ArticlePDF Available

Unique anabolic action of stem cell gene therapy overexpressing PDGFB-DSS6 fusion protein in OVX osteoporosis mouse model

Authors:

Abstract

In the present study we sought to improve the efficacy and safety of our Sca1+ PDGFB stem cell gene therapy for osteoporosis in ovariectomized (OVX) mouse model. This therapy is administered by marrow transplantation. We established the promise of this approach by previously showing that this therapy in normal mice increase bone density, increased endosteal cortical and trabecular bone formation, caused de novo trabecular bone formation, increased cortical thickness and improve bone strength. In the current study we produced a fusion gene, PDGFB-DSS6. We reasoned that the DSS6, calcium binding protein would trap the PDGFB at the bone surface and thereby limit the amount of PDGFB required to produce an optimal bone formation response, i.e. efficacy with a lower engraftment. The result shows that indeed with a very low level of engraftment we achieved a large increase in bone formation in the OVX model of bone loss. Serum analysis for biochemical marker of new bone formation showed an approximate 75% increase in alkaline phosphatase levels in Sca1+PDGFB-DSS6 group as compared to other groups. Quantitative analysis of bone by microCT showed a massive increase in trabecular bone density and trabecular connectivity of the femur in the metaphysis in Sca1+ PDGFB-DSS6 group. The increased cortical porosity produced by OVX was replaced by the Sca1+ PDGFB-DSS6 therapy but not by the positive control Sca1+ PDGFB. Additionally, an increase in the femur bone strength was also observed specifically in Sca1+ PDGFB-DSS6 as compared to other treatment groups, emphasizing the functional significance of the observed anabolic action is on bone formation. In future work we will focus on nontoxic preconditioning of our marrow transplantation procedure and also on transcriptional control of therapeutic gene expression to avoid excess bone formation.
Contents lists available at ScienceDirect
Bone Reports
journal homepage: www.elsevier.com/locate/bonr
Unique anabolic action of stem cell gene therapy overexpressing PDGFB-
DSS6 fusion protein in OVX osteoporosis mouse model
Wanqiu Chen
a,1
, Samiksha Wasnik
a,,1
, Yawen Fu
a
, Leslie Aranda
a
, Charles H. Rundle
a,b
,
Kin-Hing William Lau
a,b
, David J. Baylink
a
, Xiaobing Zhang
a
a
Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, California, USA
b
Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Aairs Medical Center, Loma Linda, CA, USA
ARTICLE INFO
Keywords:
Osteoporosis
OVX
PDGFB
DSS6
Fusion protein
Stem cell therapy
ABSTRACT
In the present study we sought to improve the ecacy and safety of our Sca1
+
PDGFB stem cell gene therapy for
osteoporosis in ovariectomized (OVX) mouse model. This therapy is administered by marrow transplantation.
We established the promise of this approach by previously showing that this therapy in normal mice increase
bone density, increased endosteal cortical and trabecular bone formation, caused de novo trabecular bone
formation, increased cortical thickness and improve bone strength. In the current study we produced a fusion
gene, PDGFB-DSS6. We reasoned that the DSS6, calcium binding protein would trap the PDGFB at the bone
surface and thereby limit the amount of PDGFB required to produce an optimal bone formation response, i.e.
ecacy with a lower engraftment. The result shows that indeed with a very low level of engraftment we
achieved a large increase in bone formation in the OVX model of bone loss. Serum analysis for biochemical
marker of new bone formation showed an approximate 75% increase in alkaline phosphatase levels in
Sca1
+
PDGFB-DSS6 group as compared to other groups. Quantitative analysis of bone by microCT showed a
massive increase in trabecular bone density and trabecular connectivity of the femur in the metaphysis in Sca1
+
PDGFB-DSS6 group. The increased cortical porosity produced by OVX was replaced by the Sca1
+
PDGFB-DSS6
therapy but not by the positive control Sca1
+
PDGFB. Additionally, an increase in the femur bone strength was
also observed specically in Sca1
+
PDGFB-DSS6 as compared to other treatment groups, emphasizing the
functional signicance of the observed anabolic action is on bone formation. In future work we will focus on
nontoxic preconditioning of our marrow transplantation procedure and also on transcriptional control of ther-
apeutic gene expression to avoid excess bone formation.
1. Introduction
In the US alone, there are 10 million non-traumatic fragility frac-
tures annually (Osteoporosis: Fragility Fracture Risk: Osteoporosis:
Assessing the Risk of Fragility Fracture, 2012). The most common osteo-
porosis-associated fracture is a vertebral compression fracture, which is
associated with substantial morbidity (Kanis, 1994). Antiresorptive
therapy has been shown to decrease vertebral fractures by 50%. Hip
fracture is much less common, but has 1458% mortality within one
year of fracture (Schnell et al., 2010;Panula et al., 2011). Anti-
resorptive therapy only decreases non-vertebral fractures by 2030%
(Kawai et al., 2011). Interestingly, anabolic therapies have been shown
to reduce non-vertebral and vertebral fractures by 3560% (Minisola
et al., 2017;Rubin and Bilezikian, 2002;Neer et al., 2001). In addition,
current anabolic therapies are expensive, require strict compliance, and
lose their eectiveness in 12 years (Kawai et al., 2011). Therefore, a
stronger anabolic agent with a prolonged activity is considered to be a
major unmet therapeutic need in the treatment of osteoporosis.
One of the most important recent and novel ndings on the pa-
thogenesis of osteoporosis is the identication of senescent bone cells,
mostly osteocytes but also osteoprogenitor cells (Farr et al., 2016).
Senescent cells are nonproliferating cells that secrete products detri-
mental to surrounding cells in tissue microenvironment. One might
expect that the accumulation of senescent cells would inhibit the action
of anabolic agents. However, this does not seem to be the case because
of the universal success of parathyroid hormone (PTH)-like medications
https://doi.org/10.1016/j.bonr.2019.100236
Received 8 May 2019; Received in revised form 6 December 2019; Accepted 9 December 2019
Corresponding author at: Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
E-mail addresses: WChen@llu.edu (W. Chen), Swasnik@llu.edu (S. Wasnik), YFu1@llu.edu (Y. Fu), Charles.Rundle@va.gov (C.H. Rundle),
William.Lau@va.gov (K.-H.W. Lau), DBaylink@llu.edu (D.J. Baylink), XZhang@llu.edu (X. Zhang).
1
These authors equally contribute to this work
Bone Reports 12 (2020) 100236
Available online 11 December 2019
2352-1872/ © 2019 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
T
(even in the elderly) to stimulate bone formation and reduce fracture
risk (Ascenzi et al., 2012;Papapoulos, 2015). Moreover, sodium
uoride, which is a bone cell mitogen, has been shown to produce a
marked and prolonged stimulation of bone formation in elderly osteo-
porotic subjects (Rubin et al., 2001;Kleerekoper and Balena, 1991).
Despite its bone anabolic action, sodium uoride impairs bone quality
and therefore is not considered an eective anabolic agent (Kleerekoper
et al., 1991;Gutteridge et al., 2002). These observations emphasize the
responsiveness of the aged osteoporotic skeleton to regeneration.
With respect to the development of stronger anabolic agents, we
have recently developed an anabolic agent, platelet-derived growth
factor-B (PDGFB) that cause a massive, prolonged increase in new en-
dosteal and trabecular bone formation in normal mice, which was at-
tended by a marked increase in bone strength (Chen et al., 2015). Re-
garding safety, PDGFB has been approved by the FDA for promotion of
wound healing (Fang and Galiano, 2008). Also in our previous study we
found no safety issues regarding PDGF administration (Chen et al.,
2015). The main concern about our approach is that toxic eect of
preconditioning needed for marrow transplantation. Marrow trans-
plantation was utilized in order to target the PDGFB to sites of bone loss
in humans (Chen et al., 2015); namely, the cortical endosteum, the
cortical canals, and trabecular bone (Seeman, 2013). The targeting
mechanism that we used was bone marrow transplantation, in which
we engineered hematopoietic stem cells (Sca1
+
cells) to overexpress
PDGFB. Total body radiation increases in the bone niche, SDF 1, which
is a chemokine for the CXCR4 receptor and expressed by Sca-1 cells.
Consequently, the Sca 1 cells localize to the hematopoietic stem cell
niche which is a site where bone is lost (Chanda et al., 2010). These Sca-
1
+
cells do not respond by proliferation to PDGFB in vitro, as men-
tioned earlier paper (Chen et al., 2015)
Total body irradiation or chemotherapy as preconditioning man-
euvers is not acceptable for a nonlethal disease, such as osteoporosis.
Therefore, we sought to further target PDGFB to the bone surface in an
attempt to reduce the Sca1
+
-PDGFB engraftment required for bone
Fig. 1. Schematics of the experimental design. Two weeks after OVX or sham surgery animals undergo irradiation and transplantation with Sca1
+
cells that were
transduced with lenti -GFP or Sca1
+
-PDGFB or Sca1
+
-PDGFB-DSS6, and bone tissues were analyzed 10 weeks later.
Fig. 2. DSS6 endows a bone surface binding ability to GFP. As mentioned in the method section, the bone slices were incubated for 1 h with 50 ng/ml of green
uorescent protein (GFP) or GFP-DSS6. The bone slices were then rinsed extensively with PBS. Representative uorescence images of the GFP-DSS6-bone slices
conjugation are shown here.
W. Chen, et al. Bone Reports 12 (2020) 100236
2
formation. This would be expected to decrease the dose of the pre-
conditioning agent. We reasoned that a calcium-binding peptide at-
tached to the PDGFB might concentrate the PDGFB at the bone surface.
In this regard, DSS6 peptide (six repetitive sequences of aspartate,
serine, and serine), is a calcium-binding peptide and has been shown to
target the bone surface (Yarbrough et al., 2010;Zhang et al., 2012). Our
proposed strategy takes advantage of the high anity of a unique DSS6
peptide for calcium and employs this peptide as a novel targeting ve-
hicle to deliver and retain an osteogenic growth factor, PDGFB (in a
fusion protein form with DSS6), at the site of bone loss. The goal of the
present study was to perform a preliminary experiment to test our hy-
pothesis that marrow transplantation with Sca1
+
PDGFB-DSS6 would
exhibit greater bone formation than with Sca1
+
PDGFB. We chose the
head of the femur for our sampling site. We sought to evaluate ecacy
before proceeding with elaborate studies of the safety.
2. Material and methods
2.1. Animal study
Female C57BL/6J mice of 6 to 8 weeks of age were purchased from
the Jackson laboratory. All experimental protocols were approved by
the Institutional Animal Care and Use Committee at Loma Linda
University and the Animal Care and Use Review Oce of the United
States Department of the Army. In conducting research using animals,
the investigators adhered to the Animal Welfare Act Regulations and
other Federal statutes relating to animals and experiments involving
animals and the principles set forth in the current version of the Guide
for Care and Use of Laboratory Animals, National Research Council.
2.2. Ovariectomy surgery
Ovariectomy (OVX) surgery was conducted on 2-month old C57BL/
6 J female mice. Mice were anesthetized by an intraperitoneal injection
of 105 mg/kg ketamine and 21 mg/kg xylazine (in a total of ~0.1 ml
volume). Body temperature was maintained by a 37 °C recirculating-
water heating pad. The back and sides of the mice were shaved and
cleaned with 70% ethanol and Betadine. Under aseptic conditions, the
pair of ovaries was removed from the mice by dorsal incision into the
region between the dorsal hump and the base of the tail. Removal of the
mbrial end of the fallopian tube was done to ensure completeness of
the ovariectomy. The muscle incision was closed with 60 silk sutures,
and skin incisions closed with 30 silk sutures. Post-operative analgesic
(0.060 mg/kg in 0.05 ml buprenorphine, subcutaneously) was ad-
ministered for each mouse. After surgery, animals were treated for two
days, twice a day, with buprenorphine, and monitored closely there-
after. The animals were observed during recovery until alert and mo-
bile. The surgical procedure for control, sham-operated mice was the
same, except that the ovaries were not removed (Thompson et al.,
1995).
2.3. Bone marrow (BM) Sca1
+
cell isolation
Bone marrow Sca1
+
cell isolation was performed as previously
described (Chen et al., 2015). Briey, bone marrow cells were har-
vested from mice femurs and tibias, and Sca 1
+
cells were puried
using Sca1
+
MACS magnetic beads (MiltenyiBiotec, cat no 130-106-
641). Before viral transduction, cells were cultured for 48 h. in Iscove's
modied Dulbecco's medium (IMDM, Invitrogen) containing 10% FBS
(Invitrogen) and 100 ng/mL each of human TPO, mouse SCF, human
Flt3L, human IL-3, and human G-CSF (Chen et al., 2015).
2.4. Lentiviral vectors and transduction of Sca1
+
cells
Human PDGFB ORF was subcloned into a lentiviral vector and fused
with DSS6 peptide under the control of the PGK promoter. Lentiviral
packaging was performed as previous described (Chen et al., 2015). SIN
lentiviral vectors were produced by transient transfection of the vector
plasmid in human 293T cells along with helper plasmid (CMVdR8.74)
and envelope plasmid (MD.G). Sca 1
+
cells were transduced once at a
MOI of 2 for 6 h. in six- well nontissue culture-treated plates precoated
with RetroNectin (Takara). From here onwards the lenti Sca1
+
-GFP-
PGK-PDGFB construct will be mentioned as Sca1
+
-PDGFB and Sca1
+
-
GFP-PGK-PDGFB-DSS6 as Sca1
+
-PDGB-DSS6 throughout the article.
2.5. Transplantation
Two weeks after ovariectomy surgery, OVX mice were irradiated
with a
60
Co source (Eldorado model, Atomic Energy of Canada) at a
single dose of 8 Gy (0.543 Gy/min) in the Department of Radiation
Medicine of the Loma Linda University. Twenty-four hours later,
1×10
6
lentiviral transduced Sca1
+
cells were resuspended in 200 μL
Fig. 3. Bone marrow transplantation and % engraftment. Two weeks after the
OVX surgery, the C57/BL6 mice were divided into 2 groups, and transplanted
with Sca1
+
cells that were transduced with, 1) Lenti GFP-PDGFB (wild-type),
and 2) Lenti GFP-PDGFB-DSS6. OVX sham surgery animals were served as
control, and transplanted with Sca1+ cells that were transduced with Lenti
GFP. To ensure engraftment of hematopoietic stem/progenitor cells, mice were
myeloablated by irradiation at 8 Gy before transplantation. Ten weeks after
transplantation, the engraftment level was evaluated by analyzing the bone
marrow cells for % GFP
+
cells by FACS. Data are means ± SEM **P< .01,
ns = not signicant.
Fig. 4. High serum ALP levels were observed in the Sca1
+
PDGFB-DSS6 treated
animals, but not in the Sca1
+
PDGFB treated animals. The measurement was
conducted 10 weeks after transplantation. Data are means ± SEM. **P < .01,
***P< .001.
W. Chen, et al. Bone Reports 12 (2020) 100236
3
IMDM and transplanted into each recipient mouse via tail vein injection
under anesthesia. The mice were restrained in the mouse restrainer
during tail vein injection, and then properly returned to their cages
(Chen et al., 2015).
2.6. μCT analysis
AμCT analysis of the femoral bone was performed using a Scanco
VivaCT 40 instrument (Scanco Medical). Femurs were scanned at an
isotropic voxel size of 10.4 μm3, and energies of 55 keV and 70 keV
were used to scan the distal metaphysis and the midshaft, respectively.
For metaphyseal analysis, the region of interest included the in-
tramedullary space inside the endosteal surface at a distance proximal
to the condylar growth plate that was normalized for femur length. The
midshaft analysis region of interest used two contours to include the
cortex and exclude the intramedullary space, and was also normalized
for any variations in bone length. Within their respective regions of
interest, trabecular bone was segmented at a density of > 220 mg/cm
3
and the cortical bone segmented at a density of > 260 mg/cm
3
.
Trabecular and cortical partial bone volume (BV/TV) was determined.
Trabecular number, thickness and spacing were also examined, as was
the trabecular connectivity density. Cortical porosity was calculated as
1BV/TV (midshaft) from the two-contour examination of the fe-
moral midshaft cortex (Chen et al., 2015).
2.7. Bone strength analysis
The mechanical strength of the femurs was evaluated at midshaft by
the three-point bending test, using an Instron DynaMight 8841 servo-
hydraulic tester (Instron). Bones were stored frozen in saline-soaked
gauze, thawed, and rehydrated in saline before testing. The femur was
positioned on the tester with the anterior aspect upwards on supports
that were 2 mm in diameter and 7 mm apart. The bone was preloaded
to 1 N at the midshaft and then loaded to failure using a blade excursion
rate of 5.0 mm/s (Chen et al., 2015).
2.8. Serum Alkaline phosphatase measurement
Serum ALP activity was measured by the QuantiChrom ALP kit
(BioAssay Systems) (Chen et al., 2015).
2.9. Eect of PDGFB-DSS6 IV injections on trabecular bone formation
At 1 month after ovariectomy, mice were received with PBS or
PDGFB-DSS6 (0.5 mg/kg or 5 mg/kg) i.v. thrice per week for 4 weeks.
Representative von Kossa staining images from L3 vertebrae showed
increased trabecular bone formation following PDGFB-DSS6 treatment.
2.10. GFP-DSS6 binding to bone chips
We choose DSS6, a six repeating sequence of AspSerSer (Fang and
Galiano, 2008;Seeman, 2013), as a fusion partner of PDGFB to allow
PDGFB to target mineralized bone surface. We rst sought to conrm
that DSS6 could eciently bind and localize DSS6 fusion protein at the
bone surface in vitro. Bone chips were rst incubated with green
uorescent protein (GFP) or GFP-DSS6 (50 ng/ml) for 1 h, and then
washed twice with 1× PBS. All images were captured with an Olympus
BX51 microscope system (Olympus).
2.11. Statistical analysis
All data were presented as mean ± SEM. OVX Sham and OVX
treated groups were compared by Student t-test or one way ANOVA. In
other parts two-way and one-way ANOVA were used. A P-value < .05
were considered statistically signicant.
3. Results
3.1. Experimental strategy
The goal of this study was to study the bone anabolic ecacy of our
Fig. 5. Representative X-ray pictures of femurs harvested from mice received Sca1
+
GFP control, Sca1
+
PDGFB or Sca1
+
PDGFB-DSS6 cells. In each group, 67 mice
underwent OVX to induce osteopenia, followed by hematopoietic stem/progenitor cell transplantation 2 weeks later. Animals were analyzed at 10 weeks after
transplantation.
W. Chen, et al. Bone Reports 12 (2020) 100236
4
stem cell gene therapy in OVX mice. We compared Sca1
+
cells that
were transduced with PDGFB (control) vs. PDGFB-DSS6 (fusion protein
with bone specic targeting) to determine whether PDGFB-DSS6 was
superior to PDGFB alone in anabolic action. The primary end-point was
the amount and distribution of the new bone formed in response to our
stem cell gene therapy. Secondary end-points include x-ray imaging of
long bones, and serum alkaline phosphatase. Animals were divided into
4 groups: OVX sham, OVX GFP (untreated), OVX Sca1
+
-PDGFB and
OVX Sca1
+
- PDGFB-DSS6 (bone targeting). Fig. 1 shows the general
experimental design using the lenti transduced Sca1
+
cells.
GFP-DSS6 binds to bone slices.
We rst determined the bone-targeting ability of DSS6. Incubation
of green uorescent protein (GFP)-DSS6 fusion protein with bone slices
produced an intense green uorescence as compared with GFP alone,
demonstrating that DSS6 fusion protein can target and bind to the bone
(Fig. 2).
3.2. Bone marrow transplantation
After total body irradiation each mouse in the stem cell therapy
group was injected with 1 million transduced Sca1
+
cells in-
travenously. To evaluate the level of engraftment of the Sca1
+
cells,
GFP
+
cells in peripheral blood was measured at 1 and 2 months after
transplantation by FACS analysis. In our Sca1
+
GFP-OVX control mice,
engraftment was about 18%, which is similar to what we have seen in
the past (Chen et al., 2015)(Fig. 3). However, in the two OVX groups
(PDGFB and PDGFBDSS6) engraftment was only ~8% (Fig. 3). These
results raise the possibility that OVX reduces the engraftment of Sca1
+
cells.
3.3. Serum ALP at 10 weeks post therapy
At 10 weeks after the stem cell gene therapy, we determined the
serum ALP levels, which is a serum biomarker of bone formation.
OVX
Sham
Sca1-
GFP
Sca1-
PDGFB
Sca1-
PDGFB-
DSS6
0.000
0.005
0.010
0.015
0.020
0.4
0.5
0.6
0.7
0.8
BV/TV%
****
0
5
10
15
Tb.N (mm-1)
****
0.00
0.02
0.04
0.06
0.08
TB-Th(mm)
*
*
****
OVX Sham
Sca1-GFP
Sca1-PDGFB
Sca1-PDGFB DSS6
0
5
10
15
20
25
600
800
1000
Conn. Den (mm-3)
****
0
2
4
6
8
Cortical Porosity (%)
ns
****
****
***
A.
B.
OVX Sham
Sca1-GFP
Sca1-PDGFB
Sca1-PDGFB DSS6
Fig. 6. A) Representative MicroCT 3D pictures
of femurs harvested from mice received GFP
control, PDGFB or PDGFB-DSS6 overexpressing
Sca 1
+
cells. In each group, OVX was performed
in 67 mice to induce osteopenia, followed by
hematopoietic stem/progenitor cell transplan-
tation 2 weeks later. Animals were analyzed at
10 weeks after transplantation. MicroCT three
dimensional bone structure analysis of femurs
from OVX osteoporosis mouse after treatment
with Sca1
+
-PDGFB or Sca1
+
-PDGFB-DSS6.
PDGFB-DSS6 is a fusion protein of PDGFB and
bone-surface binding peptide DSS6. B)
Specimens were analyzed by microCT at
10 weeks after transplantation (n=47). The
following parameters of new bone formation
from the microCT analysis are shown: trabe-
cular partial bone volume (BV/TV); con-
nectivity density (Conn. Density); trabecular
number (Tb.N); trabecular thickness (Tb.Th),
and cortical porosity. Data are means ± SEM.
****P< .0001.
W. Chen, et al. Bone Reports 12 (2020) 100236
5
Compared to the OVX sham group, there was no signicant increase in
the Sca1
+
group or the Sca1
+
-PDGFB group. However, the serum ALP
was 75% increase in the Sca1
+
-PDGFB-DSS6 group compared to the
Sca1
+
-GFP control group (Fig. 4).
3.4. X-ray of the femur long bones
We rst determined bone mineral density of femurs at 10 weeks
after therapy by X-ray imaging. X-rays showed increased trabecular
bone, particularly in the metaphysis and femoral trochanter, and cor-
tical thickening in the Sca 1
+
-PDGFB-DSS6 compared to the Sca1
+
-
PDGFB and Sca1
+
groups (Fig. 5).
3.5. μCT of femur
In the Sca1
+
PDGFB-DSS6 group there was a massive increase in
trabecular bone density of the femur, particularly in the metaphysis
compared to the Sca1
+
GFP control and the Sca1
+
- PDGFB. No dif-
ference was seen in any of the μCT parameters between the Sca1
+
GFP
control and the Sca1
+
PDGFB (Fig. 6A).Nodierences in bone density
were observed between the sham OVX control and the Sca1
+
GFP
control. However we saw highly signicant increases in cortical por-
osity in the Sca1
+
GFP group and in the Sca1
+
-PDGFB group compared
to the non-OVX group. Accordingly, in this study statistically signicant
dierences in microCT bone parameters in response to OVX were ob-
served only in cortical porosity (Fig. 6B).
Strikingly, we observed a > 20-fold increase in BV/TV in the Sca1
+
PDGFB-DSS6 compared to the Sca1
+
PDGFB or the Sca 1-GFP controls.
In addition, trabecular number, trabecular thickness and trabecular
activity were all much greater in the Sca1
+
PDGFB-DSS6 that in the
Sca1
+
PDGFB or Sca1
+
GFP (Fig. 6B).
The connectivity density after Sca1
+
-PDGFB-DSS6 treatment
was > 50-fold higher than the Sca1
+
- PDGFB group, suggesting that
our therapy has a strong ability to cause de novo bone formation; i.e.,
trabecular bone formation where there was no bone before. We found
increased cortical porosity in Sca1
+
-GFP or Sca1
+
-PDGFB treated mice,
likely due to OVX-induced bone loss. However, Sca1
+
-PDGFB-DSS6
treatment completely replenished the lost cortical bone. (Fig. 6B).
These data demonstrate that the PDGFB-DSS6 therapy induces de novo
bone formation in OVX marrow cavity and corrects cortical bone loss
due to OVX.
3.6. Bone strength analysis
We then asked whether the increased trabecular connectivity and
decreased cortical porosity would translate into increased strength.
Bone strength was measured by the 3-point bending of the right femurs.
As expected, the Sca1
+
PDGFB-DSS6 group showed a signicant in-
crease in bone strength compared to the other 3 test groups, including
the OVX sham (Fig. 7).
3.7. PDGF-DSS6 protein enhances trabecular bone formation
The striking results of PDGF-DSS6 gene therapy in OVX mice en-
couraged us to develop the PDGF-DSS6 protein to avoid marrow
transplantation. To this purpose, we have conducted preliminary stu-
dies. IV injection of PDGFB-DSS6 not only increased bone formation in
femurs (not shown), but also in lumbar vertebrae (Fig. 8) in a dose-
dependent manner. These results demonstrate that injection of PDGFB-
DSS6 protein is ecacious.
4. Discussion
The most salient nding in the present study was huge amount of
bone formation produced by our Sca1
+
PDGB-DSS6 stem cell gene
therapy, which targeted the PDGFB therapy to the bone surface by
virtue of DSS6, which is a calcium binding peptide. Here the Sca1 cells
are the source for the abundant production of PDGFB-DSS6. This ana-
bolic action of PDGF with the PDGF-DSS6 construct was achieved at a
much lower Sca1
+
cell engraftment compared to our earlier studies
(Chen et al., 2015), strongly suggesting a substantial reduction in the
ratio of PDGFB produced per unit of the amount of bone formed. The
future signicance of this lower required anabolic dose is that it should
reduce the preconditioning dose required for an optimal anabolic ac-
tion. Moreover, the overall lower synthesis of PDGFB should reduce o
target eects with HSC PDGFB-DSS6 stem cell gene therapy.
In the PDGFB-DSS6 group, there was a large increase in trabecular
bone formation. This was associated with an enormous increase in
trabecular connectivity. Also there was new trabecular bone formation
in sites where trabecular bone had not been present in the past, de novo
bone formation. Another advantage of this therapy is to decrease in
cortical porosity, which is known to be important for the preservation
of bone strength (Augat and Schorlemmer, 2006). Parathyroid hormone
like therapeutic agents are also known to produce increased trabecular
bone formation (Calvi et al., 2001); whereas, Romosozumab apparently
can form new bone, in cortical as well as trabecular regions (McClung
et al., 2018;McClung et al., 2014;Graeet al., 2015) however, the
robust anabolic eect of treatment is limited to the rst few months of
therapy(McClung, 2018). In our previous work we found that PDGFB
stem cell therapy increases bone formation in vivo in red and in fatty
marrow(Chen et al., 2015). In our earlier study we found evidence for
dierentiation capacity from the treatment of Sca1
+
cells over-
expressing PDGFB. We found an increase in BMP 2 expression, which
presented a conundrum because PDGFB alone does not cause bone cell
Fig. 7. The Sca1
+
-PDGFB-DSS6 treatment increases bone strength.
Representative loading force displacement graph presented. Three-point
bending test was used to measure bone strength at the midshaft of the femur.
Maximum load-to-failure on Sca1
+
-PDGFB-DSS6 treated femurs was sig-
nicantly greater than that of the other groups. Data are means ± SEM.
**P< .01, ***P< .001, ****P < .0001.
Fig. 8. PDGFB-DSS6 protein promotes bone formation in lumbar vertebrae in
OVX mice. At 1 month after ovariectomy, mice were received with PBS or
PDGFB-DSS6 (0.5 mg/kg or 5 mg/kg) i.v. thrice per week for 4 weeks.
Representative von Kossa staining images from L3 vertebrae showed increased
trabecular bone formation following PDGFB-DSS6 treatment.
W. Chen, et al. Bone Reports 12 (2020) 100236
6
dierentiation in vitro. We concluded that PDGFB had some type of
secondary action in the marrow space which led to an increase in BMP
2. It requires the marrow space for PDGFB to have this action because
when PDGFB was given to rats it did not increase periosteal bone for-
mation (Mitlak et al., 1996). In our previous study of the treatment of
normal mice with PDGFB via overexpressing Sca1 cells we measured
both bone formation and resorption markers. We saw evidence of a
modest increase in bone resorption but a much greater increase in bone
formation (Chen et al., 2015).
We also found that PDGFB stem cell therapy was prolonged, to the
extent that the new trabecular bone takes a considerable portion of the
marrow cavity. This is not a disadvantage, because by using tran-
scriptional genetic control, it should be possible to utilize this stem cell
gene therapy according to the monitored skeletal needs of the patient;
viz, bone density and serum bone formation markers.
In the present study and in our previous study we found that the
increase in bone formation was attended by an increase in bone
strength. Because of the eect of our therapy to increase endosteal and
trabecular bone, and because our therapy increases bone formation in
both red marrow and fatty marrow sites, it is anticipated that endosteal
and trabecular bone formation would be increased throughout the
skeleton with this novel therapeutic regimen.
There were 2 unexpected ndings in our study. First, in the 2 groups
that were OVX, the engraftment of our Sca1
+
cells was somewhat less
than we have seen previously in non-OVX mice (Chen et al., 2015;Hall
et al., 2007). In this regard, there are several studies suggesting that
estradiol has an impact on hematopoietic stem cells. Accordingly, in
mice estradiol increases the retention of hematopoietic stem cells in the
vascular niche in the bone marrow (Kim et al., 2017). Also estrogen
receptor alpha promotes mouse hematopoietic stem cell regeneration
(Chapple et al., 2018). Estrogen also promotes hematopoietic stem cell
renewal (Nakada et al., 2014). Probably relevant to the current study,
radiation signicantly decreased marrow cellularity and increased the
amount of marrow adipose tissue in OVX mice compared to intact mice
(Hui et al., 2012). In aggregate, these ndings suggest that OVX may
have modestly impaired engraftment of our Sca 1 cells. Consequently,
in the 2 groups of mice with OVX and PDGFB therapy there was a
signicant decrease in engraftment. This unexpected nding revealed
the greater potency for increases in bone density in the PDGFB-DSS6
group over the PDGFB group.
Second, we did not detect a drop in total or cortical bone density in
response to OVX control mice. We have no denitive explanation for
these negative results. This could be related to the fact that there was a
simultaneously decrease in ovarian estrogen production and a bone
repletion formation process following radiation preconditioning, which
causes bone loss (Hu et al., 2010;Georgiou et al., 2012). In any case, we
did see a statistically signicant increase in cortical porosity in the
Sca1
+
GFP in the Sca1
+
PDGFB groups, compared to the non-OVX
group. It seems likely that this is an OVX consequence. Noteworthy is
that the Sca1
+
PDGFB-DSS6 group completely eliminated the increased
porosity attributable to OVX. Thus our therapy mainly counteracts the
eects of OVX also the well-established eects of radiation bone da-
mage (Hu et al., 2010;Georgiou et al., 2012). Once marrow trans-
plantation is conducted with nontoxic preconditioning (see below) the
anabolic eects of our stem cell gene therapy may be even more sub-
stantial. One interesting potential of our stem cell gene therapy is a
possibility that mechanical loading at specic sites of the skeleton
would act to determine the distribution of new bone formation, al-
lowing for fortication of bone strength at specic sites considered to
be a risk for fracture. Considerable future work needs to be accom-
plished to optimize our stem cell gene therapy.
An alternative to our marrow transplant for targeting our anabolic
stem cell gene therapy to the sites of bone loss would be to utilize
PDGFB-DSS6 fusion protein in insulin pump type of administration or
by subcutaneous injection. However in both of these approaches there
would be a greater chance for extra skeletal eects of PDGFB than what
would occur with our Sca 1
+
PDGFB-DSS6 therapeutic. With regards to
the alternative strategy, our preliminary data with the direct IV injec-
tions of PDGFB-DSS6 protein increased the bone formation in in lumbar
vertebrae (Fig. 8) in a dose-dependent manner. These results demon-
strate that injection of PDGFB-DSS6 protein is ecacious.
In regard to safety in our previous study we saw no changes in
peripheral red blood count of white blood count and no changes in soft
tissue organs with the exception an enlarged spleen. We attributed the
increased spleen size to extra medullary hematopoiesis, because much
of the marrow cavity was overtaken by trabecular bone (Chen et al.,
2015). Regarding the adverse eects from preconditioning, there are
novel approaches in the process of development that could substantially
reduce the risk associated with preconditioning. Accordingly, an anti-
body technology to reduce the endogenous CD45 HSC, c-kit, is now in
clinical trials (Czechowicz et al., 2018). This therapy does not require
any toxic preconditioning and the only risk is a reduced white blood cell
count for approximately 2 weeks. Also, a valine decient diet, which
has a temporary nutritionally toxic eect on hematopoietic stem cells,
has shown positive results for preconditioning in both mice and humans
(Taya et al., 2016). Therefore, in the future there is a possibility that the
adverse eects of preconditioning can be minimized to the extent that
the benet to risk ratio would be suciently high to justify HSC-PDGFB
therapy for a subpopulation of severely osteoporotic patients. This
therapy would be expensive but current anabolic therapies are also
expensive (Martin, 2014).
As with any anabolic agent, there is a concern that PDGFB therapy
might cause oncogenic eects on extra skeletal bone or organ brosis.
That PDGFB does not produce heterotrophic bone formation was re-
cently demonstrated in the publication on four subjects with Kosaki
disease, which is caused by a gain of function through overexpression of
PDGFRB (Minatogawa et al., 2017;Gawlinski et al., 2018). These pa-
tients exhibited longitudinal skeletal overgrowth without heterotrophic
bone formation or evidence of oncogenesis. A soft tissue untoward ef-
fect was myobromatosis. Nonetheless, for our therapy further onco-
genic studies of and brotic actions will be required in future studies.
Only limitation of our current study is the lack of tissue distribution
analysis of PDGFB-DSS6. Relevant to this issue is that when PDGFB was
given systemically to rats, there is an increase in bone formation but
this was attended by liver and kidney brosis(Mitlak et al., 1996),
which was most likely due to an elevated blood level of PDGFB.
However, in our previous study where we targeted the PDGFB with the
bone marrow transplant, we saw no increase in brosis of soft tissue
organs and there was no increase in serum PDGFB (Chen et al., 2015).
With our current study there would be even less likelihood increased
serum PDGFB or increased soft tissue brosis because of the targeting
action of DSS6. In any case, it will be important in future studies to
perform a thorough tissue distribution of PDGFB and of PDGFB-DSS6.
In conclusion, the results of this proof of principle study together
with those ndings in our previous study (Chen et al., 2015) strongly
suggest that our robust anabolic agent coupled with successful skeletal
targeting mechanism has the potential to increase bone density and
strength at sites in the skeleton were bone is lost during osteoporosis.
For this therapy to be applied clinically, new strategies in bone marrow
transplant conditioning will need to be substituted for toxic pre-
conditioning (Hui et al., 2012).
Acknowledgements
Funding
This work was supported by the Telemedicine and Advanced
Technology Research Center (TATRC) at the US Army Medical Research
and Material Command (USAMRMC) under grant no. W81XWH-12-1-
0023 (DJB). The views, opinions and/or ndings contained in this re-
port are those of the author(s) and should not be construed as an ocial
Department of the Army position, policy or decision unless so
W. Chen, et al. Bone Reports 12 (2020) 100236
7
designated by other documentation.
Author contributions
WC, SW, YF, LA, and CHR, performed the experiments. CHR, K-
HWL, DJB, and XZ interpreted the data. XZ and DJB conceived, di-
rected, and supervised the study. K-HWL provided critical reading of
the manuscript, all authors reviewed the manuscript.
Declaration of competing interests
The authors declare that they have no competing interests.
References
Ascenzi, M.G., et al., 2012. Parathyroid hormone treatment improves the cortical bone
microstructure by improving the distribution of type I collagen in postmenopausal
women with osteoporosis. J. Bone Miner. Res. 27 (3), 702712.
Augat, P., Schorlemmer, S., 2006. The role of cortical bone and its microstructure in bone
strength. Age Ageing 35 (Suppl. 2), ii27ii31.
Calvi, L.M., et al., 2001. Activated parathyroid hormone/parathyroid hormone-related
protein receptor in osteoblastic cells dierentially aects cortical and trabecular
bone. J. Clin. Invest. 107 (3), 277286.
Chanda, D., Kumar, S., Ponnazhagan, S., 2010. Therapeutic potential of adult bone
marrow-derived mesenchymal stem cells in diseases of the skeleton. J. Cell. Biochem.
111 (2), 249257.
Chapple, R.H., et al., 2018. ERalpha promotes murine hematopoietic regeneration
through the Ire1alpha-mediated unfolded protein response. Elife 7.
Chen, W., et al., 2015. PDGFB-based stem cell gene therapy increases bone strength in the
mouse. Proc. Natl. Acad. Sci. U. S. A. 112 (29), E3893E3900.
Czechowicz, A., et al., 2018. Selective HSC-ablation using CD117 antibody-drug-con-
jugates enables safe and eective murine and human hematopoietic stem cell
transplantation. Mol. Ther. 26 (5), 2324.
Fang, R.C., Galiano, R.D., 2008. A review of becaplermin gel in the treatment of diabetic
neuropathic foot ulcers. Biologics 2 (1), 112.
Farr, J.N., et al., 2016. Identication of senescent cells in the bone microenvironment. J.
Bone Miner. Res. 31 (11), 19201929.
Gawlinski, P., et al., 2018. Phenotype expansion and development in Kosaki overgrowth
syndrome. Clin. Genet. 93 (4), 919924.
Georgiou, K.R., Hui, S.K., Xian, C.J., 2012. Regulatory pathways associated with bone loss
and bone marrow adiposity caused by aging, chemotherapy, glucocorticoid therapy
and radiotherapy. Am J Stem Cells 1 (3), 205224.
Grae, C., et al., 2015. Administration of romosozumab improves vertebral trabecular
and cortical bone as assessed with quantitative computed tomography and nite
element analysis. Bone 81, 364369.
Gutteridge, D.H., et al., 2002. A randomized trial of sodium uoride (60 mg) +/es-
trogen in postmenopausal osteoporotic vertebral fractures: increased vertebral frac-
tures and peripheral bone loss with sodium uoride; concurrent estrogen prevents
peripheral loss, but not vertebral fractures. Osteoporos. Int. 13 (2), 158170.
Hall, S.L., et al., 2007. Sca-1(+) hematopoietic cell-based gene therapy with a modied
FGF-2 increased endosteal/trabecular bone formation in mice. Mol. Ther. 15 (10),
18811889.
Hu, W.W., et al., 2010. Bone regeneration in defects compromised by radiotherapy. J.
Dent. Res. 89 (1), 7781.
Hui, S.K., et al., 2012. The inuence of therapeutic radiation on the patterns of bone
marrow in ovary-intact and ovariectomized mice. PLoS One 7 (8), e42668.
Kanis, J.A., 1994. Assessment of fracture risk and its application to screening for post-
menopausal osteoporosis: synopsis of a WHO report. WHO Study Group. Osteoporos
Int 4 (6), 368381.
Kawai, M., et al., 2011. Emerging therapeutic opportunities for skeletal restoration. Nat.
Rev. Drug Discov. 10 (2), 141156.
Kim, J.Y., et al., 2017. Multicenter phase II clinical trial of Genexol-PM(R) with gemci-
tabine in advanced biliary tract cancer. Anticancer Res. 37 (3), 14671473.
Kleerekoper, M., Balena, R., 1991. Fluorides and osteoporosis. Annu. Rev. Nutr. 11,
309324.
Kleerekoper, M., et al., 1991. A randomized trial of sodium uoride as a treatment for
postmenopausal osteoporosis. Osteoporos. Int. 1 (3), 155161.
Martin, T.J., 2014. Bone biology and anabolic therapies for bone: current status and fu-
ture prospects. J Bone Metab 21 (1), 820.
McClung, M.R., 2018. Romosozumab for the treatment of osteoporosis. Osteoporos
Sarcopenia 4 (1), 1115.
McClung, M.R., et al., 2014. Romosozumab in postmenopausal women with low bone
mineral density. N. Engl. J. Med. 370 (5), 412420.
McClung, M.R., et al., 2018. Eects of 24 months of treatment with romosozumab fol-
lowed by 12 months of Denosumab or placebo in postmenopausal women with low
bone mineral density: a randomized, double-blind, phase 2, parallel group study. J.
Bone Miner. Res. 33 (8), 13971406.
Minatogawa, M., et al., 2017. Expansion of the phenotype of Kosaki overgrowth syn-
drome. Am. J. Med. Genet. A 173 (9), 24222427.
Minisola, S., et al., 2017. New anabolic therapies for osteoporosis. Intern. Emerg. Med. 12
(7), 915921.
Mitlak, B.H., et al., 1996. The eect of systemically administered PDGF-BB on the rodent
skeleton. J. Bone Miner. Res. 11 (2), 238247.
Nakada, D., et al., 2014. Oestrogen increases haematopoietic stem-cell self-renewal in
females and during pregnancy. Nature 505 (7484), 555.
Neer, R.M., et al., 2001. Eect of parathyroid hormone (1-34) on fractures and bone
mineral density in postmenopausal women with osteoporosis. N. Engl. J. Med. 344
(19), 14341441.
Osteoporosis: Fragility Fracture Risk: Osteoporosis: Assessing the Risk of Fragility
Fracture. London.
Panula, J., et al., 2011. Mortality and cause of death in hip fracture patients aged 65 or
older: a population-based study. BMC Musculoskelet. Disord. 12, 105.
Papapoulos, S.E., 2015. Anabolic bone therapies in 2014: new bone-forming treatments
for osteoporosis. Nat Rev Endocrinol 11 (2), 6970.
Rubin, M.R., Bilezikian, J.P., 2002. New anabolic therapies in osteoporosis. Curr. Opin.
Rheumatol. 14 (4), 433440.
Rubin, C.D., et al., 2001. Sustained-release sodium uoride in the treatment of the elderly
with established osteoporosis. Arch. Intern. Med. 161 (19), 23252333.
Schnell, S., et al., 2010. The 1-year mortality of patients treated in a hip fracture program
for elders. Geriatr Orthop Surg Rehabil 1 (1), 614.
Seeman, E., 2013. Age- and menopause-related bone loss compromise cortical and tra-
becular microstructure. J. Gerontol. A Biol. Sci. Med. Sci. 68 (10), 12181225.
Taya, Y., et al., 2016. Depleting dietary valine permits nonmyeloablative mouse hema-
topoietic stem cell transplantation. Science 354 (6316), 11521155.
Thompson, D.D., et al., 1995. FDA guidelines and animal models for osteoporosis. Bone
17 (4 Suppl), 125S133S.
Yarbrough, D.K., et al., 2010. Specic binding and mineralization of calcied surfaces by
small peptides. Calcif. Tissue Int. 86 (1), 5866.
Zhang, G., et al., 2012. A delivery system targeting bone formation surfaces to facilitate
RNAi-based anabolic therapy. Nat. Med. 18 (2), 307314.
W. Chen, et al. Bone Reports 12 (2020) 100236
8
... Moreover, as a homodimer of PDGFB, plasma PDGF-BB levels are maintained by estrogen in healthy young women and play a major role in postmenopausal osteoporosis [37]. Furthermore, several studies highlighted PDGFB as possible therapeutic target for osteoporosis [38][39][40]. Finally, Ye et al. [41,42] reported that PAFAH1B1 (LIS1), another gene identified to be differentially expressed in the current study, can promote osteoclastogenesis via regulating both the differentiation and survival of osteoclast progenitors. ...
Article
Full-text available
Osteoporosis is a severe chronic skeletal disorder that affects older individuals, especially postmenopausal women. However, molecular biomarkers for predicting the risk of osteoporosis are not well characterized. The aim of this study was to identify combined biomarkers for predicting the risk of osteoporosis using machine learning methods. We merged three publicly available gene expression datasets (GSE56815, GSE13850, and GSE2208) to obtain expression data for 6354 unique genes in postmenopausal women (45 with high bone mineral density and 45 with low bone mineral density). All machine learning methods were implemented in R, with the GEOquery and limma packages, for dataset download and differentially expressed gene identification, and a nomogram for predicting the risk of osteoporosis was constructed. We detected 378 significant differentially expressed genes using the limma package, representing 15 major biological pathways. The performance of the predictive models based on combined biomarkers (two or three genes) was superior to that of models based on a single gene. The best predictive gene set among two-gene sets included PLA2G2A and WRAP73. The best predictive gene set among three-gene sets included LPN1, PFDN6, and DOHH. Overall, we demonstrated the advantages of using combined versus single biomarkers for predicting the risk of osteoporosis. Further, the predictive nomogram constructed using combined biomarkers could be used by clinicians to identify high-risk individuals and in the design of efficient clinical trials to reduce the incidence of osteoporosis.
Article
Clinical bone-morphogenetic protein 2 (BMP2) treatment for bone regeneration, often resulting in complications like soft tissue inflammation and ectopic ossification due to high dosages and non-specific delivery systems, necessitates research into improved biomaterials for better BMP2 stability and retention. To tackle this challenge, we introduced a groundbreaking bone-targeted, lipoplex-loaded, three-dimensional bioprinted bilayer scaffold, termed the polycaprolactone-bioink-nanoparticle (PBN) scaffold, aimed at boosting bone regeneration. We encapsulated BMP2 within the fibroin nanoparticle based lipoplex (Fibroplex) and functionalized it with DSS6 for bone tissue-specific targeting. 3D printing technology enables customized, porous PCL scaffolds for bone healing and soft tissue growth, with a two-step bioprinting process creating a cellular lattice structure and a bioink grid using gelatin-alginate hydrogel and DSS6-Fibroplex, shown to support effective nutrient exchange and cell growth at specific pore sizes. The PBN scaffold is predicted through in silico analysis to exhibit biased BMP2 release between bone and soft tissue, a finding validated by in vitro osteogenic differentiation assays. The PBN scaffold was evaluated for critical calvarial defects, focusing on sustained BMP2 delivery, prevention of soft tissue cell infiltration, and controlled fiber membrane pore size in vivo. The PBN scaffold demonstrated a more than eight times longer BMP2 release time than that of the collagen sponge, promoting osteogenic differentiation and bone regeneration in a calvarial defect animal. Our findings suggest that the PBN scaffold enhanced the local concentration of BMP2 in bone defects through sustained release and improved the spatial arrangement of bone formation, thereby reducing the risk of heterotopic ossification.
Article
Full-text available
Osteoporosis is a systemic metabolic bone disease with characteristics of bone loss and microstructural degeneration. The personal and societal costs of osteoporosis are increasing year by year as the ageing of population, posing challenges to public health care. Homing disorders, impaired capability of osteogenic differentiation, senescence of mesenchymal stem cells (MSCs), an imbalanced microenvironment, and disordered immunoregulation play important roles during the pathogenesis of osteoporosis. The MSC transplantation promises to increase osteoblast differentiation and block osteoclast activation, and to rebalance bone formation and resorption. Preclinical investigations on MSC transplantation in the osteoporosis treatment provide evidences of enhancing osteogenic differentiation, increasing bone mineral density, and halting the deterioration of osteoporosis. Meanwhile, the latest techniques, such as gene modification, targeted modification and co‐transplantation, are promising approaches to enhance the therapeutic effect and efficacy of MSCs. In addition, clinical trials of MSC therapy to treat osteoporosis are underway, which will fill the gap of clinical data. Although MSCs tend to be effective to treat osteoporosis, the urgent issues of safety, transplant efficiency and standardization of the manufacturing process have to be settled. Moreover, a comprehensive evaluation of clinical trials, including safety and efficacy, is still needed as an important basis for clinical translation.
Article
Full-text available
Hematopoietic stem cell transplantation (HSCT) is a curative therapy for blood and immune diseases with potential for many settings beyond current standard-of-care. Broad HSCT application is currently precluded largely due to morbidity and mortality associated with genotoxic irradiation or chemotherapy conditioning. Here we show that a single dose of a CD117-antibody-drug-conjugate (CD117-ADC) to saporin leads to > 99% depletion of host HSCs, enabling rapid and efficient donor hematopoietic cell engraftment. Importantly, CD117-ADC selectively targets hematopoietic stem cells yet does not cause clinically significant side-effects. Blood counts and immune cell function are preserved following CD117-ADC treatment, with effective responses by recipients to both viral and fungal challenges. These results suggest that CD117-ADC-mediated HSCT pre-treatment could serve as a non-myeloablative conditioning strategy for the treatment of a wide range of non-malignant and malignant diseases, and might be especially suited to gene therapy and gene editing settings in which preservation of immunity is desired.
Article
Full-text available
Over 12 months, romosozumab increased bone formation and decreased bone resorption, resulting in increased BMD in postmenopausal women with low BMD (NCT00896532). Herein we report the study extension evaluating 24 months treatment with romosozumab, discontinuation of romosozumab, alendronate followed by romosozumab, and romosozumab followed by denosumab. Postmenopausal women age 55–85 years with a lumbar spine (LS), total hip (TH), or femoral neck T‐score ≤–2.0 and ≥–3.5 were enrolled and randomly assigned to placebo, one of five romosozumab regimens (70mg, 140mg, 210mg monthly [QM]; 140mg Q3M; 210mg Q3M) for 24 months, or open‐label alendronate for 12 months followed by romosozumab 140mg QM for 12 months. Eligible participants were then re‐randomized 1:1 within original treatment groups to placebo or denosumab 60mg Q6M for an additional 12 months. Percentage change from baseline in BMD and bone turnover markers (BTMs) at months 24 and 36 and safety were evaluated. Of 364 participants initially randomized to romosozumab, placebo, or alendronate, 315 completed 24 months of treatment and 248 completed the extension. Romosozumab markedly increased LS and TH BMD through month 24, with largest gains observed with romosozumab 210mg QM (LS = 15.1%; TH = 5.4%). Women receiving romosozumab who transitioned to denosumab continued to accrue BMD, whereas BMD returned toward pretreatment levels with placebo. With romosozumab 210mg QM, bone formation marker P1NP initially increased following treatment initiation and gradually decreased to below baseline by month 12, remaining below baseline through month 24; and bone resorption marker β‐CTX rapidly decreased following treatment, remaining below baseline through month 24. Transition to denosumab further decreased both BTMs, while after transition to placebo, P1NP returned to baseline and β‐CTX increased above baseline. Adverse events were balanced between treatment groups through month 36. These data suggest that treatment effects of romosozumab are reversible upon discontinuation and further augmented by denosumab. This article is protected by copyright. All rights reserved
Article
Full-text available
Romosozumab, a specific inhibitor of sclerostin, is a unique approach to therapy for postmenopausal osteoporosis and related disorders. The elucidation of sclerostin deficiency as the molecular defect of syndromes of high bone mass with normal quality, and the pivotal role of sclerostin as a mediator of osteoblastic activity and bone formation, provided the platform for the evaluation of inhibitors of sclerostin to activate bone formation. An extensive preclinical program and 2 large fracture endpoint trials with romosozumab, a sclerostin-binding antibody, have been completed. This review will highlight the results of those studies and describe the current status of romosozumab as a potential therapy for osteoporosis. Keywords: Romosozumab, Sclerostin, Osteoporosis, Fracture risk, Manuscript category
Article
Full-text available
Activation of the unfolded protein response (UPR) sustains protein homeostasis (proteostasis) and plays a fundamental role in tissue maintenance and longevity of organisms. Long-range control of UPR activation has been demonstrated in invertebrates, but such mechanisms in mammals remain elusive. Here, we show that the female sex hormone estrogen regulates the UPR in hematopoietic stem cells (HSCs). Estrogen treatment increases the capacity of HSCs to regenerate the hematopoietic system upon transplantation and accelerates regeneration after irradiation. We found that estrogen signals through estrogen receptor α (ERα) expressed in hematopoietic cells to activate the protective Ire1α-Xbp1 branch of the UPR. Further, ERα-mediated activation of the Ire1α-Xbp1 pathway confers HSCs with resistance against proteotoxic stress and promotes regeneration. Our findings reveal a systemic mechanism through which HSC function is augmented for hematopoietic regeneration.
Article
We expand the KOGS phenotype by over 70% to include 24 unreported KOGS symptoms, in a first male patient, the third overall associated with the PDGFRB c.1751C>G, p.(Pro584Arg) mutation. 18 of these symptoms are unique to our patient, the remaining 6 are shared with other patients. Of the 24 unreported features overall, 6 show marked phenotype evolution and varying time of onset. The triangular face detected at 14 months and long palpebral fissures with lateral ectropion at 4 years are present in other members of the cohort. The remaining 4 are unique to Patient 5: pronounced macrocephaly from birth, increasingly triangular anterior skull from 14 months, camptodactyly, emerging at 4 years and worsening joint contractures from 6 years. Compilation of all new symptoms reported here with published clinical data further identifies at least 18 clinical parameters common to all cases to date, encompassing both known KOGS-associated PDGFRB mutations. We therefore propose a set of 18 core KOGS symptoms, with 16 present in early childhood. These results should also impact diagnostic/prognostic scope, intervention and outcome potential for KOGS patients, particularly for developmentally progressive conditions such as scoliosis and myofibroma.
Article
Osteoporosis is characterized by low bone mass and qualitative structural abnormalities of bone tissue, leading to increased bone fragility that results in fractures. Pharmacological therapy is aimed at decreasing the risk of fracture, mainly correcting the imbalance between bone resorption and formation at the level of bone remodeling units. Anabolic therapy has the capability to increase bone mass to a greater extent than traditional antiresorptive agents. The only currently available drug licensed is parathyroid hormone 1–34 (teriparatide); new drugs are on the horizon, targeting the stimulation of bone formation, and therefore improving bone mass, structure and ultimately skeletal strength. These are represented by abaloparatide (a 34-amino acid peptide which incorporates critical N-terminal residues, shared by parathyroid hormone and parathyroid hormone-related protein, followed by sequences unique to the latter protein) and romosozumab (an antibody to sclerostin). In the future, the availability of new anabolic treatment will allow a more extensive utilization of additive and sequential approach, with the goal of both prolonging the period of treatment and, more importantly, avoiding the side effects consequent to long-term use of traditional drugs.
Article
Skeletal overgrowth is a characteristic of several genetic disorders that are linked to specific molecular signaling cascades. Recently, we established a novel overgrowth syndrome (Kosaki overgrowth syndrome, OMIM #616592) arising from a de novo mutation in PDGFRB, that is, c.1751C>G p.(Pro584Arg). Subsequently, other investigators provided in vitro molecular evidence that this specific mutation in the juxtamembrane domain of PDGFRB causes an overgrowth phenotype and is the first gain-of-function point mutation of PDGFRB to be reported in humans. Here, we report the identification of a mutation in PDGFRB, c.1696T>C p.(Trp566Arg), in two unrelated patients with skeletal overgrowth, further confirming the existence of PDGFRB-related overgrowth syndrome arising from mutations in the juxtamembrane domain of PDGFRB. A review of all four of these patients with an overgrowth phenotype and PDGFRB mutations revealed postnatal skeletal overgrowth, premature aging, cognitive impairment, neurodegeneration, and a prominent connective tissue component to this complex phenotype. From a functional standpoint, hypermorphic mutations in PDGFRB lead to Kosaki overgrowth syndrome, infantile myofibromatosis (OMIM #228550), and Penttinen syndrome (OMIM #601812), whereas hypomorphic mutations lead to idiopathic basal ganglia calcification (OMIM #615007). In conclusion, a specific class of mutations in PDGFRB causes a clinically recognizable syndromic form of skeletal overgrowth.
Article
Patients and methods: Patients received 100 mg/m(2) Genexol-PM® and 1,000 mg/m(2) gemcitabine intravenously on days 1 and 8 every 21 days. Results: Out of 39 patients, there were 10 partial responses (25.6%) and 18 with stable diseases (46.2%) were confirmed with median response duration 4.1 months. The median progression-free survival was 5.9±1.6 months and overall survival 11.9±1.4 months. The median number of cycles administered was 4.0 (range=1-10). Grade 3 or more neutropenia occurred in 12 patients (26.7%). The most common grade 3/4 non-hematological toxicities were febrile neutropenia (13.4%) and elevation of liver enzymes (6.7%). Conclusion: Weekly Genexol-PM® combined with gemcitabine demonstrated sufficient antitumor activity to warrant further development when used as first-line chemotherapy for advanced biliary tract cancer.
Article
A specialized bone marrow microenvironment (niche) regulates hematopoietic stem cell (HSC) self- renewal and commitment. For successful donor-HSC engraftment, the niche must be emptied via myeloablative irradiation or chemotherapy. However, myeloablation can cause severe complications and even mortality. Here, we report that the essential amino acid valine is indispensable for the proliferation and maintenance of HSCs. Both mouse and human HSCs failed to proliferate when cultured in valine- depleted conditions. In mice fed a valine-restricted diet, HSC frequency fell dramatically within one week. Furthermore, dietary valine restriction emptied the mouse bone marrow niche and afforded donor-HSC engraftment without chemoirradiative myeloablation. These findings indicate a critical role for valine in HSC maintenance and suggest that dietary valine restriction may reduce iatrogenic complications in HSCT
Article
Cellular senescence is a fundamental mechanism by which cells remain metabolically active yet cease dividing and undergo distinct phenotypic alterations, including upregulation of p16(Ink4a) , profound secretome changes, telomere shortening, and decondensation of peri-centromeric satellite DNA. Because senescent cells accumulate in multiple tissues with aging, these cells and the dysfunctional factors they secrete, termed the senescence-associated secretory phenotype (SASP), are increasingly recognized as promising therapeutic targets to prevent age-related degenerative pathologies, including osteoporosis. However, the cell type(s) within the bone microenvironment that undergoes senescence with aging in vivo has remained poorly understood, largely because previous studies have focused on senescence in cultured cells. Thus, in young (6-month) and old (24-month) mice, we measured senescence and SASP markers in vivo in highly enriched cell populations, all rapidly isolated from bone/marrow without in vitro culture. In both females and males, p16(Ink4a) expression by rt-qPCR was significantly higher with aging in B cells, T cells, myeloid cells, osteoblast progenitors, osteoblasts, and osteocytes. Further, in vivo quantification of senescence-associated distension of satellites (SADS), i.e., large-scale unraveling of peri-centromeric satellite DNA, revealed significantly more senescent osteocytes in old compared to young bone cortices (11% versus 2%, p < 0.001). In addition, primary osteocytes from old mice had 6-fold more (p < 0.001) telomere dysfunction-induced foci (TIFs) than osteocytes from young mice. Corresponding with the age-associated accumulation of senescent osteocytes was significantly higher expression of multiple SASP markers in osteocytes from old versus young mice, several of which also showed dramatic age-associated upregulation in myeloid cells. These data demonstrate that with aging, a subset of cells of various lineages within the bone microenvironment become senescent, although senescent myeloid cells and senescent osteocytes predominantly develop the SASP. Given the critical roles of osteocytes in orchestrating bone remodeling, our findings suggest that senescent osteocytes and their SASP may contribute to age-related bone loss. This article is protected by copyright. All rights reserved.