ArticlePDF Available

Notch pathway inhibition controls myeloma bone disease in the murine MOPC315.BM model

Authors:
  • McGill University, Shriners Hospitals for Children-Canada

Abstract and Figures

Despite evidence that deregulated Notch signalling is a master regulator of multiple myeloma (MM) pathogenesis, its contribution to myeloma bone disease remains to be resolved. Notch promotes survival of human MM cells and triggers human osteoclast activity in vitro. Here, we show that inhibition of Notch through the gamma-secretase inhibitor XII (GSI XII) induces apoptosis of murine MOPC315.BM myeloma cells with high Notch activity. GSI XII impairs murine osteoclast differentiation of receptor activator of NF-kappaB ligand (RANKL)-stimulated RAW264.7 cells in vitro. In the murine MOPC315.BM myeloma model GSI XII has potent anti-MM activity and reduces osteolytic lesions as evidenced by diminished myeloma-specific monoclonal immunoglobulin (Ig)-A serum levels and quantitative assessment of bone structure changes via high-resolution microcomputed tomography scans. Thus, we suggest that Notch inhibition through GSI XII controls myeloma bone disease mainly by targeting Notch in MM cells and possibly in osteoclasts in their microenvironment. We conclude that Notch inhibition is a valid therapeutic strategy in MM.
Content may be subject to copyright.
OPEN
ORIGINAL ARTICLE
Notch pathway inhibition controls myeloma bone disease in the
murine MOPC315.BM model
R Schwarzer
1,9
, N Nickel
1,9
, J Godau
1,9
, BM Willie
2
, GN Duda
2
, R Schwarzer
3
, B Cirovic
4
, A Leutz
4
, R Manz
5
, B Bogen
6,7
,BDo¨ rken
1,4
and F Jundt
1,8
Despite evidence that deregulated Notch signalling is a master regulator of multiple myeloma (MM) pathogenesis, its contribution
to myeloma bone disease remains to be resolved. Notch promotes survival of human MM cells and triggers human osteoclast
activity in vitro. Here, we show that inhibition of Notch through the g-secretase inhibitor XII (GSI XII) induces apoptosis of murine
MOPC315.BM myeloma cells with high Notch activity. GSI XII impairs murine osteoclast differentiation of receptor activator of NF-kB
ligand (RANKL)-stimulated RAW264.7 cells in vitro. In the murine MOPC315.BM myeloma model GSI XII has potent anti-MM activity
and reduces osteolytic lesions as evidenced by diminished myeloma-specific monoclonal immunoglobulin (Ig)-A serum levels and
quantitative assessment of bone structure changes via high-resolution microcomputed tomography scans. Thus, we suggest that
Notch inhibition through GSI XII controls myeloma bone disease mainly by targeting Notch in MM cells and possibly in osteoclasts
in their microenvironment. We conclude that Notch inhibition is a valid therapeutic strategy in MM.
Blood Cancer Journal (2014) 4, e217; doi:10.1038/bcj.2014.37; published online 13 June 2014
INTRODUCTION
Impressive improvement in the treatment of multiple myeloma
(MM) has occurred during the past decade, as evidenced by an
increase in the number of progression-free cases and overall
survival rates of patients through the use of target-specific agents.
However, there is still a need for the identification of novel
substances, as in most cases MM remains an incurable disease.
1
High-dose therapy with the alkylating agent melphalan in
combination with autologous stem cell transplantation is used
as the standard therapy in patients, who are eligible for the
procedure.
1
In addition, novel agents such as the proteasome
inhibitor bortezomib are successfully employed to further increase
remission durations.
1
Nevertheless, still most MM patients suffer
from relapses even after many years due to intrinsic or acquired
drug resistance.
1
Recently, we showed that Notch signalling is a survival factor in
Hodgkin lymphoma, anaplastic large-cell lymphoma and particu-
larly in MM.
2–6
The evolutionarily conserved Notch signalling
pathway mediates cell–cell communication and regulates cell
growth, cell death, differentiation programs and self-renewing
processes in a context-dependent manner.
7,8
Its deregulation is
involved in developmental syndromes and cancer.
7,9
In order to
transmit a signal, Notch receptors undergo a series of proteolytic
cleavages after binding their cognate ligands of the Delta-like and
Jagged family.
7,10
Thereby the g-secretase membrane protease
complex cleaves the membrane-bound form of Notch. As a result,
the intracellular domain of Notch receptors (NIC) translocates
into the nucleus and takes part in the Notch transcriptional
complex, which in addition includes scaffold proteins of the
mastermind-like family and the DNA-binding factor RBP-Jk.
g-secretase inhibitors block the cleavage of the Notch receptors
and inhibit release of NIC.
7,8
It has been clearly established that Notch is a master regulator
of MM pathogenesis providing a rationale for evaluating anti-
Notch approaches in MM.
11
Houde et al.
12
showed that Notch
activation can be triggered by overexpressed Jagged2 in the
premalignant condition monoclonal gammopathy of
undetermined significance. Overexpression of Jagged2 can be
triggered either by promoter hypomethylation,
12
through
aberrant expression of the ubiquitin ligase Skeletrophin
13
or by
loss of SMRT/NCoR2 corepressor, which results in abnormal
acetylation of the Jagged2 promoter.
14
In MM, Notch confers
resistance to apoptotic stimuli and protects against
chemotherapy-induced toxicity.
15,16
Nefedova et al.
15
described
that Notch signalling is involved in de novo drug resistance
triggered by bone marrow stroma. Most recently, it has been
shown that Notch activation contributes to bortezomib resistance
in MM.
17
In addition, Notch might accelerate MM progression by
promoting cancer stem cell self-renewal.
18,19
Our own data
showed that activated Notch signalling promotes proliferation
and survival of MM cells.
3
The Notch1 receptor and both its
cognate ligands, Jagged1 and Jagged2, are highly expressed in
primary and cultured MM cells.
3
In addition, activated Notch
signalling is involved in the interactions between MM cells and
their microenvironment.
5
We have demonstrated that Notch
inhibition by g-secretase inhibitors might be a promising
treatment option in MM, as these inhibitors control proliferation
in cultured MM cells
3
and suppress Notch-dependent osteoclast
1
Department of Hematology, Oncology and Tumor Immunology, Campus Virchow-Klinikum, Charite
´—Universita
¨tsmedizin Berlin, Berlin, Germany;
2
Julius Wolff Institute and
Berlin-Brandenburg Center for Regenerative Therapies, Charite
´—Universita
¨tsmedizin Berlin, Berlin, Germany;
3
Institute of Biology and Molecular Biophysics, Humboldt University
Berlin, Berlin, Germany;
4
Max Delbru¨ ck Center for Molecular Medicine, Berlin, Germany;
5
Institute for Systemic Inflammation Research (ISEF), University of Lu¨ beck, Lu¨ beck,
Germany;
6
Centre for Immune Regulation, Institute of Immunology, Oslo University Hospital, Oslo, Norway;
7
Jebsen Centre for Research on Influenza Vaccines, University of Oslo,
Oslo, Norway and
8
Department of Internal Medicine II, University Hospital Wu¨rzburg, University of Wu¨ rzburg, Wu¨ rzburg, Germany. Correspondence: Professor F Jundt,
Department of Internal Medicine II, University Hospital Wu¨ rzburg, University of Wu¨ rzburg Medizinische Klinik und Poliklinik II, Oberdu¨ rrbacher Strasse 6, Wu¨ rzburg 97080,
Germany.
E-mail: jundt_f@ukw.de
9
These authors contributed equally to this work.
Received 23 April 2014; accepted 30 April 2014
Citation: Blood Cancer Journal (2014) 4, e217; doi:10.1038/bcj.2014.37
&
2014 Macmillan Publishers Limited All rights reserved 2044-5385/14
www.nature.com/bcj
activation in vitro.
5
In conclusion, we provided evidence for Notch
activation and deregulation in late-MM stages.
In this study, we used the g-secretase inhibitor XII (GSI XII) for
Notch inhibition. It has been demonstrated that GSI XII controls
the Notch pathway in human lymphoma cells,
6,20,21
and enhances
cytotoxic effects of bortezomib reducing proteasome activity in
MM.
20
To confirm its Notch-specific activity in MM, GSI XII has
been compared with other selective compounds such as the
g-secretase inhibitor DAPT and SAHM1, the dominant-negative
fragment of the Notch co-activator mastermind-like 1, which
selectively blocks the Notch transcriptional complex.
21
These
analyses revealed that GSI XII has similar anti-Notch effects as
DAPT or SAHM1, but even more effectively inhibits MM growth
and induces apoptosis, possibly due to concomitant proteasome
inhibition.
21
To test whether Notch inhibition through GSI XII affects
myeloma bone disease, we used a recently described
murine MOPC315.BM model,
22
which recapitulates the main
characteristics of human MM with particular respect to
myeloma-specific monoclonal IgA serum levels and abundant
osteolytic lesions.
MATERIALS AND METHODS
Mice
Six-week-old female BALB/c mice were obtained from Charles River
(Sulzfeld, Germany). All experiments were approved by the local
committee of the Landesamt fu¨ r Gesundheit und Soziales Berlin
(Berlin, Germany).
Cell culture and reagents
Human MM cell lines (NCI-H929 and OPM-2) were used (DSMZ,
Braunschweig, Germany). MOPC315 cells were obtained from ATCC
(Manassas, VA, USA) as an in vitro-adapted cell line and were repeatedly
injected subcutaneously into BALB/c mice, resulting in the MOPC315.4 cell
line.
23
The MOPC315.4 cell line was injected intravenously into BALB/c
mice. After nine in vivo/in vitro cycles, a cell line was generated that had a
tropism for bone marrow and was therefore named as MOPC315.BM.
22
MOPC315.BM cells produce an IgA myeloma protein, M315, which can be
measured by enzyme-linked immunosorbent assay. MOPC315.BM cells
were incubated 24–48 h with increasing doses of GSI XII, which was
obtained from Calbiochem (San Diego, CA, USA). The RAW264.7 murine
monocyte/macrophage cell line was cultured in Dulbecco’s modified
Eagle’s medium and induced to differentiate into bone resorbing
osteoclasts by 10 ng/ml receptor activator of NF-kB ligand (RANKL; 462-
TEC-010, R&D Systems, Wiesbaden, Germany) in minimal essential medium
alpha as described.
24
Immunoblotting
Whole-cell extracts were prepared and immunoblotting was performed as
described.
2
Nuclear extracts were prepared using the Nuclear Extract Kit
(Active Motif, Carlsbad, CA, USA). Blots were incubated with monoclonal
rabbit anti-Notch1 antibodies (cat# 1935-1, Epitomics, Burlingame, CA,
USA), anti-cleaved Notch1 NIC (Val1744; cat# 4147; Cell Signaling
Technologies, Frankfurt, Germany), anti-poly-(ADP-ribose) polymerase
(PARP, cat# 9532, Cell Signaling Technologies), anti-cleaved PARP
(Asp214; cat# 9541, Cell Signaling Technologies) or rabbit monoclonal
anti-tubulin antibodies (cat# 2125; Cell Signaling Technologies). Detection
was performed using Pico or Dura chemiluminescence reagents (Perbio
Science, Bonn, Germany).
RNA preparation and quantitative reverse transcription-PCR
analysis
RNA preparation and complementary DNA synthesis were performed as
described.
4
Reverse transcription-PCR analysis was performed as described
using primers and probes for murine HEY1, RANKL, NFATc1 and TRAP5.
4
As
an internal control murine hypoxanthin-guanin-phosphoribosyltransferase
was amplified. Primer sequences are available upon request.
Viability assay and assessment of apoptosis
Viability of cells was determined by CellTiter-Glo Luminescent Cell Viability
Assay (Promega, Mannheim, Germany). Each treatment was done in three
independent replicates. Luminescence was recorded; average values were
calculated and normalized to the respective dimethyl sulphoxide-treated
sample. Amount of apoptotic cells was determined using the human
AnnexinV-FITC Kit (Bender Medsystems, Vienna, Austria).
TRAP staining
After cultivation of 0.75 10
5
RAW264.7 per 12-well for 72 h, cells were
washed with phosphate-buffered saline (PBS) and fixed in 4% PBS-buffered
formaldehyde for 10 min at room temperature (RT). Cells were shown to be
tartrate-resistant acid phosphatase (TRAP)-positive by staining using the
Acid Phosphatase Leukocyte Kit (Sigma-Aldrich, Seelze, Germany) with an
adapted protocol. Staining solution was prepared with 0.2 M tartrate and
using half as much GBC (40-amino-2,30-dimethylazobenzene) solution as
described in manufacturer’s protocol. After adequate incubation with
staining solution, cells were once washed with water and stored in PBS/4%
formaldehyde for further analysis.
Differential interference contrast (DIC) microscopy
After labelling, TRAP-positive cells were subjected to DIC microscopy.
Images were collected using an inverted Olympus IX-81 microscope
(Olympus, Tokyo, Japan) equipped with a cooled monochrome CCD
camera. All images that were used for the analysis are shown in
Supplementary Figure S1. The cells were imaged using a 20 UPlanFL
air objective (numerical aperture 0.4) with a typical exposure time of 10 ms.
Cell image analysis
DIC images were analysed using the CellProfiler image analysis software
(version 2.0) using a self-provided pipeline (Supplementary Figures S2A–C).
25
Briefly, nuclei were identified by Hoechst 33258 (Sigma-Aldrich) staining and
subsequently, based on propagation from the nuclei, cell segmentation was
performed on inverted DIC images after automated editing of image
properties, such as image intensity and contrast. Then, the normalized
staining intensity of individual cells was assessed from the original, non-
processed images and saved in a spreadsheet for further analysis.
Enzyme-linked immunosorbent assay
Murine blood samples were obtained from cheeks. Blood samples were
allowed to clot for 2 h at RT before centrifugation for 10 min at 2000 g. Sera
were collected and stored at 20 1C. The M315 myeloma protein, which is
2,4-dinitrophenol specific, was detected by enzyme-linked immunosorbent
assay. Ninety-six-well plates were coated with 1 mg/ml dinitrophenol-
conjugated bovine serum albumin and incubated at 41C overnight.
Unspecific binding sites were blocked for 30 min at RT using PBS/5%
bovine serum albumin followed by three washing steps with PBS/0.1%
Tween. Serum samples were diluted 1:500–1:5000 in PBS containing 0.1 or
5% bovine serum albumin. M315 standard protein was serially diluted
within a range of 10–2560 ng/ml. Plates were incubated for 2 h at 37 1C
with serum samples and standard protein. After plates were washed three
times with PBS/0.1% Tween, 1 mg/ml biotinylated rat anti-mouse IgA (clone
C10-1, BD Pharmingen, Heidelberg, Germany) was added for 1 h at RT.
Plates were again washed three times and incubated with streptavidin-
conjugated alkaline phosphatase (1:3000; Roche, Mannheim, Germany) for
1 h at RT. After three washes, phosphatase substrate (Roche) was added at
1 mg/ml in substrate buffer and absorbance was measured at 415 nm. Sera
were collected from vehicle-treated and GSI XII-treated mice at time points
indicated (Figure 2a) or every 2–8 days (Figure 3a).
High-resolution mCT scans
For trabecular and cortical bone structural analysis, microcomputed
tomography (mCT) at an isotropic voxel size of 10.5 mm (vivaCT 40, Scanco
Medical, Bru¨ ttisellen, Switzerland; 55 kVp, 145 mA, 600 ms integration time,
no frame averaging) was performed on dissected mice tibias to assess
bone. Guidelines for assessment of bone microstructure in rodents were
applied for evaluation.
26
For each tibia, a trabecular and cortical bone
volume of interest was defined. The total tibia length was measured using
digital calipers on freshly isolated bones. The trabecular volume of interest
included secondary spongiosa in the proximal metaphysis, starting 700 mm
below the growth plate and extending distally 5% of the tibial length.
Notch in myeloma bone disease
R Schwarzer et al
2
Blood Cancer Journal &2014 Macmillan Publishers Limited
The trabecular bone volume of interest excluded the cortical shell.
Thresholds of 456 mg (14 days GXI XII treatment) and 447 mg (36 days of
GSI XII treatment) hydroxylapatite/cm
3
were used to segment proximal
trabecular bone from water and soft tissue. Trabecular (Tb.) bone outcome
parameters included: bone volume fraction (BV/TV, mm
3
/mm
3
), trabecular
thickness (Tb.Th, mm), average number of trabeculae per unit length (Tb.N,
1/mm), trabecular separation (Tb.Sp, mm) and trabecular volumetric bone
mineral density (mg hydroxylapatite/cm
3
). The second analysed volume of
interest included only cortical bone at the proximal metaphysis, excluding
trabecular bone and the marrow cavity. A 563-mg hydroxylapatite/cm
3
threshold (36 days GXI XII treatment) was used to segment cortical bone
from water and soft tissue. The following parameters were used for
analysis of cortical (Ct.) bone: cortical bone area ¼cortical volume/(number
of slices slice thickness) (Ct.Ar, mm
2
), cortical thickness (Ct.Th, mm) and
total cross-sectional area inside the periosteal envelope (Tt.Ar, mm
2
). Tt.Ar
was contoured to only include the metaphyseal cortical bone and porosity
within the bone, with the medullary canal (including trabecular bone and
bone marrow) excluded from the analysis. Also, cortical porosity
area ¼cortical porosity volume/(number of slices slice thickness)
(Ct.Po.Ar, mm
2
) and cortical volumetric bone mineral density were
measured. All quantitative analyses were performed with the system’s
software (Scanco Eval 6.5-1, Scanco Medical, Bru¨ttisellen, Switzerland).
MM model and drug treatment
MOPC315.BM cells were cultured in RPMI with 10% fetal calf serum in vitro
(37 1C, 5% CO
2
) and harvested for injection into tail veins. 5 10
5
MOPC315.BM cells were injected per mouse. The mice were killed on days
indicated or when end points were reached. Mice were treated with
10 mg/kg GSI XII intraperitoneally either for 14 days (Figure 3, dissolved in
PBS/cremophor/peanut oil, PCO) or for 36 days (Figure 4, dissolved in
dimethyl sulphoxide). Quantification of bone structure changes was
performed after 14 days and after 36 days of treatment, respectively.
Statistics
Statistical analysis was performed with Prism 5 software (GraphPad, La
Jolla, CA, USA). One- or two-tailed t-tests were used as appropriate to
analyse statistical significance. All data are shown as mean±s.d. Logrank
test was used to compare serum increases over time between vehicle-
treated and GSI XII-treated mice as shown in a Kaplan–Meier plot. P-values
o0.05 were considered as statistically significant.
RESULTS
Notch inhibition reduces viability and induces apoptosis in
MOPC315.BM cells with deregulated Notch activity in vitro
Our recent data showed aberrant Notch activation in cultured and
primary human MM cells.
3,5
In contrast, freshly isolated mature
CD19
þ
B cells and CD19
þ
B cells, which we differentiated to
CD38
þ
plasmablastic cells in vitro, were almost completely devoid
of Notch expression.
3,5
To test whether the murine MM cell line
MOPC315.BM has activated Notch signalling, we first confirmed
expression of the full-length transmembrane-bound form of the
Notch receptors (Notch1 and Notch2) and their ligands (Jagged1
and Jagged2) by immunoblotting. MOPC315.BM cells express
both Notch receptors and ligands (Figure 1a). Human NCI-H929
and OPM-2 cultured MM cells served as positive controls
(Figure 1a). To further demonstrate activated Notch signalling,
we used nuclear extracts and verified abundant expression of the
intracellular form of Notch1 (N1IC; Figure 1b). Therefore, we
confirmed activation of the Notch pathway in MOPC315.BM cells.
In addition, N1IC could be specifically downregulated through the
g-secretase inhibitor GSI XII (Figure 1b). Concomitantly, messenger
RNA expression of the Notch target gene HEY1 was suppressed
after GSI XII treatment in MOPC315.BM cells (Figure 1c). These data
indicate that activated Notch signalling was blocked in
MOPC315.BM cells by use of GSI XII.
To analyse functional consequences of Notch inhibition for
tumour cell biology, we performed viability assays, Annexin
V-staining and immunoblotting for PARP cleavage. GSI XII reduced
cell viability (Figure 1d) and induced apoptosis (Figures 1e and f)
in MOPC315.BM cells in a dose-dependent manner.
Notch inhibition impairs osteoclast differentiation of RANKL-
stimulated RAW264.7 cells
Recently, we provided evidence in vitro that Notch inhibition has
no effect on osteoblasts and their progenitors and blocks human
osteoclast activity by downregulation of TRAP5, a marker for
osteoclast differentiation and activity.
5,27
To analyse whether GSI
XII impairs murine osteoclast differentiation, we used the murine
monocyte/macrophage cell line RAW264.7.
24
RAW264.7 cells were
efficiently induced to differentiate into osteoclasts within 72 h
12 μM15μM
DMSO GSI XII
2-ΔΔCt
HEY1
MOPC315.BM
OPM-2
NCI-H929
Jagged1
Notch2
Jagged2
Notch1
α-tubulin
N1IC
PARP
DMSO
7.5 μM
10 μM
GSI
DMSO GSI XII
relative viability
0.2
0.4
0.6
0.8
1.0
1.2
0
0.2
0.4
0.6
0.8
1.0
1.2
0
1.4
cleaved
PARP
α-tubulin
DMSO
12 μM
15 μM
GSI
DMSO
PI
AnnexinV
1%
10 μM GSI
PI
AnnexinV
46%
7%
12 μM 15 μM
1%
Figure 1. Notch inhibition controls viability and induces apoptosis in
MOPC315.BM cells with activated Notch signalling. (a) Immunoblot-
ting of Jagged1, Jagged2, Notch1 and Notch2 in MOPC315.BM cells
and in the human MM cell lines NCI-H929 and OPM-2. Staining for
a-tubulin served as control for equal loading. (b) Immunoblotting of
the intracellular form of Notch1 (N1IC), and PARP (loading control) in
nuclear extracts of MOPC315.BM cells after GSI XII treatment.
(c) Quantitative reverse transcription-PCR analysis of HEY1 messenger
RNA expression in MOPC315.BM cells after GSI XII treatment.
(d) Viability of MOPC315.BM cells after GSI XII treatment. Numbers of
viable cells are given relative to dimethyl sulphoxide (DMSO)-treated
samples. (e) Immunoblotting of PARP cleavage in whole-cell extracts of
MOPC315.BM cells treated with different concentrations of GSI XII.
(f) AnnexinV/propidium iodide (PI) staining of DMSO and GSI XII-
treated cells. Percentages of early (FITC þPI ) and late apoptotic or
necrotic (F ITC þPI þ) cells as determined by AnnexinV/PI staining.
Notch in myeloma bone disease
R Schwarzer et al
3
&2014 Macmillan Publishers Limited Blood Cancer Journal
through stimulation with 10 ng/ml RANKL.
24
RANKL is known to
selectively induce NFATc1 expression during differentiation of
RAW264.7 cells into TRAP þosteoclasts.
28,29
Reverse transcription-
PCR analysis revealed that in RANKL-induced osteoclasts GSI XII
diminished messenger RNA expression of the Notch target gene
HES1 and in parallel of the transcription factor NFATc1, a master
regulator of osteoclast differentiation (Figures 2a and b;
Supplementary Figure S3A). Concomitantly, messenger RNA
expression of the osteoclast-specific gene TRAP5 was down-
regulated both on the messenger RNA (Figure 2b, right panel;
Supplementary Figure S3B) and on the protein level (Figure 2c).
TRAP5 is a known target gene of NFATc1.
28
Both NFATc1 and
TRAP5 were regulated in a dose-dependent manner by GSI XII
(Supplementary Figures S3A and B). We further performed DIC
microscopy, to quantify differences in the staining intensity of
TRAP þand TRAP þþ osteoclasts after Notch inhibition (Figures
2d and e; Supplementary Figure 4). Our data showed that GSI XII
treatment almost completely abolished RANKL-induced osteoclast
differentiation in vitro.
Notch inhibition reduces myeloma-specific paraprotein levels in
the MOPC315.BM model
Next, GSI XII efficacy was examined in the MOPC315.BM mouse
model.
22
To that end, we designed two different experiments. The
first experimental setup was designed to evaluate GSI XII
treatment in mice, which had already established MM cell
growth. Mice were inoculated with MOPC315.BM cells and
randomized into vehicle-treated mice and GSI XII-treated mice
once myeloma-specific monoclonal IgA (M315) serum levels
DMSO GSI DMSO GSI
- RANKL + RANKL
2−ΔΔCt
HES1
0
5
10
15
20
DMSO GSI DMSO GSI
- RANKL + RANKL
2-ΔΔCt
NFATc1
0
50
100
150
200
250
DMSO GSI DMSO GSI
- RANKL + RANKL
2-ΔΔCtCt
TRAP5
0
0.08
0.16
0.24
0.32
0.40
0.48
intensit
y
units
TRAP staining
TRAP+
TRAP++
100 μm
- RANKL + DMSO
100 μm
+ RANKL + GSI
100 μm
+ RANKL + DMSO
TRAP staining
DMSO GSI
- RANKL + RANKL
TRAP+
***p<0.0001
DMSO GSI
- RANKL + RANKL
TRAP++
**p=0.0011
-RANKL
+ RANKL + DMSO
+ RANKL + GSI
0.0
0.2
0.4
0.6
0.8
1.0
1.2
1.4
0
5
10
15
20
25
30
35
100
80
60
40
20
0
events (%)
events (%)
60
40
20
0
events (%)
Figure 2. Notch inhibition impairs differentiation of RAW264.7 cells into osteoclasts in vitro (aand b) Treatment of the murine monocyte/
macrophage cell line RAW264.7 without ( ) and with ( þ) 10 ng/ml RANKL for stimulation of osteoclast differentiation. Quantitative reverse
transcription-PCR analysis of HES1, NFATc1 and TRAP5 in RAW264.7 cells treated with 10 mMGSI XII. Dimethyl sulphoxide (DMSO) was used as
solvent control. (c) Osteoclast differentiation visualized by TRAP staining. RAW264.7 cells differentiated into TRAP-positive osteoclasts after
72 h of RANKL-stimulation (middle). Treatment of RAW264.7 cells with 10 mMGSI XII inhibited osteoclast differentiation and RANKL-induced
RAW264.7 cells remain TRAP negative (right). RAW.264.7 cells treated with DMSO as solvent control (left). (d) Differential interference contrast
microscopy was used to quantify differences in staining intensity of TRAP þand TRAP þþ RANKL-stimulated osteoclasts. Cells were treated
with GSI XII or DMSO as solvent control. (e) Percentages of TRAP þand TRAP þþ cells. Two-tailed t-test was used for statistical analysis.
P-values as indicated.
Notch in myeloma bone disease
R Schwarzer et al
4
Blood Cancer Journal &2014 Macmillan Publishers Limited
approximated 10–30 mg/ml at two consecutive time points
(Figure 3a). Threshold corresponded to the lowest detectable
IgA levels. GSI XII was then administered at a dose of 10 mg/kg for
14 days (Figure 3a). GSI XII-treated mice showed a strong
reduction in M315 levels as compared with vehicle-treated mice
over time (Figure 3b). In addition, progression-free survival of GSI
XII-treated mice was significantly increased (Figure 3c). Progres-
sion was determined when M315 levels exceeded 150 mg/ml.
If bone structure changes were already apparent, it would be
difficult to detect changes in bone mass and volume during a
treatment period of only 14 days. Therefore, we designed a
second experiment (Figure 4a) and focused our analysis on IgA
levels (Figure 4b) and bone structure changes employing mCT
scans (Figures 4c and d; Tables 1 and 2). MOPC315.BM cells were
transplanted at day 0 and mice were randomized into two groups
(vehicle- and GSI XII-treated mice) at the same day. GSI XII was
administered at a dose of 10 mg/kg for 36 days (Figure 4a). GSI XII
treatment reduced tumour burden as evidenced by diminished
M315 levels at day of death (Figure 4b). We conclude that GSI XII
reduces MM development and progression but is not able to
completely cure treated mice. In GSI-treated groups still a few
mice develop MM, however, with lower tumour load and slower
increase of IgA levels over time as compared with control animals.
Notch inhibition diminishes osteolytic lesions in the MOPC315.BM
mice with GSI XII
We recently demonstrated osteolytic lesions in the MOPC315.BM
model by use of mCT analysis, TRAP staining for osteoclasts in the
bone marrow and measurement of serum Ca2 þlevels in BALB/c
mice.
22
To determine quantitative bone structural changes
between vehicle-treated and GSI XII-treated mice, we performed
mCT analysis and used recently published guidelines for
assessment of bone microstructure in rodents.
26
Representative
data of proximal tibiae of mice are shown (Figures 4c and d;
Tables 1 and 2). Transversal sections (Figure 4c) and three-
dimensional reconstruction (Figure 4d) of tibiae revealed
increased cortical porosity (Ct.Po.Ar; Figure 4e; Table 1) and
diminished trabecular structures (Table 2) in vehicle-treated mice
as compared with GSI XII-treated mice. Quantitative analysis of
bone structural changes showed significant changes between the
two groups, particularly in trabecular bone structures (Table 2). For
example, bone volume fraction (BV/TV), trabecular number (Tb.N)
as well as volumetric bone mineral density were higher in GSI XII-
treated mice as compared with vehicle-treated mice (Table 2),
indicating that Notch inhibition through GSI XII reduces bone loss
and diminishes osteolytic lesions in the MOPC315.BM model.
DISCUSSION
In this study, we used our recently described MOPC315.BM MM
model to investigate whether Notch inhibition through the
g-secretase inhibitor GSI XII has anti-MM activity in vivo.
22
We
showed that MOPC315.BM cells are characterized by aberrant
Notch activity in vitro, which was specifically downregulated
through GSI XII. As a result, viability of murine MM cells was
reduced and apoptosis was induced. Tumour biologic relevance of
these findings was revealed by our in vivo studies: (1) decreased
MOPC315.BM
injection
d0
IgA=10-30 μg/ml
d5 d8 d10 d12 d14 d15 - serum IgA analysis
sacrifice
bone analysis
* p=0.0113
daily treatment
day 0
1.5
3
4.5
6
0
0
25
50
75
100
0
IgA [mg/ml]
day 8 day 10 day 12
vehicle GSI vehicle GSI vehicle GSI vehicle GSI
n. s. * p=0.023 * p=0.036 * p=0.035
51015
GSI
vehicle
days
progression-free [%]
Figure 3. Notch inhibition reduces myeloma-specific paraprotein levels in the MOPC315.BM MM model. (a)Schemeindicatestimepointsof
MOPC315.BM injection, serum collection for measurement of MM-specific M315 IgA levels and GSI XII treatment. GSI XII treatment (10 mg/kg,
daily) for 14 days started after M315 serum levels reached B10–30 mg/ml. (b) M315 serum levels of vehicle-treated (n¼13) and GSI XII-treated
(n ¼9) mice on days 0, 8, 10 and 12 of treatment. Two-tailed t-test was used for statistical analysis. (c) Kaplan–Meier plot for time points when
150 mg/ml M315 serum levels were reached. Logrank test was used for statistical analysis. P-values as indicated.
Notch in myeloma bone disease
R Schwarzer et al
5
&2014 Macmillan Publishers Limited Blood Cancer Journal
myeloma-specific IgA serum levels and (2) decreased myeloma
bone disease indicated diminished tumour burden in the
MOPC315.BM MM model. We have previously shown that the
MOPC315.BM model closely resembles human disease, as
mice experience generalized bone loss and focal osteolytic bone
lesions.
22
Our data here showed that Notch inhibition reduced
bone loss, specifically in the trabecular compartment and curtailed
the presence of osteolytic lesions. Particularly, cortical porosity
was less pronounced in GSI XII-treated mice indicative of
diminished changes in cortical bone structures. We suggest that
Notch inhibition efficiently targets MM cells and their
microenvironment in the MOPC315.BM model.
There is increasing evidence that Notch signalling is involved in
differentiation and activation of osteoclasts.
30–33
Activation of
Table 1. mCT analyses of cortical bone of vehicle-treated and GSI XII-treated MOPC315.BM mice (36 days of GSI XII treatment)
Variable GSI XII n¼9 s.d. Vehicle n¼13 s.d. P-value Signif. diff.
Ct.area (mm
2
) 0.78 0.07 0.78 0.04 0.918 NS
Tt.area (mm
2
) 0.95 0.07 0.97 0.04 0.444 NS
Ct.Po.Ar (mm
2
) 0.16 0.02 0.18 0.02 0.018 Yes
Ct.Th (mm) 0.11 0.01 0.11 0.01 0.589 NS
Ct. vBMD (mg HA/cm
3
) 917 30 902 24 0.234 NS
Abbreviations: Ar, area; Ct., cortical; mCT, microcomputed tomography; GSI, g-secretase inhibitor; HA, hydroxylapatite; Po., porosity; Th, thickness; Tt., total;
vBMD, volumetric bone mineral density. Two-tailed t-test was used for statistical analysis. Significant P-value is indicated in bold.
MOPC315.BM
injection
sacrifice
bone analysis
d37
daily treatment
serum IgA
analysis
0.1
0.15
0.2
0.25
vehicle GSI
Ct.Po.Ar (mm2)
1 mm 1 mm
* p=0.04
vehicle GSI
1
2
3
4
5
0
vehicle GSI
vehicle GSI
day of death
IgA [mg/ml]
* p=0.018
d0
Figure 4. Osteolytic lesions are diminished after treatment of MOPC315.BM mice with GSI XII. (a) Scheme indicates time points of
MOPC315.BM injection, serum collection for measurement of MM-specific M315 IgA levels and GSI XII treatment. After intravenous inoculation
of MOP315.BM cells, mice were randomized into two groups (vehicle-treated versus GSI XII-treated mice). Treatment started immediately
and was discontinued after 36 days. M315 serum levels were measured at the time of death and latest at day 37. (b) M315 serum levels of
vehicle-treated (n¼13) and GSI XII-treated (n¼9) mice on the day of death. One-tailed t-test with Welsh’s correction was used for statistical
analysis. (c) Transversal sections through representative proximal tibiae of vehicle-treated mice and GSI XII-treated mice. Arrows indicate
osteolytic lesions. (d) Three-dimensional reconstruction of proximal tibiae demonstrate reduced wall thickness, diminished trabecular
structures and holes in vehicle-treated mice as compared with GSI XII-treated mice. (e) Box plot for values of the cortical porosity area (in mm
2
)
in vehicle-treated and GSI XII-treated mice. Two-tailed t-test was used for statistical analysis. P-values as indicated.
Table 2. mCT analyses of the trabecular bone of vehicle-treated and GSI XII-treated MOPC315.BM mice (36 days of GSI XII treatment)
Variable GSI XII n¼9 s.d. Vehicle n¼13 s.d. P-value Signif. diff.
Tb. BV/TV (mm
3
/mm
3
) 0.066 0.025 0.044 0.016 0.016 Yes
Tb.Th (mm) 0.046 0.003 0.046 0.004 0.814 NS
Tb.Sp (mm) 0.352 0.079 0.417 0.053 0.032 Yes
Tb.N(1/mm) 3 0.6 2.5 0.3 0.009 Yes
Tb. vBMD (mg HA/cm
3
) 144 31 119 18 0.03 Yes
Abbreviations: BV, bone volume; mCT, microcomputed tomography; GSI, g-secretase inhibitor; HA, hydroxylapatite; Sp, separation; Tb., trabecular; Th, thickness;
TV, total volume; vBMD, volumetric bone mineral density. Two-tailed t-test was used for statistical analysis. Significant P-values are indicated in bold.
Notch in myeloma bone disease
R Schwarzer et al
6
Blood Cancer Journal &2014 Macmillan Publishers Limited
Notch through Jagged1 enhances the osteoclast transcription
factor NFATc1 promoter activity and expression, and thereby
promotes osteoclastogenesis.
32,34
Recently, a mechanism
of this regulation has been proposed, demonstrating that
calcium/calmodulin-dependent protein kinase IV (CaMKIV)
bindstoNIC,stabilizestheproteinandinhibitsitsproteasomal
degradation.
31
Thus, promoter activity of the Notch-dependent
target gene NFATc1 is enhanced.
31
In the present study we
provided evidence that Notch inhibition through GSI XII
transcriptionally downregulated NFATc1 expression in RANKL-
induced RAW264.7 cells in vitro. Concomitantly, the
osteoclast differentiation marker TRAP5 was suppressed.
Our current data indicate that Notch inhibition impairs
osteoclast differentiation in vitro. They are in accordance with a
previous study that used small hairpin RNA for Notch2 and
GSI X for inhibition of Notch in osteoclast progenitors and
demonstrated decreased expression of NFATc1 resulting in
inhibition of osteoclastogenesis.
32
We speculate that this
mechanism presumably contributes to the in vivo efficacy of GSI
XII in MOPC315.BM mice with decreased osteolytic lesions and
generalized bone loss. However, we cannot exclude that Notch
inhibition through GSI XII exerts its effects in vivo mainly through
induction of MM cell apoptosis.
Recent evidence from a breast cancer mouse model indicated
that the Jagged1/Notch pathway promotes osteolytic bone
disease of breast cancer through engagement of Notch
signalling in osteoblasts and osteoclasts.
35
Notch inhibition
reverses the Jagged/Notch-mediated bone effects and
reduces development of bone metastasis.
35
In MM, tumour
cells originate from transformed post-germinal center B cells,
which selectively migrate to the bone marrow to establish
bone disease.
11
Our data provided evidence that in mice,
which received GSI XII immediately after MOPC315.BM cell
injection, tumour burden and bone lesions were significantly
reduced as compared with controls (Figure 4). We hypothesize
that Notch inhibition controls MOPC315.BM cell growth and
interferes with MM cell homing and/or interaction with the bone
marrow microenvironment. Supporting this hypothesis it has
most recently been demonstrated that anti-Notch treatment
prevents bone marrow infiltration of human MM cells in a
mouse non-obese diabetic/severe combined immunodeficient
xenograft model by modulation of the chemokine receptor
CXCR4/stromal cell-derived factor-1 system.
21
This is consistent
with the observation that MOPC315.BM cells similar to human
MM cells express CXCR4, which interacts with stromal cell-
derived factor-1 on stromal cells for their recruitment to the
bone marrow.
36
In conclusion, our study provided preclinical evidence
in vivo for Notch inhibition through GSI XII as a valid
therapeutic strategy against myeloma bone disease.
Single use of GSI XII does not eradicate the tumour.
However, it is has been shown that g-secretase inhibitors,
namely GSI XII, increase sensitivity to drugs such as bortezomib,
17
which have recently essentially improved therapeutic outcomes
in MM.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
ACKNOWLEDGEMENTS
This work was supported by the Deutsche Forschungsgemeinschaft (DFG, TRR54,
TPB6 to FJ and BD), the Deutsche Krebshilfe (108658 to FJ and RM); and the
Wilhelm-Sander Stiftung (2011.018.1 to FJ). It was further supported by the
Norwegian Cancer Society (BB) and the Multiple Myeloma Research Foundation
(BB). We thank Katharina Pardon and Alexander Haake for excellent technical
assistance.
AUTHOR CONTRIBUTIONS
RS conceived the hypothesis, designed and performed experiments and
analysed the data. NN, JG, RS and BC performed experiments and analysed the
data. BMW, GND, AL, RM, BB and BD analysed data and contributed to writing
the manuscript. FJ conceived the hypothesis, designed experiments and wrote
the manuscript.
REFERENCES
1 Palumbo A, Bringhen S, Ludwig H, Dimopoulos MA, Blade J, Mateos MV et al.
Personalized therapy in multiple myeloma according to patient age and
vulnerability: a report of the European Myeloma Network (EMN). Blood 2011; 118:
4519–4529.
2 Jundt F, Anagnostopoulos I, Forster R, Mathas S, Stein H, Dorken B. Activated
Notch1 signaling promotes tumor cell proliferation and survival in Hodgkin and
anaplastic large cell lymphoma. Blood 2002; 99: 3398–3403.
3 Jundt F, Probsting KS, Anagnostopoulos I, Muehlinghaus G, Chatterjee M, Mathas S
et al. Jagged1-induced Notch signaling drives proliferation of multiple myeloma
cells. Blood 2004; 103: 3511–3515.
4 Jundt F, Acikgoz O, Kwon SH, Schwarzer R, Anagnostopoulos I, Wiesner B et al.
Aberrant expression of Notch1 interferes with the B-lymphoid phenotype of
neoplastic B cells in classical Hodgkin lymphoma. Leukemia 2008; 22:
1587–1594.
5 Schwarzer R, Kaiser M, Acikgoez O, Heider U, Mathas S, Preissner R et al.
Notch inhibition blocks multiple myeloma cell-induced osteoclast activation.
Leukemia 2008; 22: 2273–2277.
6 Schwarzer R, Dorken B, Jundt F. Notch is an essential upstream regulator
of NF-kappaB and is relevant for survival of Hodgkin and Reed-Sternberg cells.
Leukemia 2012; 26: 806–813.
7 Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the
activation mechanism. Cell 2009; 137: 216–233.
8 Guruharsha KG, Kankel MW, Artavanis-Tsakonas S. The Notch signalling system:
recent insights into the complexity of a conserved pathway. Nat Rev Genet 2012;
13: 654–666.
9 Radtke F, Fasnacht N, Macdonald HR. Notch signaling in the immune system.
Immunity 2010; 32: 14–27.
10 Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: cell fate control and
signal integration in development. Science 1999; 284: 770–776.
11 Colombo M, Mirandola L, Platonova N, Apicella L, Basile A, Figueroa AJ et al.
Notch-directed microenvironment reprogramming in myeloma: a single path to
multiple outcomes. Leukemia 2013; 27: 1009–1018.
12 Houde C, Li Y, Song L, Barton K, Zhang Q, Godwin J et al. Overexpression of the
NOTCH ligand JAG2 in malignant plasma cells from multiple myeloma patients
and cell lines. Blood 2004; 104: 3697–3704.
13 Takeuchi T, Adachi Y, Ohtsuki Y. Skeletrophin, a novel ubiquitin ligase to the
intracellular region of Jagged-2, is aberrantly expressed in multiple myeloma.
Am J Pathol 2005; 166: 1817–1826.
14 Ghoshal P, Nganga AJ, Moran-Giuati J, Szafranek A, Johnson TR, Bigelow AJ et al.
Loss of the SMRT/NCoR2 corepressor correlates with JAG2 overexpression in
multiple myeloma. Cancer Res 2009; 69: 4380–4387.
15 Nefedova Y, Cheng P, Alsina M, Dalton WS, Gabrilovich DI. Involvement of Notch-1
signaling in bone marrow stroma-mediated de novo drug resistance of myeloma
and other malignant lymphoid cell lines. Blood 2004; 103: 3503–3510.
16 Nefedova Y, Sullivan DM, Bolick SC, Dalton WS, Gabrilovich DI. Inhibition of
Notch signaling induces apoptosis of myeloma cells and enhances sensitivity to
chemotherapy. Blood 2008; 111: 2220–2229.
17 Xu D, Hu J, De Bruyne E, Menu E, Schots R, Vanderkerken K et al. Dll1/Notch
activation contributes to bortezomib resistance by upregulating CYP1A1 in
multiple myeloma. Biochem Biophys Res Commun 2012; 428: 518–524.
18 Chiron D, Maiga S, Descamps G, Moreau P, Le Gouill S, Marionneau S et al. Critical
role of the NOTCH ligand JAG2 in self-renewal of myeloma cells. Blood Cells Mol
Dis 2012; 48: 247–253.
19 Xu D, Hu J, Xu S, De Bruyne E, Menu E, Van Camp B et al. Dll1/Notch activation
accelerates multiple myeloma disease development by promoting CD138 þ
MM-cell proliferation. Leukemia 2012; 26: 1402–1405.
20 Chen F, Pisklakova A, Li M, Baz R, Sullivan DM, Nefedova Y. Gamma-secretase
inhibitor enhances the cytotoxic effect of bortezomib in multiple myeloma. Cell
Oncol (Dordr) 2011; 34: 545–551.
21 Mirandola L, Apicella L, Colombo M, Yu Y, Berta DG, Platonova N et al. Anti-Notch
treatment prevents multiple myeloma cells localization to the bone marrow via
the chemokine system CXCR4/SDF-1. Leukemia 2013; 27: 1558–1566.
22 Hofgaard PO, Jodal HC, Bommert K, Huard B, Caers J, Carlsen H et al. A novel
mouse model for multiple myeloma (MOPC315.BM) that allows noninvasive
spatiotemporal detection of osteolytic disease. PLoS One 2012; 7: e51892.
Notch in myeloma bone disease
R Schwarzer et al
7
&2014 Macmillan Publishers Limited Blood Cancer Journal
23 Lauritzsen GF, Bogen B. The role of idiotype-specific, CD4 þT cells in tumor
resistance against major histocompatibility complex class II molecule negative
plasmacytoma cells. Cell Immunol 1993; 148: 177–188.
24 Collin-Osdoby P, Osdoby P. RANKL-mediated osteoclast formation from murine
RAW 264.7 cells. Methods Mol Biol 2012; 816: 187–202.
25 Carpenter AE, Jones TR, Lamprecht MR, Clarke C, Kang IH, Friman O et al.
CellProfiler: image analysis software for identifying and quantifying cell pheno-
types. Genome Biol 2006; 7: R100.
26 Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Muller R.
Guidelines for assessment of bone microstructure in rodents using micro-com-
puted tomography. J Bone Miner Res 2010; 25: 1468–1486.
27 Zavrski I, Krebbel H, Wildemann B, Heider U, Kaiser M, Possinger K et al.
Proteasome inhibitors abrogate osteoclast differentiation and osteoclast function.
Biochem Biophys Res Commun 2005; 333: 200–205.
28 Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H et al. Induction and
activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in
terminal differentiation of osteoclasts. Dev Cell 2002; 3: 889–901.
29 Ishida N, Hayashi K, Hoshijima M, Ogawa T, Koga S, Miyatake Y et al. Large scale
gene expression analysis of osteoclastogenesis in vitro and elucidation of NFAT2
as a key regulator. J Biol Chem 2002; 277: 41147–41156.
30 Yamada T, Yamazaki H, Yamane T, Yoshino M, Okuyama H, Tsuneto M et al.
Regulation of osteoclast development by Notch signaling directed to osteoclast
precursors and through stromal cells. Blood 2003; 101: 2227–2234.
31 Choi YH, Ann EJ, Yoon JH, Mo JS, Kim MY, Park HS. Calcium/calmodulin-
dependent protein kinase IV (CaMKIV) enhances osteoclast differentiation via
the up-regulation of Notch1 protein stability. Biochim Biophys Acta 2013; 1833:
69–79.
32 Fukushima H, Nakao A, Okamoto F, Shin M, Kajiya H, Sakano S et al. The asso-
ciation of Notch2 and NF-kappaB accelerates RANKL-induced osteoclastogenesis.
Mol Cell Biol 2008; 28: 6402–6412.
33 Duan L, de Vos P, Fan M, Ren Y. Notch is activated in RANKL-induced osteoclast
differentiation and resorption. Front Biosci 2008; 13: 7064–7071.
34 Asagiri M, Sato K, Usami T, Ochi S, Nishina H, Yoshida H et al. Autoamplification of
NFATc1 expression determines its essential role in bone homeostasis. J Exp Med
2005; 202: 1261–1269.
35 Sethi N, Dai X, Winter CG, Kang Y. Tumor-derived JAGGED1 promotes osteolytic
bone metastasis of breast cancer by engaging notch signaling in bone cells.
Cancer Cell 2011; 19: 192–205.
36 Riedel SS, Mottok A, Brede C, Bauerlein CA, Jordan Garrote AL, Ritz M et al. Non-
invasive imaging provides spatiotemporal information on disease progression
and response to therapy in a murine model of multiple myeloma. PLoS One 2012;
7: e52398.
This work is licensed under a Creative Commons Attribution 3.0
Unported License. The images or other third party material in this
article are included in the article’s Creative Commons license, unless indicated
otherwise in the credit line; if the material is not included under the Creative Commons
license, users will need to obtain permission from the license holder to reproduce
the material. To view a copy of this license, visit http://creativecommons.org/licenses/
by/3.0/
Supplementary Information accompanies this paper on Blood Cancer Journal website (http://www.nature.com/bcj)
Notch in myeloma bone disease
R Schwarzer et al
8
Blood Cancer Journal &2014 Macmillan Publishers Limited
... Other studies suggest that MM cell skeletal infiltration may be due to events promoted by NOTCH, including MM cell recruitment at the BM [17], mitogenic or anti-apoptotic effect [17][18][19], or MM stem cell self-renewal [20]. Additionally, MM infiltration of BM niche is also associated with the activation of NOTCH signaling in the tumor niche, which promotes angiogenesis [16,21], osteoclastogenesis [22][23][24], and bone marrow stromal cell (BMSC) release of cytokines involved in these events (IL-6, VEGF, IGF-1, SDF-1, RANKL, etc.) [16,18,19,25]. ...
... We and other groups previously reported that MM cells affect the surrounding BM microenvironment inducing osteoclastogenesis [22][23][24] and tumor angiogenesis [16,21] in a NOTCH-dependent way. ...
... The evidence that NOTCH signaling activation, mediated by MM cell heterotypic interaction with the surrounding BM cells, plays a key role in tumor angiogenesis [16,21] and osteoclastogenesis [22][23][24] prompted us to investigate whether NOTCH signaling contributes to determine the impact of MM-EV on these processes. ...
Article
Full-text available
Multiple myeloma (MM) is an incurable hematologic neoplasm, whose poor prognosis is deeply affected by the propensity of tumor cells to localize in the bone marrow (BM) and induce the pro-tumorigenic activity of normal BM cells, leading to events associated with tumor progression, including tumor angiogenesis, osteoclastogenesis, and the spread of osteolytic bone lesions. The interplay between MM cells and the BM niche does not rely only on direct cell-cell interaction, but a crucial role is also played by MM-derived extracellular vesicles (MM-EV). Here, we demonstrated that the oncogenic NOTCH receptors are part of MM-EV cargo and play a key role in EV pro-tumorigenic ability. We used in vitro and in vivo models to investigate the role of EV-derived NOTCH2 in stimulating the protumorigenic behaviour of endothelial cells and osteoclast progenitors. Importantly, MM-EV can transfer NOTCH2 between distant cells and increase NOTCH signaling in target cells. MM-EV stimulation increases endothelial cell angiogenic ability and osteoclast differentiation in a NOTCH2 dependent way. Indeed, interfering with NOTCH2 expression in MM cells may decrease the amount of NOTCH2 also in MM-EV and affect their angiogenic and osteoclastogenic potential. Finally, we demonstrated that the pharmacologic blockage of NOTCH activation by b-Secretase inhibitors may hamper the biological effect of EV derived by MM cell lines and by the BM of MM patients. These results provide the first evidence that targeting the NOTCH pathway may be a valid therapeutic strategy to hamper the pro-tumorigenic role of EV in MM as well as other tumors.
... These initial observations suggest that the main outcome of Notch activation in MM is increased tumor growth. Indeed, in vitro and animal studies show that both homotypic (among MM cells) and heterotypic (between MM cells and host cells) Notch activation increases MM cell proliferation and decreases apoptosis in both human and murine MM cell lines and primary cells from patients [40,43,[48][49][50][51][52] . The increased levels of Notch ligands and receptors in MM cells facilitate physical communication with other neighboring MM cells, leading to increased Cyclin D1 expression and Il-6 production, which in turn stimulates proliferation and promotes survival [43,53] . ...
... In vitro, genetic knockdown of Notch receptor 1/2 or blockade of Notch signaling with GSI decreased angiogenesis induced by MM cells. Further, GSI treatment reduced the secretion of pro-angiogenic cytokines in conditioned media and decreased angiogenesis in animal models of MM [48,78,79] . Until recently, MM cells or stromal cells have been considered the main source of angiogenic factors in the MM niche. ...
Article
Full-text available
Multiple myeloma (MM) is a hematologic cancer characterized by uncontrolled growth of malignant plasma cells in the bone marrow and currently is incurable. The bone marrow microenvironment plays a critical role in MM. MM cells reside in specialized niches where they interact with multiple marrow cell types, transforming the bone/bone marrow compartment into an ideal microenvironment for the migration, proliferation, and survival of MM cells. In addition, MM cells interact with bone cells to stimulate bone destruction and promote the development of bone lesions that rarely heal. In this review, we discuss how Notch signals facilitate the communication between adjacent MM cells and between MM cells and bone/bone marrow cells and shape the microenvironment to favor MM progression and bone disease. We also address the potential and therapeutic approaches used to target Notch signaling in MM.
Preprint
Full-text available
Osteocytes are bone-embedded cells connected via canaliculi and they regulate the bone resorption/formation balance. Osteocyte function is altered in skeletal disorders including cancer. Multiple myeloma (MM) is a hematological malignancy, whereby plasma cells disrupt the bone homeostasis and induce excessive resorption of the mineralized extracellular matrix (ECM), as evidenced by clinical computed tomography (CT). However, morphometric analyses of the mineralized ECM and bone microporosity are not yet performed in patients with MM, nor in the precursor conditions of monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM (SMM). Here, we characterize changes in 3D osteocyte lacunar morphology in bone biopsy samples obtained from patients with MGUS, SMM and newly diagnosed MM, using high resolution microCT. In the small lacunae 20-900 um3 range, we found a trend for lower lacunar density and increasing 50% cutoff of the lacunar volume cumulative distribution for diseased samples. In the larger lacunae 900-3000 um3 range, we found significantly higher lacunar density and microporosity in the MM group compared to the MGUS/SMM group. Regarding the shape distribution, the MGUS/SMM group showed a trend for flatter, more elongated and anisotropic osteocyte lacunae compared to the control group, though the differences were not statistically significant. Altogether, our findings suggest that osteocytes in human MM bone disease undergo changes in their lacunae morphology, which could be an indicator for impaired bone quality. Future studies are needed to show whether these are consistent within comparable anatomical sites and could serve as a diagnostic or prognostic marker.
Article
Full-text available
We here defined the impacts of γ-secretase inhibitors (GSIs) on T-cell-dependent BCMA-specific multiple myeloma (MM) cell lysis and immunomodulatory effects induced by bispecific antibodies (BisAbs). GSIs-induced membrane BCMA (mBCMA) accumulation reached near maximum within 4 h and sustained over 42h-study period on MM cell lines and patient MM cells. GSIs, i.e., 2 nM LY-411575 or 1 μM DAPT, robustly increased mBCMA densities on CD138+ but not CD3+ patient cells, concomitantly with minimum soluble/shed BCMA (sBCMA) in 1 day-culture supernatants. In ex vivo MM-T-cell co-cultures, GSIs overcame sBCMA-inhibited MM cell lysis and further enhanced autologous patient MM cell lysis induced by BCMAxCD3 BisAbs, accompanied by significantly enhanced cytolytic markers (CD107a, IFNγ, IL2, and TNFα) in patient T cells. In longer 7 day-co-cultures, LY-411575 minimally affected BCMAxCD3 BisAb (PL33)-induced transient expression of checkpoint (PD1, TIGIT, TIM3, LAG3) and co-stimulatory (41BB, CD28) proteins, as well as time-dependent increases in % effector memory/central memory subsets and CD8/CD4 ratios in patient T cells. Importantly, LY41157 rapidly cleared sBCMA from circulation of MM-bearing NSG mice reconstituted with human T cells and significantly enhanced anti-MM efficacy of PL33 with prolonged host survival. Taken together, these results further support ongoing combination BCMA-targeting immunotherapies with GSI clinical studies to improve patient outcome.
Article
Full-text available
In multiple myeloma (MM), communication via Notch signaling in the tumor niche stimulates tumor progression and bone destruction. We previously showed that osteocytes activate Notch, increase Notch3 expression, and stimulate proliferation in MM cells. We show here that Notch3 inhibition in MM cells reduced MM proliferation, decreased Rankl expression, and abrogated the ability of MM cells to promote osteoclastogenesis. Further, Notch3 inhibition in MM cells partially prevented the Notch activation and increased proliferation induced by osteocytes, demonstrating that Notch3 mediates MM-osteocyte communication. Consistently, pro-proliferative and pro-osteoclastogenic pathways were upregulated in CD138⁺ cells from newly diagnosed MM patients with high vs. low NOTCH3 expression. These results show that NOTCH3 signaling in MM cells stimulates proliferation and increases their osteoclastogenic potential. In contrast, Notch2 inhibition did not alter MM cell proliferation or communication with osteocytes. Lastly, mice injected with Notch3 knock-down MM cells had a 50% decrease in tumor burden and a 50% reduction in osteolytic lesions than mice bearing control MM cells. Together, these findings identify Notch3 as a mediator of cell communication among MM cells and between MM cells and osteocytes in the MM tumor niche and warrant future studies to exploit Notch3 as a therapeutic target to treat MM.
Article
Full-text available
Multiple myeloma is a hematologic malignancy of plasma cells that causes bone-destructive lesions and associated skeletal-related events (SREs). The pathogenesis of myeloma-related bone disease (MBD) is the imbalance of the bone-remodeling process, which results from osteoclast activation, osteoblast suppression, and the immunosuppressed bone marrow microenvironment. Many important signaling cascades, including the RANKL/RANK/OPG axis, Notch signaling, the Wnt/β-Catenin signaling pathways, and signaling molecules, such as DKK-1, sclerostin, osteopontin, activin A, chemokines, and interleukins are involved and play critical roles in MBD. Currently, bisphosphonate and denosumab are the gold standard for MBD prevention and treatment. As the molecular mechanisms of MBD become increasingly well understood, novel agents are being thoroughly explored in both preclinical and clinical settings. Herein, we will provide an updated overview of the pathogenesis of MBD, summarize the clinical management and guidelines, and discuss novel bone-modifying therapies for further management of MBD.
Chapter
Multiple myeloma is a B cell neoplasm caused by the monoclonal expansion and infiltration of malignant plasma cells in the bone marrow. Myeloma growth can significantly disrupt normal bone function by cell-to-cell interactions and by the secretion of factors that promote osteoclastic bone resorption and inhibit osteoblastic bone formation. This imbalance can lead to the development of osteolytic lesions, hypercalcemia, and susceptibility to bone fractures. Ultimately a vicious cycle is created that increases bone damage, releasing more factors that in turn enhance myeloma cell growth. In addition, other cells within the bone marrow microenvironment, including osteocytes, adipocytes, and immune cells, can influence myeloma cell colonization, survival, and subsequent growth or dormancy. Myeloma in vivo models, patient studies, and clinical trials support these findings. In this chapter we will discuss the biological relationship between cells in the bone marrow microenvironment and myeloma cells, describing key mechanisms that disrupt normal bone function and current bone modulating therapies.
Article
Full-text available
Lytic bone disease remains a life-altering complication of multiple myeloma, with up to 90% of sufferers experiencing skeletal events at some point in their cancer journey. This tumour-induced bone disease is driven by an upregulation of bone resorption (via increased osteoclast (OC) activity) and a downregulation of bone formation (via reduced osteoblast (OB) activity), leading to phenotypic osteolysis. Treatments are limited, and currently exclusively target OCs. Despite existing bone targeting therapies, patients successfully achieving remission from their cancer can still be left with chronic pain, poor mobility, and reduced quality of life as a result of bone disease. As such, the field is desperately in need of new and improved bone-modulating therapeutic agents. One such option is the use of bone anabolics, drugs that are gaining traction in the osteoporosis field following successful clinical trials. The prospect of using these therapies in relation to myeloma is an attractive option, as they aim to stimulate OBs, as opposed to existing therapeutics that do little to orchestrate new bone formation. The preclinical application of bone anabolics in myeloma mouse models has demonstrated positive outcomes for bone repair and fracture resistance. Here, we review the role of the OB in the pathophysiology of myeloma-induced bone disease and explore whether novel OB targeted therapies could improve outcomes for patients.
Article
Systemic inhibition of Notch with γ-secretase inhibitors (GSI) decreases multiple myeloma tumor growth, but the clinical use of GSI is limited due to its severe gastrointestinal toxicity. In this study, we generated a GSI Notch inhibitor specifically directed to the bone (BT-GSI). BT-GSI administration decreased Notch target gene expression in the bone marrow, but it did not alter Notch signaling in intestinal tissue or induce gut toxicity. In mice with established human or murine multiple myeloma, treatment with BT-GSI decreased tumor burden and prevented the progression of multiple myeloma-induced osteolytic disease by inhibiting bone resorption more effectively than unconjugated GSI at equimolar doses. These findings show that BT-GSI has dual anti-myeloma and anti-resorptive properties, supporting the therapeutic approach of bone-targeted Notch inhibition for the treatment of multiple myeloma and associated bone disease. Significance Development of a bone-targeted Notch inhibitor reduces multiple myeloma growth and mitigates cancer-induced bone destruction without inducing the gastrointestinal toxicity typically associated with inhibition of Notch.
Article
Osteocytes, former osteoblasts encapsulated by mineralized bone matrix, are far from being passive and metabolically inactive bone cells. Instead, osteocytes are multifunctional and dynamic cells capable of integrating hormonal and mechanical signals and transmitting them to effector cells in bone as well as in distant tissues. Osteocytes are a significant source of molecules that regulate bone homeostasis by integrating mechanical cues and hormonal signals that coordinate the differentiation and function of osteoclasts and osteoblasts. Osteocyte function is altered in rare and common bone diseases, suggesting that osteocyte dysfunction is directly involved in the pathophysiology of disorders affecting the skeleton. Advances in osteocyte biology initiated the development of novel therapeutics interfering with osteocyte secreted molecules. Moreover, osteocytes are targets and key distributors of biological signals mediating the beneficial effects of various bone therapeutics used in the clinic. Herein, we review the most recent discoveries in osteocyte biology demonstrating that osteocytes regulate bone homeostasis and bone marrow fat via paracrine signaling, influence body composition and energy metabolism via endocrine signaling, and contribute to the damaging effects of diabetes mellitus and hematological and metastatic cancers in the skeleton.
Article
Full-text available
Multiple myeloma is a deadly hematopoietic malignancy. Despite therapeutic advances such as autologous stem-cell transplantation and novel chemotherapeutics, multiple myeloma remains incurable. Multiple myeloma cell localization in the bone marrow and the cross-talk with the bone niche trigger dramatic alterations in the bone marrow microenvironment critical for tumor progression, resistance to therapies and osteolytic bone destruction. It does not surprise that the molecular bases of such fatal interaction are under examination as source of novel potential pharmacological targets. Among these, the Notch family of receptors and ligands has gained growing interest in the recent years due to their early deregulation in multiple myeloma and their ability to affect multiple features of the disease, including tumor cell growth, drug resistance, angiogenesis and bone lesions. This review will explore the evidences of Notch deregulation in multiple myeloma, the state of the art of the currently known roles of its signaling in the fatal interaction between multiple myeloma cells, extracellular matrix and cells in the bone marrow stroma. Finally we will present recent finding concerning the arguments for or against a therapy addressed to Notch signaling inhibition in the cure of multiple myeloma.Leukemia accepted article preview online, 11 January 2013; doi:10.1038/leu.2013.6.
Article
Full-text available
Background: Multiple myeloma (MM) is a B-cell malignancy, where malignant plasma cells clonally expand in the bone marrow of older people, causing significant morbidity and mortality. Typical clinical symptoms include increased serum calcium levels, renal insufficiency, anemia, and bone lesions. With standard therapies, MM remains incurable; therefore, the development of new drugs or immune cell-based therapies is desirable. To advance the goal of finding a more effective treatment for MM, we aimed to develop a reliable preclinical MM mouse model applying sensitive and reproducible methods for monitoring of tumor growth and metastasis in response to therapy. Material and methods: A mouse model was created by intravenously injecting bone marrow-homing mouse myeloma cells (MOPC-315.BM) that expressed luciferase into BALB/c wild type mice. The luciferase in the myeloma cells allowed in vivo tracking before and after melphalan treatment with bioluminescence imaging (BLI). Homing of MOPC-315.BM luciferase+ myeloma cells to specific tissues was examined by flow cytometry. Idiotype-specific myeloma protein serum levels were measured by ELISA. In vivo measurements were validated with histopathology. Results: Strong bone marrow tropism and subsequent dissemination of MOPC-315.BM luciferase(+) cells in vivo closely mimicked the human disease. In vivo BLI and later histopathological analysis revealed that 12 days of melphalan treatment slowed tumor progression and reduced MM dissemination compared to untreated controls. MOPC-315.BM luciferase(+) cells expressed CXCR4 and high levels of CD44 and α4β1 in vitro which could explain the strong bone marrow tropism. The results showed that MOPC-315.BM cells dynamically regulated homing receptor expression and depended on interactions with surrounding cells. Conclusions: This study described a novel MM mouse model that facilitated convenient, reliable, and sensitive tracking of myeloma cells with whole body BLI in living animals. This model is highly suitable for monitoring the effects of different treatment regimens.
Article
Full-text available
Multiple myeloma (MM) is a lethal human cancer characterized by a clonal expansion of malignant plasma cells in bone marrow. Mouse models of human MM are technically challenging and do not always recapitulate human disease. Therefore, new mouse models for MM are needed. Mineral-oil induced plasmacytomas (MOPC) develop in the peritoneal cavity of oil-injected BALB/c mice. However, MOPC typically grow extramedullary and are considered poor models of human MM. Here we describe an in vivo-selected MOPC315 variant, called MOPC315.BM, which can be maintained in vitro. When injected i.v. into BALB/c mice, MOPC315.BM cells exhibit tropism for bone marrow. As few as 10(4) MOPC315.BM cells injected i.v. induced paraplegia, a sign of spinal cord compression, in all mice within 3-4 weeks. MOPC315.BM cells were stably transfected with either firefly luciferase (MOPC315.BM.Luc) or DsRed (MOPC315.BM.DsRed) for studies using noninvasive imaging. MOPC315.BM.Luc cells were detected in the tibiofemoral region already 1 hour after i.v. injection. Bone foci developed progressively, and as of day 5, MM cells were detected in multiple sites in the axial skeleton. Additionally, the spleen (a hematopoietic organ in the mouse) was invariably affected. Luminescent signals correlated with serum myeloma protein concentration, allowing for easy tracking of tumor load with noninvasive imaging. Affected mice developed osteolytic lesions. The MOPC315.BM model employs a common strain of immunocompetent mice (BALB/c) and replicates many characteristics of human MM. The model should be suitable for studies of bone marrow tropism, development of osteolytic lesions, drug testing, and immunotherapy in MM.
Article
Full-text available
Notch signalling links the fate of one cell to that of an immediate neighbour and consequently controls differentiation, proliferation and apoptotic events in multiple metazoan tissues. Perturbations in this pathway activity have been linked to several human genetic disorders and cancers. Recent genome-scale studies in Drosophila melanogaster have revealed an extraordinarily complex network of genes that can affect Notch activity. This highly interconnected network contrasts our traditional view of the Notch pathway as a simple linear sequence of events. Although we now have an unprecedented insight into the way in which such a fundamental signalling mechanism is controlled by the genome, we are faced with serious challenges in analysing the underlying molecular mechanisms of Notch signal control.
Article
Full-text available
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.
Article
Multiple myeloma is a deadly hematopoietic malignancy characterized by proliferation of malignant plasma cells in the bone marrow and bone disease. Interactions between myeloma and bone marrow cells facilitate tumor progression and resistance to therapies. CXCR4 and its ligand SDF-1 play a primary role in this process and are associated with poor prognosis. The Notch pathway is active in myeloma cells, resulting in increased proliferation, resistance to apoptosis and osteolytic activity. We hypothesized that the CXCR4/SDF-1 axis mediates the effects of Notch signals in myeloma cells. Here we show that Notch positively controls CXCR4/SDF-1 expression and functions in myeloma cell lines, and that forced CXCR4 activation partially rescues tumor cells from the outcomes of Notch inhibition. Additionally, we provide evidences that Notch blocking in vivo significantly reduces bone marrow infiltration by human myeloma cells in mouse xenografts. This is the first evidence that a Notch-targeted approach effectively prevents multiple myeloma cell migration, proliferation and resistance to apoptosis by reducing CXCR4 and SDF-1 levels.Leukemia accepted article preview online, 28 January 2013;. doi:10.1038/leu.2013.27.
Article
The Notch signaling pathway plays a crucial role in the regulation of cell fate decision, and is also a key regulator of cell differentiation, including bone homeostasis, in a variety of contexts. However, the role of Notch1 signaling in osteoclast differentiation is still controversial. In this study, we show that Receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation is promoted by the Notch1 intracellular domain (Notch1-IC) and Ca(2+)/Calmodulin dependent protein kinase IV (CaMKIV) signaling. Notch1-IC protein level was augmented by CaMKIV through escape from ubiquitin dependent protein degradation. In addition, CaMKIV remarkably increased Notch1-IC stability, and the kinase activity of CaMKIV was essential for facilitating Notch1 signaling. CaMKIV directly interacted with Notch1-IC and phosphorylates Notch1-IC, thereby decreasing proteasomal protein degradation through F-box and WD repeat domain-containing 7 (Fbw7). We also found that Notch1-IC prevented inhibition of osteoclast differentiation by KN-93 but not the phosphorylation deficient form of Notch1-IC. These results suggest that phosphorylated Notch1-IC by CaMKIV increases Notch1-IC stability, which enhances osteoclast differentiation.
Article
Extensive research efforts over the years have provided us with great insights into how bone-resorbing osteoclasts (OCs) develop and function and, based on such work, valuable antiresorptive therapies have been developed to help combat the excessive bone loss that occurs in numerous skeletal disorders. The RAW 264.7 murine cell line has proven to be an important tool for in vitro studies of OC formation and function, having particular advantages over the use of OCs generated from primary bone marrow cell populations or directly isolated from murine bones. These include their ready access and availability, simple culture for this pure macrophage/pre-OC population, sensitive and rapid development into highly bone-resorptive OCs expressing hallmark OC characteristics following their RANKL stimulation, abundance of RAW cell-derived OCs that can be generated to provide large amounts of study material, relative ease of transfection for genetic and regulatory manipulation, and close correlation in characteristics, gene expression, signaling, and developmental or functional processes between RAW cell-derived OCs and OCs either directly isolated from murine bones or formed in vitro from primary bone marrow precursor cells. Here, we describe methods for the culture and RANKL-mediated differentiation of RAW cells into bone-resorptive OCs as well as procedures for their enrichment, characterization, and general use in diverse analytical assays.