCs, showed no obvious

change in the apoptotic ratio between WT and Aspp1�/� cells

(Figure S4A). Importantly, there was no significant decrease of

apoptosis in Aspp1�/� LT-HSCs under steady state (Figure 3C).

Immunofluorescence revealed that p53 was normally stabilized

in Aspp1�/� LSK cells and LT-HSCs upon IR (Figure S4C).

Nevertheless, expression profiling of apoptosis-related genes

revealed that the expressions of multiple proapoptotic genes,

whether direct transcriptional targets of p53 or not, were signif-

icantly decreased in Aspp1�/� LSK cells upon IR (Figures 3D

and S4D). Likewise, Aspp1�/� LT-HSCs showed a marked

reduction in the expression levels of proapoptotic genes (Figures

3E and S4E), corresponding with the decreased apoptosis in

Aspp1�/� HSC populations. These results clearly demonstrate

that Aspp1 is an important intrinsic regulator of HSC apoptosis

especially under genotoxic conditions.

Apoptosis is an important mechanism for eliminating severely

damaged cells. To elucidate the relationship between Aspp1

deficiency and DNA damage, LSK cells from irradiated WT and

Aspp1�/� mice were cultured in serum-free medium and immu-

nostained for 53BP1, which forms subnuclear foci at the site of

DSBs and participates mainly in NHEJ-based DSB repair (Moh-

rin et al., 2010; Schultz et al., 2000). Immediately after LSK cells

were isolated from irradiated mice, comparable numbers of

53BP1 foci formed in both WT and Aspp1�/� LSK cells (Figures

3F and 3G). However, a larger portion (�12.2%) of the Aspp1�/�

LSK cells still survived with considerable (>10) 53BP1 foci even

at 36 hr post-irradiation, whereas the majority of the cells have

only a few foci at that time (Figure 3G). These findings support

our hypothesis that Aspp1may play a significant role in removing

severely damaged HSCs and that Aspp1�/� HSCs cannot be

eliminated when suffering from unrepairable DNA damage

because of defective apoptosis.

A

B

C D

Figure 2. Sustained Self-Renewal Capacity

of Aspp1–/– HSCs

(A) Serial competitive BM transplantation usingWT

and Aspp1�/� LT-HSCs. Two hundred LT-HSCs

isolated from adult WT and Aspp1�/� mice

(CD45.2) were transplanted into lethally irradiated

recipient mice (CD45.1) with 2 3 105 competitor

BM MNCs (CD45.1). Four months later, 2 3 106

BM MNCs of primary recipients were re-trans-

planted into lethally irradiated recipient mice.

Tertiary transplantation was performed in the

same way as secondary transplantation. The

percentage of donor-derived cells in PB was

measured monthly (n = 5–6/group).

(B) Expression levels of HSC-related genes in WT

andAspp1�/� LSK cells before (pre) and 16 hr after

(post) transplantation were measured using the

multiplex qRT-PCR array (50 LSK cells/sample,

n = 16–30).

(C) In vivo BrdU short labeling analysis ofAspp1�/�

mice. The percentages of BrdU+ LT-HSCs are

shown (n = 10).

(D) Flow cytometric analysis of cell cycle of donor-

derivedWT and Aspp1�/� LSK cells 4months after

primary (1st) or secondary (2nd) BM trans-

plantation. Representative flow cytometry profiles

(left) and the average percentages ± SD of G0, G1,

and S/G2/M fractions in donor-derived LSK cells

(right) are shown (n = 6).

The horizontal lines represent the mean in (A), (B),

and (C). Data shown are mean ± SD in (D). *p <

0.05, **p < 0.01. See also Figure S3.

26 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.

Page 6: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

Aspp1 Coordinates with p53 to Regulate Self-Renewaland Persistent DNA Damage of HSCsAspp1 is a p53-binding protein that activates p53-dependent

apoptosis (Samuels-Lev et al., 2001). However, several recent

studies have demonstrated multiple p53-independent roles for

Aspp1 (Hirashima et al., 2008; Wang et al., 2013). To clarify the

relationship between Aspp1 and p53 in the regulation of HSCs

in vivo, we evaluated HSCs doubly deficient for Aspp1 and p53

(Aspp1�/�p53�/�). Because Aspp1�/�p53�/� offspring were

difficult to obtain from conventional mating (Table S3), we bred

Aspp1�/� mice with both conditional p53flox/flox mice (Marino

et al., 2000) andMx1-Cremice (Kuhn et al., 1995). Cre recombi-

nase activity was induced by the administration of polyinosinic-

polycytidylic acid (pIpC) to obtain Aspp1�/�p53�/�BM cells. BM

cellularity was slightly elevated in Aspp1�/�p53�/� mice (Fig-

ure 4A). Furthermore, flow cytometric analysis revealed that

the numbers of LSK cells and LT-HSCs were most significantly

A B C

D

E

F

G

Figure 3. Impaired Apoptosis and Persistent

DNA Damage Foci in Aspp1–/– HSCs

(A) Flow cytometric analysis of apoptosis of donor-

derivedWT and Aspp1�/� LSK cells 4 months after

primary (1st) or secondary (2nd) BM trans-

plantation. Representative flow cytometry profiles

are shown.

(B) The average percentages ± SD of Annexin V+

fraction in donor-derived LSK cells (n = 6).

(C) Apoptosis analysis of WT and Aspp1�/� LT-

HSC cells before and after exposure to in vivo

ionizing radiation (IR). WT and Aspp1�/� mice

treated with 4 Gy irradiation were analyzed at 16 hr

post-irradiation. The average percentages ± SD

of Annexin V+ DAPI� fraction in LT-HSCs are

shown (n = 6).

(D) Representative heat map of the gene expres-

sion levels collected bymultiplex qRT-PCR array in

LSK cells from WT and Aspp1�/� mice 1 hr after

4 Gy irradiation. LSK cells from nonirradiated mice

were used as controls.

(E) Expression levels of proapoptotic genes in

nonirradiated and irradiated LT-HSCs derived from

WT and Aspp1�/� mice were analyzed by qRT-

PCR (n = 4). Irradiated LT-HSCswere isolated 16 hr

after 4 Gy irradiation in vivo.

(F) Immunofluorescence of irradiated WT and

Aspp1�/� LSK cells using antibodies against

53BP1. Representative pictures of 53BP1 staining

of LSK cells collected immediately (0 h) or 36 hr

after irradiation are shown.

(G) Histograms displaying the number of 53BP1

foci per cell in LSK cells after 4 Gy irradiation.

A total of 90–100 nuclei were scored and the per-

centages of nuclei expressing more than 10 foci

were calculated for each time point.

Data shown are mean ± SD. *p < 0.05, **p < 0.01.

See also Figure S4.

increased in Aspp1�/�p53�/� mice

compared with other congenic controls

(Figures 4B and 4C). Notably, the number

of LT-HSCs, especially CD150high LT-

HSCs, was significantly increased in

Aspp1�/�p53�/� mice compared with

WT, as well as in p53�/� single mutant mice (Figures 4C

and S5A).

We next performed single-cell culture using Aspp1�/�p53�/�

HSCs ex vivo to evaluate proliferation and apoptosis (Ema

et al., 2006). Single-cell culture revealed that proliferation capac-

ity was not significantly changed in Aspp1�/�p53�/� LT-HSCs

(Figures S5C and S5D). Importantly, Aspp1�/�p53�/� LT-HSCs

did not show a significant decrease in the ratio of negative wells,

which are derived from negative sorting or apoptosis (Fig-

ure S5C), indicating that apoptosis may not be a main cause

for the increased number of LT-HSCs. To evaluate self-renewal

capacity of Aspp1�/�p53�/� HSCs, we performed serial

competitive transplantation using Aspp1�/�p53�/� LSK cells.

Despite comparable proportions of LT-HSCs in the LSK

population in Aspp1�/�, p53�/�, and Aspp1�/�p53�/� mice (Fig-

ure S5B), Aspp1�/�p53�/� LSK cells showed enhanced engraft-

ment potential compared with Aspp1�/� or p53�/� single mutant

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 27

Page 7: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

LSK cells, especially after secondary transplantation (Figure 4D).

These results were consistent with the results in LSK cells

with p53, suggesting that Aspp1 has a p53-independent role in

the regulation of HSC self-renewal. Furthermore, competitive

reconstitution analysis using LT-HSCs with limiting dilution

revealed that Aspp1�/�p53�/� LT-HSCs have enhanced HSC

activity compared with single mutant LT-HSCs, and the long-

term competitive repopulation unit in Aspp1�/�p53�/� BM was

3.4-fold higher compared with 1.6-fold higher in Aspp1�/� and

p53�/� BM (Figure 4E). Thus, these results indicate that Aspp1

negatively regulates the self-renewal of HSCs that lack p53

and acts coordinately with p53 as a limiter for HSC self-renewal

upon stress stimuli.

We then addressed the question of whether Aspp1 deficiency

facilitates the survival of damaged HSCs in a p53-deficient

background by analyzing gH2AX foci as a marker of DSB sites

(Rogakou et al., 1998). Consistent with the 53BP1 staining,

Aspp1 deficiency also caused a significant increase in the

number of LSK cells with numerous (>10) gH2AX foci at 36 hr

A B

C

D

E

Figure 4. Aspp1 and p53 Coordinately

Regulate HSC Pool Size and Self-Renewal

(A) BM cellularity of adult WT, Aspp1�/�, p53�/�,and Aspp1�/�p53�/� mice (n = 3).

(B) Representative flow cytometry profiles of LSK

cells and LT-HSCs in adult WT, Aspp1�/�, p53�/�,and Aspp1�/�p53�/�BM cells. Data shown are the

mean fractions of LSK cells and LT-HSCs in BM

MNCs.

(C) Absolute numbers of LSK and LT-HSC pop-

ulations in the BM of adult WT, Aspp1�/�, p53�/�,and Aspp1�/�p53�/� mice (n = 3; �p < 0.05, ��p <

0.01 [versus WT]; #p < 0.05 [versus p53�/�]).(D) Serial competitive BM transplantation using

Aspp1�/�, p53�/�, and Aspp1�/�p53�/� LSK cells.

The percentages of donor-derived cells in PB

measured monthly after primary and secondary

transplantation are shown (n = 4–6/group).

(E) Limiting dilution analysis using LT-HSCs. Three

different doses of LT-HSCs (WT and Aspp1�/�:4, 10, and 30; p53�/� and Aspp1�/�p53�/�: 2, 5,and 15; n = 9–10/group) were transplanted into

lethally irradiated recipient mice together with

2 3 105 competitor BM MNCs. Recipients were

considered reconstituted if the percentage of

donor-derived cells in PB showed no less than 1%

at 4months post-transplantation. The frequency of

non-reconstituted mice was plotted against the

dose of transplanted LT-HSCs (left). The frequency

of functional HSCs was calculated using ELDA

software at http://bioinf.wehi.edu.au/software/

elda (middle), and the numbers of functional

HSCs per mouse were calculated using the fre-

quency (right).

The horizontal lines represent the mean in (D). Data

shown are mean ± SD in (A), (C), and (E). *p < 0.05,

**p < 0.01. See also Figure S5.

post-IR (Figures 5A, S6A, and S6B).

Notably, loss of p53 also significantly

increased the number of damaged LSK

cells, and double deletion of Aspp1 and

p53 further enhanced the survival of

severely damaged LSK cells (Figures 5A, S6A, and S6B).

Furthermore, the tolerance for DNA damage in the mutant cells

was still observed in LSK cells and LT-HSCs isolated 4 weeks af-

ter in vivo IR (Figures 5B and 5C). Consistent with the results

of ex vivo analysis, the percentages of severely damaged cells

in LSK and LT-HSC populations were significantly increased

in Aspp1�/� and p53�/� mice and were further increased in

Aspp1�/�p53�/� mice (Figure 5D), indicating that Aspp1 still

contributes to the removal of severely damaged HSCs in vivo

in the absence of p53. Taken together, these results highlight

the coordinated regulation of DNA damage accumulation in the

HSC pool by Aspp1 and p53.

Aspp1 and p53 Double-Deficient HSCs DevelopHematological MalignancyThe coordinated regulation of self-renewal and persistent DNA

damage prompted us to investigate tumorigenicity. To focus on

the development of hematological disorders, we followed up

overall survival and tumor development after competitive BM

28 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.

Page 8: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

transplantation. Aspp1-deficient LSK cells did not cause a

significant increase in susceptibility to malignant lymphoma or

leukemia development (Figure 6A). However, within 6 months

after transplantation, whereas 46% of the recipients receiving

p53�/� LSK cells died of thymic lymphoma, recipient mice

transplanted with Aspp1�/�p53�/� LSK cells showed 100%

lethality (Figure 6A). These mice succumbed to an acute illness,

presenting rapid weight loss, lethargy, and dyspnea. The ma-

jority of the diseased mice exhibited thymic enlargement

(Figure S7C) and some displayed splenomegaly (Figure S7D).

Histological analysis revealed massive growths of lympho-

blast-like cells in PB, BM, and thymus (Figure 6B). Flow

cytometric analysis confirmed that the tumor cells were all

donor-derived cells, and in most cases (88%), they expressed

both CD4 and CD8 or CD8 alone (Figure 6C), revealing that

the illness was akin to T cell lymphoblastic lymphoma/

leukemia. We also observed B cell lymphoblastic leukemia

(6%) and myeloproliferative neoplasia (6%) (Figure 6D and

Table S4), indicating that the hematological disorders caused

by Aspp1�/�p53�/� LSK cells were heterogeneous. These find-

ings clearly demonstrate that double deletion of Aspp1 and p53

in HSCs eventually results in leukemia and lymphoma, and

A

B

C

D

Figure 5. Coordinated Regulation of

Persistent DNA Damage Foci in HSCs by

Aspp1 and p53

(A) Immunofluorescence of irradiated WT,

Aspp1�/�, p53�/�, and Aspp1�/�p53�/� LSK cells

using antibodies against gH2AX. The percentages

of nuclei expressing more than 10 foci immediately

(0 h) or 36 hr after 4 Gy irradiation are shown (n = 3).

(B) Representative pictures of gH2AX immunoflu-

orescence of LSK cells and LT-HSCs isolated from

WT, Aspp1�/�, p53�/�, and Aspp1�/�p53�/� mice

4 weeks after 4 Gy irradiation.

(C) Histograms displaying the number of gH2AX

foci per cell in LSK cells and LT-HSCs of each

genotype 4 weeks after 4 Gy irradiation. Approxi-

mately 102–109 nuclei were scored and the

percentages of nuclei with more than 10 foci are

displayed.

(D) The percentages of nuclei expressing more

than 10 foci 4 weeks after 4 Gy irradiation (n = 3).

Data shown are mean ± SD. *p < 0.05, **p < 0.01.

See also Figure S6.

Aspp1 protects HSCs from malignant

transformation in conjunction with p53.

To delineate the mechanism by which

Aspp1 coordinately regulates HSCs with

p53, we conducted microarray analysis

using Aspp1�/�p53�/� LSK cells. Gene

ontology (GO) analysis revealed sig-

nificant enrichment of GO terms mainly

in downregulated genes in mutant

mice (Figure 7A). GO terms associated

with phosphorylation, inflammation, and

apoptosis were highly enriched in down-

regulated genes in Aspp1�/�p53�/� LSK

cells compared with p53�/� LSK cells

(Figure 7B). We speculated that the

downregulated genes with significant enrichment of GO terms

could offer functional signatures in each comparison, and we

extracted 38 gene sets as a common signature of Aspp1 defi-

ciency by analyzing two sets of the genes that could be downre-

gulated by Aspp1 deficiency in the presence or absence of p53

(Figure 7A and Table S5). Gene set enrichment analysis (GSEA)

revealed that 9 out of the 38 gene sets were downregulated in

all three mutant LSK cells compared with WT LSK cells and

also in Aspp1�/�p53�/� LSK cells compared with p53�/� LSK

cells (Table S5). These gene sets, which are likely candidate

targets of coordinated regulation by Aspp1 and p53, include

genes associatedwith HSCs and angioimmunoblastic T cell lym-

phoma signature (Figure 7C), suggesting that Aspp1 functions

with p53 to suppress transcriptional programs associated with

HSC activity and T cell malignancy. Furthermore, corresponding

with the GO analysis, our screening also identified significant

downregulation of several immune response signatures in

Aspp1�/�p53�/� LSK cells (Figure 7C), suggesting a possible

involvement of the immune response in the p53-independent

regulation by Aspp1. Collectively, these data highlight p53-

independent pathways through which Aspp1 regulates HSC

self-renewal and tumorigenicity in coordination with p53.

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 29

Page 9: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

DISCUSSION

Accumulation of DNA damage in HSCs is correlated with aging,

hematopoietic failure, and development of hematological malig-

nancies (Beerman et al., 2014; Blanpain et al., 2011; Garaycoe-

chea et al., 2012). Thus, the management of DNA damage in

HSCs is a matter of critical importance for lifelong hematopoie-

sis. Here, we revealed that Aspp1, which is preferentially ex-

pressed in the HSC population, regulates both the size and the

soundness of the HSC pool by restricting HSC self-renewal

and quiescence in steady state, along with activating p53-

dependent apoptosis of HSCs after cellular stress. Our results

also revealed a p53-independent function of Aspp1 in HSC

self-renewal and DNA damage tolerance. Our findings under-

score the importance of the tumor suppressor network at the

stem cell level for prevention of tumorigenesis, as HSCs deficient

for both Aspp1 and p53massively accumulate DNA damage and

subsequently develop hematological malignancies.

Our findings provide amolecular basis for the preferential acti-

vation of p53-mediated apoptosis in HSCs. Although HSCs are

deemed apoptosis resistant and survive after BM injury (Mohrin

et al., 2010), HSPCs react more strongly than myeloid progenitor

cells against irradiation as shown by increased p53 phosphory-

lation and upregulation of p53 target genes ex vivo (Mohrin

et al., 2010). In addition, our results further confirmed that

LT-HSCs show even stronger activation of p53 and preferential

induction of its proapoptotic targets after exposure to severe

DNA damage in vivo. These observations indicate that p53 activ-

ity is highly potentiated and thus DNA damage is rigorously

controlled through p53 in the HSC pool, once HSCs are exposed

to cellular stress. Our results demonstrate that HSCs deficient

for Aspp1 show impaired induction of apoptosis and proapopto-

tic genes after irradiation, suggesting that Aspp1 contributes to

the potent upregulation of p53-dependent proapoptotic genes

in the HSC population after DNA damage.

A B

C

D

Figure 6. Aspp1 Coordinates with p53 to

Prevent Malignant Transformation

(A) Kaplan-Meier survival curves of mice trans-

planted withWT (n = 16), Aspp1�/� (n = 17), p53�/�

(n = 20), and Aspp1�/�p53�/� (n = 32) LSK cells.

(B) Representative histology of PB, BM, and

thymus in mice transplanted with WT and

Aspp1�/�p53�/� LSK cells.

(C) Representative flow cytometry profiles for CD4

and CD8, or CD45.1 and CD45.2, in cells from

thymus of the recipient mice transplanted with WT

or Aspp1�/�p53�/� LSK cells.

(D) Frequency of each tumor type observed in

the recipients of Aspp1�/�p53�/� LSK cells (left,

n = 16) and frequency of each immunophenotype

of thymic tumor cells observed in the recipients of

Aspp1�/�p53�/� LSK cells (right, n = 11). *p < 0.05,

**p < 0.01. See also Figure S7.

Our study highlights a differential role of

Aspp1 in fate decision of HSCs under

basal and genotoxic conditions. We

showed that Aspp1 restricts the size of

the HSC pool both phenotypically and

functionally. This is likely attributed to limited self-renewal of

HSCs, as Aspp1 does not have a major impact on their prolifer-

ation capacity and apoptosis rate during ex vivo culture,

although we cannot exclude the possibility that some changes

observed might be still attributable to potentially reduced fre-

quency of apoptotic events in the HSC compartment that is

under the detection limit. Furthermore, Aspp1 facilitates exit

from quiescence, which itself causes DNA damage that in turn

induces differentiation and attrition of HSCs (Walter et al.,

2015; Wang et al., 2012). Thus, Aspp1 regulates pool size and

self-renewal of HSCs through a cell-cycle-induced DNA damage

response. Our data also suggest that Aspp1 induces apoptosis

in the HSPC population, particularly after serial transplantation,

indicating that Aspp1 potentially regulates cell death of HSCs

after replication stress, which is a potent driver of functional

decline in aging HSCs (Flach et al., 2014). Furthermore, Aspp1

contributes to the removal of severely damaged cells from the

HSC pool by the induction of apoptosis after severe DNA dam-

age. This is reminiscent of the recent study of iAspp in HSCs

and leukemia, which revealed that overexpression of iAspp

results in blockage of apoptosis and an increase in the propor-

tion of damaged HSPCs after irradiation (Jia et al., 2014). Thus,

the balance among Aspp family members may be critical to

the fine-tuned control of DNA damage accumulation in the

HSC pool.

Our study also uncovered a p53-independent role of Aspp1 in

the regulation of HSCs. In addition to effects similar to those of

p53 deficiency, Aspp1 deficiency further increased the number

of functional HSCs in vivo, enhanced self-renewal capacity of

HSCs, and facilitated survival of HSCs carrying considerable

DNA lesions in the absence of p53. Thus, Aspp1 regulates

HSCs in both p53-dependent and -independent fashions. Our

microarray analyses identified multiple potential molecular

pathways that could be responsible for the p53-independent

function of Aspp1. We extracted several pathways relating to

30 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.

Page 10: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

phosphorylation, immune response, and hypoxia-induced

response by GO analysis and GSEA. Aspp family members

can associate with protein phosphatase 1 (Helps et al., 1995),

suggesting a possibility for Aspp1 to regulate the dephosphory-

lation of various proteins. TNF-a signaling, which has a negative

impact on HSC self-renewal (Pronk et al., 2011), activates

NF-kB, and its major component RelA/p65 can associate with

Aspp1 (Yang et al., 1999). In addition, our microarray indicates

a possible involvement of the hypoxia-induced response, which

is another key component of HSC regulation and regulates the

protein level of the Aspp1-paralogue Aspp2 (Kim et al., 2014).

Whether these pathways are involved in the p53-independent

regulation of HSCs by Aspp1 should be examined in future

studies.

Finally, our results offer direct evidence that Aspp1 coordi-

nates with p53 to prevent HSCs from developing hematological

malignancies. In humans, loss of ASPP1 expression due to

hypermethylation of its promoter region has been observed in

approximately 25% of ALL patients, mostly in T cell leukemia,

and has been proven by multivariate analysis to be an indepen-

dent poor prognosis factor in ALL patients (Agirre et al., 2006;

Roman-Gomez et al., 2005). In line with this, we have biologically

proven that Aspp1 functions as a tumor suppressor for hemato-

logical malignancies in the context of p53-deficiency-driven

tumorigenesis. Aspp1 deficiency alone did not increase the

incidence of hematological tumors, seemingly because of

the contribution of p53. The coordinated tumor-suppressive

function of Aspp1 can possibly be mediated by its ability to

activate the proapoptotic function of p73 (Bergamaschi et al.,

2004), which has been implicated in a variety of hematological

A B

C

Figure 7. p53-Independent Regulation of

HSCs by Aspp1

(A) Schematic procedure of microarray analysis.

Differentially expressed genes (more than 1.5-fold)

were subjected to gene ontology (GO) analysis.

Genes downregulated in Aspp1�/� compared with

WT LSK cells and Aspp1�/�p53�/� to p53�/� LSK

cells were then subjected to the ‘‘compute over-

lap’’ tool at http://www.broadinstitute.org/gsea/

index.jsp to screen gene sets highly relevant to

the downregulated genes. The top 100 gene sets

were obtained, and overlapped gene sets are listed

in Table S5. Red-colored numbers in left-hand

boxes show the number of upregulated genes and

blue-colored numbers show the number of down-

regulated genes. Asterisks indicate significant

enrichment of GO terms in the indicated genes

revealed by GO analysis.

(B) Top 10 GO terms significantly enriched in genes

downregulated in Aspp1�/�p53�/� compared with

p53�/� LSK cells.

(C) Coordinated changes in Aspp1�/�p53�/� LSK

cells compared with p53�/� LSK cells determined

by gene set enrichment analysis (GSEA) using the

overlapped gene sets.

malignancies (Alexandrova and Moll,

2012). The preference toward T cell

malignancies was underpinned by our

microarray analysis showing that Aspp1

coordinately downregulates the signature associated with

angioimmunoblastic T cell lymphoma, a common type of mature

T cell lymphoma with poor prognosis (Palomero et al., 2014;

Sakata-Yanagimoto et al., 2014; Yoo et al., 2014), in the HSPC

population. However, several of the tumors developed from

Aspp1�/�p53�/� LSK cells were of B cell or myeloid lineage.

Because Aspp1 and p53 deficiency causes non-homogeneous

tumors, it is possible that loss of both Aspp1 and p53 may

trigger eventual acquisition of driver mutations because of ge-

netic instability in HSCs during expansion rather than through

differentiation.

Together our findings reveal that Aspp1 and p53 coordinately

maintain integrity of the HSC pool and prevent hematological

malignancies. Our study highlights Aspp1 as well as p53 as a

potential target for stem cell-directed therapy of leukemia and

lymphoma. Our BM transplantation model using HSCs deficient

for both Aspp1 and p53 may provide beneficial findings for a

comprehensive understanding of how HSCs acquire mutations

and expand their clones before the onset of hematological

malignancies.

EXPERIMENTAL PROCEDURES

Mice

Aspp1�/� mice were generated as described (Hirashima et al., 2008) and

backcrossed to C57BL/6 mice for more than six generations. p53�/� mice

(Zhang et al., 2004), Mx1-Cre mice (Kuhn et al., 1995), and p53flox/flox mice

(Marino et al., 2000) were used to obtain p53-deficient LSK cells. Deletion of

floxed p53 alleles was induced as described (Yamashita et al., 2013). Litter-

mates or age- and gender-matched mice were used as controls unless other-

wise indicated. C57BL/6 mice congenic for the CD45 locus (C57BL/6-CD45.1)

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 31

Page 11: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

were purchased from Sankyo Labo Service (Tokyo, Japan). All procedures

were performed in accordance with the guidelines for animal and recombinant

DNA experiments of Keio University.

Flow Cytometry Analysis and Isolation of Hematopoietic Cells

Cell staining and enrichment for isolation of HSCs and other hematopoietic

cells were performed as described (Arai et al., 2004; Nitta et al., 2011). Cell-

cycle analysis using BrdU and anti-Ki67 antibody and apoptosis detection

using Annexin V were performed as described (Nitta et al., 2011). For detection

of b-gal activity, BM MNCs were stained with antibodies for HSC surface

markers and then loaded with FDG (Molecular Probes) by hypotonic shock

as described (Nolan et al., 1988).

BM Transplantation

C57BL/6-CD45.1 mice were used as recipients and also as a source of

competitor BM MNCs. Serial competitive reconstitution assays were per-

formed as described (Nitta et al., 2011). For cell-cycle and apoptosis analysis

after transplantation, 200 LT-HSCs were transplanted competitively into

lethally irradiated recipient mice and 2 3 106 BM MNCs from the primary

recipients were transplanted into lethally irradiated secondary recipients.

For gene expression analysis, 2 3 107 BM MNCs were injected into lethally

irradiated recipient mice and CD45.2+ LSK cells were isolated from BM of

the recipients at 16 hr post-transplantation.

Gene Expression Analysis

RNA extraction, cDNA synthesis, and qRT-PCR analysis were performed

as described (Yamashita et al., 2013). qRT-PCR arrays after BM transplan-

tation and after irradiation in vivo were performed with a BioMark 96.96

Dynamic Array and BioMark 48.48 Dynamic Array, respectively (Fluidigm)

using the TaqMan Gene Expression Assay (Applied Biosystems) as

described (Nakamura et al., 2010). For microarray analysis, isolated RNA

from approximately 3 3 105 LSK cells of each genotype was amplified,

labeled, and hybridized to an Agilent SurePrint G3 Mouse GE 8x60K

Microarray. Normalization of signals and initial GO analyses were performed

using Agilent GeneSpring version 12.6.1. GO analysis using DAVID, and

GSEAs were performed as described (Huang et al., 2009; Subramanian

et al., 2005).

Immunofluorescence Microscopy and Western Blotting

Immunofluorescence staining of hematopoietic cells was performed as

described (Yamashita et al., 2013). Briefly, cells were attached onto a MAS-

coated glass slide (Matsunami Glass), fixed with ice-cold methanol followed

by ice-cold acetone, and stainedwith anti-phospho-p53 (Ser15) (Cell Signaling

Technology; 1:400), anti-53BP1 (Novus Biologicals; 1:200), and anti-gH2AX

(Millipore; 1:500) antibodies. Nuclei were identified by being stained with

DAPI or TOTO-3 (Invitrogen). For western blotting, antibodies against mouse

Aspp1 were generated in rabbits against a peptide corresponding to amino

acid 87–106 (IBL). Cell lysates were separated by electrophoresis and trans-

ferred to polyvinylidene fluoride (PVDF) membranes (Millipore), which were

then incubated with antibodies against Aspp1 (1:10,000) and b-actin (Sigma-

Aldrich; 1:5,000) followed by horseradish-conjugated secondary antibodies.

Protein bands were visualized using ECL Prime (GE Healthcare) and

LAS4000 (Fuji Film).

Statistical Analysis

Unpaired two-tailed Student’s t test was used for comparison between two

groups. The survival rates of mice were compared using a log-rank nonpara-

metric test. Data are shown as the means ± SD. The value of p < 0.05 was

considered significant and p < 0.01 was considered highly significant.

ACCESSION NUMBERS

The microarray data reported in this manuscript have been deposited in

the GEO databank (http://www.ncbi.nlm.nih.gov/geo) under the accession

number GEO: GSE69032.

SUPPLEMENTAL INFORMATION

Supplemental Information for this article includes seven figures, five tables,

and Supplemental Experimental Procedures and can be found with this article

online at http://dx.doi.org/10.1016/j.stem.2015.05.013.

AUTHOR CONTRIBUTIONS

M.Y. and E.N. designed the study and performed the experiments. M.Y.

analyzed and interpreted the data and wrote the manuscript. E.N. and T.S.

assisted in manuscript preparation. T.S. directed the research and secured

funding.

ACKNOWLEDGMENTS

We thank Dr. Tohru Minamino (currently Niigata University, Niigata, Japan) for

providing the p53flox/flox mice; Dr. John E. Dick (University of Toronto, Toronto,

Canada), Dr. Michael Milyavsky (Tel Aviv University, Ramat Aviv, Israel), and

Dr. Masanori Hirashima (Kobe University, Kobe, Japan) for insights and

constructive suggestions; and all members of the Suda laboratory for com-

ments and discussion. We are grateful to the Collaborative Research

Resources, School of Medicine, Keio University for technical support and re-

agents. This work was supported by a Grant-in-Aid for Scientific Research

and a Grant-in-Aid for Scientific Research on Innovative Areas ‘‘Cancer

Stem Cells’’ from the Ministry of Education, Culture, Sports, Science and

Technology (MEXT) of Japan; the Global COE program ‘‘Education and

Research Center for Stem Cell Medicine’’ of Keio University; a Grant-in-Aid

for Japan Society for the Promotion of Science Fellows; and a grant-in-aid

from Takeda Science Foundation. M.Y. and E.N. are Research Fellows of

the Japan Society for the Promotion of Science.

Received: July 25, 2014

Revised: April 2, 2015

Accepted: May 26, 2015

Published: June 25, 2015

REFERENCES

Agirre, X., Roman-Gomez, J., Jimenez-Velasco, A., Garate, L., Montiel-Duarte,

C., Navarro, G., Vazquez, I., Zalacain, M., Calasanz, M.J., Heiniger, A., et al.

(2006). ASPP1, a common activator of TP53, is inactivated by aberrant

methylation of its promoter in acute lymphoblastic leukemia. Oncogene 25,

1862–1870.

Alexandrova, E.M., andMoll, U.M. (2012). Role of p53 family members p73 and

p63 in human hematological malignancies. Leuk. Lymphoma 53, 2116–2129.

Arai, F., Hirao, A., Ohmura, M., Sato, H., Matsuoka, S., Takubo, K., Ito, K., Koh,

G.Y., and Suda, T. (2004). Tie2/angiopoietin-1 signaling regulates hematopoi-

etic stem cell quiescence in the bone marrow niche. Cell 118, 149–161.

Beerman, I., Bhattacharya, D., Zandi, S., Sigvardsson, M., Weissman, I.L.,

Bryder, D., and Rossi, D.J. (2010). Functionally distinct hematopoietic stem

cells modulate hematopoietic lineage potential during aging by a mechanism

of clonal expansion. Proc. Natl. Acad. Sci. USA 107, 5465–5470.

Beerman, I., Seita, J., Inlay, M.A., Weissman, I.L., and Rossi, D.J. (2014).

Quiescent hematopoietic stem cells accumulate DNA damage during aging

that is repaired upon entry into cell cycle. Cell Stem Cell 15, 37–50.

Bergamaschi, D., Samuels, Y., O’Neil, N.J., Trigiante, G., Crook, T., Hsieh,

J.K., O’Connor, D.J., Zhong, S., Campargue, I., Tomlinson, M.L., et al.

(2003). iASPP oncoprotein is a key inhibitor of p53 conserved from worm to

human. Nat. Genet. 33, 162–167.

Bergamaschi, D., Samuels, Y., Jin, B., Duraisingham, S., Crook, T., and Lu, X.

(2004). ASPP1 and ASPP2: common activators of p53 family members. Mol.

Cell. Biol. 24, 1341–1350.

Blanpain, C., Mohrin, M., Sotiropoulou, P.A., and Passegue, E. (2011).

DNA-damage response in tissue-specific and cancer stem cells. Cell Stem

Cell 8, 16–29.

32 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.

Page 12: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

Cabezas-Wallscheid, N., Klimmeck, D., Hansson, J., Lipka, D.B., Reyes, A.,

Wang, Q., Weichenhan, D., Lier, A., von Paleske, L., Renders, S., et al.

(2014). Identification of regulatory networks in HSCs and their immediate prog-

eny via integrated proteome, transcriptome, and DNA Methylome analysis.

Cell Stem Cell 15, 507–522.

Corces-Zimmerman, M.R., Hong, W.J., Weissman, I.L., Medeiros, B.C., and

Majeti, R. (2014). Preleukemic mutations in human acute myeloid leukemia

affect epigenetic regulators and persist in remission. Proc. Natl. Acad. Sci.

USA 111, 2548–2553.

de Laval, B., Pawlikowska, P., Petit-Cocault, L., Bilhou-Nabera, C., Aubin-

Houzelstein, G., Souyri, M., Pouzoulet, F., Gaudry, M., and Porteu, F. (2013).

Thrombopoietin-increased DNA-PK-dependent DNA repair limits hematopoi-

etic stem and progenitor cell mutagenesis in response to DNA damage. Cell

Stem Cell 12, 37–48.

Ema, H., Morita, Y., Yamazaki, S., Matsubara, A., Seita, J., Tadokoro, Y.,

Kondo, H., Takano, H., and Nakauchi, H. (2006). Adult mouse hematopoietic

stem cells: purification and single-cell assays. Nat. Protoc. 1, 2979–2987.

Flach, J., Bakker, S.T., Mohrin, M., Conroy, P.C., Pietras, E.M., Reynaud, D.,

Alvarez, S., Diolaiti, M.E., Ugarte, F., Forsberg, E.C., et al. (2014).

Replication stress is a potent driver of functional decline in ageing haemato-

poietic stem cells. Nature 512, 198–202.

Garaycoechea, J.I., Crossan, G.P., Langevin, F., Daly, M., Arends, M.J., and

Patel, K.J. (2012). Genotoxic consequences of endogenous aldehydes on

mouse haematopoietic stem cell function. Nature 489, 571–575.

Helps, N.R., Barker, H.M., Elledge, S.J., and Cohen, P.T. (1995). Protein phos-

phatase 1 interacts with p53BP2, a protein which binds to the tumour suppres-

sor p53. FEBS Lett. 377, 295–300.

Hirashima,M., Bernstein, A., Stanford,W.L., and Rossant, J. (2004). Gene-trap

expression screening to identify endothelial-specific genes. Blood 104,

711–718.

Hirashima, M., Sano, K., Morisada, T., Murakami, K., Rossant, J., and Suda, T.

(2008). Lymphatic vessel assembly is impaired in Aspp1-deficient mouse em-

bryos. Dev. Biol. 316, 149–159.

Huang, W., Sherman, B.T., and Lempicki, R.A. (2009). Systematic and integra-

tive analysis of large gene lists using DAVID bioinformatics resources. Nat.

Protoc. 4, 44–57.

Jia, Y., Peng, L., Rao, Q., Xing, H., Huai, L., Yu, P., Chen, Y., Wang, C., Wang,

M., Mi, Y., et al. (2014). Oncogene iASPP enhances self-renewal of hematopoi-

etic stem cells and facilitates their resistance to chemotherapy and irradiation.

FASEB J. 28, 2816–2827.

Kim, H., Claps, G., Moller, A., Bowtell, D., Lu, X., and Ronai, Z.A. (2014). Siah2

regulates tight junction integrity and cell polarity through control of ASPP2

stability. Oncogene 33, 2004–2010.

Kuhn, R., Schwenk, F., Aguet, M., and Rajewsky, K. (1995). Inducible gene tar-

geting in mice. Science 269, 1427–1429.

Lerner, C., and Harrison, D.E. (1990). 5-Fluorouracil spares hemopoietic stem

cells responsible for long-term repopulation. Exp. Hematol. 18, 114–118.

Liu, Y., Elf, S.E., Miyata, Y., Sashida, G., Liu, Y., Huang, G., Di Giandomenico,

S., Lee, J.M., Deblasio, A., Menendez, S., et al. (2009). p53 regulates hemato-

poietic stem cell quiescence. Cell Stem Cell 4, 37–48.

Marino, S., Vooijs, M., van Der Gulden, H., Jonkers, J., and Berns, A. (2000).

Induction ofmedulloblastomas in p53-null mutantmice by somatic inactivation

of Rb in the external granular layer cells of the cerebellum. Genes Dev. 14,

994–1004.

Milyavsky, M., Gan, O.I., Trottier, M., Komosa, M., Tabach, O., Notta, F.,

Lechman, E., Hermans, K.G., Eppert, K., Konovalova, Z., et al. (2010). A

distinctive DNA damage response in human hematopoietic stem cells reveals

an apoptosis-independent role for p53 in self-renewal. Cell Stem Cell 7,

186–197.

Mohrin, M., Bourke, E., Alexander, D., Warr, M.R., Barry-Holson, K., Le Beau,

M.M., Morrison, C.G., and Passegue, E. (2010). Hematopoietic stem cell

quiescence promotes error-prone DNA repair and mutagenesis. Cell Stem

Cell 7, 174–185.

Morita, Y., Ema, H., and Nakauchi, H. (2010). Heterogeneity and hierarchy

within the most primitive hematopoietic stem cell compartment. J. Exp.

Med. 207, 1173–1182.

Nakamura, Y., Arai, F., Iwasaki, H., Hosokawa, K., Kobayashi, I., Gomei, Y.,

Matsumoto, Y., Yoshihara, H., and Suda, T. (2010). Isolation and characteriza-

tion of endosteal niche cell populations that regulate hematopoietic stem cells.

Blood 116, 1422–1432.

Nitta, E., Yamashita, M., Hosokawa, K., Xian, M., Takubo, K., Arai, F., Nakada,

S., and Suda, T. (2011). Telomerase reverse transcriptase protects ATM-defi-

cient hematopoietic stem cells from ROS-induced apoptosis through a telo-

mere-independent mechanism. Blood 117, 4169–4180.

Nolan, G.P., Fiering, S., Nicolas, J.F., and Herzenberg, L.A. (1988).

Fluorescence-activated cell analysis and sorting of viable mammalian cells

based on beta-D-galactosidase activity after transduction of Escherichia coli

lacZ. Proc. Natl. Acad. Sci. USA 85, 2603–2607.

Orkin, S.H., and Zon, L.I. (2008). Hematopoiesis: an evolving paradigm for

stem cell biology. Cell 132, 631–644.

Palomero, T., Couronne, L., Khiabanian, H., Kim, M.Y., Ambesi-Impiombato,

A., Perez-Garcia, A., Carpenter, Z., Abate, F., Allegretta, M., Haydu, J.E.,

et al. (2014). Recurrent mutations in epigenetic regulators, RHOA and FYN ki-

nase in peripheral T cell lymphomas. Nat. Genet. 46, 166–170.

Pronk, C.J., Veiby, O.P., Bryder, D., and Jacobsen, S.E. (2011). Tumor necro-

sis factor restricts hematopoietic stem cell activity in mice: involvement of two

distinct receptors. J. Exp. Med. 208, 1563–1570.

Quivoron, C., Couronne, L., Della Valle, V., Lopez, C.K., Plo, I., Wagner-Ballon,

O., Do Cruzeiro, M., Delhommeau, F., Arnulf, B., Stern, M.H., et al. (2011). TET2

inactivation results in pleiotropic hematopoietic abnormalities in mouse and is

a recurrent event during human lymphomagenesis. Cancer Cell 20, 25–38.

Rogakou, E.P., Pilch, D.R., Orr, A.H., Ivanova, V.S., and Bonner, W.M. (1998).

DNA double-stranded breaks induce histone H2AX phosphorylation on serine

139. J. Biol. Chem. 273, 5858–5868.

Roman-Gomez, J., Jimenez-Velasco, A., Agirre, X., Prosper, F., Heiniger, A.,

and Torres, A. (2005). Lack of CpG island methylator phenotype defines a clin-

ical subtype of T-cell acute lymphoblastic leukemia associated with good

prognosis. J. Clin. Oncol. 23, 7043–7049.

Rossi, D.J., Jamieson, C.H., and Weissman, I.L. (2008). Stems cells and the

pathways to aging and cancer. Cell 132, 681–696.

Sakata-Yanagimoto, M., Enami, T., Yoshida, K., Shiraishi, Y., Ishii, R., Miyake,

Y., Muto, H., Tsuyama, N., Sato-Otsubo, A., Okuno, Y., et al. (2014). Somatic

RHOA mutation in angioimmunoblastic T cell lymphoma. Nat. Genet. 46,

171–175.

Samuels-Lev, Y., O’Connor, D.J., Bergamaschi, D., Trigiante, G., Hsieh, J.K.,

Zhong, S., Campargue, I., Naumovski, L., Crook, T., and Lu, X. (2001). ASPP

proteins specifically stimulate the apoptotic function of p53. Mol. Cell 8,

781–794.

Schultz, L.B., Chehab, N.H., Malikzay, A., and Halazonetis, T.D. (2000). p53

binding protein 1 (53BP1) is an early participant in the cellular response to

DNA double-strand breaks. J. Cell Biol. 151, 1381–1390.

Shlush, L.I., Zandi, S., Mitchell, A., Chen, W.C., Brandwein, J.M., Gupta, V.,

Kennedy, J.A., Schimmer, A.D., Schuh, A.C., Yee, K.W., et al.; HALT

Pan-Leukemia Gene Panel Consortium (2014). Identification of pre-leukaemic

haematopoietic stem cells in acute leukaemia. Nature 506, 328–333.

Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, B.L.,

Gillette, M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lander, E.S., and

Mesirov, J.P. (2005). Gene set enrichment analysis: a knowledge-based

approach for interpreting genome-wide expression profiles. Proc. Natl.

Acad. Sci. USA 102, 15545–15550.

Trigiante, G., and Lu, X. (2006). ASPP [corrected] and cancer. Nat. Rev. Cancer

6, 217–226.

Van Zant, G. (1984). Studies of hematopoietic stem cells spared by 5-fluoro-

uracil. J. Exp. Med. 159, 679–690.

Walter, D., Lier, A., Geiselhart, A., Thalheimer, F.B., Huntscha, S., Sobotta,

M.C., Moehrle, B., Brocks, D., Bayindir, I., Kaschutnig, P., et al. (2015). Exit

Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc. 33

Page 13: Aspp1 Preserves Hematopoietic Stem Cell Pool Integrity and ... · Data shown are mean ± SD. *p < 0.05, **p < 0.01. See also Figures S1 and S2. S1A and S1C). LSK cells and LT-HSCs

from dormancy provokes DNA-damage-induced attrition in haematopoietic

stem cells. Nature 520, 549–552.

Wang, J., Sun, Q., Morita, Y., Jiang, H., Gross, A., Lechel, A., Hildner, K.,

Guachalla, L.M., Gompf, A., Hartmann, D., et al. (2012). A differentiation

checkpoint limits hematopoietic stem cell self-renewal in response to DNA

damage. Cell 148, 1001–1014.

Wang, Y., Godin-Heymann, N., Dan Wang, X., Bergamaschi, D., Llanos, S.,

and Lu, X. (2013). ASPP1 and ASPP2 bind active RAS, potentiate RAS sig-

nalling and enhance p53 activity in cancer cells. Cell Death Differ. 20,

525–534.

Wilson, A., Laurenti, E., Oser, G., van der Wath, R.C., Blanco-Bose, W.,

Jaworski, M., Offner, S., Dunant, C.F., Eshkind, L., Bockamp, E., et al.

(2008). Hematopoietic stem cells reversibly switch from dormancy to self-

renewal during homeostasis and repair. Cell 135, 1118–1129.

Yamashita, M., Nitta, E., Nagamatsu, G., Ikushima, Y.M., Hosokawa, K., Arai,

F., and Suda, T. (2013). Nucleostemin is indispensable for the maintenance

and genetic stability of hematopoietic stem cells. Biochem. Biophys. Res.

Commun. 441, 196–201.

Yang, J.P., Hori, M., Sanda, T., and Okamoto, T. (1999). Identification of a

novel inhibitor of nuclear factor-kappaB, RelA-associated inhibitor. J. Biol.

Chem. 274, 15662–15670.

Yoo, H.Y., Sung, M.K., Lee, S.H., Kim, S., Lee, H., Park, S., Kim, S.C., Lee, B.,

Rho, K., Lee, J.E., et al. (2014). A recurrent inactivating mutation in RHOA

GTPase in angioimmunoblastic T cell lymphoma. Nat. Genet. 46, 371–375.

Yoshihara, H., Arai, F., Hosokawa, K., Hagiwara, T., Takubo, K., Nakamura, Y.,

Gomei, Y., Iwasaki, H., Matsuoka, S., Miyamoto, K., et al. (2007).

Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence

and interaction with the osteoblastic niche. Cell Stem Cell 1, 685–697.

Zhang, D., Hirota, T., Marumoto, T., Shimizu, M., Kunitoku, N., Sasayama, T.,

Arima, Y., Feng, L., Suzuki, M., Takeya, M., and Saya, H. (2004). Cre-loxP-

controlled periodic Aurora-A overexpression induces mitotic abnormalities

and hyperplasia in mammary glands of mouse models. Oncogene 23, 8720–

8730.

34 Cell Stem Cell 17, 23–34, July 2, 2015 ª2015 Elsevier Inc.