Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

HEMATOPOIESIS AND STEM CELLS

Salidroside stimulates DNA repair enzyme Parp-1 activity in mouse HSC


maintenance
Xue Li,1,2 Jared Sipple,1 Qishen Pang,1,3 and Wei Du1
1Division
of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; 2College of Life Sciences, South China
Normal University, Guangzhou, China; and 3Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH

Salidroside is a phenylpropanoid glyco- duces hydrogen peroxide–induced DNA- blasts. PARP-1 activation by salidroside
side isolated from the medicinal plant strand breaks in bone marrow cells protects quiescent HSCs from oxidative
Rhodiola rosea, which has potent antioxi- enriched for HSCs. We tested whether stress–induced cycling in native animals
dant properties. Here we show that salidroside enhances oxidative DNA dam- and self-renewal defect in transplanted
salidroside prevented the loss of hemato- age repair in mice deficient for 5 DNA recipients, which was abrogated by ge-
poietic stem cells (HSCs) in mice under repair pathways known to be involved in netic ablation or pharmacologic inhibi-
oxidative stress. Quiescent HSCs were oxidative DNA damage repair; we found tion of PARP-1. Together, these findings
recruited into cell cycling on in vivo chal- that salidroside activated poly(ADP- suggest that activation of PARP-1 by
lenge with oxidative stress, which was ribose)polymerase-1 (PARP-1), a compo- salidroside could affect the homeostasis
blocked by salidroside. Surprisingly, nent of the base excision repair pathway, and function of HSCs and contribute to
salidroside does not prevent the produc- in mouse bone marrow HSCs as well as the antioxidant effects of salidroside.
tion of reactive oxygen species but re- primary fibroblasts and human lympho- (Blood. 2012;119(18):4162-4173)

Introduction
Hematopoietic stem cells (HSCs) are a rare population of ROS in hematopoietic tissues.8,14 There is strong evidence that
pluripotent cells that can self-renew and differentiate into HSCs are activated and thus functionally exhausted by oxidative
various types of cells of the blood lineage.1 Under steady stress. Mice with mutations in the ATM or FOXO genes, as well as
physiologic conditions, the most primitive HSCs are in a various DNA repair genes, exhibit premature exhaustion of HSCs
quiescent state and reside in the bone marrow (BM) niche where because of accumulation of ROS or DNA damage, indicating that
they preserve the capacity to self-renew and to continue to cellular balance between ROS and antioxidant defense, as well as
produce all types of blood cells throughout a prolonged life span DNA repair, is crucial for the maintenance of HSC self-renewal and
without depleting the regenerative cell pool.2,3 In response to hematopoietic function.15,16
stress or stimulation, the HSCs can move out of the BM niche, Salidroside is a phenylpropanoid glycoside isolated from the
entering cell cycle and undergoing division. In addition, the medicinal plant Rhodiola rosea that grows in high altitude or
cycling HSCs may return to the BM niche and regain their cold regions of the world and was used as a folk medicine in
quiescent state.4 Disruption of HSC quiescence prematurely France, Germany, and many European countries to fight fatigue
exhausts the stem cell pool and causes hematologic failure under in the 19th century.17 In present days, extract of R rosea has been
various stresses, such as oxidative stress, cell cycling, and used to enhance both the physical and mental performance.18,19
aging.5,6 Salidroside (2-[4-hydroxyphenyl]ethyl ␤-D-glucopyranoside) has
Oxidative stress, defined as an imbalance between the produc- been reported to have anti-aging, anti-cancer, anti-inflammatory,
tion of reactive oxygen species (ROS) and antioxidant defense, is and antioxidative functions.20-23 A recent study shows that
most evident in states of aging and diseases such as BM failure and salidroside promotes erythropoiesis and up-regulates the level
cancer.7,8 Even in a healthy state, HSCs are exposed to various of antioxidative enzymes glutathione peroxidase-1 and thiore-
ROS, which are routinely generated during metabolic or inflamma- doxin-1 to counteract oxidative stress.24 In this study, we show
tory process.9 ROS induce a variety of responses in HSCs, that salidroside prevented quiescent HSCs and progenitor cells
including cellular proliferation and apoptosis.10,11 ROS can also from being recruited into cell cycling on in vivo challenge with
cause DNA damage and drive HSCs into cell division, which is oxidative stress in mice. Using several mouse models deficient
essential for DNA repair processes.12,13 Similar to stem cells from for DNA repair pathways known to be involved in oxidative
other tissues, HSCs have developed several mechanisms to prevent DNA damage repair (ODDR), we demonstrate that salidroside
the damage induced by oxidative stress. Antioxidant enzymes, protects quiescent hematopoietic stem progenitor cells (HSPCs)
including superoxide dismutases, catalase, glutathione peroxi- from oxidative stress–induced cycling through stimulation activ-
dases, and peroxiredoxins, can directly eliminate ROS.10 Other ity of poly(ADP-ribose)polymerase-1 (PARP-1), a component
cellular enzymes can function to repair DNA damage induced by of the base excision repair pathway.

Submitted October 20, 2011; accepted March 6, 2012. Prepublished online as Blood The publication costs of this article were defrayed in part by page charge
First Edition paper, March 16, 2012; DOI 10.1182/blood-2011-10-387332. payment. Therefore, and solely to indicate this fact, this article is hereby
marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
The online version of this article contains a data supplement. © 2012 by The American Society of Hematology

4162 BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18


From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4163

salidroside on oxidative stress in HSC in mice, we first determined


Methods the optimal dose of hydrogen peroxide (H2O2) and found that
0.25 ␮mol/g body weight was the most effective dose that
Mice and treatments effectively induced DNA damage without causing death (supple-
Parp1⫺/⫺ or Xpc⫺/⫺ mice were generated by backcrossing 129S- mental Figure 1B). In addition, this range of H2O2 doses gave an
Parp1tm1zqw/J or B6;129-Xpctm1Ecf/J (The Jackson laboratory), respectively, inverse correlation between DNA damage and HSC repopulation
with wild-type (WT) C57BL/6 mice. Fancd2⫺/⫺ mice (provided by (supplemental Figure 1C). We then used this dose of H2O2 to treat
Dr Markus Grompe, Oregon Health & Science University) or DNA- the mice and found that salidroside effectively antagonized H2O2-
PKcs3A/3A mice (a gift from Benjamin P. C. Chen, University of Texas induced effect on WT HSCs in vivo (Figure 1). Although both
Southwestern Medical Center at Dallas)25 were generated by interbreeding salidroside and H2O2 treatments did not change absolute number of
heterozygous Fancd2⫾ or DNA-PKcs⫹/3A mice, respectively. Brca2⫺/⫺
total BM cells (supplemental Figure 3), H2O2-treated mice had a
mice were generated by Cre-mediated deletion of floxed alleles by crossing
Brca2tm1Brn (NCI)26 with a Cre-ERT2 strain.27
much higher frequency of Lin-c-kit⫹Sca-1⫹ (LSK) cells in the BM
For H2O2 treatment, mice were first screened with increasing doses of than untreated mice, and salidroside partially limited this expansion
H2O2 (Sigma-Aldrich; 0, 0.05, 0.15, 0.25, 0.35, and 0.50 ␮mol/g body (Figure 1A-B). Further analysis of the LSK compartment indicated
weight), and that the optimal dose (0.25 ␮mol/g body weight) was chosen that H2O2-treated mice had much lower LSK CD34⫺Flt3⫺ cells, a
for further experiments. For antioxidant treatment, mice were injected with population enriched for long-term (LT)–HSCs,29,30 than untreated
salidroside (PhytoLab; 75␮g/g body weight), manganese (III) tetrakis control mice (Figure 1C-D). Significantly, salidroside almost
(4-benzoic acid) porphyrin (MnTBAP, Sigma-Aldrich; 10 ␮g/g body
completely abrogated H2O2-induced loss of LT-HSCs. We also
weight), N-acetyl-L-cysteine (NAC; Sigma-Aldrich; 50 ␮g/g body weight)
or saline vehicle, intraperitoneally followed by H2O2 (0.25 ␮mol/g body determined the effect of salidroside on the BM HSC compartment
weight). For NU1025 treatment, mice were injected with NU1025 (25 mg/kg using the CD150 and CD48-based immunophenotyping.31,32 Simi-
body weight; Sigma-Aldrich) followed by H2O2 treatment. All experimental larly, we observed that salidroside effectively rescued LT-HSCs
procedures conducted in this study were approved by the Institutional Animal (CD150⫹CD48⫺ LSK) in H2O2-treated mice (Figure 1E-F). More-
Care and Use Committee of Cincinnati Children’s Hospital Medical Center. over, salidroside protected H2O2-induced HSC loss in mice defi-
cient for the FA gene, Fanca (Fanca⫺/⫺) or Fancc (Fancc⫺/⫺;
Parp1 activity supplemental Figure 2). Together, these data suggest that salidro-
Parp1 activity was detected by flow cytometry as previously described.28 side plays a selective role in the maintenance of LT-HSCs.
Briefly, cells were centrifuged and resuspended in 100% ethanol and left at
⫺20°C for at least 20 minutes. Cells were then resuspended in ⬃10 mL
Salidroside enhances the ability of stressed HSCs to
buffer A (10mM Tris-HCl pH 7.8, 1mM EDTA [ethylenediaminetetraacetic
acid], 4mM MgCl2, and 30mM 2-mercaptoethanol). Then cells were repopulate mouse hematopoietic system
centrifuged again and resuspended in buffer A again and transferred to a
V-shaped 96-well plate on ice for at least 5 minutes. Then 20 ␮L of 3X
We next assessed whether salidroside improved the repopulating
reaction buffer (with or with NAD⫹) plus 13 ␮L of 15mM NaCl ability of oxidative stressed HSCs. We transplanted LSK cells from
incorporating were added to the reaction mix followed by 37°C incubation H2O2-treated WT C57BL/6 mice (CD45.2⫹) pretreated with or
for 10 minutes. Then second fixation was done by adding 60 ␮L of 4% without salidroside, along with BM cells (depleted of c-Kit⫹ cells
formaldehyde/phosphate-buffered saline (PBS) for 20 minutes at room to provide short-term hematopoiesis after irradiation) obtained
temperature. PBS was then added to quench the reaction. Cells were then from congenic WT Boy J mice (CD45.1⫹), into lethally irradiated
centrifuged and resuspended in 100 ␮L primary antibody diluted in congenic recipients (CD45.1⫹). We observed a significant increase
fluorescence-activated cell sorter (FACS) buffer and incubated at 37°C for 1 in both repopulated hematopoiesis in the peripheral blood (Figure
hour or overnight at 4°C. Then the cells were washed and resuspended in
2A) and HSC-enriched LSK cells in the BM of recipients that
100 ␮L of diluted secondary antibody (Alexa 488–conjugated goat
anti–mouse; Invitrogen) followed by 37°C incubation for 30 minutes. Cells received LSK cells from salidroside-treated mice (Figure 2B) at
were washed and resuspended for flow cytometry analysis. 4 months after transplantation. We also performed BM transplanta-
For flow cytometry and cell cycle analysis, BrdU incorporation, tion using total BM from stressed WT mice, which shows similar
determination of ROS production, comet assay, and BM transplantation, see results (supplemental Figure 4). Further, we performed competitive
supplemental Methods (available on the Blood Web site; see the Supplemen- reconstitution using limiting dilutions of LSK cells from stressed
tal Materials link at the top of the online article). mice with defined ratios of WT LSK competitor cells, and analyzed
hematopoietic reconstitution in recipient mice by donor cells at
8 and 16 weeks after transplantation. The results show that
Results donor-repopulated hematopoiesis was greater in the recipients that
received all doses of LSK cells from salidroside-treated mice than
Salidroside prevents oxidative stress–induced HSC loss in in those that received LSK cells from untreated mice (Figure 2C).
mice Furthermore, the recipients transplanted with cells from salidroside-
treated mice showed greater abundance of donor-derived LSK cells
In an attempt to search for new chemopreventive and antioxidant
agents that are effective and less toxic in hematopoietic improve- in the BM (Figure 2D). Analysis of the frequency of CD45.2⫹
ment for patients with BM failure syndromes, such as Fanconi lymphoid and myeloid as well as erythroid lineage cells in the
anemia (FA), in which oxidative stress is identified as a physiologic peripheral blood showed that both groups of donor LSK cells had a
mediator of HSC loss,7 we investigated the antioxidant effect of similar multilineage-reconstitution capacity (supplemental Figure
salidroside on HSC maintenance. Salidroside (2-[4-hydroxyphenyl- 5), indicating that salidroside does not affect cell fate or differentia-
]ethyl ␤-D-glucopyranoside; supplemental Figure 1A), a phenylpro- tion potential of HSCs. To investigate the long-term-repopulation
panoid glycoside found in the medicinal plant R rosea, has a wide abilities of salidroside-conditioned HSCs, we performed serial
range of biologic activities, such as antiaging, anticancer, anti- transplantation experiments by transplanting primary recipients of
inflammatory, and antioxidative functions.20-23 To test the effect of salidroside-treated or untreated mouse BM at 16 weeks after
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

4164 LI et al BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18

Figure 1. Salidroside prevents oxidative stress–


induced HSC loss. (A) salidroside partially limits H2O2-
induced LSK expansion. BM cells from WT C57BL/6
mice pretreated with or without salidroside (75␮g/g body
weight) followed by H2O2 (0.25 ␮mol/g body weight)
injection were harvested for LSK (Lin-Sca1⫹c-kit⫹) cell
frequency assessment using flow cytometry. (B) Quantifi-
cation of LSK frequency in mice described in panel A.
(C) SD prevents H2O2-induced LSK CD34-Flt3- cell loss.
BM cells described in panel A were subjected to clow
cytometry analysis for LSK CD34⫺Flt3⫺ frequency.
(D). Quantification of LSK CD34⫺Flt3⫺ frequency in mice
described in panel A. (E) SD prevents H2O2-induced LSK
CD150⫹CD48⫺ cell loss. BM cells described in panel A
were subjected to Flow Cytometry analysis for LSK
CD150⫹CD48⫺ frequency. (F) Quantification of
CD150⫹CD48⫺ frequency in mice described in panel A.
Results are means ⫾ standard deviation (SD) of
3 independent experiments (n ⫽ 9 per group).

transplantation into lethally irradiated secondary recipients. Six- apoptosis of HSCs in stressed animals. We observed increased
teen weeks after transplantation, we analyzed hematopoiesis de- apoptosis of LSK cells in H2O2-treated WT C57BL/6 mice
rived from donor (CD45.2⫹) cells. The results show that donor- compared with untreated mice, and salidroside partially reduced
derived hematopoiesis from primary recipients of salidroside- the stress-induced apoptosis (Figure 3A). However, H2O2 treatment
treated BM was significantly greater in the secondary recipients did not induce significant apoptosis in the more primitive CD34⫺
than that from primary recipients of untreated BM (Figure 2E). LSK cell compartment29 and salidroside had no effect on the
Notably, this was correlated with an increased number of LSK cells apoptosis in the cell population (Figure 3B). This finding suggests
in the bone marrow (Figure 2F). Together, these results demon- that the loss of HSCs in stressed mice may be because of the
strate that salidroside enhances the ability of stressed HSCs to recruitment of quiescent HSCs into cell cycle on challenge with
repopulate the hematopoietic system. oxidative stress. To test whether salidroside played a role in
Salidroside prevents oxidative stress–induced HSC cycling in vivo
preventing quiescent HSCs from entering cell cycling or prolifera-
tion, we first analyzed the cell cycle profile of CD34⫺ LSK cells by
To identify the mechanism of salidroside in HSC maintenance, we staining RNA and DNA with pyronin Y and Hoechst 33342,
asked whether salidroside prevented oxidative stress–induced respectively. Quiescent cell populations can be identified by
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4165

Figure 2. Salidroside enhances HSC repopulation.


(A) Salidroside enhances the repopulating ability in recipi-
ent mice. LSK (Lin-Sca1⫹c-kit⫹) cells from WT C57BL/6
mice (CD45.2⫹) pretreated with or without salidroside
(75␮g/g body weight) followed by H2O2 (0.25 ␮mol/g
body weight) treatment were isolated by cell sorting using
FlowAria II. One thousand sorted LSK cells plus 1 million
c-kit–depleted competitors (CD45.1⫹) were injected to
lethally irradiated recipients. Donor chimerism was exam-
ined at 4 months after transplantation using peripheral
blood from recipients. Representative images (left) and
quantifications (right) were shown. Results are means
⫾ SD of 3 independent experiments (n ⫽ 15 per group).
(B) Salidroside increases HSC-enriched LSK cells in
recipient mice. One thousand sorted LSK cells from WT
C57BL/6 mice (CD45.2⫹) pretreated with or without
salidroside followed by H2O2 treatment were injected
along with 1 million c-kit–depleted competitors (CD45.1⫹)
to lethally irradiated recipients. BM cells from recipient
mice were harvested and subjected to flow cytometry
analysis for LSK frequency. (C) Salidroside enhances
competitive reconstitution of stressed HSCs. Various
numbers of donor (CD45.2⫹) LSK cells (50, 100, or
1000) from stressed WT C57BL/6 mice were mixed
with 1000 competitor LSK (CD45.1⫹) cells and the
mixtures were transplanted intravenously into lethally
irradiated congenic (CD45.1 ⫹) recipients. Donor-
derived hematopoietic reconstitution was analyzed by
flow cytometry 8 and 16 weeks after transplantation
using peripheral blood from recipients. (D) Salidroside
increases HSC-enriched LSK in recipient mice. BM
cells from recipient mice described in panel C were
subjected to flow cytometry analysis for LSK fre-
quency. (E-F) Salidroside enhances LT-HSC repopula-
tion. BM cells from primary recipient mice described in
panel B were used for secondary transplantation by
injecting 5 ⫻ 106 BM cells to lethally irradiated recipi-
ents. Donor-derived chimerism (E) and LSK (F) were
analyzed by flow cytometry 16 weeks after BMT.
Results are means ⫾ SD of 2 independent experi-
ments (n ⫽ 10 per group).

negative to low pyronin Y staining; whereas actively cycling cells incorporated BrdU was elevated in H2O2-treated mice compared
are positive for pyronin Y.33 There was a marked decrease in the with the unstressed animals, which was largely abrogated by
number of quiescent (pyronin Y-negative; G0) and an increase in salidroside (Figure 3D). These data suggest that salidroside pre-
the number of cycling (pyronin Y-positive; G1⫹S) CD34⫺ LSK vents quiescent HSCs from oxidative stress–induced cycling.
cells in H2O2-treated mice compared with the unstressed animals
(Figure 3C). Salidroside treatment significantly decreased cycling, Salidroside reduces H2O2-induced DNA-strand breaks in
and at the same time, increased quiescent (G0) CD34⫺ LSK cells in HSPCs
the stressed mice. We also performed in vivo BrdU labeling in mice
to determine the proliferative status of HSCs in the BM. In line Because salidroside has antioxidative activity,20,21,24 we tested
with the cell-cycle data, the percentage of CD34⫺ LSK cells that whether salidroside could function as a free radical scavenger in
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

4166 LI et al BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18

Figure 3. Salidroside prevents HSC cycling in vivo. (A) Salidroside partially reduces H2O2-induced LSK cell apoptosis. BM cells were harvested from WT C57BL/6 mice
pretreated with or without salidroside (75␮g/g body weight) followed by H2O2 (0.25 ␮mol/g body weight), and gated for LSK population. Apoptotic LSK cells were determined by
annexin-V and 7AAD staining. (B) Salidroside and H2O2 have no effect on apoptosis of CD34-LSK cells. CD34 negative LSK cells were gated for apoptosis analysis.

BM HSCs of mice stressed with H2O2, which is a potent producer To further elucidate the action of mechanism by salidroside, we
of reactive oxygen species (ROS). We measured ROS by staining determined whether salidroside played a role in preventing oxida-
BM CD34⫺ LSK cells from H2O2-injected mice with CM-H2 tive DNA damage in HSPCs of the H2O2-treated mice. We used the
DCFDA, a cell-permeable fluorescent dye that reacts to a broad comet assay35 using a Fpg-FLARE (fragment length analysis using
spectrum of ROS. To accurately evaluate the effect of salidroside repair enzymes) assay kit, which measures specifically oxidative
on ROS, we used 2 small antioxidant molecules, NAC, and DNA damage including single and double-strand DNA breaks.36
manganese (III) tetrakis (4-benzoic acid) porphyrin (MnTBAP). There was significant accumulation of DNA damage in BM Lin-
NAC stimulates the formation of the endogenous reducing agent cells freshly isolated from H2O2-treated mice, which was largely
glutathione, which cells use to scavenge H2O2.10 MnTBAP has eliminated by salidroside, as well as by NAC or MnTBAP (Figure
catalytic activities similar to the ROS-scavenging enzymes super- 4B). Consistent with this, H2O2 induced expression of ␥-H2AX, a
oxide dismutase (SOD) and catalase.34 We pretreated the mice with robust indicator of DNA strand breaks,37 and all 3 antioxidants
salidroside, NAC, or MnTBAP, and then concomitantly with H2O2. suppressed ␥-H2AX expression in BM cells from H2O2-treated
Compared with NAC and MnTBAP, both eliminated most of the mice (supplemental Figure 6). A similar increase in 8-oxo-
ROS generated in the BM LSK cells of H2O2-injected mice, deoxyguanosin (8-oxodG), an established marker of oxidative
salidroside did not prevent the production of ROS in these cells DNA damage,9 was also demonstrated in BM cells from H2O2-
(Figure 4A). This finding suggests that salidroside antagonizes injected mice (Figure 4C). Cotreatment of H2O2-injected mice with
oxidative stress through a distinct mechanism that does not involve salidroside, or the ROS scavenger NAC or MnTBAP reduced the
ROS-scavenging. accumulation of 8-oxodG (Figure 4C).
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4167

Figure 3. (continued) (C-D) Salidroside prevents HSC cycling in vivo. Cells described in panel B were subjected to Hochest and pyronin Y (C) or BrdU (D) staining.
Representative images (top) and quantifications (bottom) were shown. Results are means ⫾ SD of 3 independent experiments (n ⫽ 9 per group).

To distinguish the antioxidant property of salidroside from that mechanism from that of the ROS scavenger NAC or MnTBAP in
of NAC or MnTBAP in the context of oxidative DNA damage repair of oxidative DNA damage.
repair, we pretreated fresh-isolated low-density BM cells with
salidroside, NAC, or MnTBAP for 2 hours, followed by H2O2 for Salidroside stimulates PARP-1 activity
additional 2 hours, and then measured the remaining amounts of
8-oxodG and DNA strand breaks for a period of 12 hours after To identify the mechanism by which salidroside antagonists the effect of
H2O2 treatment. There was significant difference between the effect oxidative stress on HSC maintenance, we next tested whether salidro-
of salidroside and that of NAC or MnTBAP in terms of the kinetics side enhanced ODDR in mice deficient for 5 different DNA repair
of 8-oxodG removal. Specifically, the level of oxidative DNA pathways known to be involved in ODDR, including homologous
damage was not decreased until 4 hours after salidroside treatment recombination (HR), nonhomologous end-joining (NHEJ), base exci-
with almost no 8-oxodG remaining at 12 hours (Figure 4D). sion repair (BER), nucleotide excision repair (NER), and FA path-
However, NAC and MnTBAP reduced the level of 8-oxodG at each ways.7,38-40 We observed reduction of H2O2-induced DNA strand breaks
time point after H2O2 incubation. This result indicates that the ROS by salidroside in BM Lin- cells from mice deficient for genes function-
scavengers NAC and MnTBAP have no effect on the repair of ing in HR (Brca2⫺/⫺),41 NHEJ (DNA-PKcs3A/3A),42 NER (Xpc⫺/⫺),43 and
8-oxodG, which is consistent with their function in reducing ROS FA (Fancd2⫺/⫺)44 pathways (Figure 5A). However, salidroside failed to
formation and preventing DNA damage. We observed similar reduce H2O2-induced DNA strand breaks in HSPCs from mice deleted
repair kinetics of DNA strand breaks in a comet assay, in which for the gene encoding poly(ADP-ribose)polymerase-1 (PARP-1), a
DNA strand breaks remained high in salidroside-treated cells component of the BER pathway (Figure 5A). Analysis of H2O2-induced
during 8 hours after H2O2 incubation (supplemental Figure 7). 8-oxodG in these gene-knockout BM cells showed similar results, with
Together, these results suggest that salidroside may use different salidroside accelerating clearance of the oxidative DNA adducts in
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

4168 LI et al BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18

Figure 4. Salidroside reduces DNA strand breaks but


not ROS. (A) Salidroside does not reduce H2O2-induced
ROS. WT C57BL/6 mice were pretreated with one dose
of salidroside (75␮g/g body weight), NAC (50␮g/g body
weight), or MnTBAP (10␮g/g body weight), followed by
H2O2 (0.25 ␮mol/g body weight). BM cells were then
harvested and labeled with CM-H2DCFDA for flow cytom-
etry analysis of ROS in the LSK-gated cells. Representa-
tive images (left) and quantifications (right) were shown.
(B) Salidroside reduces DNA strand breaks. BM cells
from mice described in panel A were isolated by magnetic
beads depletion, and then subjected to analysis for DNA
strand breaks by the comet assay. The mean tail moment
of untreated vehicle sample is expressed as 100%.
Larger tail moment represents higher levels of DNA
damage. For each treatment, 50 cells were scored from
random sampling. (C) Salidroside reduces 8-oxodG.
Protein extractions were prepared using low density BM
cells from mice described in panel A. Whole cell lysates
were then subjected to SDS-PAGE and immunoblotted
with Western Blot antibodies for 8-oxodG and ␤-actin.
(D) Repair kinetics. Low-density BM cells were pre-
treated with salidroside (250␮M), NAC (500␮M), or
MnTBAP (250␮M) for 2 hours, followed by H2O2 for
additional 2 hours, and then released for indicated time
intervals. Whole cell lysates were prepared and sub-
jected to SDS-PAGE and immunoblotted with antibodies
for 8-oxodG and ␤-actin.

Brca2⫺/⫺, DNA-PKc3A/3A, Fancd2⫺/⫺, and Xpc⫺/⫺ cells but having no salidroside in these MEFs (supplemental Figure 8A). We also found that
effect in Parp-1⫺/⫺ cells (Figure 5B). These results suggest that salidroside enhanced PARP-1 activity in human lymphoblasts (supple-
salidroside may specifically target PARP-1 in response to oxidative mental Figure 8B). To substantiate these observations, we performed
DNA damage. To test this notion, we used a flow-cytometric assay to immunoprecipitation to determine the stimulatory effect of salidroside
determine the effect of salidroside on the enzyme activity of PARP-1 in on PARP1 activity in vivo. We cotreated fresh-isolated BM cells with
BM LSK cells from WT or knockout mice deficient for each of the H2O2 and salidroside, and subjected the cell lysates to immunoprecipita-
5 repair pathways. We found that salidroside stimulated PARP-1 activity tion with PAR antibodies followed by Western blot with PARP1
in cells from all except Parp-1 knockout mice treated with or without antibodies. Consistent with the results obtained from the flow-
H2O2 (Figure 5C). We next determined whether salidroside stimulated cytometric assay, we observed enhanced PARP1 poly-ADP-ribosylation
PARP-1 activity in other cell types. We isolate mouse embryonic by salidroside in both stressed and unstressed cells (Figure 5D).
fibroblasts (MEFs) from WT mice and cotreated the cells with salidro- Together, these results indicate that salidroside reduces oxidative DNA
side and H2O2. We observed similar stimulation of PARP-1 activity by damage through stimulation of PARP-1 activity.
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4169

Figure 5. Salidroside stimulates PARP-1 activity. (A) Salidroside fails to reduce H2O2-induced DNA strand breaks in Parp-1⫺/⫺ cells. Lin⫺ cells were isolated from Brca2⫺/⫺,
DNA-PKcs3A/3A, Fancd2⫺/⫺, Parp-1⫺/⫺, or Xpc⫺/⫺ mice. Cells were treated with or without salidroside, NAC, or MnTBAP in the presence of H2O2, and subjected to Comet
Assay. The mean tail moment of H2O2-treated WT sample is expressed as 100%. For each sample, 50 cells were scored from random sampling. (B) Salidroside fails to reduce
H2O2-induced 8-oxodG in Parp-1⫺/⫺ cells. Protein lysates were prepared from low-density BM cells treated as described in panel A. Whole cell lysates were subjected to
SDS-PAGE and immunoblotted with antibodies against 8-oxodG and ␤-actin. (C) Salidroside stimulates Parp-1 activity. Low-density BM cells isolated from mice described in
panel A were treated with or without H2O2 and salidroside, and BM cells were isolated and subjected to flow cytometry analysis for Parp-1 activity in the LSK population using
antibody against PARP-1. (D) Salidroside activates Parp-1 in vivo. Low-density BM cells were isolated from WT mice, and treated with or without SD and H2O2 for 2 hours.
Whole cell lysates were subjected to immunoprecipitation using PAR antibody. Precipitated samples were resolved by SDS-PAGE and blotted with antibody specific for
PARP-1.

Inhibition of PARP-1 abrogates the effect of salidroside on HSC which salidroside increased quiescent HSC pool significantly,
maintenance salidroside had no discernible effect on oxidative stress–induced
We next sought to test whether PARP-1 activation by salidroside HSC cycling in Parp-1⫺/⫺ mice (Figure 6A). To substantiate this
protected quiescent HSCs from oxidative stress–induced cycling. finding, we made use of a specific small molecule inhibitor of
We took 2 approaches: genetic ablation of the Parp-1 gene and PARP-1, 8-hydroxy-2-methylquinazolinone (NU1025).45 Consis-
pharmacologic inhibition of PARP-1 enzymatic activity in oxida- tent with PARP-1 knockout, NU1025 treatment abolished salidroside-
tive stressed mice to further elucidate the specific action of mediated increase in quiescent HSC frequency in stressed mice
salidroside in PARP-1 stimulation. Compared with WT mice in (Figure 6B). As expected, both salidroside and NU1025 had no
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

4170 LI et al BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18

Figure 6. Inhibition of PARP-1 abrogates the effect of salidroside on HSC maintenance. (A) Deletion of Parp-1 abolishes salidroside-mediated increase in quiescent HSC
frequency in stressed mice. Parp1⫺/⫺ mice as well as their WT littermates were pretreated with or without salidroside (75 ␮g/g body weight) followed by H2O2 (0.25 ␮mol/g body
weight). BM cells were subjected to flow cytometry analysis for quiescent HSC (G0 phase). Results are means ⫾ SD of 3 independent experiments (n ⫽ 9 per group).
(B) NU1025 treatment abolishes salidroside-mediated increase in quiescent HSC frequency in stressed mice. H2O2-treated (0.25 ␮mol/g body weight) WT mice were treated
with or without salidroside (SD; 75␮g/g body weight) and NU1025 (25 mg/kg body weight). BM cells were subjected to flow cytometry analysis for quiescent HSC. Results are
means ⫾ SD of 3 independent experiments (n ⫽ 9 per group). (C) The maintenance of HSC quiescence by salidroside requires Parp-1. 1000 LSK cells from WT or Parp1⫺/⫺
mice were injected to lethally irradiated WT recipients. Recipient mice were then subjected to salidroside and H2O2 treatments. Cycling donor-derived (CD45.2⫹) cells were
assessed by Flow Cytometry analysis by pyronin Y staining. Representative images (top) and quantifications (bottom) were shown. Results are means ⫾ SD of 3 independent
experiments (n ⫽ 9 per group). (D) The enhancing effect of salidroside on the long-term repopulation abilities of stressed HSCs requires Parp1. Cells from primary recipients
described in panel C were used for second transplantation by injecting 10 million whole BM cells to lethally irradiated WT recipients. Donor-derived cells were determined by
flow cytometry analysis. Representative images (top) and quantifications (bottom) were shown. Results are means ⫾ SD of 3 independent experiments (n ⫽ 9 per group).

effect on oxidative stress–induced HSC cycling in Parp-1⫺/⫺ mice To provide functional evidence that HSC maintenance by
(supplemental Figure 9). Collectively, these results provided con- salidroside under oxidative stress requires PARP-1, we assessed the
vincing evidence to show that protection of quiescent HSCs by ability of salidroside to preserve the self-renewal and hematopoi-
salidroside from oxidative stress–induced cycling requires PARP-1. etic reconstitution capacity of HSCs in the context of PARP-1
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4171

function. We transplanted BM LSK cells from WT or Parp-1⫺/⫺ the HSC population, in mice deficient in several oxidative damage
mice into lethally irradiated WT recipient mice, which were treated response and repair pathways.15,46,47 It is in this context that we
with salidroside and H2O2 at 4 months after transplantation. On identified salidroside as a potent small molecule that prevents
injection with H2O2, we observed that cell cycle progression exhaustion of HSCs from oxidative stress–induced uncontrolled
induced by oxidative stress was not prevented by salidroside in cell-cycle entry and excessive proliferation.
donor-derived HSCs deficient for Parp-1 (Figure 6C), which Another interesting finding of our study is that unlike other
suggests that the action of salidroside in maintaining quiescent antioxidants, such as NAC, MnTBAP, and quercetin that function
HSCs requires PARP-1. To determine whether the effect of as free radical scavengers, salidroside does not prevent the
salidroside on the long-term repopulation abilities of HSCs also production of ROS but acts to eliminate oxidative DNA damage in
required PARP-1, we did serial transplantation experiments by BM cells enriched for HSCs, as demonstrated by a robust reduction
transplanting BM cells from the salidroside and H2O2-treated of DNA-stranded breaks and expression of ␥-H2AX and 8-oxodG.
primary recipients of Parp-1⫹/⫹ or Parp-1⫺/⫺ BM cells into More importantly, we present evidence of distinct kinetics of
lethally irradiated WT secondary recipients. Four months after 8-oxodG clearance by salidroside compared with those by the ROS
transplantation, we obtained peripheral blood from the secondary scavenger NAC or MnTBAP. That is, the effect of salidroside on
recipient mice and analyzed hematopoietic reconstitution derived repair kinetics consistently lags those of NAC and MnTBAP. We
from donor (CD45.2⫹) cells. Our analysis indicated that salidroside propose that salidroside acts directly in the process of oxidative
failed to enhance donor-derived hematopoiesis in recipients trans- DNA damage repair. Oxidative DNA damage by increased ROS
planted with Parp-1⫺/⫺ HSCs (Figure 6D). These findings suggest accumulation in HSCs has been documented in several recent
that the effect of salidroside on the maintenance of quiescent HSCs studies using age-related murine HSCs15,16 and more recently
under oxidative stress requires PARP-1. human HSCs.13 Although these studies suggest a correlation
between oxidative DNA damage and impaired HSC maintenance,
little is known about the mechanism by which oxidative DNA
damage influences the function of HSCs in disease states, such as
Discussion BM failure and leukemia, which are commonly accompanied by
defective DNA repair. Our results suggest that oxidative DNA
Oxidative stress has been linked to aging and cancer as well as
damage drives HSCs into cell cycling, which may be a prerequisite
other major human health problems. A promising strategy for
for DNA repair to proceed. Inefficient repair of oxidative DNA
preventing oxidative damage to the cell is to use readily available
damage in these disease conditions may prolong cell cycling,
natural compounds isolated from vegetables, fruits, and herbs.
which leads to HSC exhaustion. In addition, patients with BM
Many of the phytochemicals have already been identified to have
failure and leukemia often have an extremely low number of HSCs,
chemopreventive potential, capable of intervening in tumorigen-
which critically limits the success of stem cell therapy. Therefore,
esis. We investigated the potential of the natural antioxidant
salidroside as a new chemopreventive and antioxidant agent that one of the key issues in these disease states is to identify conditions
has therapeutic value for patients with BM failure or leukemia. to increase the number of HSCs under stressful conditions, either
Salidroside has been reported to have antiaging, anticancer, anti- in vivo or during ex vivo growth cultures. We show here that
inflammatory, and antioxidative functions.20-23 In this study, we salidroside promotes HSC self-renewal and preservation under
demonstrate that salidroside promotes the maintenance of mouse oxidative stress and thus may be valuable for HSC expansion.
HSCs under oxidative stress. There are several findings that Although further studies are required to reveal the underlying
highlight the significance of our study: first, salidroside prevents molecular mechanisms, it is intriguing that salidroside promotes
quiescent HSCs from oxidative stress–induced activation; second, HSC maintenance by reducing oxidative DNA damage through
salidroside does not function as a ROS scavenger but enhances stimulation of PARP-1 activity. Studies conducted using mutant
oxidative DNA damage repair through a mechanism involving mice have underscored the importance of DNA repair pathways in
stimulation of repair enzyme PARP-1 activity; and third, PARP-1 maintaining the functionality of HSCs. A number of studies
activation by salidroside protects quiescent HSCs from oxidative demonstrate hematopoietic defects in mice deficient for HR, NHEJ,
stress–induced cycling and repopulating defect. Thus, salidroside NER, and telomere maintenance pathways.15,16 In the context of
plays a significant role in inhibiting oxidative DNA damage; and oxidative DNA damage response/repair in HSC maintenance,
thereby regulates the homeostasis of HSCs. recent studies in mice deficient for the ATM kinase or the Foxo
Quiescence has been postulated to prevent HSC exhaustion.3 In transcription factors showed impaired HSC function as a result of
the BM, HSCs are kept in a low proliferative, relatively quiescent increased accumulation of ROS in HSCs.47-50 Cumulatively, these
state within the BM microenvironment termed niches. Whereas studies suggest that genomic DNA integrity is a limiting factor in
numerous molecular factors that contribute to quiescence exist in the maintenance of functional HSCs. Indeed we observed defective
the HSCs and the BM niche, HSCs are inevitably exposed to stress, long-term hematopoietic repopulation by oxidative stressed HSCs
such as accumulation of ROS and DNA damage, which can drive from mice lacking the BER enzyme Parp-1. Furthermore, our
HSCs into uncontrolled cell-cycle entry and excessive prolifera- results indicate that salidroside reduces oxidative DNA damage
tion. Indeed, we found that on challenge with oxidative stress, through stimulation of PARP-1 activity, and that inhibition of
quiescent HSCs were recruited into cell cycle, evidenced by a PARP-1 abrogates the beneficial effect of salidroside on HSC
marked decrease in the number of quiescent and an increase in the maintenance. These data suggest that stimulation of PARP-1
number of cycling CD34⫺ LSK cells in H2O2-treated mice activity by salidroside could account for the accelerated repair of
compared with the unstressed animals. We confirmed this finding the oxidative DNA damage and enhanced repopulating capacity of
by showing elevated BrdU-positive CD34⫺ LSK cells in H2O2- the stressed HSCs.
treated mice. Functionally, we demonstrate that oxidative stress Another interesting finding of this study is the observation that
impaired long-term repopulation abilities of HSCs. These results salidroside was able to maintain the oxidative stressed HSCs from
are consistent with the recent reports describing functional exhaus- WT but not Parp-1⫺/⫺ mice in quiescent state in transplanted mice.
tion of HSCs, because of uncontrolled accumulation of ROS within Cell-cycle analysis shows that on challenge with oxidative stress,
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

4172 LI et al BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18

donor-derived HSCs exited quiescence and underwent cycling. Texas Southwestern Medical Center at Dallas) for DNA-PKcs⫹/3A
However, these proliferating HSCs were able to regain quiescence mice, Dr Liang Li for technical assistance, and the Comprehensive
in recipient mice conditioned with salidroside. This phenomenon Mouse and Cancer Core of the Cincinnati Children’s Research
was probably because of Parp-1 stimulation by salidroside, because Foundation (Cincinnati Children’s Hospital Medical Center) for
donor-derived HSCs deficient for Parp-1 failed to regain quiescent BM transplantation service.
in salidroside-conditioned recipient mice. These results might This work was supported by a Visiting Scholarship from South
suggest that cell-cycle status of HSCs could reversibly change from China Normal University (X.L.) and partially by National Institutes
oxidative stress–induced activation back to quiescence after repair of Health (NIH) grants R01 HL076712 and R01 CA157537. Q.P. is
of the oxidative DNA damage. This finding raises important
supported by a Leukemia & Lymphoma Scholar award. W.D. is
questions as to whether activation is required for efficient repair of
supported by an NIH T32 training grant.
oxidative DNA damage in the stressed HSCs and whether elimina-
tion of the DNA damage is sufficient for the activated HSCs to
return to quiescent state. Although further evidence needs to be
provided, it is tempting to speculate that reversibility between
quiescence and activation may be a physiologic function of Authorship
activated HSCs at the interface between damage repair and
Contribution: X.L. designed research and performed research and
reestablishment of homeostasis. In this context, it is noteworthy
that recent studies in both mice and human show that HSCs can analyzed data; J.S. performed research; Q.P. designed research and
reversibly switch between dormancy and self-renewal at the analyzed data; and W.D. designed research, analyzed data, and
interface between homeostasis and repair.12,13 wrote the paper.
Conflict-of-interest disclosure: The authors declare no compet-
ing financial interests.
Acknowledgments Correspondence: Wei Du, Division of Experimental Hematol-
ogy and Cancer Biology, Cincinnati Children’s Hospital Medical
The authors thank Dr Markus Grompe (Oregon Health & Science Center, 3333 Burnet Ave, Cincinnati, OH 45229; e-mail:
University) for Fancd2⫾ mice, Benjamin Chen (University of wei.du@cchmc.org.

References
1. Orkin SH, Zon LI. Hematopoiesis: an evolving 14. Fruehauf JP, Meyskens FL Jr. Reactive oxygen Wasiutyński A, et al. The influence of Rhodiola
paradigm for stem cell biology. Cell. 2008;132(4): species: a breath of life or death? Clin Cancer quadrifida 50% hydro-alcoholic extract and
631-644. Res. 2007;13(3):789-794. salidroside on tumor-induced angiogenesis in
2. Morrison SJ, Uchida N, Weissman IL. The biology 15. Rossi DJ, Bryder D, Seita J, Nussenzweig A, mice. Pol J Vet Sci. 2008;11(2):97-104.
of hematopoietic stem cells. Annu Rev Cell Dev Hoeijmakers J, Weissman IL. Deficiencies in 24. Qian EW, Ge DT, Kong SK. Salidroside promotes
Biol. 1995;11:35-71. DNA damage repair limit the function of haemato- erythropoiesis and protects erythroblasts against
3. Orford KW, Scadden DT. Deconstructing stem poietic stem cells with age. Nature. 2007; oxidative stress by up-regulating glutathione per-
cell self-renewal: genetic insights into cell-cycle 447(7145):725-729. oxidase and thioredoxin. J Ethnopharmacol.
regulation. Nat Rev Genet. 2008;9(2):115-128. 16. Nijnik A, Woodbine L, Marchetti C, et al. DNA re- 2011;133(2):308-314.
4. Wilson A, Trumpp A. Bone-marrow hematopoi- pair is limiting for haematopoietic stem cells dur- 25. Zhang S, Yajima H, Huynh H, et al. Congenital
etic-stem-cell niches. Nat Rev Immunol. 2006; ing ageing. Nature. 2007;447(7145):686-690. bone marrow failure in DNA-PKcs mutant mice
6(2):93-106. 17. Spasov AA, Wikman GK, Mandrikov VB, Mi- associated with deficiencies in DNA repair. J Cell
5. Cheng T, Rodrigues N, Shen H, et al. Hematopoi- ronova IA, Neumoin VV. A double-blind, placebo- Biol. 2011;193(2):295-305.
etic stem cell quiescence maintained by p21cip1/ controlled pilot study of the stimulating and adap- 26. Jonkers J, Meuwissen R, van der Gulden H,
waf1. Science. 2000;287(5459):1804-1808. togenic effect of Rhodiola rosea SHR-5 extract on Peterse H, van der Valk M, Berns A. Synergistic
the fatigue of students caused by stress during an tumor suppressor activity of BRCA2 and p53 in a
6. Hock H, Hamblen MJ, Rooke HM, et al. Gfi-1 re- examination period with a repeated low-dose
stricts proliferation and preserves functional in- conditional mouse model for breast cancer. Nat
regimen. Phytomedicine. 2000;7(2):85-89. Genet. 2001;29(4):418-425.
tegrity of haematopoietic stem cells. Nature.
2004;431(7011):1002-1007. 18. Darbinyan V, Kteyan A, Panossian A, Gabrielian E,
27. Ventura A, Kirsch DG, McLaughlin ME, et al. Res-
Wikman G, Wagner H. Rhodiola rosea in stress in-
7. Du W, Adam Z, Rani R, Zhang X, Pang Q. Oxida- toration of p53 function leads to tumour regres-
duced fatigue-a double blind cross-over study of a
tive stress in Fanconi anemia hematopoiesis and sion in vivo. Nature. 2007;445(7128):661-665.
standardized extract SHR-5 with a repeated low-
disease progression. Antioxid Redox Signal. dose regimen on the mental performance of healthy 28. Kunzmann A, Liu D, Annett K, et al. Flow-
2008;10(11):1909-1921. physicians during night duty. Phytomedicine. 2000; cytometric assessment of cellular poly(ADP-ribo-
8. Warren LA, Rossi DJ. Stem cells and aging in the 7(5):365-371. syl)ation capacity in peripheral blood lympho-
hematopoietic system. Mech Ageing Dev. 2009; cytes. Immun Ageing. 2006;3:8.
19. Shevtsov VA, Shevtsov VA, Zholus BI, et al. A
130(1-2):46-53. randomized trial of two different doses of a 29. Osawa M, Hanada K, Hamada H, Nakauchi H.
9. Klaunig JE, Kamendulis LM. The role of oxidative SHR-5 Rhodiola rosea extract versus placebo Long-term lymphohematopoietic reconstitution by
stress in carcinogenesis. Annu Rev Pharmacol and control of capacity for mental work. Phyto- a single CD34-low/negative hematopoietic stem
Toxicol. 2004;44:239-267. medicine. 2003;10(2-3):95-105. cell. Science. 1996;273(5272):242-245.
10. Pervaiz S, Taneja R, Ghaffari S. Oxidative stress 20. Kanupriya Prasad D, Sai Ram M, et al. Cytopro- 30. Christensen JL, Weissman IL. Flk-2 is a marker in
regulation of stem and progenitor cells. Antioxid tective and antioxidant activity of Rhodiola imbri- hematopoietic stem cell differentiation: a simple
Redox Signal. 2009;11(11):2777-2789. cata against tert-butyl hydroperoxide induced oxi- method to isolate longterm stem cells. Proc Natl
11. Eliasson P, Jönsson JI. The hematopoietic stem dative injury in U-937 human macrophages. Mol Acad Sci U S A. 2001;98(25):14541-14546.
cell niche: low in oxygen but a nice place to be. Cell Biochem. 2005;275(1-2):1-6.
31. Kiel MJ, Yilmaz OH, Iwashita T, Terhorst C,
J Cell Physiol. 2010;222(1):17-22. 21. Yu P, Hu C, Meehan EJ, Chen L. X-ray crystal Morrison SJ. SLAM family receptors distinguish
12. Wilson A, Laurenti E, Oser G, et al. Hematopoi- structure and antioxidant activity of salidroside, a hematopoietic stem and progenitor cells and re-
etic stem cells reversibly switch from dormancy to phenylethanoid glycoside. Chem Biodivers. 2007; veal endothelial niches for stem cells. Cell. 2005;
self-renewal during homeostasis and repair. Cell. 4(3):508-513. 121(7):1109-1121.
2008;135(6):1118-1129. 22. Wang H, Ding Y, Zhou J, Sun X, Wang S. The in 32. Yilmaz OH, Kiel MJ, Morrison SJ. SLAM family
13. Yahata T, Takanashi T, Muguruma Y, et al. Accu- vitro and in vivo antiviral effects of salidroside markers are conserved among hematopoietic
mulation of oxidative DNA damage restricts the from Rhodiola rosea L. against coxsackievirus stem cells from old and reconstituted mice and
self-renewal capacity of human hematopoietic B3. Phytomedicine. 2009;16(2-3):146-155. markedly increase their purity. Blood. 2006;
stem cells. Blood. 2011;118(11):2941-2950. 23. Skopińska-Rózewska E, Malinowski M, 107(3):924-930.
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

BLOOD, 3 MAY 2012 䡠 VOLUME 119, NUMBER 18 SALIDROSIDE STIMULATES PARP-1 ACTIVITY 4173

33. Lacorazza HD, Yamada T, Liu Y, et al. The tran- 39. Shuck SC, Short EA, Turchi JJ. Eukaryotic nucle- killing of BRCA2-deficient tumours with inhibitors
scription factor MEF/ELF4 regulates the quies- otide excision repair: from understanding mecha- of poly(ADP-ribose) polymerase. Nature. 2005;
cence of primitive hematopoietic cells. Cancer nisms to influencing biology. Cell Res. 2008; 434(7035):913-917.
Cell. 2006;9(3):175-187. 18(1):64-72.
46. Naka K, Hirao A. Maintenance of genomic integ-
34. Melov S, Schneider JA, Day BJ, et al. A novel 40. Svilar D, Goellner EM, Almeida KH, Sobol RW. rity in hematopoietic stem cells. Int J Hematol.
neurologic phenotype in mice lacking mitochon- Base excision repair and lesion-dependent sub- 2011;93(4):434-439.
drial manganese superoxide dismutase. Nature pathways for repair of oxidative DNA damage.
Genet. 1998;18(2):159-163. Antioxid Redox Signal. 2011;14(12):2491-2507. 47. Ito K, Hirao A, Arai F, et al. Reactive oxygen spe-
cies act through p38 MAPK to limit the lifespan of
35. Fairbairn DW, Olive PL, O’Neill KL. The comet 41. Helleday T. Homologous recombination in cancer
hematopoietic stem cells. Nat Med. 2006;12(4):
assay: a comprehensive review. Mutat Res. development, treatment and development of drug
446-451.
1995;339(1):37-59. resistance. Carcinogenesis. 2010;31(6):955-960.
42. Dobbs TA, Tainer JA, Lees-Miller SP. A structural 48. Miyamoto K, Araki KY, Naka K, et al. Foxo3a is
36. Olive PL, Banáth JP, Durand RE. Heterogeneity
model for regulation of NHEJ by DNA-PKcs auto- essential for maintenance of the hematopoietic
in radiation-induced DNA damage and repair in
phosphorylation. DNA Repair (Amst). 2010;9(12): stem cell pool. Cell Stem Cell. 2007;1(1):101-112.
tumor and normal cells measured using the
“comet” assay. Radiat Res. 1990;122(1):86-94. 1307-1314. 49. Tothova Z, Kollipara R, Huntly BJ, et al. FoxOs
37. Celeste A, Difilippantonio S, Difilippantonio MJ, et 43. van Hoffen A, Balajee AS, van Zeeland AA, are critical mediators of hematopoietic stem cell
al. H2AX haploinsufficiency modifies genomic Mullenders LH. Nucleotide excision repair and its resistance to physiologic oxidative stress. Cell.
stability and tumor susceptibility. Cell. 2003; interplay with transcription. Toxicology. 2003; 2007;128(2):325-339.
114(3):371-383. 193(1-2):79-90. 50. Yalcin S, Zhang X, Luciano JP, et al. Foxo3 Is es-
38. Goodarzi AA, Jeggo P, Lobrich M. The influence 44. Moldovan GL, D’Andrea AD. How the fanconi sential for the regulation of ataxia telangiectasia
of heterochromatin on DNA double strand break anemia pathway guards the genome. Annu Rev mutated and oxidative stress–mediated homeo-
repair: Getting the strong, silent type to relax. Genet. 2009;43:223-249. stasis of hematopoietic stem cells. J Biol Chem.
DNA Repair (Amst). 2010;9(12):1273-1282. 45. Bryant HE, Schultz N, Thomas HD, et al. Specific 2008;283(37):25692-25705.
From www.bloodjournal.org by guest on September 29, 2018. For personal use only.

2012 119: 4162-4173


doi:10.1182/blood-2011-10-387332 originally published
online March 16, 2012

Salidroside stimulates DNA repair enzyme Parp-1 activity in mouse HSC


maintenance
Xue Li, Jared Sipple, Qishen Pang and Wei Du

Updated information and services can be found at:


http://www.bloodjournal.org/content/119/18/4162.full.html
Articles on similar topics can be found in the following Blood collections
Hematopoiesis and Stem Cells (3524 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://www.bloodjournal.org/site/misc/rights.xhtml#repub_requests

Information about ordering reprints may be found online at:


http://www.bloodjournal.org/site/misc/rights.xhtml#reprints

Information about subscriptions and ASH membership may be found online at:
http://www.bloodjournal.org/site/subscriptions/index.xhtml

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly by the American Society
of Hematology, 2021 L St, NW, Suite 900, Washington DC 20036.
Copyright 2011 by The American Society of Hematology; all rights reserved.

You might also like