Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

TISSUE-SPECIFIC STEM CELLS

Human Bone Marrow-Derived Mesenchymal Stem Cells Suppress


Human Glioma Growth Through Inhibition of Angiogenesis
IVY A.W. HO,a HAN C TOH,b WAI H. NG,c YUAN L. TEO,a,d CHANG M. GUO,e KAM M. HUI,f,h PAULA Y.P. LAMa,g,h
a
Laboratory of Cancer Gene Therapy; bLaboratory of Cell Therapy and Cancer Vaccine, and fBek Chai Heah
Laboratory of Cancer Genomics, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore;
c
Department of Neurosurgery, National Neuroscience Institute, Singapore; dNational Institute of Education,
Singapore; eDepartment of Orthopedic, Singapore General Hospital, Singapore; gDepartment of Physiology,
National University of Singapore, Singapore; hCancer and Stem Cells Biology Program, Duke-NUS Graduate
Medical School, Singapore

Key Words. Angiogenesis • Mesenchymal stem cells • Glioma • Platelet derived growth factor • Interleukin-1b

ABSTRACT
Tumor tropism of human bone marrow-derived mesenchy- MSC suppressed tumor angiogenesis through the release of
mal stem cells (MSC) has been exploited for the delivery of antiangiogenic factors. Further studies using antibody
therapeutic genes for anticancer therapy. However, the array showed reduced expression of platelet-derived growth
exact contribution of these cells in the tumor microenviron- factor (PDGF)-BB and interleukin (IL)-1b in MSC/glioma
ment remains unknown. In this study, we examined the bio- coculture when compared with controls. In MSC/glioma
logical effect of MSC on tumor cells. The results showed coculture, PDGF-BB mRNA and the corresponding pro-
that MSC inhibited the growth of human glioma cell lines teins (soluble and membrane bound forms) as well as the
and patient-derived primary glioma cells in vitro. Coadmi- receptors were found to be significantly downregulated
nistration of MSC and glioma cells resulted in significant when compared with that of glioma cocultured with normal
reduction in tumor volume and vascular density, which was human astrocytes or glioma monoculture. Furthermore,
not observed when glioma was injected with immortalized IL-1b, phosphorylated Akt, and cathepsin B proteins were
normal human astrocytes. Using endothelial progenitor also reduced in MSC/glioma. Taken together, these data
cells (EPC) from healthy donors and HUVEC endothelial indicated that the antitumor effect of MSC may be medi-
cells, the extent of EPC recruitment and capacity to form ated through downregulation of PDGF/PDGFR axis, which
endothelial tubes was significantly impaired in conditioned is known to play a key role in glioma angiogenesis. STEM
media derived from MSC/glioma coculture, suggesting that CELLS 2013;31:146–155
Disclosure of potential conflicts of interest is found at the end of this article.

cells [2]. MSC could also cause a direct effect through inter-
INTRODUCTION cellular signaling via physical contacts with neighboring cells,
which may consist of tumor cells, stromal fibroblast, infiltrat-
Human mesenchymal stem cells (MSC) are a population of ing immune cells, blood, and lymphatic networks that ulti-
nonhematopoeitic stem cells in the bone marrow microenvir- mately determine the fate of the tumor growth kinetics.
onment that have the ability to self-renew and differentiate Angiogenesis, the formation of new blood vessels from
into multiple lineages. MSC are increasingly being developed pre-existing vessels, is essential for normal development and
for potential clinical usage due to the ease of isolation from maintenance of homeostasis. Tumor angiogenesis is also cru-
adult donors, thus obviating the ethical issues that plague cial for tumor growth; a tumor cannot grow beyond the size
embryonic stem cell research [1]. Because of its strong che- of 2–3 mm3 without blood supply [3]. Tumor angiogenesis
motactic response to injury, MSC have emerged as an attrac- could arise from the recruitment of existing endothelial cells
tive homing vehicle for the delivery of therapeutic genes to (EC) or endothelial progenitor cells (EPC) in response to
tumors and diseased tissues. MSC secrete a variety of cyto- proangiogenic factors. In the latter example, vascular endothe-
kines that have both paracrine and autocrine functions in the lial growth factor (VEGF) [4], fibroblast growth factor (FGF)
tumor milieu. These include suppression of local immune [4], granulocyte macrophage-colony stimulating factor [5],
response, inhibition of fibrosis and apoptosis, modulation of platelet derived growth factor (PDGF) [6], hypoxia-induced
angiogenesis, and stimulation of mitosis and differentiation of macrophage migration inhibitory factor [7] have been shown

Author contributions: I.A.W.H.: conception and design, data analysis and interpretation, and manuscript writing; H.C.T., C.M.G., and
W.H.N: provision of study material; Y.L.T.: collection and assembly of data; K.M.H.: administrative support; P.Y.P.L.: conception and
design, data analysis and interpretation, manuscript writing, financial support, and final approval of manuscript.
Correspondence: Paula Y.P. Lam, Ph.D., Laboratory of Cancer Gene Therapy, Division of Cellular and Molecular Research, Humphrey
Oei Institute of Cancer Research, National Cancer Centre, 11, Hospital Drive, Singapore 169610, Singapore. Telephone: 65-64368357;
Fax: 65-62265694; e-mail: cmrlyp@nccs.com.sg Received February 2, 2012; accepted for publication August 1, 2012; first published
online in STEM CELLS EXPRESS October 3, 2012. V
C AlphaMed Press 1066-5099/2012/$30.00/0 doi: 10.1002/stem.1247

STEM CELLS 2013;31:146–155 www.StemCells.com


Ho, Toh, Ng et al. 147

to mobilize EPC. Alternatively, cytokine-stimulated MSC [8] Biosciences, San Jose, CA, http://www.bdbiosciences.com) and
and MSC that have undergone transdifferentiation into EC or then subjected to centrifugation. The collected cells were cultured
smooth muscles cells [9–11] have been shown to participate in complete ABM.
in tumor angiogenesis and growth.
Having said this, MSC are known for their unique biologi- Isolation and Characterization of EPC
cal property of expressing low levels of major histocompatibil- EPC was isolated according to Brunt et al. [17]. Briefly, periph-
ity complex antigens, which may contribute to their low immu- eral blood mononuclear cells were isolated by centrifugation
nogenicity and anti-inflammation effect when introduced (400g for 20 minutes) in the presence of Histopaque-1077 (1.077
exogenously [2]. Using chemically burned corneas in rat as a g/ml, Sigma, http://www.sigma-aldrich.com). After centrifugation,
model, Oh et al. [12] showed that MSC mediated an anti- the buffy coat was isolated, rinsed twice with phosphate buffered
saline (PBS), and resuspended in endothelial growth medium
inflammatory and antiangiogenic effects on the injured corneal
(EGM-2MV) containing 5% serum, VEGF, FGF-2, EGF, IGF,
surface. Secchiero et al. [13] demonstrated that intraperitoneal
hydrocortisone, and ascorbic acid. Cells were cultured at 37 C
injection of MSC improved the survival of lymphoma-bearing with 5% CO2 in a humidified incubator for 3 days. Nonadherent
mice through induction of EC apoptosis. Murine MSC was cells were removed after 3 days, and adherent cells were cultured
observed to intercalate into the capillary networks of EC and for a further 10–20 days until colonies appeared.
caused capillary degeneration in a MSC/EC coculture system in EPC was characterized by the expression of CD133, CD34,
vitro and abrogated melanoma growth in vivo [14]. More Tie2, vascular endothelial (VE)-cadherin (CD144), VEGF recep-
recently, the antitumor effect of cord blood-derived MSC was tor (VEGFR)-2 and for the uptake of acetylated-low density lipo-
also demonstrated by Dasari et al. [15] in a glioma tumor protein (Ac-LDL) [17, 18]. Briefly, DiI-labeled acetylated-LDL
model. Given the numerous conflicting reports on the ability of (DiI-Ac-LDL; 10 lg/ml; Invitrogen, Carlsbad, CA, http://www.
MSC to support tumor growth, it is important to further explore invitrogen.com) was added to live cells and incubated at 37 C for
the role of MSC in the tumor microenvironment. 1 hour. Cells were rinsed twice with PBS to remove excess dye.
In this study, we demonstrated that MSC elicited an antitu- The number of DiI-Ac-LDL positive cells was counted under a
mor effect in patient-derived primary glioma cells as well as in wide-field fluorescence microscope equipped with a CCD camera
human glioma xenograft model. The antitumor effect is medi- (Nikon Eclipse 90i; Nikon, Tokyo, Japan, http://www.nikon.com).
ated through the secretion of soluble factors that inhibit EPC
Flow Cytometry
recruitment and impaired tumor angiogenesis. Using antibody
array, we have identified and subsequently confirmed that the For quantification of the glioma cells in the coculture experi-
antiangiogenic effect of MSC might be mediated through the ments, carboxyfluorescein diacetate N-succinimidyl ester (CFDA-
downregulation of PDGF/PDGF Receptor (PDGFR) axis, SE)-labeled-DGli36 glioma cells were coincubated with carboxy-
which is known to play a key role in glioma angiogenesis methyl-DiI-labeled MSC for 48 hours. Labeling of the cells was
described previously [16]. Cells were detached with 0.25% tryp-
sin/EDTA and washed twice with PBS. Cell pellet was then
resuspended in 1 ml of PBS. Fluorescence-activated cell sorting
MATERIALS AND METHODS analysis was performed using Cell Quest.
In Vivo Tumor Model
Cell Culture To establish the subcutaneous tumor model for monitoring the effect
Human MSC were isolated, cultured, and characterized as previ- of MSC on glioma growth, immunodeficient BALB/c-nu/nu mice
ously described [16]. The culturing of human glioma cells (Animal Resource Center, Canningvale, Western Australia, http://
DGli36 was described previously [16]. Normal human astrocytes www.arc.wa.gov.au) were inoculated with equal ratio of DGli36 and
(NHA) was purchased from Lonza (Basel, Switzerland, http:// carboxymethyl-DiI-labeled-MSC in the presence of Matrigel (BD
www.lonza.com) and cultured in astrocyte basal medium (ABM) Biosciences). Tumor volume was calculated using the formula: tu-
supplemented with recombinant human EGF, insulin, ascorbic mor volume (mm3) ¼ 0.52  (width [mm2]  (length [mm]) [19].
acid, gentamycin sulfate, amphotericin, L-glutamine, and fetal bo- All animal experiments were performed according to the guidelines
vine serum (FBS) as recommended by the supplier. Immortalized and protocols approved by the Institutional Animal Care and Use
NHAs that overexpress E6, E7, and human telomerase reverse Committee at the Singapore General Hospital, Singapore.
transcriptase (hTERT) were kindly provided by R.O. Pieper (Uni-
versity of California, San Francisco, CA). Human ECs, Human Immunofluorescence and Immunohistochemistry
umbilical vein endothelial cells (HUVEC), was purchased from Staining
Lonza and cultured in endothelial growth medium supplemented Slides were visualized using wide-field microscopy using an
with recombinant epidermal growth factor (EGF), hydrocortisone, upright microscope (Eclipse 90i; Nikon), and images were
VEGF, basic FGF, insulin growth factor (IGF), heparin, ascorbic acquired on a CCD color digital camera (DS-U2; Nikon) using
acid, gentamycin sulfate, amphotericin-B, and 2% FBS. NIS Element AR v2.3 software (Nikon). To assess the cell local-
ization with higher resolution, images were obtained with a con-
Isolation of Primary Human Glioma Cells focal system (LSM 510 Meta; Carl Zeiss, G€ottingen, Germany,
This study has been approved by the SingHealth Centralized http://www.zeiss.com). Images were obtained using either a 40/
Institutional Review Board, Singapore. Primary human glioma 0.75 numerical aperture (N.A.) Plan-Neofluar or a 63/1.25 N.A.
cells were isolated, after informed consent, from the brain tumor Plan-Neofluar oil immersion objective (Carl Zeiss). The following
tissues of patients undergoing brain tumor surgery at the National antibodies were used for the various staining: CD31 (1 lg/ml;
Neuroscience Institute, Singapore. The harvested tissue was sepa- BD Biosciences), cathepsin B (5 lg/ml; R&D Systems, Minneap-
rated into small pieces in the presence of complete medium olis, MN, http://www.rndsystems.com), single-stranded DNA
(ABM) supplemented with 10% FBS, penicillin/streptomycin, (ssDNA; 1 lg/ml; clone F7-26, Millipore, Temecula, CA, http://
normocin, and L-glucose (Cambrex Bio Science Walkersville, www.millipore.com), a-smooth muscle actin (SMA; 1:100 dilu-
Inc., Walkersville, MD, http://www.cambrex.com). The tissue tion; Sigma Aldrich, St. Louis, MO, http://www.sigmaaldrich.
suspensions were first passed through a 5 ml serological pipette, com), CD34 (1 lg/ml), CD133 (1:100 dilution), Tie2 (1 lg/ml),
followed by a 1 ml pipette and finally a flame-polished pasteur VE-cadherin (1 lg/ml) and VEGFR-2 (1 lg/ml) (Santa Cruz Bio-
pipette until no clumps were visible. Following trypsin digestion, technology, Santa Cruz, CA, http://www.scbt.com), and CD45
the homogenate was filtered through a 70-lm cell strainer (BD (1:100 dilution; BD Biosciences). Mouse IgG1 (Dako Denmark
www.StemCells.com
148 MSC Restrict Glioma Angiogenesis

Figure 1. Coculture of MSC with glioma cell lines or patient-derived glioma cells induced tumor cell death in vitro. (A): Equal ratio of
CFDA-SE-labeled-DGli36 glioma cells were cocultured with either carboxymethyl-DiI-labeled-MSC or carboxymethyl-DiI-labeled-NHA. Flow
cytometry FL1-H and FL2-H analysis of the DGli36/MSC coculture demonstrating the separation of the two populations by color. Total number
of CFDA-SE-labeled-DGli36 human glioma cells was counted using fluorescence-activated cell sorting analysis after 48 hours. (B): Equal ratio
of patient-derived glioma cells NNI32 were cocultured with either carboxymethyl-DiI-labeled-MSC (MSC-1 and MSC-8) or carboxymethyl-DiI-
labeled-iNHA . Images were captured from 10 random fields under 100 original magnification using Nikon TE300 wide-field microscope
equipped with a CCD camera after 48 hours. Total number of NNI32 glioma cells was counted using NIS Elements version 3.0. Data shown are
averages of triplicates 6 SEM. Abbreviations: CFDA-SE, carboxyfluorescein diacetate N-succinimidyl ester; iNHA, immortalized NHA; MSC,
mesenchymal stem cell; NHA, normal human astrocytes. **, p < 0.01; ***, p < 0.0001.

A/S, Glostrup, Denmark, http://www.dakocytomation.com) and tubulin (1:5,000 dilution; Sigma Aldrich), and interleukin (IL)-1R1
rabbit serum (Dako Denmark A/S) were used as isotype control. (1:100 dilution; Epitomics, Burlingame, CA, http://www.epitomics.
Mice were sacrificed 21 days post-tumor implantation and com). Chemiluminescence detection was performed using Western
immediately perfused through the heart with ice-cold saline. Lightning chemiluminescence kit (Perkin Elmer, Waltham, MA,
Tumors were harvested and kept in 4% paraformaldehyde (PFA) http://www.perkinelmer.com). The band intensity of specific proteins
for 4 hours at 4 C, transferred to 30% sucrose in PBS overnight was quantified after normalization with the density of b-tubulin with
at 4 C, and embedded in Jung Tissue Freezing Medium (Leica MetaVue imaging software (version 6.1; Sunnyvale, CA).
Microsystems Nussloch, Heidelberg, Germany, http://www.leica-
microsystems.com), snap-frozen in isopentane immersed in liquid Cytokine Array Analysis
nitrogen, and stored at 80 C. RayBio Human Cytokine Array C2000 (RayBiotech, Inc., Nor-
For immunofluorescence staining, frozen sections were fixed cross, GA, http://www.raybiotech.com) that contains antibodies
with ice-cold acetone or 4% PFA, blocked for 30 minutes with targeting to 174 proteins were used to detect proteins that are dif-
10% goat serum, and incubated for 1 hour at room temperature ferentially expressed in DGli36/MSC-CM in comparison to that
with primary antibodies. To assess the pericyte coverage of of DGli36-CM and MSC-CM. One hundred microgram of condi-
microvessels, tumor sections were double-stained for a-SMA tioned media (CM) was used according to manufacturer’s instruc-
expression to detect pericytes and CD31 (1 lg/ml) to detect ECs. tions. Signals were detected by chemiluminescence reaction and
To determine the microvessels density (MVD), the number of quantified using Quantity-One software (Bio-Rad Laboratories,
CD31þ cells was counted. To express MVD counts microscope- Hercules, CA, http://www.bio-rad.com).
independent, counts were transformed and expressed as the num-
ber of microvessels per mm2. ECs Tube Formation Assay
For immunohistochemistry staining, following incubation Tube formation assay was performed according to manufacturer’s
with primary antibodies, sections were stained with goat anti- instructions. Briefly, on the day of the assay, 10 ll of EC matrix
mouse or anti-rabbit polymer, followed by detection using 3,30 (Millipore) was added to the ibidi l-Slide angiogenesis dish (ibidi,
diaminobenzidine substrate solution (Dako Denmark A/S). Anti- Munich, Germany, http://www.ibidi.de) and incubated for 1 hour
body detection in sections stained with mouse monoclonal anti- at 37 C with 5% CO2. During the incubation time, HUVEC was
bodies was performed using the Zenon mouse IgG labeling kit harvested. The cells were counted and 50 ll of cells suspension
(Invitrogen). Sections were then washed and coverslipped in containing 5,000 cells was added to the wells containing the EC
mounting medium. Parallel samples were incubated with no pri- matrix. The slides were then incubated at 37 C for 6 hours. The
mary antibody or an isotype as a specificity control. tubes formed are visualized and captured using the Zeiss 200M
phase contrast microscope at 50 original magnification (Carl
Immunoblot Analysis Zeiss). Tube formation was quantified after 6 hours by measuring
Tumor lysates (DGli36, DGli36/iNHA, and DGli36/MSC) (50 lg) the total tube length using ImageJ software (National Institutes of
were harvested for immunoblot analysis. The following antibodies Health, Bethesda, MD, http://rsb.info.nih.gov/ij/). For checking the
were used: total and activated PDGFR-b (1:1,000 dilution; Cell Sig- effect of the CM on HUVEC tube formation abilities, cells were
naling Technology, Beverly, MA, http://www.cellsignal.com), total resuspended in the respective medium, namely, DGli36-CM, MSC-
and activated Akt (1:1,000 dilution; Cell Signaling Technology), b- CM, NHA-CM, DGli36/NHA-CM, and DGli36/MSC-CM.
Ho, Toh, Ng et al. 149

Figure 2. Antitumor effect of MSC in glioma/MSC coculture in vivo. (A): DGli36 cells (5  105), MSC (5  105), iNHA (5  105), DGli36/
iNHA (5  105:5  105), DGli36/MSC-1 (5  105:5  105), and DGli36/MSC-7 (5  105:5  105) were implanted into the flank of nude mice
in the presence of Matrigel. Tumor volume was monitored at various time points. Data shown are averages of seven mice 6 SEM. *, p < .05;
**, p < .01 (B): Tumor weight comparison between DGli36 and DGli36/MSC-1 tumors. (C): Photomicrograph of DGli36 and DGli36/MSC
tumors. H&E staining was performed on the tumor sections. Blue arrow indicated the presence of pockets of DGli36 tumor cells. Scale bar ¼
100 lm. (D): Photomicrograph of DGli36 tumor coinjected with carboxymethyl-DiI-labeled-MSC. Sections were counterstained with DAPI nu-
clear stain; red color indicates the presence of carboxymethyl-DiI-labeled-MSC. Photomicrograph showed composite image from three separate
images (original magnification 40, scale bar ¼ 250 lm.). I, inset, showed the localization of carboxymethyl-DiI-labeled-MSC (original magnifi-
cation 200; scale bar ¼ 100 lm). Images were captured using Nikon TE300 wide-field microscope equipped with a CCD camera. (E): Photomi-
crograph of tumor sections stained for the presence of apoptotic cells using anti-single-stranded DNA in DGli36, DGli36/MSC, and DGli36/iNHA
tumor. Sections were counterstained with DAPI nuclear stain (blue); green color indicates anti-single-stranded-DNA-positive cells. Sections were
shown at original magnification 200. Scale bar ¼ 50 lm. Abbreviations: H&E, hematoxylin and eosin; iNHA, immortalized NHA; MSC, mes-
enchymal stem cell; N, necrotic region; NHA, normal human astrocytes; T, tumor; DAPI, 40 ,6-diamidino-2-phynylindole.

Transwell EPC Recruitment Assay and IL-1b was quantified using QuantiTech SYBR Green PCR
4 kit (Qiagen, Hilden, Germany, http://www1.qiagen.com). All
For determining the recruitment potential of EPC, EPC (1  10 )
qPCR reactions were performed in duplicate. Standard curves for
were cultured in EBM-2MV media with 0.5% FBS in a 24-well
each gene were generated independently. The relative copy num-
tissue culture insert with a 8-lm pore size membrane (BD Bio-
ber of each sample was calculated according to the corresponding
sciences). CM from DGli36 human glioma cells (DGli36-CM),
standard curve using RotorGene software version 6.0. The rela-
MSC (MSC-CM), or DGli36/MSC (DGli36/MSC-CM) coculture
tive expression levels were calculated by arbitrarily designating
were added to the bottom well. After 6 hours, the filter membrane
the lowest normalized value to 1. Quantification of PDGF-BB,
was fixed with 4% PFA, and cells were mounted in mounting
IGF-1, FGF-2, and IL-1b protein expression was performed using
medium containing propidium iodide (PI, 100 lg/ml; Sigma-
Quantikine human ELISA kit (R&D Systems) according to manu-
Aldrich) containing RNase A. Migration of EPC was determined
facturer’s suggestions.
by counting the number of PI-stained nuclei on the underside of
the membrane under 200 magnification.
Statistical Analysis
Quantitative Real Time Polymerase Chain Reaction Statistical analysis was performed using Prism 3.0 (Graphpad
and ELISA Software Inc., San Diego, CA, http://www.graphpad.com). One-
Quantitative real time polymerase chain reaction (qPCR) was per- way ANOVA followed by Bonferroni multiple comparisons test
formed as described previously [20]. Primers used are listed in were used for comparing statistical significance for more than
supporting information Table S1. Primers for the amplification of two groups. Two-way ANOVA was used for comparing statistical
PDGF-BB and IL-1b were purchased from Qiagen (QuantiTech significance for more than two factors. p value <.05 was consid-
Primer Assay). The expression level of PDGF-BB, IGF-1, FGF-2, ered statistically significant.
www.StemCells.com
150 MSC Restrict Glioma Angiogenesis

Figure 3. Reduction of angiogenesis in DGli36 cocultured with MSC. (A): Angiogenic vessels were identified by immunostaining using anti-
body against CD31 in DGli36, DGli36/MSC, and DGli36/iNHA tumor. Microvessel density of the tumor sections were determined by expressing
the cumulative CD31þ vessels to the area of the tumor sections. Data shown are averages of five randomly selected fields 6 SEM. *, p < .05;
**, p < .01. Scale bar ¼ 100 lm. (B): DGli36 and DGli36/MSC tumor sections were double immunostained with CD31 (green) and a-SMA
(blue). White arrow indicates colocalization of CD31 and a-SMA staining. Scale bar ¼ 20 lm. (C): Confocal images of DGli36/MSC tumor sec-
tions stained for the colocalization of CD31 (green), a-SMA (blue), and carboxymethyl-DiI-labeled MSC (red). Scale bar ¼ 20 lm. Abbrevia-
tions: iNHA, immortalized NHA; NHA, normal human astrocytes; MSC, mesenchymal stem cells; SMA, a-smooth muscle actin.

Fig. 2A). By contrast, there was no statistically significant dif-


RESULTS ference in the tumor volumes between DGli36/MSC-1 and
DGli36/MSC-7 groups. It is important to note that none of the
MSC Induced Cell Death in Glioma Cells in an In MSC isolates became tumorigenic; tumor nodule was not
Vitro and In Vivo System of Coculture detectable in mice injected with the same MSC cell number
To evaluate the influence of MSC on the growth of human in the presence of Matrigel up to 1 month. Gross anatomy of
glioma cells, CFDA-SE-labeled MSC (green fluorescence) the tumors clearly showed that DGli36/MSC tumors were
was cocultured with human glioma DGli36 cells prelabeled visibly less vascularized than those of the DGli36 control
with carboxymethyl-DiI (red fluorescence) at equal ratio for tumors (Fig. 2C). Hematoxylin and eosin (H&E) staining per-
48 hours. Using flow cytometry analysis, we detected a formed on the cocultured tumor sections revealed a peripheral
69.4% reduction in the number of glioma cells in DGli36/ rim of tumor cells cuffing a zone of necrosis (N) (Fig. 2C).
MSC coculture in comparison to that of DGli36/NHA cocul- Furthermore, analysis using antibodies against ssDNA indi-
ture (DGli36/NHA; Fig. 1A). Similar findings were observed cated that the regions of the apoptotic cells in the cocultured
in primary glioma cells derived from human tumor biopsy tumor (Fig. 2E) coincided with the localization of the car-
materials (denoted as NNI32 henceforth). In comparison to boxymethyl-DiI-labeled-MSC (Fig. 2D, inset (I)), thus sug-
NNI32/immortalized NHA (iNHA) coculture, the percentage gesting that the presence of MSC causes cell death in the gli-
of NNI32 was greatly reduced by 52.3% and 56.6% when the oma. On the contrary, apoptosis was not detected in DGli36
cells were cocultured with two independent MSC isolates, tumor and DGli36/iNHA tumor (Fig. 2E; supporting informa-
MSC-1 and MSC-8, respectively (Fig. 1B), suggesting that tion Fig. S1). Taken together, the results demonstrated that
MSC exert an inhibitory effect on glioma cells. specific interactions between MSC and DGli36 human glioma
Because in vitro coculture conditions are not representa- cells induced apoptosis of the glioma cells.
tive of the tumor microenvironment, we performed in vivo
coculture experiment to monitor the effect of MSC on the MSC Restrict Vascular Growth in Glioma Cells In
tumorigenicity of DGli36 glioma cells. For ease of monitoring Vivo and Reduced Angiogenesis In Vitro
changes in the tumor volume, both cell types were implanted Tumors obtained from coinjection of MSC and DGli36 human
in equal ratio subcutaneously. The tumor volumes and tumor glioma cells were further evaluated for vessel morphology
weights in mice injected with equal ratio of MSC and DGli36 and microvessel density. The microvessel density as revealed
were significantly smaller than control animals injected with by CD31 staining was significantly higher in DGli36 and
either DGli36 cells or DGli36/iNHA coculture (Fig. 2A, 2B, DGli36/iNHA tumor when compared with that of DGli36/
respectively). This phenomenon was consistently observed in MSC (p < .01; Fig. 3A). To assess the functional status of
two isolates of MSC, thus indicated that the antitumor effect the tumor neovasculature, tumor sections were simultaneously
was not pertaining to a particular MSC isolates. Two-way stained for CD31 expression and pericytes coverage (a-SMA).
ANOVA analysis indicated that the differences among the six The results showed the presence of CD31þ cells in regional
groups (DGli36, iNHA, MSC, DGli36/iNHA, DGli36/MSC-1, areas of the representative tumor sections of DGli36 and
and DGli36/MSC-7) were statistically significant (p < .0001; DGli36/MSC. Expression of a-SMAþ cells could only be
Ho, Toh, Ng et al. 151

Figure 4. Conditioned medium from DGli36/MSC coculture prevented recruitment of EPC and endothelial tube formation. (A): EPCs were
immunostained with antibodies against CD133, CD34, VE-cadherin, VEGFR-2, Tie2, and CD45. Mouse IgG1 and rabbit serum were used as neg-
ative control for the first five antibodies and CD45, respectively. Scale bar ¼ 100 lm. (B): EPC and MSC were assessed for their ability to
uptake DiI-Ac-LDL (red fluorescence). Cells were counterstained with DAPI (blue fluorescence). Representative images were presented. Scale
bar ¼ 50 lm. (C): EPC recruitment assay was performed using DGli36-CM, MSC-CM, and DGli36/MSC-CM. The number of recruited cells to
the bottom chamber were counterstained with propidium iodide and counted. Data shown are averages of triplicates 6 SEM. *, p < .05. (D):
Angiogenesis assay was performed to determine whether DGli36-CM, MSC-CM, NHA-CM, DGli36/NHA-CM, or DGli36/MSC-CM could induce
tube formation of HUVEC. 50 ng/ml of recombinant human VEGF was used as positive control. Tubes formed were visualized and captured
using the Zeiss 200M wide-field microscope at 50 original magnification. Scale bar ¼ 200 lm. (E): Tube formation index was scored using
ImageJ software (National Institutes of Health, Bethesda, MD). Data were obtained from four replicates of two independent experiments and pre-
sented as ratio of coculture to monoculture. Abbreviations: CM, conditioned medium; EPC, endothelial progenitor cells; LDL, low density lipo-
protein; NHA, normal human astrocytes; MSC, mesenchymal stem cell; VE, vascular endothelial; VEGFR, vascular endothelial growth factor
receptor; DAPI, 40 ,6-diamidino-2-phenylindole; HUVEC, Human umbilical vein endothelial cells.

found in DGli36/MSC but not in DGli36 (Fig. 3B), indicating impaired by DGli36/MSC-CM in comparison to DGli36-CM.
the presence of a reactive microvasculature in the DGli36 Similar findings were obtained with another isolate of MSC
tumors. In DGli36/MSC tumors, a-SMAþ pericytes were (data not shown), indicating that the suppressive effect of
present within the tumor vasculature. These cells did not MSC on EPC recruitment is not specific to one MSC isolate.
overlap with the staining against CD31 (Fig. 3B). As shown Interestingly, the extent of EPC recruitment in MSC-CM was
in Figure 3C, MSC (red cells) could be detected close to similar to those in DGli36/MSC-CM, thus suggesting that the
CD31þ regions, suggesting a possible role in angiogenesis. observed reduced tumor vasculature in the presence of MSC
To further investigate whether MSC may be involved in is likely due to an inhibition on the recruitment of EPC and
EPC recruitment during angiogenesis, we isolated EPC from its associated angiogenic inducing factors.
peripheral blood obtained from healthy donor using density Next, we investigated whether the antitumor effect was
gradient centrifugation. EPC expressed high level of CD133, mediated by antiangiogenic factors in DGli36/MSC coculture
which is indicative of early population of EPC [17]. In addi- using in vitro angiogenesis assay. Our results showed that
tion, the isolated cells were immune-positive for CD34, Tie2, incubation of HUVECs in DGli36-CM, MSC-CM, NHA-CM,
VE-cadherin (CD144), VEGFR2 (CD309) but expressed very and DGli36/NHA-CM stimulated tube formation (Fig. 4D),
low level of CD45 (Fig. 4A; supporting information Fig. S2). which is comparable with the known angiogenic molecule,
Moreover, cellular uptake of DiI-Ac-LDL, which is indicative recombinant human VEGF (data not shown). As shown in
of functional EPC, was also observed in these cells and was Figure 4E, increased tube formation index was observed in
absent in MSC (Fig. 4B). Collectively, these markers con- DGli36/NHA coculture when compared with that of NHA
firmed the endothelial lineage of the isolated cells [17, 18]. monoculture (ratio of DGli36/NHA vs. NHA is 1.5), whereas
As shown in Figure 4C, the recruitment of EPC was severely tube formation index is similar for DGli36/NHA and DGli36
www.StemCells.com
152 MSC Restrict Glioma Angiogenesis

ture (ratio of DGli36/MSC vs. DGli36 or MSC is 0.5), thus


Table 1. Cytokines that are differentially expressed in DGli36/ indicating that MSC impaired tumor angiogenesis through the
mesenchymal stem cells coculture as determined by antibody release of antiangiogenic factors.
array analysis
Fold change MSC Downregulate the Expression of Angiogenic
(normalized Molecules in Glioma Cells
Protein to DGli36-CM) Biological process
To determine which factors are expressed by DGli36/MSC co-
IGF-1 0.431 Angiogenesis, culture, a proteome array was performed using CM harvested
differentiation from DGli36, MSC, and DGli36/MSC. Cytokines that are dif-
PDGF-BB 3.576 Angiogenesis, ferentially expressed in DGli36/MSC coculture were shown in
cell migration, Table 1. These candidates included activated leukocyte cell
and proliferation
adhesion molecule (also known as CD166 antigen), PDGF-
IL-1b 0.06 Angiogenesis,
inflammatory response BB, FGF-2, macrophage inflammatory protein-1-a, Eotaxin-3
MIP-1a (CCL3) 1.26 Chemotaxis, (also known as CCL26), regulated on activation normal T-cell
inflammatory response expressed and secreted (also known as CCL5), tissue inhibi-
Eotaxin-3 (CCL-26) 0.251 Chemotaxis, tors of metalloproteinases-2, interleukin-1b (IL-1b), and IGF-
inflammatory response 1 (Table 1). Because angiogenic index was reduced in the in
RANTES (CCL-5) 0.293 Chemotaxis, vivo coculture, we narrowed down the candidate proteins to
inflammatory response those that were downregulated in angiogenesis. The mRNA
TIMP2 1.473 Cell growth and expression of these genes was further validated by qPCR in
proliferation, ECM
the coculture cells. Significant downregulation of PDGF-BB
disassembly
ALCAM 88.51 Cell adhesion, and IL-1b mRNA expression was observed in DGli36/MSC
axon guidance coculture in comparison to that of DGli36/NHA and DGli36
FGF-2 0.389 Angiogenesis, (Fig. 5A). In line with the qPCR results, lower level of
differentiation PDGF-BB proteins were also detected in DGli36/MSC when
compared with DGli36/NHA and DGli36, thus confirmed our
Fold change shown was normalized to DGli36-CM.
antibody array results (Fig. 5B). However, IGF-1, IL-1b, and
The contribution from MSC-CM was removed by subtracting the
values obtained from that of DGli36/MSC-CM. FGF-2 protein expression failed to show similar trend as
For purpose of completeness, the expression of these cytokines observed in qPCR. IGF-1 and FGF-2 protein expressions were
was also normalized to basal levels in MSC-CM. The results are lower in DGli36/MSC when compared with the controls,
IGF-1 (3.74); PDGF-BB (1.53); IL-1b (4.86); MIP-1a while the expression of IL-1b protein was similar for the
(1.1); Eotaxin-3 (3.42); RANTES (8.53); TIMP2 (0.48); three culture conditions (Fig. 5B).
ALCAM (18.41); FGF-2 (4.08)
Abbreviations: ALCAM, activated leukocyte cell adhesion Impaired IL-1b and PDGF Signaling Cascades in
molecule; CM, conditioned medium; ECM, Extracellular Matrix; Glioma Cocultured with MSC
FGF-2, fibroblast growth factor-2; IL-1b, interleukin 1b; IGF-1,
insulin growth factor-1; PDGF, platelet-derived growth factor; IL-1b, a proinflammatory cytokine, binds to its cell-surface
MIP, macrophage inflammatory protein; RANTES, regulated on receptor and initiates signaling cascade that involves the acti-
activation normal T-cell expressed and secreted; TIMP, tissue vation of nuclear factor kappa B, which in turn activates the
inhibitors of metalloproteinases. cysteine protease, cathepsin B. Cathepsin B has been shown
to be overexpressed in malignant gliomas, which are charac-
terized by aberrant neovascularization [21]. Thus, we first
(ratio of DGli36/NHA vs. DGli36 is 1.0). However, the pres- examined whether MSC may downregulate angiogenesis
ence of MSC in the DGli36 coculture inhibited tube formation through IL-1b signaling. IL-1b protein expression was 40%
by 50% when compared with either DGli36 or MSC monocul- and 80% lower in DGli36/MSC tumor in comparison to that

Figure 5. Crosstalk between MSC and DGli36 human glioma cells downregulate the expression of angiogenic molecules. (A): QPCR was per-
formed on total RNA isolated from DGli36, DGli36/MSC, and DGli36/NHA coculture using primers against PDGF-BB, IGF-1, IL-1b, and FGF-
2. (B): ELISA was performed on CM harvested from DGli36, DGli36/MSC, and DGli36/NHA coculture. For both qPCR and ELISA, the results
shown are normalized to DGli36. Data shown are averages of triplicate 6 SEM. *, p < .05. Abbreviations: CM, conditioned medium; FGF-2,
fibroblast growth factor-2; IL-1b, interleukin-1b; IGF, insulin growth factor; NHA, normal human astrocytes; PDGF-BB, platelet-derived growth
factor.
Ho, Toh, Ng et al. 153

Figure 6. Glioma/MSC coculture resulted in downregulation of PDGF and IL-1b signaling. (A): IL-1b and IL1-R1 expression were determined
using ELISA and Western blot, respectively, on tumor lysate harvested from DGli36, DGli36/MSC, and DGli36/iNHA tumors. Amount of protein
loaded was normalized against b-tubulin. (B): Photomicrograph of tumor sections stained for the presence of cathepsin B in DGli36, DGli36/
MSC, and DGli36/iNHA tumor. Sections were counterstained with hematoxylin and shown at original magnification 200. Scale bar ¼ 50 lm.
(C): PDGF-BB expression was determined using ELISA on DGli36, DGli36/MSC, and DGli36/iNHA tumor lysates. Data shown are averages of
triplicate 6 SEM. (D): Immunoblot was performed on tumor lysates harvested from DGli36, DGli36/MSC, and DGli36/iNHA tumors using
antibodies against Akt, phospho-Akt, PDGFR-b, and phospho-PDGFR-b. Amount of protein loaded was normalized against b-tubulin. *, p < .05.
Immunoblots were performed using tumor lysates harvested from two representative animals per group. Abbreviations: IL-1b, interleukin-1b;
iNHA, immortalized NHA; MSC, mesenchymal stem cells; PDGF-BB, platelet-derived growth factor.

of DGli36/iNHA and DGli36, respectively (Fig. 6A). How- the involvement of PDGF/PDGFR signaling in MSC-induced
ever, using a representative of two animals per group, we tumor suppression.
detected significantly lower level of IL1-R1 in one of the
DGli36/MSC sample (Fig. 6A). Immunohistochemistry stain-
ing confirmed that cathepsin B expression was lost in the
DGli36/MSC tumor, whereas abundant cathepsin B expression DISCUSSION
was detected in DGli36 and DGli36/iNHA tumor alone (Fig.
6B), indicating that MSC exert an inhibitory effect on tumor In the field of cancer therapy, the most attractive feature of
vasculature, in part, through the downregulation of IL-1b sig- MSC is its inherent ability to track tumor cells and its immu-
naling cascade. nosuppressive nature, suggesting that MSC modified with the
In a recent report, the downregulation of cathepsin B appropriate therapeutic gene of interest could home to micro-
was shown to decrease the expression of PDGFR-b expres- scopic tumors and deliver long-term transgene expression
sion and correspondingly increase the apoptotic index of gli- prior to eradication by the immune system. However, the mul-
oma cells [22]. Since PDGF-BB is also known to play an tifaceted role of MSC in the tumor microenvironment is still
important role in glioma angiogenesis [23], we further exam- not fully understood. Results of this study suggested that the
ined whether the reduction in microvessel density may be inhibitory effect of MSC on glioma tumor angiogenesis might
modulated through PDGF-BB/PDGFR-b signaling. As shown be mediated through paracrine pathways that resulted in
in Figure 6C, PDGF-BB protein expression was significantly impaired EPC recruitment and downregulation of proangio-
reduced in the DGli36/MSC tumor section when compared genic factors such as PDGF-BB, IGF-1, FGF-2, and IL-1b.
with that of DGli36 and DGli36/iNHA tumors (Fig. 6C). Im- MSC has been shown to have the capacity to transdifferentiate
munoblot analysis further showed that the levels of activated into EC-like [10] and pericytes-like cells [24] to stimulate angio-
PDGFR-b were correspondingly reduced in DGli36/MSC tu- genesis and promote tumor growth [25]. Direct coculture of adi-
mor in comparison to control tumors (using a representative pose-derived MSC (AMSC) with prostate cancer was shown to
of two animals per group as shown in Fig. 6D). This reduced enhance tumor growth through increase in tumor vascularity
PDGFR-b activation correlated with downregulation of mediated by FGF-2 [26] and differentiation of AMSC into endo-
activated Akt (Fig. 6D) in DGli36/MSC tumors, suggesting thelial-like cells [27]. By contrast, Otsu et al. [14] showed that
www.StemCells.com
154 MSC Restrict Glioma Angiogenesis

coculture containing 1:1 ratio of EC to MSC resulted in EC apo- which may produce synergistic effects in downstream signaling.
ptosis and degeneration of EC capillaries due to the generation of Moreover, IL-1b signaling regulates cathepsin B expression via
reactive oxygen species. In our system, neither differentiation of the NF-jB pathway. Cathepsin B has been shown to promote
MSC into EC (Fig. 3) nor pericytes-like cells (Fig. 3) was remodeling of the ECM to permit neovascularization [40, 41].
observed in glioma/MSC cocultured tumors. Rather, our results Because cathepsin B cleaves and processes IGF-1 following re-
showed decreased microvessel density in vivo that correlated with ceptor internalization [42], a process that is required for cell sig-
reduced tube formation index and recruitment of EPC in vitro, naling via the Shc/MAP kinase pathway, the marked inhibition
suggestive of an antiangiogenic response elicited by the MSC. of cathepsin B expression by IL-1b signaling suggested that
The presence of the a-SMAþ cells, which is indicative of peri- MSC in the tumor milieu prevented ECM remodeling that
cytes, and corresponding reduction of CD31þ microvessel density results in increase invasion and metastasis. Additionally, down-
in glioma/MSC when compared to glioma alone suggested that regulation of cathepsin B, either alone or with urokinase-type
the presence of MSC in the EC environment reduces vascular per- plasminogen activator receptor silencing, decrease the expres-
meability, a finding that is consistent with that of Pati et al. [28]. sion of PDGFR-b, increase the apoptotic index of glioma cells
Pericytes are known to contribute to the integrity of mature [22], and downregulate integrin expression [43]. Furthermore,
blood vessels. Absence of pericytes results in a more reactive ves- disruption of the tube-like structure of EC in vitro and reduced
sel that is irregular, leaky and supports metastasis [29, 30]. It is microvasculature in vivo was also observed in cathepsin B-
interesting to note that although a-SMAþ cells are present in gli- silenced SNB19 glioma cells [44], thus supports our results that
oma/MSC coculture, the level of PDGF-BB, which has been reduced cathepsin B expression retarded ECM-dependent tumor
shown to recruit pericytes to glioma [23], is downregulated. expansion and associated microvascular growth.
PDGF-BB is expressed in the endothelial tip cells or cell that In summary, our results showed that MSC could prevent EPC
leads the new sprout at the forefront during angiogenesis, which recruitment to tumor vasculature, and at the same time, releases
stimulates both recruiting pericytes with PDGFR-b expressed on soluble factors that inhibit ECs tube formation and reduced
their surface and the subsequent creation of the wall of the newly microvessel density. The antitumor effect might be mediated, in
formed capillary [31, 32]. However, PDGF-BB is not the only part, through paracrine pathways that resulted in impaired EPC
factor that mediates pericyte recruitment because mice that are recruitment and downregulation of proangiogenic factors such as
deficient in either PDGF or PDGFR-b have been shown to have PDGF-BB, IGF-1, FGF-2, and IL-1b. Although there are many
normal pericyte coverage in the perisinusoidal capillaries [33], factors that could influence the fate of the tumor growth kinetic,
suggesting the involvement of other factors, including secreted it is important to continue in-depth investigations toward the
protein acidic and rich in cysteine [34] and endothelial nitric ox- understanding of MSC biology as they could serve as an invalu-
ide [35]. On the other hand, the reduced PDGF expression in the able tool in the detection or possibly, treatment of cancer when
presence of MSC may be a consequence of the downregulation of modified with appropriate therapeutic gene.
angiogenesis pathways. Gliomas are highly invasive and malig-
nant tumors and displayed aberrant PDGF signaling [36]. PDGFs
bind and signal through the receptor tyrosine kinases, PDGFR-a CONCLUSION
and b. Binding of PDGFs to its receptors trigger receptor dime-
rization that leads to receptor autophosphorylation and subse-
Our results have clearly demonstrated that MSC exhibited an
quent signal propagation. Inhibition of PDGF signaling with the
receptor tyrosine kinase inhibitor SU6668 [37] or SU5416 [38] antiglioma effect. Analysis indicated that PDGF signaling,
has been shown to inhibit tumor growth through vessel regression which is known to play a key role in glioma angiogenesis, is
one of the pathways that is likely involved in the observed
with or without pericyte involvement.
Inflammation and angiogenesis are pivotal processes in tu- reduced glioma angiogenesis.
mor progression. Inflammatory molecules such as IL-1b are fre-
quently upregulated in cancer cells and macrophages found in
the tumor microenvironment. In the coculture of DGli36/MSC, ACKNOWLEDGMENTS
the IL-1b mRNA levels were significantly reduced when com-
pared with that of DGli36 or DGli36/NHA (Fig. 5A). Although This research is supported by grants from the SingHealth Foun-
we observed a slight decrease in IL-1b proteins in the CM dation and National Medical Research Council, Singapore.
derived from DGli36/MSC when compared with the controls,
the difference was not significant possibly due to the relatively
low levels of proteins (8.74–9.93 pg/ml). However, the lower
IL-1b proteins in DGli36/MSC versus DGli36 or DGli36/NHA DISCLOSURE OF POTENTIAL
were statistically significant in vivo (Fig. 6A). IL-1b has been CONFLICTS OF INTEREST
shown to interact with PDGF-BB to induce sustained phospho-
rylation of PDGFR-b and its association with IL1-R1 [39], The authors indicate no potential conflicts of interest.

5 Cho HJ, Kim HS, Lee MM et al. Mobilized endothelial progenitor


REFERENCES cells by granulocyte-macrophage colony-stimulating factor accelerate
reendothelialization and reduce vascular inflammation after intravascu-
lar radiation. Circulation 2003;108:2918–2925.
1 Aboody KS, Najbauer J, Danks MK. Stem and progenitor cell-medi- 6 Li X, Tjwa M, Moons L et al. Revascularization of ischemic tissues
ated tumor selective gene therapy. Gene Ther 2008;15:739–752. by PDGF-CC via effects on endothelial cells and their progenitors. J
2 Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. Clin Invest 2005;115:118–127.
J Cell Biochem 2006;98:1076–1084. 7 Simons D, Grieb G, Hristov M et al. Hypoxia-induced endothelial
3 Folkman J. Tumor angiogenesis: Therapeutic implications. N Engl J secretion of macrophage migration inhibitory factor and role in endo-
Med 1971;285:1182–1186. thelial progenitor cell recruitment. J Cell Mol Med 2011;15:668–678.
4 Poole TJ, Finkelstein EB, Cox CM. The role of FGF and VEGF in 8 Liu Y, Han ZP, Zhang SS et al. Effects of inflammatory factors on
angioblast induction and migration during vascular development. Dev mesenchymal stem cells and their role in the promotion of tumor
Dyn 2001;220:1–17. angiogenesis in colon cancer. J Biol Chem 2011;286:25007–25015.
Ho, Toh, Ng et al. 155

9 Annabi B, Naud E, Lee YT et al. Vascular progenitors derived from 26 Lin G, Yang R, Banie L et al. Effects of transplantation of adipose tis-
murine bone marrow stromal cells are regulated by fibroblast growth sue-derived stem cells on prostate tumor. Prostate 2010;70:1066–1073.
factor and are avidly recruited by vascularizing tumors. J Cell Bio- 27 Prantl L, Muehlberg F, Navone NM et al. Adipose tissue-derived stem
chem 2004;91:1146–1158. cells promote prostate tumor growth. Prostate 2010;70:1709–1715.
10 Oswald J, Boxberger S, Jorgensen B et al. Mesenchymal stem cells 28 Pati S, Khakoo AY, Zhao J et al. Human Mesenchymal stem cells in-
can be differentiated into endothelial cells in vitro. Stem Cells 2004; hibit vascular permeability by modulating vascular endothelial cad-
22:377–384. herin/beta-catenin signaling. Stem Cells Dev 2011;20:89–101.
11 Lozito TP, Kuo CK, Taboas JM et al. Human mesenchymal stem cells 29 Gerhardt H, Semb H. Pericytes: Gatekeepers in tumour cell metasta-
express vascular cell phenotypes upon interaction with endothelial cell sis? J Mol Med 2008;86:135–144.
matrix. J Cell Biochem 2009;107:714–722. 30 Xian X, Hakansson J, Stahlberg A et al. Pericytes limit tumor cell me-
12 Oh JY, Kim MK, Shin MS et al. The anti-inflammatory and anti- tastasis. J Clin Invest 2006;116:642–651.
angiogenic role of mesenchymal stem cells in corneal wound healing 31 Bergers G, Song S. The role of pericytes in blood-vessel formation
following chemical injury. Stem Cells 2008;26:1047–1055. and maintenance. Neuro Oncol 2005;7:452–464.
13 Secchiero P, Zorzet S, Tripodo C et al. Human bone marrow mesen- 32 Gerhardt H, Betsholtz C. Endothelial–pericyte interactions in angio-
chymal stem cells display anti-cancer activity in SCID mice bearing genesis. Cell Tissue Res 2003;314:15–23.
disseminated non-Hodgkin’s lymphoma xenografts. PLoS One 2010;5: 33 Hellstrom M, Kalen M, Lindahl P et al. Role of PDGF-B and
e11140. PDGFR-beta in recruitment of vascular smooth muscle cells and peri-
14 Otsu K, Das S, Houser SD et al. Concentration-dependent inhibition cytes during embryonic blood vessel formation in the mouse. Devel-
of angiogenesis by mesenchymal stem cells. Blood 2009;113: opment 1999;126:3047–3055.
4197–4205. 34 Rivera LB, Brekken RA. SPARC promotes pericyte recruitment via
15 Dasari VR, Kaur K, Velpula KK et al. Upregulation of PTEN in gli- inhibition of endoglin-dependent TGF-beta1 activity. J Cell Biol 2011;
oma cells by cord blood mesenchymal stem cells inhibits migration 193:1305–1319.
via downregulation of the PI3K/Akt pathway. PLoS One 2010;5: 35 Yu J, deMuinck ED, Zhuang Z et al. Endothelial nitric oxide synthase
e10350. is critical for ischemic remodeling, mural cell recruitment, and blood
16 Ho IA, Chan KY, Miao L et al. HSV-1 amplicon viral vector-medi- flow reserve. Proc Natl Acad Sci USA 2005;102:10999–11004.
ated gene transfer to human bone marrow-derived mesenchymal stem 36 Calzolari F, Malatesta P. Recent insights into PDGF-induced glioma-
cells. Cancer Gene Ther 2008;15:553–562. genesis. Brain Pathol 2010;20:527–538.
17 Brunt KR, Hall SR, Ward CA et al. Endothelial progenitor cell and 37 Laird AD, Christensen JG, Li G et al. SU6668 inhibits Flk-1/KDR
mesenchymal stem cell isolation, characterization, viral transduction. and PDGFRbeta in vivo, resulting in rapid apoptosis of tumor vascula-
Methods Mol Med 2007;139:197–210. ture and tumor regression in mice. FASEB J 2002;16:681–690.
18 Richardson MR, Yoder MC. Endothelial progenitor cells: Quo vadis? 38 Erber R, Thurnher A, Katsen AD et al. Combined inhibition of VEGF
J Mol Cell Cardiol 2011;50:266–272. and PDGF signaling enforces tumor vessel regression by interfering
19 Bergers G, Javaherian K, Lo KM et al. Effects of angiogenesis inhibi- with pericyte-mediated endothelial cell survival mechanisms. FASEB
tors on multistage carcinogenesis in mice. Science 1999;284:808–812. J 2004;18:338–340.
20 Ho IA, Chan KY, Ng WH et al. Matrix metalloproteinase 1 is neces- 39 Chen CN, Li YS, Yeh YT et al. Synergistic roles of platelet-derived
sary for the migration of human bone marrow-derived mesenchymal growth factor-BB and interleukin-1beta in phenotypic modulation of
stem cells toward human glioma. Stem Cells 2009;27:1366–1375. human aortic smooth muscle cells. Proc Natl Acad Sci USA 2006;
21 Rempel SA, Rosenblum ML, Mikkelsen T et al. Cathepsin B expres- 103:2665–2670.
sion and localization in glioma progression and invasion. Cancer Res 40 Cavallo-Medved D, Rudy D, Blum G et al. Live-cell imaging demon-
1994;54:6027–6031. strates extracellular matrix degradation in association with active ca-
22 Malla R, Gopinath S, Alapati K et al. Downregulation of uPAR and thepsin B in caveolae of endothelial cells during tube formation. Exp
cathepsin B induces apoptosis via regulation of Bcl-2 and Bax and in- Cell Res 2009;315:1234–1246.
hibition of the PI3K/Akt pathway in gliomas. PLoS One 2010;5: 41 Wang M, Tang J, Liu S et al. Expression of cathepsin B and micro-
e13731. vascular density increases with higher grade of astrocytomas. J Neuro-
23 Guo P, Hu B, Gu W et al. Platelet-derived growth factor-B enhances oncol 2005;71:3–7.
glioma angiogenesis by stimulating vascular endothelial growth factor 42 Authier F, Kouach M, Briand G. Endosomal proteolysis of insulin-like
expression in tumor endothelia and by promoting pericyte recruitment. growth factor-I at its C-terminal D-domain by cathepsin B. FEBS Lett
Am J Pathol 2003;162:1083–1093. 2005;579:4309–4316.
24 Ball SG, Shuttleworth AC, Kielty CM. Direct cell contact influences 43 Veeravalli KK, Chetty C, Ponnala S et al. MMP-9, uPAR and cathep-
bone marrow mesenchymal stem cell fate. Int J Biochem Cell Biol sin B silencing downregulate integrins in human glioma xenograft
2004;36:714–727. cells in vitro and in vivo in nude mice. PLoS One 2010;5:e11583.
25 Suzuki K, Sun R, Origuchi M et al. Mesenchymal stromal cells pro- 44 Yanamandra N, Gumidyala KV, Waldron KG et al. Blockade of ca-
mote tumor growth through the enhancement of neovascularization. thepsin B expression in human glioblastoma cells is associated with
Mol Med 2011;17:579–587. suppression of angiogenesis. Oncogene 2004;23:2224–2230.

See www.StemCells.com for supporting information available online.

www.StemCells.com

You might also like