ArticlePDF Available

AMD3100 improves ovariectomy-induced osteoporosis in mice by facilitating mobilization of hematopoietic stem/progenitor cells

Authors:

Abstract and Figures

Inhibition of an increase of osteoclasts has become the most important treatment for osteoporosis. The CXCR4 antagonist, AMD3100, plays an important role in mobilization of osteoclast precursors within bone marrow (BM). However, the actual therapeutic impact of AMD3100 in osteoporosis has not yet been ascertained. Here we demonstrate the therapeutic effect of AMD3100 in treatment of ovariectomy-induced osteoporosis in mice. We found that treatment with AMD3100 resulted in direct induction of release of SDF-1 from BM to blood and mobilization of hematopoietic stem/progenitor cells (HSPCs) in an osteoporosis model. AMD3100 prevented bone density loss after ovariectomy by mobilization of HSPCs, suggesting a therapeutic strategy to reduce the number of osteoclasts on bone surfaces. These findings support the hypothesis that treatment with AMD3100 can result in efficient mobilization of HSPCs into blood through direct blockade of the SDF-1/CXCR4 interaction in BM and can be considered as a potential new therapeutic intervention for osteoporosis.
Content may be subject to copyright.
BMB
Re
p
orts
BMB Rep. 2014; 47(8): 439-444
www.bmbreports.org
*Corresponding author. Tel: +82-53-420-4815; Fax: +82-53-424-
3349; E-mail: jsbae@knu.ac.kr
#
These authors contributed equally to this work.
http://dx.doi.org/10.5483/BMBRep.2014.47.8.159
Received 10 July 2013, Revised 22 July 2013,
Accepted 19 November 2013
Keywords: AMD3100, Hematopoietic stem/Progenitor cell,
Mobilization, Osteoclast, Osteoporosis
ISSN: 1976-670X (electronic edition)
Copyright 2014 by the The Korean Society for Biochemistry and Molecular Biology
This is an open-access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/li-
censes/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
AMD3100 improves ovariectomy-induced osteoporosis in mice by
facilitating mobilization of hematopoietic stem/progenitor cells
Jin Young Im
1,2,3,#
, Woo-Kie Min
4,#
, Min Hee Park
1,2,3
, NamOh Kim
1,2,3
, Jong Kil Lee
1,2,3
, Hee Kyung Jin
1,5
, Je-Yong Choi
3,6
,
Shin-Yoon Kim
4
& Jae-sung Bae
1,2,3,
*
1
Stem Cell Neuroplasticity Research Group, Kyungpook National University,
2
Department of Physiology, Cell and Matrix Research
Institute, School of Medicine, Kyungpook National University,
3
Department of Biomedical Science, BK21 Plus KNU Biomedical
Convergence Program, Kyungpook National University, Daegu 700-842,
4
Department of Orthopaedic Surgery, Kyungpook National
University Hospital, Daegu 700-721,
5
Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National
University, Daegu 700-721,
6
Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu
702-701, Korea
Inhibition of an increase of osteoclasts has become the most
important treatment for osteoporosis. The CXCR4 antagonist,
AMD3100, plays an important role in the mobilization of
osteoclast precursors within bone marrow (BM). However, the
actual therapeutic impact of AMD3100 in osteoporosis has not
yet been ascertained. Here we demonstrate the therapeutic
effect of AMD3100 in the treatment of ovariectomy-induced
osteoporosis in mice. We found that treatment with AMD3100
resulted in direct induction of release of SDF-1 from BM to
blood and mobilization of hematopoietic stem/progenitor cells
(HSPCs) in an osteoporosis model. AMD3100 prevented bone
density loss after ovariectomy by mobilization of HSPCs,
suggesting a therapeutic strategy to reduce the number of
osteoclasts on bone surfaces. These findings support the
hypothesis that treatment with AMD3100 can result in efficient
mobilization of HSPCs into blood through direct blockade of
the SDF-1/CXCR4 interaction in BM and can be considered as
a potential new therapeutic intervention for osteoporosis.
[BMB Reports 2014; 47(8): 439-444]
INTRODUCTION
Bone remodeling is required for balance between bone-form-
ing osteoblasts and bone resorbing osteoclasts (1, 2). Destruc-
tion of this balance may lead to the development of bone dis-
ease, such as osteoporosis, which is characterized clinically by
low bone density and a consequent increase in the risk of frac-
ture, and pathologically by a decrease in bone mineral density
(BMD) and an increase in osteoclastic bone resorption (3, 4).
 Osteoclasts are somatic cells that differentiate from hema-
topoietic stem/progenitor cells (HSPCs) (5). Previous study has
shown that sphingosine-1-phosphate (S1P), a lipid mediator
enhanced in blood, regulates the recruitment of osteoclast pre-
cursors from bone marrow (BM) into blood. Treatment with
the S1P agonist resulted in relief of bone loss through the re-
duction of osteoclast deposition onto bone surfaces (6). This
study led us to speculate that a more effective approach to the
recruitment of endogenous HSPCs into blood in patients with
osteoporosis could be via an important therapy involving the
decline of osteoclasts differentiated from HSPCs in BM.
 Chemokine stromal cell-derived factor-1 (SDF-1, also termed
CXCL12), the most powerful chemoattractant of both human
and murine HSPCs, and its major receptor, CXCR4, are key
players in HSPC mobilization (6, 7). The number of circulating
stem cells can be significantly increased by mobilizing them
from the BM to the peripheral blood with cytokines, chemo-
kines, or small molecule inhibitors (8). Due to its potency, gran-
ulocyte colony-stimulating factor (G-CSF) is currently the most
widely used agent for induction of HSPC mobilization (9).
However, mobilization of HSPCs by G-CSF occurs through sup-
pression of osteoblast cells and decreasing expression of SDF-1
on osteoblasts, resulting in major impairment of HSC retention
in BM and reduced bone formation (9, 10). It is also associated
with a clinically significant osteopenia, characterized by an in-
crease in osteoclast activity and a decrease in BMD (11-13). In
order to overcome these obstacles, we used AMD3100, which
inhibits SDF-1-mediated mobilization through direct blockade
of the chemokine binding to its receptor, CXCR4, without im-
pairment of osteoblast function (10, 14, 15) in a model of post-
menopausal osteoporosis. AMD3100 has recently been re-
ported to enhance recruitment of CXCR4-dependent HSPCs by
inducing an increase in the level of SDF-1 in blood secreted
HSPCs mobilization by AMD3100 in osteoporosis
J
in Young Im, et al.
440 BMB Reports http://bmbreports.org
Fig. 1. SDF-1 level increased in blood and did not affect
osteoblasts. (A) Timeline of the experimental design of this study.
Twelve-week-old C57BL/6 mice (n = 40) were divided into fou
r
groups (Sham/PBS group [n = 10]; Sham/AMD3100 group [n =
10]; OVX/PBS group [n = 10]; OVX/AMD3100 group [n = 10]).
(B, C) Steady-state homeostasis fold change in levels of SDF-1
was evaluated in mouse plasma and BM supernatants after admin-
istration of PBS or AMD3100. AMD3100 induced release of func-
tional SDF-1 to plasma. (D) Expression of Osteocalcin, PTHR1,
Osterix, and Runx2 was analyzed by Real-Time PCR from BM o
f
OVX and sham mice. Data represent mean ± SEM (Student’s
t-test. n = 4-5 per group). *P 0.05 compared with AMD3100
treated mice or matched control.
Fig. 2. AMD3100 mobilized HSPCs to blood in an OVX model. (A)
We analyzed the effects of AMD3100 on HSPCs mobilization by
CFU assay in blood. (B) Flow cytometry analysis of Lin
-
Sca-1
+
c-kit
+
cells in BM of PBS-treated OVX and AMD3100-treated OVX mice.
Data represent mean ± SEM (Student’s t-test. n = 4-5 per group). *P
0.05 compared with AMD3100 treated mice or matched control.
from BM stromal cells (15, 16).
 Based on these concepts and findings, this study was de-
signed to determine whether AMD3100 has the capacity to
mobilize HSPCs, resulting in a decrease in the number of os-
teoclasts in BM. If so, use of this molecule might have a benefi-
cial effect on ovariectomy (OVX)-induced osteoporosis in mice.
RESULTS
To determine the effect of AMD3100 on OVX mice, we in-
jected OVX mice with AMD3100. The injection protocol is de-
scribed in Fig. 1A. Recent studies have demonstrated direct in-
duction of release of SDF-1 into the circulation and an in-
crease in mobilization of HSPCs by treatment with AMD3100
(14). Therefore, we performed an analysis in order to de-
termine whether treatment with AMD3100 could induce re-
lease of SDF-1 and whether it is associated with recruitment of
HSPCs. Consistent with previous results (14), we found that
levels of functional SDF-1 decreased in BM and increased in
plasma of the AMD3100-treated group (Fig. 1B and C). These
results indicate that treatment with AMD3100 resulted in an
increase in SDF-1 in blood and might affect HSPCs mobi-
lization from BM to blood. Next, we investigated osteoblast
lineage-specific genes, including Osteocalcin, PTHR1, Osterix,
and Runx2 in order to determine whether AMD3100 has an
effect on osteoblast. However, no significant differences in the
levels of osteoblast lineage-specific genes were observed be-
tween the groups (Fig. 1D). These results confirmed that
AMD3100 induced an increase in levels of SDF-1 in blood
and did not alter osteoblasts of OVX mice.
 To determine whether AMD3100 can mobilize HSPCs from
BM in OVX mice, we administered AMD3100 or phos-
phate-buffered saline (PBS) to OVX for 21 days. As shown in
Fig. 2A, the number of hematopoietic colony-forming unit
(CFU) cells showed a significant increase in AMD3100-treated
OVX mice compared to PBS-treated OVX mice. Flow cyto-
metric analysis was performed for evaluation of the percentage
of HSPCs in BM. As expected, the percentage of Lin
-
Sca-1
+
c-Kit
+
(LSK) cells showed a decrease in AMD3100-treated
OVX mice, compared with PBS-treated OVX mice in BM, al-
though this did not reach statistical significance (Fig. 2B).
These data indicate that AMD3100 induces mobilization of
HSPCs from BM to blood in a model of OVX-induced
osteoporosis.
 To investigate the effect of AMD3100 on different bone pa-
HSPCs mobilization by AMD3100 in osteoporosis
Jin Young Im, et al.
441http://bmbreports.org BMB Reports
Fig. 3. AMD3100 relieves OVX-induced bone loss through reduction of osteoclast number onto the bone surface. (A) Representative mi-
cro-CT images of the distal femurs in each group are shown. (B) Each graph represents BMD, BVF, BMC, TMD, Tb.N., Tb.Sp., Cr.BMD
and Cr.BMC in total analysis of cortical and trabecular bone. (C) The effects of AMD3100 on the expression of genes associated with os-
teoclast differentiation by quantitative real-time PCR. Total RNA was extracted from BM cultures treated with or without RANKL in the pres-
ence or absence of 25 μg/ml of AMD3100 for 3 days. Data represent mean ± SEM (Anova, Tukey’s HSD test. n = 3 per group). *P
0.05 compared with AMD3100-treated media or matched control. (D) Representative pictures showing osteoclasts in the trabecular region o
f
OVX mice. Reduction of multi-nucleated TRAP
+
osteoclasts was detected in BM. Arrowheads indicate active TRAP
+
osteoclasts stained in
red (original magnification, ×20, Scale bar 100 μm). (E) Histogram representing the osteoclast number/bone surface [N.Oc/BS (/mm)] and (F)
the osteoblast number/bone surface [N.Ob/BS (/mm)]. Data indicate mean ± SEM (Student’s t-test. n= 4-5 per group). *P 0.05 compared
with PBS-treated OVX mice.
rameters, we performed micro-CT analysis for the assessment
of BMD, bone mineral content (BMC), bone volume fraction
(BVF), tissue mineral density (TMD), trabecular number
(Tb.N.), trabecular separation (Tb.Sp.), cortical bone mineral
density (Cr.BMD) and cortical bone mineral content (Cr.BMC).
Increased bone density was observed in AMD3100-treated
OVX mice compared with PBS-treated OVX mice (Fig. 3A).
BMD showed an increase in AMD3100-treated OVX mice
compared with PBS-treated OVX mice; however, BVF, BMC,
TMD, Tb.N., Tb.Sp., Cr.BMD and Cr.BMC were similar be-
tween the groups (Fig. 3B).
  Osteoporosis is likely the result of osteoclastic deposition
rather than osteoblastic defects (3, 4). Therefore, in order to
determine whether mobilization of AMD3100 affected osteo-
clast differentiation, we first examined HSPCs differentiation
into mature functional osteoclasts by treatment with
AMD3100 (17). According to the result, AMD3100 did not af-
fect osteoclast differentiation (Fig. 3C). We also performed tar-
trate resistant acid phosphatase (TRAP) staining for detection of
osteoclasts in the trabecular region of OVX mice. The number
and size of TRAP
+
active osteoclasts (black arrowhead)
showed a decrease in the trabecular region in AMD3100-treat-
ed OVX mice (Fig. 3D). Histomorphometric analysis was per-
formed for determination of the number of osteoclasts. A de-
crease in the number of osteoclasts was observed in
AMD3100-treated OVX mice compared with PBS-treated OVX
mice (Fig. 3E). Next, we performed H&E staining in order to
determine the effect of AMD3100 on the number of
osteoblasts. Our findings showed that osteoblast numbers did
not change significantly among the AMD treatment groups
(Fig. 3F). Taken together, the data presented here demonstrate
that treatment with AMD3100 induced mobilization of BM-de-
rived HSPCs into blood, leading to amelioration of bone loss
in a model of OVX-induced osteoporosis by reducing the num-
ber of osteoclasts attached to the bone surface.
DISCUSSION
Previous studies have demonstrated a unique role of
AMD3100 in the rapid mobilization of osteoclast precursors,
HSPCs from BM to blood (9, 10). However, the actual ther-
apeutic impact of AMD3100 on pathology of osteoporosis and
its mechanism of action have not been determined.
AMD3100, a small bicyclam derivative, selectively inhibits
binding of SDF-1 in osteoblasts to its receptor, CXCR4, in
HSPCs, resulting in rapid mobilization of HSPCs (14).
AMD3100-induced levels of SDF-1 can also induce mobi-
lization of HSPCs (18). The data presented here suggest that
HSPCs mobilization by AMD3100 in osteoporosis
J
in Young Im, et al.
442 BMB Reports http://bmbreports.org
AMD3100 can improve bone loss in an OVX mice model by
decreasing the number of osteoclasts differentiated from
BM-derived HSPCs.
 We found that administration of AMD3100 resulted in an
increase in the functional levels of SDF-1 in plasma compared
with BM. This result suggests that release of HSPCs from BM to
blood occurred in response to increasing levels of plasma
SDF-1 in blood following administration of AMD3100. In ad-
dition, no changes in the levels of osteoblast markers were de-
tected after treatment with AMD3100. These findings indicate
that mobilization of HSPCs by AMD3100 occurs through di-
rect blockade of CXCR4-mediated sensing of the SDF-1 che-
motactic gradient in BM and by promoting the release of
SDF-1 from BM stromal cells into the circulation, similar to
previous reports (10, 19).
 Notably, we demonstrated that the total number of CFUs in
blood was increased by treatment with AMD3100 in Sham
and OVX mice. We also confirmed that the LSK compartment
was decreased by administration of AMD3100 in BM of OVX
mice. Consistent with these results, micro-CT analyses con-
firmed an increase in bone thickness as a result of the in-
hibition of osteoclastic bone resorption. These results indicate
that AMD3100 prevented bone loss in OVX mice through re-
duction of osteoclast number onto bone surfaces. However, al-
though mobilization and levels of SDF-1 were increased in the
plasma of Sham mice, treatment with AMD3100 did not result
in significant change of BMD. This is because AMD3100 has
been shown to very rapidly mobilize HSPCs into blood, with-
out altering BM niche function, with restoration to normal
steady state within hours (9, 15). These results suggest that
AMD3100 had no effect on bone density under normal
steady-state BM environment not in osteoporosis.
 In summary, results of this study demonstrated that
AMD3100, a novel small-molecule antagonist of CXCR4, had
no influence on osteoblasts and directly targeted distribution
of the interaction of CXCL12/CXCR4, resulting in rapid mobi-
lization of HSPCs into the blood circulation and prevented
bone loss induced by osteoporosis. In this study, for the first
time, we link bone remodeling with the regulation of
AMD3100-induced mobilization of HPSCs and propose that
the delicate balance of osteoblasts and osteoclasts is also a ma-
jor regulator of hematopoiesis. Therefore, we suggest the po-
tential for use of AMD3100 as an alternative therapy for treat-
ment of osteoporosis.
MATERIALS AND METHODS
Animals and treatments
Mouse studies were approved by the Kyungpook National
University Institutional Animal Care and Use Committee
(IACUC). Female C57BL/6 mice were purchased from Jackson
Laboratory (Bar Harbor, ME). Mice were housed in an air-con-
ditioned room with a 12 hr light/dark cycle at a temperature of
22 ± 2
o
C and humidity of 45-65% and given free access to
food and tap water. Mice underwent either sham surgery or
OVX at 12 weeks of age and were sacrificed at 16 weeks of
age. One week after surgery, sham and OVX mice received an
intraperitoneal injection with AMD3100 (Sigma-Aldrich
#A5602, St. Louis, MO, Sigma Aldrich.com) (5 mg/kg/day) or
PBS for 21 days. At the end of treatment, the mice were sacri-
ficed, and blood samples were collected by cardiac puncture
for the CFU assay. Femora were removed, fixed with 4% paraf-
ormaldehyde in PBS solution (pH 7.4) for 16 hr, and then stor-
ed (4
o
C) at 80% ethanol for measurement of bone density.
Quantitative Real-Time PCR
RNA samples were extracted from whole BM cells of four in-
dividual animals per group and isolated from the cultured cells
using the RNeasy Mini kit (Qiagen, Hilden, Germany), and the
concentration was determined using a Nanodrop ND-1000
spectrophotometer. A total of 5 μg of each RNA was converted
to cDNA using the sprint RT complete-oligo (dT) 18 (Clontech,
MountainView, CA) according to the manufacturer’s guide.
The cDNA was quantified using the QuantiTect SYBR Green
PCR Kit (Qiagen). For each investigated transcript, a mixture of
the following reaction components was prepared to the in-
dicated end-concentration: forward primer (5 pM), reverse pri-
mer (5 pM), and QuantiTect SYBR Green PCR Master mix. The
10 μl master-mix was added to a 0.1 ml tube, and 5 μl vol-
ume, containing 100 ng reverse transcribed total RNA, was
added as polymerase chain reaction (PCR) template. The tubes
were closed, centrifuged, and placed into the Corbett research
RG-6000 real-time PCR machine (Corbett LifeScience, Sydney,
Australia). The following primers were used: Osteocalcin
(Forward 5’-GGGCAATAAGGTAGTGAACAG-3’, Reverse 5’-G
CAGCACAGGTCCTAAATAGT-3’), Osterix (Forward 5’-GCGT
ATGGCTTCTTTGTGCCT-3’, Reverse 5’-AGCTCACTATGGCT
CCAGTCC-3’), Runt-related transcription factor 2 (RUNX2)
(Forward 5’-ATACTGGGATGAGGAATGCG-3’, Reverse 5’-CC
AAGAAGGCACAGACAGAA-3’), parathyroid receptor-1 (PTHR1)
(Forward: 5’-GATTCTGGTGGAGGGACTGT-3’, Reverse 5’-GGA
TGATCCACTTCTTGTGC-3’), Atp6v0d2 (Forward 5’-CGGAAA
AGAACTCGTGAAGA-3’, Reverse 5’-CTGGAAGCCCAGTAAA
CAGA-3’), NFATc-1 (Forward 5’-AGGTGACACTAGGGGACA
CA-3’, Reverse 5’-AGTCCCTTCCAAGTTTCCAC-3’), TRAP
(Forward 5’-ACTTCCCCAGCCCTTACTAC-3’, Reverse 5’-TCA
GCACATAGCCCACACCG-3’).
Enzyme-Linked Immunosorbent Assay (ELISA)
Murine plasma was collected by cardiac puncture in tubes (a 1
ml syringe containing 50 μl of 100 mM EDTA), and BM was
flushed with PBS. After centrifugation, plasma and BM super-
natants were collected, and used for detection of SDF-1 pro-
tein by ELISA (R&D Systems, Minneapolis, MN, USA).
Hematopoietic CFU assay
Single-cell suspensions of peripheral blood (PB) after ammo-
nium chloride lysis were plated into 35 mm dishes (3 × 10
5
HSPCs mobilization by AMD3100 in osteoporosis
Jin Young Im, et al.
443http://bmbreports.org BMB Reports
cells/plate) with MethoCult GF M3434 (StemCell Technolog-
ies). Hematopoietic colonies were counted and scored after in-
cubation for 12-14 days at 37
o
C, 5% CO
2
, as instructed by the
manufacturer.
Flow cytometry
BM cells from the femurs and tibias were collected by flushing
with 20 ml PBS passed through a 25-gauge needle. After cen-
trifugation at 1,300 rpm for 5 min, the supernatant was re-
moved and the cells were then washed by ammonium chlor-
ide lysis. Cells were incubated first using a Lineage Cell
Depletion Kit magnetic labeling system with the biotinylated
lineage antibody cocktail (CD5, CD45R [B220], CD11b, Gr-1
[Ly-6G/C], and Ter-119) for 10 min at 4
o
C and anti-biotin
MicroBeads (Milt-enyi Biotec) for an additional 20 min at 4
o
C.
Positive immunoselection was performed with PE/Cy7-conju-
gated anti-Sca-1 (BD Pharmingen), APC-conjugated anti-mouse
CD117 (c-Kit) (BD Pharmingen), SAV-PB, and a FACS Aria (BD
Biosciences) using a flow cytometer.
Micro-computed tomography
For micro-computed tomography (micro-CT) in vivo imaging,
we sacrificed and then scanned each group of mice at 8 μm
resolution using the eXplore Locus scanner (GE Healthcare). In
the femora, scanning regions were confined to the distal meta-
physis, extending proximally 1.7 mm from the proximal tip of
the primary spongiosa. BMD, BMC, BVF, TMD, Tb.N., Tb.Sp.,
Cr.BMD and Cr.BMC were applied for performance of quanti-
tative analysis using software provided with 2.0+ ABA
Micro-view of the micro-CT system.
Osteoclast differentiation
BM cells were prepared by removal from the femurs and tibias
of seven-week-old mice. The bone marrow suspension was
added to plates along with macrophage colony stimulating fac-
tor (M-CSF; 30 ng/ml). After culture for 24 hr, the non-adherent
cells were collected and resuspended in α-MEM containing
10% FBS. For the osteoclastogenesis experiments, BM-derived
macrophages were plated into 6-well plates at a density of 2 ×
10
6
cells/well in α-MEM with 10% FBS, receptor activator for
nuclear factor κB ligand (RANKL; 100 ng/ml) and M-CSF (30
ng/ml) in the presence or absence of AMD3100 (25 μg/ml) for
3 days.
Histological analysis
Femurs were fixed in 4% paraformaldehyde for 24 hr; the tis-
sues were then decalcified in 10% EDTA for one week, dehy-
drated in ethanol, embedded in paraffin, sectioned to 4-μm
thickness and stained with hematoxylin and eosin (H&E). For
TRAP staining, sections were stained with 225 μM Naphthol
AS-MX phosphate (Sigma-Aldrich, St. Louis, MO, USA), 0.84%
N, N-dimethylformamide (Sigma-Aldrich), and 1.33 mM Fast
Red Violet LB Salt (Sigma-Aldrich) in 50 mM sodium acetate
(pH 5.0) containing 50 mM sodium tartrate, and incubated for
30 min. After incubation, sections were washed in distilled
water and counterstained with 1% methyl green. We per-
formed histomorphometric analysis using the Bioquant
OSTEOII Program (BIOQUANT Image Analysis Corporation,
Nashville, TN, USA).
Statistical analysis
The Student’s t-test was used for comparison of two groups,
whereas Tukey’s HSD test and Repeated Measures Analysis of
Variance test were used for multi group comparisons accord-
ing to the SAS statistical package (Release 9.1; SAS Institute
Inc., Cary, NC). P 0.05 was considered significant.
ACKNOWLEDGEMENTS
This work was supported by the Basic Science Research
Program (2013R1A1A2008239) and Bio & Medical Technology
Development Program (2012M3A9C6049913) through the
National Research Foundation of Korea (NRF) funded by the
Ministry of Education, Science and Technology, Republic of
Korea. Additional support for this work was provided by
Biomedical Research Institute grant, Kyungpook National
University Hospital (2012).
REFERENCES
1. Usui, M., Yoshida, Y., Tsuji, K., Oikawa, K., Miyazono, K.,
Ishikawa, I., Yamamoto, T., Nifuji, A. and Noda, M. (2004)
Tob deficiency superenhances osteoblastic activity after
ovariectomy to block estrogen deficiency-induced
osteoporosis. Proc. Natl. Acad. Sci. U S A. 101, 6653-6658.
2. Mansour, A., Abou-Ezzi, G., Sitnicka, E., Jacobsen, S. E.,
Wakkach, A. and Blin-Wakkach, C. (2012) Osteoclasts
promote the formation of hematopoietic stem cell niches
in the bone marrow. J. Exp. Med. 209, 537-549.
3. van den Bergh, J. P., van Geel, T. A. and Geusens, P. P.
(2012) Osteoporosis, frailty and fracture: implications for
case finding and therapy. Nat. Rev. Rheumatol. 8,
163-172.
4. Kim, T., Ha, H., Shim, K. S., Cho, W. K. and Ma, J. Y.
(2013) The anti-osteoporotic effect of Yijung-tang in an
ovariectomized rat model mediated by inhibition of osteo-
clast differentiation. J. Ethnopharmacol. 146, 83-89.
5. Takayanagi, H. (2007) Osteoimmunology: shared mecha-
nisms and crosstalk between the immune and bone
systems. Nat. Rev. Immunol. 7, 292-304.
6. Ishii, M., Egen, J. G., Klauschen, F., Meier-Schellersheim,
M., Saeki, Y., Vacher, J., Proia, R. L. and Germain, R. N.
(2009) Sphingosine-1-phosphate mobilizes osteoclast pre-
cursors and regulates bone homeostasis. Nature 458,
524-528.
7. M. Z., Lee, H., Wysoczynski, M., Wan, W., Marlicz, W.,
Laughlin, M. J., Kucia, M., Janowska-Wieczorek, A. and
Ratajczak, J. (2010) Novel insight into stem cell mobi-
lization-plasma sphingosine-1-phosphate is a major che-
moattractant that directs the egress of hematopoietic stem
progenitor cells from the bone marrow and its level in pe-
HSPCs mobilization by AMD3100 in osteoporosis
J
in Young Im, et al.
444 BMB Reports http://bmbreports.org
ripheral blood increases during mobilization due to acti-
vation of complement cascade/membrane attack complex.
Leukemia 24, 976-985.
8. Nervi, B., Ramirez, P., Rettig, M. P., Uy, G. L., Holt, M.
S., Ritchey, J. K., Prior, J. L., Piwnica-Worms, D., Bridger,
G., Ley, T. J. and DiPersio, J. F. (2009) Chemosensitiza-
tion of acute myeloid leukemia (AML) following mobi-
lization by the CXCR4 antagonist AMD3100. Blood 113,
6206-6214.
9. Rettig, M. P., Ansstas, G. and DiPersio, J. F. (2012)
Mobilization of hematopoietic stem and progenitor cells
using inhibitors of CXCR4 and VLA-4. Leukemia 26,
34-53.
10. Winkler, I. G., Pettit, A. R., Raggatt, L. J., Jacobsen, R. N.,
Forristal, C. E., Barbier, V., Nowlan, B., Cisterne, A.,
Bendall, L. J., Sims, N. A. and Levesque, J. P. (2012)
Hematopoietic stem cell mobilizing agents G-CSF, cyclo-
phosphamide or AMD3100 have distinct mechanisms of
action on bone marrow HSC niches and bone formation.
Leukemia 26, 1594-1601.
11. Christopher, M. J. and Link, D. C. (2008) Granulocyte col-
ony-stimulating factor induces osteoblast apoptosis and in-
hibits osteoblast differentiation. J. Bone. Miner. Res. 23,
1765-1774.
12. Dale, D. C., Cottle, T. E., Fier, C. J., Bolyard, A. A.,
Bonilla, M. A., Boxer, L. A., Cham, B., Freedman, M. H.,
Kannourakis, G., Kinsey, S. E., Davis, R., Scarlata, D.,
Schwinzer, B., Zeidler, C. and Welte, K. (2003) Severe
chronic neutropenia: treatment and follow-up of patients
in the severe chronic neutropenia international registry.
Am. J. Hematol. 72, 82-93.
13. Lee, M. Y., Fukunaga, R., Lee, T. J., Lottsfeldt, J. L. and
Nagata, S. (1991) Bone modulation in sustained hema-
topoietic stimulation in mice. Blood 77, 2135-2141.
14. Dar, A., Schajnovitz, A., Lapid, K., Kalinkovich, A., Itkin,
T., Ludin, A., Kao, W. M., Battista, M., Tesio, M., Kollet,
O., Cohen, N. N., Margalit, R., Buss, E. C., Baleux, F.,
Oishi, S., Fujii, N., Larochelle, A., Dunbar, C. E.,
Broxmeyer, H. E., Frenette, P. S. and Lapidot, T. (2011)
Rapid mobilization of hematopoietic progenitors by
AMD3100 and catecholamines is mediated by
CXCR4-dependent SDF-1 release from bone marrow stro-
mal cells. Leukemia 25, 1286-1296.
15. Golan, K., Vagima, Y., Ludin, A., Itkin, T., Cohen-Gur, S.,
Kalinkovich, A., Kollet, O., Kim, C., Schajnovitz, A.,
Ovadya, Y., Lapid, K., Shivtiel, S., Morris, A. J., Ratajczak,
M. Z. and Lapidot, T. (2012) S1P promotes murine pro-
genitor cell egress and mobilization via S1P1-mediated
ROS signaling and SDF-1 release. Blood 119, 2478-2488.
16. Pusic, I. and DiPersio, J. F. (2010) Update on clinical ex-
perience with AMD3100, an SDF-1/CXCL12-CXCR4 in-
hibitor, in mobilization of hematopoietic stem and pro-
genitor cells. Curr. Opin. Hematol. 17, 319-326.
17. De Klerck, B., Geboes, L., Hatse, S., Kelchtermans, H.,
Meyvis, Y., Vermeire, K., Bridger, G., Billiau, A., Schols,
D. and Matthys, P. (2005) Pro-inflammatory properties of
stromal cell-derived factor-1 (CXCL12) in collagen-in-
duced arthritis. Arthritis. Res. Ther. 7, R1208-1220.
18. Kollet, O., Dar, A., Shivtiel, S., Kalinkovich, A., Lapid, K.,
Sztainberg, Y., Tesio, M., Samstein, R. M., Goichberg, P.,
Spiegel, A., Elson, A. and Lapidot, T. (2006) Osteoclasts
degrade endosteal components and promote mobilization
of hematopoietic progenitor cells. Nat. Med. 12, 657-664.
19. Paganessi, L. A., Walker, A. L., Tan, L. L., Holmes, I.,
Rich, E., Fung, H. C. and Christopherson, K. W., 2nd.
(2011) Effective mobilization of hematopoietic progenitor
cells in G-CSF mobilization defective CD26-/- mice
through AMD3100-induced disruption of the CXCL12-
CXCR4 axis. Exp. Hematol. 39, 384-390.
... Previous studies have demonstrated that AMD3100-induced SDF-1 can induce HSPC mobilization and ameliorate bone loss in an ovariectomy-induced osteoporosis mouse model by decreasing the number of osteoclasts differentiated from BM-derived HSPCs (16). Moreover, recruitment of osteoclast precursors from the BM to the blood relieves bone loss in ovariectomized mice by reducing osteoclast accumulation on bone surfaces (17). ...
... To search for more effective sequences from the full-length NPY for HSPC mobilization, we recombined full length NPY (1-36) into four different peptides: NPY (1-15), NPY (6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)(19)(20), NPY (11)(12)(13)(14)(15)(16)(17)(18)(19)(20)(21)(22)(23)(24)(25), and NPY (21)(22)(23)(24)(25)(26)(27)(28)(29)(30)(31)(32)(33)(34)(35)(36). Recent studies demonstrated that HSPC maintenance factors such as SDF-1, Kitl, Angpt1, Vcam1, IL7, and Spp1 (10)(11)(12)(13)(14) are important for maintaining HSPCs with niche cells in the BM, and a reduction of these factors induces HSPC mobilization (15,24). ...
... To search for more effective sequences from the full-length NPY for HSPC mobilization, we recombined full length NPY (1-36) into four different peptides: NPY (1-15), NPY (6)(7)(8)(9)(10)(11)(12)(13)(14)(15)(16)(17)(18)(19)(20), NPY (11)(12)(13)(14)(15)(16)(17)(18)(19)(20)(21)(22)(23)(24)(25), and NPY (21)(22)(23)(24)(25)(26)(27)(28)(29)(30)(31)(32)(33)(34)(35)(36). Recent studies demonstrated that HSPC maintenance factors such as SDF-1, Kitl, Angpt1, Vcam1, IL7, and Spp1 (10)(11)(12)(13)(14) are important for maintaining HSPCs with niche cells in the BM, and a reduction of these factors induces HSPC mobilization (15,24). ...
Article
Full-text available
Ovariectomy-induced bone loss is related to an increased deposition of osteoclasts on bone surfaces. We reported that the 36-amino-acid-long neuropeptide Y (NPY) could mobilize hematopoietic stem/progenitor cells (HSPCs) from the bone marrow to the peripheral blood by regulating HSPC maintenance factors and that mobilization of HSPCs ameliorated low bone density in an ovariectomy-induced osteoporosis mouse model by reducing the number of osteoclasts. Here, we demonstrated that new NPY peptides, recombined from the cleavage of the full-length NPY, showed better functionality for HSPC mobilization than the full-length peptide. These recombinant peptides mediated HSPC mobilization with greater efficiency by decreasing HSPC maintenance factors. Furthermore, treatment with these peptides reduced the number of osteoclasts and relieved ovariectomy-induced bone loss in mice more effectively than treatment with full-length NPY. Therefore, these results suggest that peptides recombined from full-length NPY can be used to treat osteoporosis. © 2017 by the The Korean Society for Biochemistry and Molecular Biology.
... AMD3100 (cat. 3299; TOCRIS, CXCR4 antagonist) was intraperitoneally injected 1 h before application of force at a dosage of 5 μg/g once a day until sacrifice (Im et al. 2014). Other groups were injected with the same volume of saline. ...
... Furthermore, ORR and elevated M1/M2 ratio were attenuated by blocking the CXCL12/CXCR4 axis with AMD3100. The therapeutic effect of AMD3100 on bone resorption has been described in other pathological bone loss models, including ovariectomy-induced osteoporosis (Im et al. 2014) and traumatic osteoarthritis (Dong et al. 2016). Overall, these results indicate that CXCL12/CXCR4 is a mechanosensitive chemokine axis and might mediate ORR by M1 enrichment. ...
... Previous studies have indicated that CXCL12 enhances osteoclastogenesis by affecting osteoprogenitor cells and increasing bone resorption in several pathological conditions and that antagonists of CXCR4 improve ovariectomy-induced osteoporosis and multiple myeloma-mediated osteoclastogenesis. [324][325][326][327] However, according to research by Ponte et al., the indirect restraint of CXCL12 on bone remodeling through inhibiting osteogenesis and the osteoclastogenesis support provided by cells of the osteoblast lineage exceeds its direct pro-osteoclastogenic effect. 328 Additionally, Pont et al. further exemplified that CXCL12 deletion greatly attenuated the loss of cortical bone caused by estrogen deficiency, suggesting that CXCL12 may contribute to estrogen deficiency-induced bone loss. ...
Article
Full-text available
Since the proposal of Paul Ehrlich's magic bullet concept over 100 years ago, tremendous advances have occurred in targeted therapy. From the initial selective antibody, antitoxin to targeted drug delivery that emerged in the past decades, more precise therapeutic efficacy is realized in specific pathological sites of clinical diseases. As a highly pyknotic mineralized tissue with lessened blood flow, bone is characterized by a complex remodeling and homeostatic regulation mechanism, which makes drug therapy for skeletal diseases more challenging than other tissues. Bone-targeted therapy has been considered a promising therapeutic approach for handling such drawbacks. With the deepening understanding of bone biology, improvements in some established bone-targeted drugs and novel therapeutic targets for drugs and deliveries have emerged on the horizon. In this review, we provide a panoramic summary of recent advances in therapeutic strategies based on bone targeting. We highlight targeting strategies based on bone structure and remodeling biology. For bone-targeted therapeutic agents, in addition to improvements of the classic denosumab, romosozumab, and PTH1R ligands, potential regulation of the remodeling process targeting other key membrane expressions, cellular crosstalk, and gene expression, of all bone cells has been exploited. For bone-targeted drug delivery, different delivery strategies targeting bone matrix, bone marrow, and specific bone cells are summarized with a comparison between different targeting ligands. Ultimately, this review will summarize recent advances in the clinical translation of bone-targeted therapies and provide a perspective on the challenges for the application of bone-targeted therapy in the clinic and future trends in this area.
... Consistent with these findings, the CXCR4 antagonist AMD3100 has been shown to improve ovariectomy-induced bone loss by facilitating mobilization of haematopoietic progenitor cells. 34 Moreover, treatment with vascular endothelial growth factor and AMD3100 can mobilize mesenchymal stem cells towards fracture healing, leading to bone formation in a delayed union osteotomy model. 35 CXCL12, a major ligand for CXCR4, is not only linked to the severity of postmenopausal osteoporosis 36 but also acts as a proinflammatory factor during progression of collagen-induced osteoarthritis by attracting inflammatory cells to joints and by activating osteoclasts. ...
Article
Full-text available
Background: Mitochondrial oxidative phosphorylation (OxPhos) is a critical regulator of skeletal muscle mass and function. Although muscle atrophy due to mitochondrial dysfunction is closely associated with bone loss, the biological characteristics of the relationship between muscle and bone remain obscure. We showed that muscle atrophy caused by skeletal muscle-specific CR6-interacting factor 1 knockout (MKO) modulates the bone marrow (BM) inflammatory response, leading to low bone mass. Methods: MKO mice with lower muscle OxPhos were fed a normal chow or high-fat diet and then evaluated for muscle mass and function, and bone mineral density. Immunophenotyping of BM immune cells was also performed. BM transcriptomic analysis was used to identify key factors regulating bone mass in MKO mice. To determine the effects of BM-derived CXCL12 (C-X-C motif chemokine ligand 12) on regulation of bone homeostasis, a variety of BM niche-resident cells were treated with recombinant CXCL12. Vastus lateralis muscle and BM immune cell samples from 14 patients with hip fracture were investigated to examine the association between muscle function and BM inflammation. Results: MKO mice exhibited significant reductions in both muscle mass and expression of OxPhos subunits but increased transcription of mitochondrial stress response-related genes in the extensor digitorum longus (P < 0.01). MKO mice showed a decline in grip strength and a higher drop rate in the wire hanging test (P < 0.01). Micro-computed tomography and von Kossa staining revealed that MKO mice developed a low mass phenotype in cortical and trabecular bone (P < 0.01). Transcriptomic analysis of the BM revealed that mitochondrial stress responses in skeletal muscles induce an inflammatory response and adipogenesis in the BM and that the CXCL12-CXCR4 (C-X-C chemokine receptor 4) axis is important for T-cell homing to the BM. Antagonism of CXCR4 attenuated BM inflammation and increased bone mass in MKO mice. In humans, patients with low body mass index (BMI = 17.2 ± 0.42 kg/m2 ) harboured a larger population of proinflammatory and cytotoxic senescent T-cells in the BMI (P < 0.05) and showed reduced expression of OxPhos subunits in the vastus lateralis, compared with controls with a normal BMI (23.7 ± 0.88 kg/m2 ) (P < 0.01). Conclusions: Defects in muscle mitochondrial OxPhos promote BM inflammation in mice, leading to decreased bone mass. Muscle mitochondrial dysfunction is linked to BM inflammatory cytokine secretion via the CXCL12-CXCR4 signalling axis, which is critical for inducing low bone mass.
... (10)(11)(12) Consistent with this evidence, antagonists of CXCR4 attenuate the pathologic bone loss at sites of inflammation in the synovium and bone in collagen-induced arthritis, (13) osteoclastogenesis associated with multiple myeloma, (14) and the skeletal effects of ovariectomy (OVX) in mice. (15) Estrogens contribute to the maintenance of bone mass by decreasing osteoclast numbers and bone resorption. These effects are the result of direct estrogen actions on osteoclast precursors of the myeloid lineage and mature osteoclasts, as well as estrogen actions on osteoblast precursors of the mesenchymal lineage that suppress the production of osteoclastogenic cytokines. ...
Article
CXCL12 is abundantly expressed in reticular cells associated with the perivascular niches of the bone marrow (BM) and is indispensable for B‐lymphopoiesis. Cxcl12 promotes osteoclastogenesis and has been implicated in pathologic bone resorption. We had shown earlier that estrogen receptor α deletion in osteoprogenitors and estrogen deficiency in mice increase Cxcl12 mRNA and protein levels in the BM plasma, respectively. We have now generated female and male mice with conditional deletion of a Cxcl12 allele in Prrx1 targeted cells (Cxcl12∆Prrx1) and show herein that they have a 90% decrease in B lymphocytes, but increased erythrocytes and adipocytes in the marrow. Ovariectomy increased the expression of Cxcl12 and B cell number in the Cxcl12f/f control mice, but these effects were abrogated in the Cxcl12∆Prrx1 mice. Cortical bone mass was not affected in Cxcl12∆Prrx1 mice. Albeit, the cortical bone loss caused by ovariectomy was greatly attenuated. Most unexpectedly, the rate of bone turnover in sex steroid sufficient female or male Cxcl12∆Prrx1 mice was dramatically increased, as evidenced by a more than two‐fold increase in several osteoblast‐ and osteoclast‐ specific mRNAs, as well as increased mineral apposition and bone formation rate and increased osteoclast number in the endosteal surface. The magnitude of the Cxcl12∆Prrx1 induced changes were much greater than those caused by ovariectomy or orchidectomy in the Cxcl12f/f mice. These results strengthen the evidence that CXCL12 contributes to the loss of cortical bone mass caused by estrogen deficiency. Moreover, they reveal for the first time that in addition to its effects on hematopoiesis, CXCL12 restrains bone turnover – without changing the balance between resorption and formation – by suppressing osteoblastogenesis and the osteoclastogenesis support provided by cells of the osteoblast lineage. This article is protected by copyright. All rights reserved.
... To discriminate between these two possibilities, an in vivo competition experiment was designed in which the CXCR4 antagonist, AMD3100 [42][43][44][45][46][47], was used to block tumor accumulation of the administered protein materials. As observed (Fig. 4 A, B), the antagonist dramatically minimized the occurrence of T22-GFP-H6 nanoparticles in tumoral tissues, supporting again the role of T22 in active targeting for CXCR4 + cancers. ...
Article
By the appropriate selection of functional peptides and proper accommodation sites, we have generated a set of multifunctional proteins that combine selectivity for CXCR4⁺ cell binding and relevant endosomal escape capabilities linked to the viral peptide HA2. In particular, the construct T22-GFP-HA2-H6 forms nanoparticles that upon administration in mouse models of human, CXCR4⁺ colorectal cancer, accumulates in primary tumor at levels significantly higher than the parental T22-GFP-H6 HA2-lacking version. The in vivo application of a CXCR4 antagonist has confirmed the prevalence of the CXCR4⁺ tumor tissue selectivity over unspecific cell penetration, upon systemic administration of the material. Such specificity is combined with improved endosomal escape, what overall results in a precise and highly efficient tumor biodistribution. These data strongly support the functional recruitment as a convenient approach to generate protein materials for clinical applications. More precisely, they also support the unexpected concept that enhancing the unspecific membrane activity of a protein material does not necessarily compromise, but it can even improve, the selective cell targeting offered by an accompanying functional module. Statement of Significance We have shown here that the combination of cell-penetrating and tumor cell-targeting peptides dramatically enhances precise tumor accumulation of protein-only nanoparticles intended for selective drug delivery, in mouse models of human colorectal cancer. This fact is a step forward for the rational design of multifunctional protein nanomaterials for improved cancer therapies.
... However, signiicant efects on bone turnover have only been reported after prolonged treatment with AMD3100. Im et al. reported that AMD3100 treatment (5 mg/kg/day) for 21 consecutive days induced release of SDF-1, preventing ovariectomy-induced bone loss in female C57BL/6 mice by decreasing the number of osteoclasts diferentiated from BM-derived precursors [43]. In contrast, short term administration of 5 mg/kg/day AMD3100 to C57BL/6 mice for 3 days is reported not to afect bone mineral density in vivo [44]. ...
Article
Full-text available
Background: Bone metastasis is one of the most common complications of advanced breast cancer. During dissemination to bone, breast cancer cells locate in a putative 'metastatic niche', a microenvironment that regulates the colonisation, maintenance of tumour cell dormancy and subsequent tumour growth. The precise location and composition of the bone metastatic niche is not clearly defined. We have used in vivo models of early breast cancer dissemination to provide novel evidence that demonstrates overlap between endosteal, perivascular, HSC and the metastatic niche in bone. Methods: Estrogen Receptor (ER) +ve and -ve breast cancer cells were labelled with membrane dyes Vybrant-DiD and Vybrant-CM-DiI and injected via different routes in BALBc/nude mice of different ages. Two-photon microscopy was used to detect and quantitate tumour cells and map their location within the bone microenvironment as well as their distance to the nearest bone surface compared to the nearest other tumour cell. To investigate whether the metastatic niche overlapped with the HSC niche, animals were pre-treated with the CXCR4 antagonist AMD3100 to mobilise hematopoietic (HSCs) prior to injection of breast cancer cells. Results: Breast cancer cells displayed a characteristic pattern of homing in the long bones, with the majority of tumour cells seeded in the trabecular regions, regardless of the route of injection, cell-line characteristics (ER status) or animal age. Breast cancer cells located in close proximity to the nearest bone surface and the average distance between individual tumour cells was higher than their distance to bone. Mobilisation of HSCs from the niche to the circulation prior to injection of cell lines resulted in increased numbers of tumour cells disseminated in trabecular regions. Conclusion: Our data provide evidence that homing of breast cancer cells is independent of their ER status and that the breast cancer bone metastasis niche is located within the trabecular region of bone, an area rich in osteoblasts and microvessels. The increased number of breast cancer cells homing to bone after mobilisation of HSCs suggests that the HSC and the bone metastasis niche overlap.
Article
Mechanical force–induced external root resorption is a major clinical side effect of orthodontic treatment. Recent work has revealed that M1 macrophages play a vital role in promoting orthodontic root resorption (ORR), but the mechanism of how mechanical force stimulation increases the M1/M2 macrophage ratio in periodontal tissue is poorly understood. In the current study, we showed that C-X-C motif chemokine 12 (CXCL12) ⁺ periodontal ligament cells (PDLCs) and C-X-C chemokine receptor type 4 (CXCR4) ⁺ monocytes in the periodontal ligament (PDL) were significantly increased after force application with ongoing root resorption, and these effects were partially rescued after force removal in mice. The expression of CXCL12 in PDLCs was increased by force stimulation in a time- and intensity-dependent manner in vitro. Blockage of the CXCL12/CXCR4 axis using CXCR4 antagonist AMD3100 was sufficient to alleviate ORR and reverse the force-enhanced M1/M2 macrophage ratio. Further mechanism exploration showed that Ly6C hi inflammatory monocytes homed in a CXCL12/CXCR4 axis-dependent manner. The number and proportion of CD11b ⁺ Ly6C hi inflammatory monocytes in cervical lymph nodes were significantly increased by force loading, accompanied by decreased CD11b ⁺ Ly6C hi monocytes in the blood. These changes were blunted by intraperitoneal injection of AMD3100. In addition, blockage of the CXCL12/CXCR4 axis effectively reversed M2 suppression and promoted M1 polarization. Collectively, results indicate that force-induced CXCL12/CXCR4 axis mediates ORR by increasing the M1/M2 ratio in periodontal tissues through attracting Ly6C hi inflammatory monocytes and modulating macrophage polarization. The results also imply that AMD3100 is potentially inhibitory to root resorption.
Article
Metastasis is the main cause of death in cancer patients; there is currently no effective treatment for cancer metastasis. This is primarily due to our insufficient understanding of the metastatic mechanisms in cancer. An increasing number of studies have shown that the C-X-C motif chemokine ligand 12 (CXCL12) is overexpressed in various tissues and organs. It is a key niche factor that nurtures the pre-metastatic niches (tumorigenic soil) and recruits tumor cells (oncogenic “seeds”) to these niches, thereby fostering cancer cell aggression and metastatic capabilities. However, the C-X-C motif chemokine receptor 4 (CXCR4) is aberrantly overexpressed in various cancer stem/progenitor cells and functions as a CXCL12 receptor. CXCL12 activates CXCR4 as well as multiple downstream multiple tumorigenic signaling pathways, promoting expression of various oncogenes. Activation of the CXCL12-CXCR4 signaling axis promotes epithelial-mesenchymal transition (EMT) and mobilization of cancer stem/progenitor cells to pre-metastatic niches. It also nurtures cancer cells with high motility, invasion, and dissemination phenotypes, thereby escalating multiple proximal or distal cancer metastasis; this results in poor patient prognosis. Based on this evidence, recent studies have explored either CXCL12- or CXCR4-targeted anti-cancer therapeutics and have achieved encouraging results in the preclinical trials. Further exploration of this new strategy and its potent therapeutics effect against metastatic cancer via the targeting of the CXCL12-CXCR4 signaling axis may lead to a novel therapy that can clean up the tumor microenvironment (“soil”) and kill the cancer cells, particularly the cancer stem/progenitor cells (“seeds”), in cancer patients. Ultimately, this approach has the potential to effectively treat metastatic cancer.
Article
Musculoskeletal disorders are the leading cause of disability worldwide; two of the most prevalent of which are osteoporosis and sarcopenia. Each affect millions in the aging population across the world and the associated morbidity and mortality contributes to billions of dollars in annual healthcare cost. Thus, it is important to better understand the underlying pathologic mechanisms of the disease process. Regulatory chemokine, CXCL12, and its receptor, CXCR4, are recognized to be essential in the recruitment, localization, maintenance, development and differentiation of progenitor stem cells of the musculoskeletal system. CXCL12 signaling results in the development and functional ability of osteoblasts, osteoclasts, satellite cells and myoblasts critical to maintaining musculoskeletal homeostasis. Interestingly, one suggested pathologic mechanism of osteoporosis and sarcopenia is a decline in the regenerative capacity of musculoskeletal progenitor stem cells. Thus, because CXCL12 is critical to progenitor function, a disruption in the CXCL12 signaling axis might play a distinct role in these pathological processes. Therefore, in this article, we perform a review of CXCL12, its physiologic and pathologic function in bone and muscle, and potential targets for therapeutic development.
Article
Full-text available
Formation of the hematopoietic stem cell (HSC) niche in bone marrow (BM) is tightly associated with endochondral ossification, but little is known about the mechanisms involved. We used the oc/oc mouse, a mouse model with impaired endochondral ossification caused by a loss of osteoclast (OCL) activity, to investigate the role of osteoblasts (OBLs) and OCLs in the HSC niche formation. The absence of OCL activity resulted in a defective HSC niche associated with an increased proportion of mesenchymal progenitors but reduced osteoblastic differentiation, leading to impaired HSC homing to the BM. Restoration of OCL activity reversed the defect in HSC niche formation. Our data demonstrate that OBLs are required for establishing HSC niches and that osteoblastic development is induced by OCLs. These findings broaden our knowledge of the HSC niche formation, which is critical for understanding normal and pathological hematopoiesis.
Article
Full-text available
The CXCR4 antagonist AMD3100 is progressively replacing cyclophosphamide (CYP) as adjuvant to granulocyte colony-stimulating factor (G-CSF) to mobilize hematopoietic stem cells (HSC) for autologous transplants in patients who failed prior mobilization with G-CSF alone. It has recently emerged that G-CSF mediates HSC mobilization and inhibits bone formation via specific bone marrow (BM) macrophages. We compared the effect of these three mobilizing agents on BM macrophages, bone formation, osteoblasts, HSC niches and HSC reconstitution potential. Both G-CSF and CYP suppressed niche-supportive macrophages and osteoblasts, and inhibited expression of endosteal cytokines resulting in major impairment of HSC reconstitution potential remaining in the mobilized BM. In sharp contrast, although AMD3100 was effective at mobilizing HSC, it did not suppress osteoblasts, endosteal cytokine expression or reconstitution potential of HSC remaining in the mobilized BM. In conclusion, although G-CSF, CYP and AMD3100 efficiently mobilize HSC into the blood, their effects on HSC niches and bone formation are distinct with both G-CSF and CYP targeting HSC niche function and bone formation, whereas AMD3100 directly targets HSC without altering niche function or bone formation.
Article
Full-text available
In almost all patients with incident fractures, the absolute risk of subsequent fracture and mortality is highest immediately after the fracture is incurred; the risk is substantially increased in frail elderly patients. The risk factors for incident fractures, such as bone fragility, tendency to fall and the presence of metabolic bone disease, remain underdiagnosed and undertreated. Here, we review the evidence that demonstrates the influence of these risk factors on susceptibility to subsequent fracture and mortality after an incident fracture, and discuss the tools available to predict these outcomes. In this Review, we also propose a systematic, coordinator-based approach to assessment of risk, allocation of treatment and follow-up in all patients over 50 years of age who present with a fracture. The aim of this proposed multistep procedure is to improve the prevention of secondary fracture, decrease mortality rates and reduce patient undertreatment or overtreatment.
Article
Full-text available
Steady-state egress of hematopoietic progenitor cells can be rapidly amplified by mobilizing agents such as AMD3100, the mechanism, however, is poorly understood. We report that AMD3100 increased the homeostatic release of the chemokine stromal cell derived factor-1 (SDF-1) to the circulation in mice and non-human primates. Neutralizing antibodies against CXCR4 or SDF-1 inhibited both steady state and AMD3100-induced SDF-1 release and reduced egress of murine progenitor cells over mature leukocytes. Intra-bone injection of biotinylated SDF-1 also enhanced release of this chemokine and murine progenitor cell mobilization. AMD3100 directly induced SDF-1 release from CXCR4(+) human bone marrow osteoblasts and endothelial cells and activated uPA in a CXCR4/JNK-dependent manner. Additionally, ROS inhibition reduced AMD3100-induced SDF-1 release, activation of circulating uPA and mobilization of progenitor cells. Norepinephrine treatment, mimicking acute stress, rapidly increased SDF-1 release and progenitor cell mobilization, whereas β2-adrenergic antagonist inhibited both steady state and AMD3100-induced SDF-1 release and progenitor cell mobilization in mice. In conclusion, this study reveals that SDF-1 release from bone marrow stromal cells to the circulation emerges as a pivotal mechanism essential for steady-state egress and rapid mobilization of hematopoietic progenitor cells, but not mature leukocytes.
Article
To understand the etiology of bone modulation and hypercalcemia observed in granulocytosis of a tumor-bearing animal model and to gain insight into the implication of sustained hematopoietic stimulation on the bone tissue, in vivo responses of normal mouse hematopoietic and bone tissues to long-term injections of recombinant human and murine granulocyte colony-stimulating factor (G-CSF), murine granulocyte- macrophage CSF (GM-CSF), and human erythropoietin were quantitatively analyzed. Osteoclast activation was estimated by the osteoclast- endosteal ratio, determined by morphometric analyses of femoral sections. Medullary and bone areas were measured on transverse ground bone sections of the tibia. Recombinant murine G-CSF provoked marked granulocytosis associated with significant increases in the number of marrow granulocytes and their progenitors, and caused expansion of granulopoietic marrow into fatty marrow. The bone of G-CSF-treated mice showed a significant increase in endosteal osteoclast numbers with medullary area enlargement and a reduction in the bone thickness; indicative of endosteal bone resorption. Although GM-CSF had little effect on granulopoiesis, it caused peritoneal macrophages to increase and induced similar bone changes as those observed in G-CSF treatment. Enhanced erythropoiesis stimulated by erythropoietin was also associated with evidence of endosteal bone resorption. Bone changes induced by these growth factors were not associated with hypercalcemia. These animal studies document association of bone modulation in sustained stimulation of hematopoiesis, and implicate important physiologic effects of hematopoietic growth factors on skeletal tissue in vivo.
Article
Ethnopharmacological relevance: Yijung-tang (YJ), a traditional Asian medicine, is used to treat various diseases. However, its anti-osteoporotic effect and mechanism of action remain unclear. The aim of the present study was to evaluate the anti-osteoporotic effect of YJ in ovariectomized (OVX) rats. Materials and methods: Sprague-Dawley rats were divided into five groups as follows: sham-operated, ovariectomized (OVX), OVX rats treated with 100 g/kg/day 17-estradiol, and OVX rats treated with 0.3 and 1.0 g/kg/day YJ for 12 weeks. Trabecular bone mineral density (BMD) and bone microarchitecture were evaluated by microcomputed tomography. The effects of YJ on osteoblast and osteoclast formation were also investigated in an in vitro model using primary murine bone marrow-derived macrophages and murine calvarial preosteoblasts. mRNA expression of osteoclast differentiation-related genes was measured by real-time quantitative reverse transcription-polymerase chain reaction. Activation of the mitogen-activated protein kinases and nuclear factor-κB (NF-κB) were determined by Western blot. Results: The decrease of BMD and destruction of bone microarchitecture were significantly reduced in the OVX-induced osteoporosis rat model after 12 weeks of YJ treatment. The anti-osteoporotic effect of YJ on bone loss was due to inhibition of osteoclast differentiation through down-regulation of the NF-κB pathway. In addition, YJ suppressed the induction of nuclear factor of activated T-cells, cytoplasmic 1 and c-Fos following receptor activator of nuclear factor kappa-B ligand stimulation. Conclusions: These results suggest that YJ possess potent anti-osteoporotic activity in OVX rats and may be a useful remedy for the treatment of postmenopausal osteoporosis.
Article
The mechanisms of hematopoietic progenitor cell egress and clinical mobilization are not fully understood. Herein, we report that in vivo desensitization of Sphingosine-1-phosphate (S1P) receptors by FTY720 as well as disruption of S1P gradient toward the blood, reduced steady state egress of immature progenitors and primitive Sca-1(+)/c-Kit(+)/Lin(-) (SKL) cells via inhibition of SDF-1 release. Administration of AMD3100 or G-CSF to mice with deficiencies in either S1P production or its receptor S1P(1), or pretreated with FTY720, also resulted in reduced stem and progenitor cell mobilization. Mice injected with AMD3100 or G-CSF demonstrated transient increased S1P levels in the blood mediated via mTOR signaling, as well as an elevated rate of immature c-Kit(+)/Lin(-) cells expressing surface S1P(1) in the bone marrow (BM). Importantly, we found that S1P induced SDF-1 secretion from BM stromal cells including Nestin(+) mesenchymal stem cells via reactive oxygen species (ROS) signaling. Moreover, elevated ROS production by hematopoietic progenitor cells is also regulated by S1P. Our findings reveal that the S1P/S1P(1) axis regulates progenitor cell egress and mobilization via activation of ROS signaling on both hematopoietic progenitors and BM stromal cells, and SDF-1 release. The dynamic cross-talk between S1P and SDF-1 integrates BM stromal cells and hematopoeitic progenitor cell motility.
Article
Successful hematopoietic stem cell transplant requires the infusion of a sufficient number of hematopoietic stem/progenitor cells (HSPCs) that are capable of homing to the bone marrow cavity and regenerating durable trilineage hematopoiesis in a timely manner. Stem cells harvested from peripheral blood are the most commonly used graft source in HSCT. Although granulocyte colony-stimulating factor (G-CSF) is the most frequently used agent for stem cell mobilization, the use of G-CSF alone results in suboptimal stem cell yields in a significant proportion of patients. Both the chemokine receptor CXCR4 and the integrin α(4)β(1) (very late antigen 4 (VLA-4)) have important roles in the homing and retention of HSPCs within the bone marrow microenvironment. Preclinical and/or clinical studies have shown that targeted disruption of the interaction of CXCR4 or VLA-4 with their ligands results in the rapid and reversible mobilization of hematopoietic stem cells into the peripheral circulation and is synergistic when combined with G-CSF. In this review, we discuss the development of small-molecule CXCR4 and VLA-4 inhibitors and how they may improve the utility and convenience of peripheral blood stem cell transplantation.
Article
Leukemia is one of the leading journals in hematology and oncology. It is published monthly and covers all aspects of the research and treatment of leukemia and allied diseases. Studies of normal hemopoiesis are covered because of their comparative relevance.