Вы находитесь на странице: 1из 11

European Journal of Cell Biology 95 (2016) 153163

Contents lists available at ScienceDirect

European Journal of Cell Biology


journal homepage: www.elsevier.com/locate/ejcb

Research paper

Effects of mesenchymal stem cell-derived cytokines on the functional


properties of endothelial progenitor cells
Witchayaporn Kamprom a,b , Pakpoom Kheolamai b,c,d , Yaowalak U-Pratya b,e ,
Aungkura Supokawej f , Methichit Wattanapanitch g , Chuti Laowtammathron b ,
Surapol Issaragrisil b,e,
a
Department of Immunology, Faculty of Medicine Siriraj hospital, Mahidol University, Bangkok, Thailand
b
Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
c
Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
d
Center of Excellence in Stem Cell Research, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
e
Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
f
Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
g
Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand

a r t i c l e i n f o a b s t r a c t

Article history: Human mesenchymal stem cell (hMSC) is a potential source for cell therapy due to its property to promote
Received 27 August 2015 tissue repair. Although, it has been known that hMSCs promote tissue repair via angiogenic cytokines, the
Received in revised form interaction between hMSC-derived cytokines and the endothelial progenitor cells (EPCs), which play an
28 December 2015
important role in tissue neovascularization, is poorly characterized. We investigate the effect of cytokine
Accepted 3 February 2016
released from different sources of hMSCs including bone marrow and gestational tissues on the EPC
functions in vitro. The migration, extracellular matrix invasion and vessel formation of EPCs were studied
Keywords:
in the presence or absence of cytokines released from various sources of hMSCs using transwell culture
Mesenchymal stem cell
Neovascularization
system. The migration of EPCs was highest when co-culture with secretory factors from placenta-derived
Gestational tissues hMSCs (PL-hMSCs) compared to those co-culture with other sources of hMSCs. For invasion and vessel
formation, secretory factors from bone marrow-derived hMSCs (BM-hMSCs) could produce the maximal
enhancement compared to other sources. We further identied the secreted cytokines and found that
the migratory-enhancing cytokine from PL-hMSCs was PDGF-BB while the enhancing cytokine from BM-
hMSCs on invasion was IGF-1. For vessel formation, the cytokines released from BM-hMSCs were IGF1
and SDF-1. In conclusion, hMSCs can release angiogenic cytokines which increase the migration, invasion
and vessel forming capacity of EPCs. We can then use hMSCs as a source of angiogenic cytokines to induce
neovascularization in injured/ischemic tissues.
2016 Elsevier GmbH. All rights reserved.

Abbreviations: AM-hMSCs, amnion-derived human mesenchymal stem cells; 1. Introduction


ANGPT1, angiopoietin 1; ANGPT2, angiopoietin 2; BM-hMSCs, bone marrow-derived
human mesenchymal stem cells; CFSE, 5-(6)-carboxyuorescein diacetate succin- Human mesenchymal stem cells (hMSCs) are multipotent
imidyl ester; EPCs, endothelial progenitor cells; FGF2, broblast growth factor 2;
stem/progenitor cells which are present in bone marrow, adipose
GAPDH, glyceraldehydes-3-phosphate dehydrogenase; HUVECs, human umbilical
vein endothelial cells; IGF1, insulin-like growth factor 1; IGF2, insulin-like growth tissue, and postnatal sources including umbilical cord, placenta, the
factor 2; IL6, interleukin 6; IL8, interleukin 8; PDGF, platelet-derived growth factor Whartons jelly, and amnion (Bunnell et al., 2008; Int Anker et al.,
beta; PL-hMSCs, placenta-derived human mesenchymal stem cells; PlGF, placenta 2004; Lee et al., 2004; Wang et al., 2004). hMSCs are considered to
growth factor; SDF1, stromal cell-derived factor 1; TGF, transforming growth factor be a potential source for cellular therapy in several disorders such
beta; UC-hMSCs, umbilical cord-derived human mesenchymal stem cells; VEGFA,
as acute graft-versus-host disease (acute GvHD), myocardial infarc-
vascular endothelial growth factor A; VEGFR2, vascular endothelial growth factor
receptor 2; vWF, Von willebrand factor; WJ-hMSCs, Whartons jelly-derived human tion, and neurological diseases (Uccelli et al., 2008; Williams et al.,
mesenchymal stem cells. 2013) due to their immunomodulatory property and multilineage
Corresponding author at: Division of Hematology, Department of Medicine, differentiation capacity (Jiang et al., 2002; Nauta and Fibbe, 2007;
Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand. Pittenger et al., 1999). The therapeutic effects of hMSCs are believed
Tel.: +66 2 419 4448 50; fax: +66 2 411 2012.
due to soluble factors or cytokines released from hMSCs rather
E-mail address: surapolsi@gmail.com (S. Issaragrisil).

http://dx.doi.org/10.1016/j.ejcb.2016.02.001
0171-9335/ 2016 Elsevier GmbH. All rights reserved.
154 W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163

than their direct effect (Meirelles Lda et al., 2009). hMSC-derived 2.3. Characterization of cultured hMSCs by ow cytometry
soluble factors can exert immunomodulatory effect, reduce tissue
inammation and prevent apoptosis of several cell types in injured The 3rd5th passage of hMSCs were characterized for hMSC sur-
tissues which might be the underlying mechanism in the improve- face markers by incubation with the following mouse anti-human
ment of tissue repair and inammatory disease (Gnecchi et al., antibodies: anti-CD45-PerCP (BD Pharmingen, USA), anti-CD34-PE
2008; Wang et al., 2014). hMSCs can stimulate neovascularization (Biolegend, USA), anti-CD90-FITC (AbD Serotec, USA), anti-CD73-
in the injured ischemic tissue via pro-angiogenic cytokines which PE (BD Pharmingen, USA), and anti-CD105-PE (Miltenyi Biotec,
enhance endothelial cell proliferation and migration leading to neo- Germany) for 30 min at 4 C in the dark. After incubation with the
vascularization and tissue restoration (Hung et al., 2007; Kamihata antibodies, cell pellets were washed twice with PBS and xed with
et al., 2001; Li et al., 2010). It was the belief that mature endothelial 1% (w/v) paraformaldehyde in PBS. Flow cytometry was performed
cells in the local vasculature are responsible for postnatal neovascu- by FACS caliburTM Flow cytometer using CellQuestTM software
larization. However, recent studies have shown the important role (Becton Dickinson, USA).
of endothelial progenitor cells (EPCs) in vascular homeostasis and
postnatal neovascularization in both physiological and pathological 2.4. Osteogenic and adipogenic differentiation of cultured hMSCs
conditions (Asahara et al., 1999; Melero-Martin et al., 2007; Tepper
et al., 2005). There are no reported information on the effects of The 3rd5th passage hMSCs were used to assess their adi-
cytokines released from hMSCs on the endothelial progenitor cells. pogenic and osteogenic differentiation potentials. For adipogenic
In order to better understand the therapeutic effect of hMSCs on differentiation, 5 104 cells were cultured in NH AdipoDiff
neovascularization, we investigated the effects of soluble factors Medium (Miltenyi Biotec, Germany). Medium was replaced every
released from hMSCs on the EPC functions including proliferation, 3 days according to the manufacturers instruction. After culture
migration, invasion and vessel formation capacity in vitro, and also for 3 weeks, cells were stained with 0.5% (w/v) Oil Red O (Sigma
identied those factors that play roles on various EPC functions. Sol- Aldrich, USA), in isopropanol for 20 min at room temperature, to
uble factors released from gestational tissues-derived hMSCs were determine the number of hMSC-derived adipocytes in culture.
chosen to study due to easily and non-invasive collection compared For osteogenic differentiation, 5 104 cells were cultured in
to bone marrow-derived hMSCs (BM-hMSCs). NH OsteoDiff Medium (Miltenyi Biotec, Germany). Medium was
replaced every 3 days according to the manufacturers instruction.
After culture for 3 weeks, cells were stained with 40 mM Alizarin
2. Materials and methods
Red S (Sigma Aldrich, USA) for 20 min at room temperature to deter-
mine the number of hMSC-derived osteocytes in culture.
2.1. Subjects
2.5. Cultures and characterization of EPCs from umbilical cord
This study was approved by the Siriraj Institutional Review
blood
Board, Faculty of Medicine Siriraj Hospital, Mahidol University
which was in accordance with the Declaration of Helsinki, the
Twenty milliliter of heparinized umbilical cord blood was col-
Belmont Report, CIOMS Guidelines, and ICH-GCP. Human bone
lected for EPC isolation. Umbilical cord blood-derived mononuclear
marrow samples were obtained from healthy volunteers after giv-
cells were then isolated using IsoPrep (Robbins Scientic Corpora-
ing written informed consent. The gestational tissues (umbilical
tion, USA) density gradient centrifugation, washed twice with PBS
cord, Whartons jelly, placenta, and amnion) were obtained from
(GIBCOTM , Invitrogen Corporation, USA), re-suspended in endothe-
healthy newborns after receiving written informed consent from
lial cell growth medium [endothelial basal medium-2 (LONZA,
their mothers.
Germany), supplemented with EGM-2 single aliquots (LONZA,
Germany) containing 2% (v/v) fetal bovine serum (FBS), 5 g/ml
2.2. Isolation and culture of hMSCs epidermal growth factor, 200 g/ml hydrocortisone, 0.5 g/ml vas-
cular endothelial growth factor, 10 g/ml basic broblast growth
Ten milliliter of heparinized bone marrow was collected for factor, 20 g/ml long R3 Insulin-like growth factor 1 and 1 mg/ml
hMSC isolation. Bone marrow-derived mononuclear cells were ascorbic acid], and plated in an individual well of 6-well plate
then isolated using IsoPrep (Robbins Scientic Corporation, coated with 10 g/ml human bronectin (Amersham Biosciences,
USA) density gradient centrifugation, washed twice with PBS USA) at a density of 1 106 cells/well. After culture for 3 days, the
(GIBCOTM , Invitrogen Corporation, USA), and re-suspended in com- non-adherent cells were removed and fresh medium was added.
plete medium which is Dulbeccos Modied Eagle Medium (DMEM) Cultures were maintained at 37 C in a humidied atmosphere con-
(GIBCOTM , Invitrogen Corporation, USA) supplemented with 10% taining 5% CO2 and medium was replaced every 3 days throughout
(v/v) Fetal Bovine Serum (FBS) (Lonza, USA), 100 U/ml penicillin the entire culture period.
(General Drug House CO., Ltd, Thailand), and 100 g/ml strepto- The cultured cells were characterized for EPC surface markers
mycin (General Drug House CO., Ltd, Thailand). Cell suspensions by incubation with the following mouse anti-human antibodies:
were then plated in 25 cm2 culture ask (Corning, USA) at a den- anti-CD34-PE (R&D Systems, USA), anti-VEGFR2-PE (R&D Systems,
sity of 2 105 cells/cm2 . Cultures were maintained at 37 C in a USA), anti-CD146-FITC (R&D Systems, USA), and anti-vWF-FITC
humidied atmosphere containing 5% CO2 and the medium was (R&D Systems, USA) for 15 min at 4 C in the dark. Cell pellets were
replaced every 3 days throughout the entire culture period. then washed twice with PBS and xed with 1% (v/v) paraformalde-
For hMSC isolation from gestational tissues, umbilical cord, hyde in PBS. Flow cytometry was performed by FACScaliburTM ow
Whartons jelly, placenta, and amnion were cut into small cytometer (Becton Dickinson, USA) using CellQuest software.
pieces and digested by incubation with 0.25% (w/v) trypsin-EDTA
(GIBCOTM , Invitrogen Corporation, USA) for 30 min at 37 C. Cell 2.6. Study of hMSC-derived cytokines on the functional properties
suspensions were washed twice with PBS, re-suspended in com- of EPCs
plete medium and plated in 25 cm2 culture ask (Corning, USA).
Cultures were maintained at 37 C in a humidied atmosphere 2.6.1. Effect on proliferation
containing 5% CO2 and the medium was replaced every 3 days 2 105 hMSCs derived from ve distinct tissues were cultured
throughout the entire culture period. in 2 ml DMEM supplemented with 2% (v/v) FBS for 48 h to generate
W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163 155

Table 1
The sequences of primers for qRT-PCR.

Gene name Sequence of forward primer Sequence of reverse primer

VEGFA 5 -TCCTCACACCATTGAAACCA-3 5 -GATCCTGCCCTGTCTCTCTG-3


ANGPT1 5 -AGGAACCAGCCTCCTCTCTC-3 5 -TTCTCCAGCAGCTGTATCTCAA-3
ANGPT2 5 -AACCAGACGGCTGTGATGAT-3 5 -TTGTCGAGAGGGAGTGTTCC-3
FGF2 5 -AGCGGCTGTACTGCAAAAAC-3 5 -AGCCAGGTAACGGTTAGCAC-3
SDF1 5 -GTGGTCGTGCTGGTCCTC-3 5 -ATCTGAAGGGCACAGTTTGG-3
IGF1 5 -CCGGAGCTGTGATCTAAGGA-3 5 -CCTGCACTCCCTCTACTTGC-3
PlGF 5 -GTTCAGCCCATCCTGTGTCT-3 5 -AACGTGCTGAGAGAACGTCA-3
IGF2 5 -TGCTGGTGCTTCTCACCTTC-3 5 -AGACGAACTGGAGGGTGTCC-3
IL6 5 -CAGGAGCCCAGCTATGAACT-3 5 -GTGAGTGGCTGTCTGTGTGG-3
IL8 5 -AAGAAACCACCGGAAGGAAC-3 5 -AAATTTGGGGTGGAAAGGTT-3
TGF 5 -GAGCCTGAGGCCGACTACTA-3 5 -CACGTGCTGCTCCACTTTTA-3
PDGF 5 -CCGGAGTCGGCATGAATC-3 5 -CGTTGGAGATCATCAAAGGAG-3
GAPDH 5 -GTCAACGGATTTGGTCGTATTG-3 5 -CATGGGTGGAATCATATTGGAA-3

hMSC conditioned media for proliferation assay. Briey, 2.5 104 (Invitrogen Corporation, USA). MicroAmp fast optical 96-well
EPCs were cultured in an individual well of culture E-Plate (Roche reaction plate (Applied Biosystem, USA) was used for qRT-PCR. Each
Applied Science, USA) containing 100 l hMSC conditioned media well contained 3 l cDNA, 1 l 10 M forward and reverse primer
for 7 days. The number of EPCs in each well was continuously mix, and 10 l SYBR Green PCR Mastermix (Applied Biosystem,
monitored throughout the entire culture period by xCELLigence USA). The plate was then sealed with MicroAmp clear adhesive
Real-Time Cell Analyzer (Roche Applied Science, USA) and reported lm (Applied Biosystem, USA) to prevent evaporation of the reac-
as cell index (CI). EPCs cultured in DMEM containing 2% (v/v) FBS tant. PCR was performed using 7500 Fast Real-time PCR system
served as controls. (Applied Biosystem, USA) using the following protocol: 95 C ini-
tial denaturation for 10 min, followed by 40 cycles of denaturation
2.6.2. Effect on migration and invasion (95 C, 10 s), annealing (60 C, 10 s), and extension (72 C, 40 s). The
Five distinct sources of hMSCs were co-cultured with EPCs relative quantity of a target gene was calculated by normaliza-
through 8 m transwell inserts (Corning, USA) to assess the effect of tion with glyceraldehyde-3-phosphate dehydrogenase (GAPDH),
hMSC-derived cytokines on the migratory and invasive capacity of using the 7500 software version 2.0.5 (Applied Biosystem, USA).
EPCs. For migration assay, hMSCs were seeded in the lower cham- The primer sequences were described in Table 1.
ber containing 600 l DMEM supplemented with 2% (v/v) FBS at
the density of 2.5 104 cells/cm2 , while 4 104 EPCs were seeded
2.8. Study of PDGF-BB on EPC migration
in the transwell inserts. After 6 h of co-culture, numbers of EPCs
that migrate to the other side of the transwells membrane were
4 104 EPCs were seeded into 8 m transwell inserts that were
determined by hematoxylin staining.
placed into an individual well of 24-well plates containing 600 l
The procedure of invasion assay was similar to that of migra-
DMEM supplemented with 2% (v/v) FBS and various concentra-
tion assay, except that the membrane of the transwell inserts were
tions (0, 20, 50, and 100 ng/ml) of platelet-derived growth factor-BB
pre-coated with 0.4 g/l Matrigel (BD Biosciences, USA) to serve
(PDGF-BB; R&D system, USA). After 6 h of culture, the effect of
as an extracellular matrix barrier. The numbers of invasive EPCs
PDGF-BB on EPC migration was determined by migration assay as
which are able to degrade coated Matrigel and migrate to the other
previously described.
side of the transwells membrane were determined by hematoxylin
To further determine the role of PDGF- on EPC migration,
staining.
4 104 EPCs were treated with 10 g/ml anti-PDGFR- neutraliz-
ing antibody (R&D system, USA) for 30 min to inhibit the action
2.6.3. Effect on vessel-forming capacity
of PDGF-BB before co-cultured with hMSCs through 8 m tran-
Five distinct sources of hMSCs were co-cultured with EPCs
swell. After 6 h of culture, the migration assay was performed as
through 0.4 m transwell inserts (Corning, USA) to assess the
previously described.
effect of hMSC-derived cytokines on the vessel-forming capac-
ity of EPCs using an in vitro vessel formation assay as previously
described (Jiraritthamrong et al., 2012). Briey, 3.5 104 EPCs 2.9. Study of SDF1, IGF1 and PlGF on EPC invasion
labeled with 5-(6)-carboxyuorescein diacetate succinimidyl ester
(CFSE, CellTraceTM ; Invitrogen, USA) were mixed with an equal 4 104 EPCs were seeded into 8 m Matrigel-coated transwell
number of non-labeled human umbilical vein endothelial cells inserts that were placed into an individual well of 24-well plates
(HUVECs) and plated into an individual well of 6-well plate pre- containing 600 l DMEM with 2% (v/v) FBS which were supple-
coated with MatrigelTM (BD Bioscience, USA) while hMSCs were mented with either 100 ng/ml stromal derived factor1 (SDF1) (R&D
seeded in the transwell inserts at the density of 1 104 cells/cm2 . system, USA), 100 ng/ml insulin-like growth factor1 (IGF1; R&D
After 12 h of co-culture, the extent of incorporated EPCs in the system, USA), 100 ng/ml placenta growth factor (PlGF; R&D sys-
generated capillary-like structure was determined by uorescent tem, USA), or combination of those three factors (100 ng/ml each).
microscopy (Nikon, Japan) using NIS-Elements D software (Nikon, After culture for 18 h, the effect of those factors on capacity of EPC
Japan). invasion was determined by invasion assay as previously described.
To further determine the roles of SDF1, IGF1 and PlGF on
2.7. Expression levels of angiogenic-related genes in hMSCs by EPC invasion, 4 104 EPCs were pre-treated with either 10 M
quantitative real-time PCR (qRT-PCR) AMD3100 octahydrochloride (inhibitor of SDF1; Tocris Bioscience,
UK), 10 g/ml anti-hIGF1R (inhibitor of IGF1; R&D system, USA),
Total RNA were isolated from hMSCs using TRIzol reagent 5 g/ml anti-hVEGFR1 (inhibitor of PlGF; R&D Systems, USA), or
(Invitrogen Corporation, USA). cDNA was synthesized from the combination of all those three inhibitors for one hour before co-
2 g of RNA using the SuperScriptTM III Reverse Trancriptase culture with hMSCs through 8 m Matrigel-coated transwell. After
156 W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163

culture for 18 h, the invasion assay was performed as previously 3.3. Characteristics of umbilical cord blood-derived endothelial
described. progenitor cells (EPCs)

EPCs were cultured from mononuclear cell populations that


2.10. Study of SDF1, IGF1 and PlGF on vessel-forming capacity of had been separated from umbilical cord blood by density gradi-
EPCs ent centrifugation. After culture in endothelial growth medium for
710 days, several EPC-like colonies of cobblestone morphology
3.5 104 CFSE-labeled EPCs were co-cultured with an equal were observed (Fig. 3A). These cells expressed typical EPC surface
number of non-labeled HUVECs in endothelial basal medium sup- markers, including CD34, CD146, Von Willebrand factor (vWF) and
plemented with either 100 ng/ml SDF1, 100 ng/ml IGF1, 100 ng/ml vascular endothelial growth factor receptor 2 (VEGFR2) (Fig. 3B),
PlGF, or the combination of all those three factors (100 ng/ml each). and were able to form capillary-like structures on MatrigelTM
The mixtures of cells were then seeded into an individual well (Fig. 3C).
of Matrigel-coated 24-well plate. After 12 h of culture, an in vitro
vessel formation assay was performed as previously described. 3.4. Effects of hMSC-derived soluble factors on the functional
To further verify the roles of SDF1 and IGF1 on vessel-forming properties of EPCs
capacity of EPCs, 105 EPCs were treated with either 10 M
AMD3100 octahydrochloride (SDF1 inhibitor; Tocris Bioscience, 3.4.1. Effect of hMSC secreted factors on EPC proliferation
UK) or 10 g/ml anti-hIGF1R (IGF1 inhibitor; R&D system, USA) for The conditioned medium derived from hMSCs from various
one hour before co-culture with BM-hMSCs through transwell cul- sources were collected and used to culture EPCs. Similar to con-
ture system. An in vitro vessel formation assay was performed as trol group (cultured in DMEM supplemented with 2% [v/v] FBS), the
previously described. numbers of EPCs cultured in various hMSC-conditioned media were
increased initially during the rst 48 h of culture before rapidly
decreasing toward the baseline level at the end of culture (Fig. 4),
2.11. Statistical analysis
indicating that the soluble factors secreted from bone marrow and
gestational tissue-derived hMSCs did not promote EPC prolifera-
Data were presented as mean standard error of the mean
tion.
(SEM). The paired Students t-test and one-way ANOVA were used
to assess the signicance of differences between observed data.
3.4.2. Effect of hMSC secreted factors on EPC migration
P < 0.05 was considered to be statistically signicant.
EPCs were co-cultured with hMSC from various sources using
the transwell culture system (Fig. 5A). The numbers of EPCs that
3. Results migrate toward various hMSC sources were determined after 6 h
of co-culture. Interestingly, only the soluble factors derived from
3.1. Characteristics of hMSCs derived from gestational tissues and PL-hMSCs signicantly enhanced EPC migration in comparison to
bone marrow controls (306.2 60% vs. 100% of control, P < 0.001) (Fig. 5BC). In
contrast, the number of EPCs, that migrated after co-culture with
hMSCs derived from gestational tissues, including placenta BM-hMSCs (98.8 11.9% vs. 100% of control), UC-hMSCs (80 5.2%
(PL-hMSCs), umbilical cord (UC-hMSCs), Whartons jelly (WJ- vs. 100% of control), WJ-hMSCs (107.1 22.8% vs. 100% of control),
hMSCs) and amnion (AM-hMSCs) exhibited similar characteristics and AM-hMSCs (80.3 10% vs. 100% of control), were not statis-
to those of bone marrow-derived hMSCs (BM-hMSCs). The gesta- tically different from their controls, suggesting that the soluble
tional tissue-derived hMSCs displayed broblast-like morphology factors which were uniquely released from PL-hMSCs enhanced
(Fig. 1AE), expressed typical hMSC surface markers (positive for EPC migration.
CD73, CD90, CD105 and negative for hematopoietic markers CD34 According to the gene expression results (Fig. 2), PDGF- is a
and CD45; Fig. 1P). Moreover, the gestational tissue-derived hMSCs possible candidate for the EPC migration-inducing factor. To deter-
could differentiate toward adipocyte- and osteocyte-lineages as mine the effect of PDGF- on EPC migration, various concentrations
demonstrated by Oil Red O (Fig. 1FJ) and Alizarin Red S staining of PDGF-BB were added to the lower chamber of the transwell
(Fig. 1KO), respectively. system (Fig. 5A) to induce EPC migration (Fig. 5C). As shown in
Fig. 5D, 20-, 50-, and 100 ng/ml PDGF-BB signicantly increased
the numbers of EPC migrants compared to control (120.01 4.76%,
3.2. Expression of pro-angiogenic genes in gestational tissue and 137.26 7.31%, and 125.39 2.04% vs. 100% of control). All the
bone marrow-derived hMSCs various concentrations of PDGF-BB used in this study enhanced
EPC migration at the equivalent levels (Fig. 5D). EPCs were also
The expression levels of pro-angiogenic genes among vari- treated with PDGFR- neutralizing antibody for 30 min before co-
ous sources of hMSCs were determined by quantitative real-time culture with PL-hMSCs and the anti-PDGFR- signicantly reduced
PCR. The expression levels of stromal cell-derived factor 1 (SDF1), the ability of PL-hMSCs to induce EPC migration compared to con-
insulin-like growth factor 1 (IGF1), and placental growth factor trols (71.62 7.37% vs. 100% of control) (Fig. 5E). Taken together,
(PlGF) genes in the BM-hMSCs were signicantly higher than those the results demonstrate that PL-hMSCs enhanced the EPC migra-
of other hMSC sources (Fig. 2). In contrast, the expression level tion and this enhancing effect can be mediated, at least in part, by
of platelet-derived growth factor beta (PDGF-) was signicantly PDGF-BB.
higher in PL-hMSCs than the rest of the hMSC sources. Apart from
those 4 genes, the expression levels of several pro-angiogenic 3.4.3. Effect of hMSC secreted factors on EPC invasion
genes, including vascular endothelial growth factor A (VEGF- EPCs were co-cultured with various sources of hMSCs using
A), broblast growth factor 2 (FGF2), angiopoietin 1 (ANGPT1), the transwell culture system (Fig. 6A). The ability of EPCs to
angiopoietin 2 (ANGPT2), insulin-like growth factor 2 (IGF2), inter- degrade MatrigelTM and migrate toward various hMSC sources
leukin 6 (IL6), interleukin 8 (IL8), and transforming growth factor was determined after 18 h of co-culture. In contrast to the
beta (TGF-) were not signicantly different among the various migration assay, only the soluble factors released from BM-
hMSC sources. hMSCs signicantly enhanced EPC invasion in comparison to
W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163 157

Fig. 1. Characteristics of hMSCs derived from bone marrow and gestational tissues. (A)(E) Fibroblast-like morphology of the various hMSC sources (Scale bar: 500 m). (F)(J)
Adipogenic differentiation of various hMSC sources as determined by Oil Red O staining (Scale bar: 100 m). (K)(O) Osteogenic differentiation of various hMSC sources as
determined by Alizarin Red S staining (Scale bar: 500 m). (P) Immunophenotypes of various hMSC sources as determined by ow cytometry.

controls (223.3 44.3% vs. 100% of control) (Fig. 6BC). The inva- AM-hMSCs (84.15 2.29% vs. 100% of control) was not statisti-
sive capacity of EPCs which were co-cultured with PL-hMSCs cally different in comparison to controls. Soluble factors, uniquely
(100.9 6.4% vs. 100% of control), UC-hMSCs (89.7 10% vs. 100% released from BM-hMSCs appear to signicantly enhance EPC
of control), WJ-hMSCs (59.46 11.08% vs. 100% of control), and invasion.
158 W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163

Fig. 2. Expression of angiogenic genes in various hMSC sources. Total RNA were isolated from hMSCs using TRIzol reagent and expression levels of various angiogenic genes
were determined by qRT-PCR. The relative quantity of a target gene was calculated by normalization with glyceraldehyde-3-phosphate dehydrogenase (GAPDH), using the
7500 software version 2.0.5. Data are presented as mean SEM of three independent experiments. One-way ANOVA was used to assess the signicance of differences between
observed data. * P < 0.05 vs. other hMSC sources.

Fig. 3. Characteristics of umbilical cord blood-derived EPCs. (A) EPC colonies with a cobblestone-like appearance (Scale bar: 500 m). (B) Immunophenotype of EPCs as
determined by ow cytometry. (C) The capillary-like structures generated from EPCs which were cultured on MatrigelTM (Scale bar: 200 m).
W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163 159

Fig. 4. Effect of hMSC-derived cytokines on EPC proliferation. The number of EPCs, which were cultured in conditioned medium derived from various hMSC sources, as
determined by xCELLigence Real-Time Cell Analyzer and reported as cell index (CI) were plotted against culture time. Data are presented as mean SEM of three independent
experiments.

According to the gene expression results (Fig. 2), SDF1, IGF1 when compare with the effect of IGF1 alone (157.9 20.4% of
and PlGF are the possible candidates for the EPC invasion- control vs. 164.8 10.7% of control) (Fig. 6D). Additionally, EPCs
inducing factor. To determine the effect of those three factors were treated with AMD3100, antibody to hIGF1R and antibody to
on EPC invasion, 100 ng/ml SDF1, 100 ng/ml IGF1, and 100 ng/ml hVEGFR1, either separately or in combination, before co-culture
PlGF were added to the lower chamber of the transwell sys- with BM-hMSCs to perform invasion assay. AMD3100, anti-
tem, either separately or in combination, to determine their hIGF1R, anti-hVEGFR1 or the combination of all three inhibitors
capacity to induce EPC invasion. Indeed, 100 ng/ml IGF1 sig- failed to inhibit the enhancing effects of BM-hMSCs on EPC
nicantly increase the number of invaded EPC in comparison invasion (157.2 12.6% vs. 100% of control for AMD3100 treat-
to controls (164.8 10.72% vs. 100% of control). In contrast, ment, 119.8 8.5% vs. 100% of control for anti-hIGF1R treatment,
the number of invaded EPCs that are treated with 100 ng/ml 165.3 29.5% vs. 100% of control for anti-hVEGFR1 treatment, and
SDF1 (116.4 14.4% vs. 100% of control) and 100 ng/ml PlGF 174.4 10.4% vs. 100% of control for the combination of three
(120.5 21.9% vs. 100% of control) were not signicantly different inhibitors) (Fig. 6E), indicating that BM-hMSCs enhanced EPC inva-
when compared to controls (Fig. 6D). Moreover, the combination sion and that enhancing effect was mediated, at least in part, by
of all three factors did not show any additional benecial effect IGF1.

Fig. 5. Effect of hMSC-derived cytokines on EPC migration. (A) The transwell culture system used for migration assay. (B) EPCs which migrated to the other side of transwell
membrane after co-culture with various hMSC sources as determined by hematoxylin staining (Scale bar: 100 m.). (C) Levels of EPCs migration after co-culture with various
hMSC sources. One-way ANOVA was used to assess the signicance of differences between observed data. * P < 0.05 vs. other hMSC sources. (D) Effect of various concentrations
of PDGF-BB on the levels of EPC migration in the absence of hMSCs. Paired Students t-test was used to assess the signicance of differences between observed data. (E)
Effect of 10 g/ml anti-PDGFR- (PDGF- inhibitor) on the migration of EPCs after co-culture with placenta-derived hMSCs. Paired Students t-test was used to assess the
signicance of differences between observed data. (Scale bar: 100 m) Data are presented as mean SEM of three independent experiments.
160 W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163

Fig. 6. Effect of hMSC-derived cytokines on EPC invasion. (A) The transwell culture system used for invasion assay. (B) EPCs which invade the coated Matrigel and migrate to the
other side of transwell membrane after co-culture with various hMSC sources as determined by hematoxylin staining. (Scale bar: 200 m). (C) Levels of EPC invasion after
co-culture with various hMSC sources. One-way ANOVA was used to assess the signicance of differences between observed data. * P < 0.05 vs. other MSC sources. (D) Effect
of SDF1, IGF1, and PlGF on the levels of EPC invasion in the absence of hMSCs. Paired Students t-test was used to assess the signicance of differences between observed data.
(E) Effect of SDF1 inhibitor (AMD3100), IGF1 inhibitor (anti-hIGFIR), and PlGF inhibitor (anti-hVEGFR1) on the invasion of EPCs after co-culture with bone marrow-derived
hMSCs. One-way ANOVA was used to assess the signicance of differences between observed data. Data are presented as mean SEM of three independent experiments.

3.4.4. Effect of hMSC secreted factors on vessel-forming capacity combination of all three factors (157.9 10.9% vs. 100% of con-
of EPC trol, P < 0.05) while the addition of PlGF alone did not increase the
To investigate the effect of hMSC-derived soluble factors on vessel-forming capacity of EPCs (122.37 12.93% vs. 100% of con-
the vessel-forming capacity of EPCs, EPCs were co-cultured with trol) (Fig. 7DE), indicating that BM-hMSCs enhance vessel-forming
hMSCs from various sources using the transwell culture sys- capacity of EPCs and that enhancing effect was mediated, at least
tem and then subjected to an in vitro vessel formation assay in part, by IGF1 and SDF1.
(Fig. 7A). The vessel-forming capacity of EPCs co-cultured with The effect of IGF1 and SDF1 on vessel-forming capacity of
BM-hMSCs was signicantly higher than those co-cultured with EPCs was further determined by treating EPCs with either anti-
PL-hMSCs (121.3 9.7% of control vs. 96.9 4.7% of control) and hIGF1R (IGF1 inhibitor) or AMD3100 (SDF1 inhibitor) before
WJ-hMSCs (121.3 9.7% of control vs. 82.4 8.4% of control) co-culture with BM-hMSCs (Fig. 7A). SDF1 inhibitor could signif-
(Fig. 7BC), suggesting that there are soluble factors, uniquely icantly reduce the ability of BM-hMSCs to promote vessel-forming
released from BM-hMSCs to enhance the vessel-forming capacity capacity of EPCs (85.62 2.44% vs. 100% of control, P < 0.05). IGF1
of EPCs. inhibitor could diminish the enhancing effect of BM-hMSCs on
According to the gene expression results (Fig. 2), SDF1, IGF1 and EPC vessel-forming capacity but the difference was not signicant
PlGF are possible candidate factors. To determine their effects on (90.31 0.61% vs. 100% of control, P > 0.05) (Fig. 7FG). These nd-
the vessel-forming capacity of EPCs, 100 ng/ml SDF1, 100 ng/ml ings indicate that BM-hMSCs enhance the vessel-forming capacity
IGF1, and 100 ng/ml PlGF were added to the lower chamber of EPCs, at least in part, through the secreted SDF1.
of the transwell system, either separately or in combination,
before performing the vessel formation assay. Indeed, the vessel- 4. Discussion
forming capacity of EPCs was signicantly increased by the addition
of 100 ng/ml SDF1 (150.7 13.3% vs. 100% of control, P < 0.05), Recently, hMSCs have been used to treat tissue injury caused
100 ng/ml IGF1 (144.2 12.2% vs. 100% of control, P < 0.05) and the by ischemia and inammation. Despite evidence suggesting that
W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163 161

Fig. 7. Effect of hMSC-derived cytokines on the vessel-forming capacity of EPCs. (A) The competitive vessel formation assay used to determine the vessel-forming capacity of EPCs.
(B) Extent of CFSE-labeled EPCs in the capillary-like structures after co-culture with various hMSC sources as determined by uorescent microscopy. (Scale bar: 200 m). (C)
The vessel-forming capacity of EPCs after co-culture with various hMSC sources. Paired Students t-test was used to assess the signicance of differences between observed
data. (D) Effect of SDF1, IGF1, and PlGF on the extent of CFSE-labeled EPCs in capillary-like structures in the absence of hMSCs. (Scale bar: 200 m). (E) Effect of SDF1, IGF1,
and PlGF on the vessel-forming capacity of EPCs in the absence of hMSCs. Paired Students t-test was used to assess the signicance of differences between observed data. (F)
Effect of SDF1 inhibitor (AMD3100) and IGF1 inhibitor (anti-hIGFIR) on the extent of CFSE-labeled EPCs in capillary-like structures after co-culture with bone marrow-derived
hMSCs. (Scale bar: 200 m). (G) Effect of SDF1 inhibitor (AMD3100) and IGF1 inhibitor (anti-hIGFIR) on vessel-forming capacity of EPCs. One-way ANOVA was used to assess
the signicance of differences between observed data. Data are presented as mean SEM of three independent experiments.

MSCs could differentiate to endothelial cells (ECs) and smooth HGF, IGF1, MCP1, IL-6 and PlGF which play important roles during
muscle cells, which are essential parts of the newly generated the processes of neovascularization and wound healing (Kinnaird
vessels (Davani et al., 2003), most studies suggest that the major et al., 2004; Wang et al., 2006; Wu et al., 2007), the effect of those
mechanism underlying the effects of MSCs is the induction of factors on the functional properties of EPCs during neovascular-
neovascularization in the injured/ischemic tissues by angiogenic ization processes has yet to be characterized. In the present study,
factors released from MSCs (Au et al., 2008; Kinnaird et al., 2004; we used in vitro culture model to study the effect of hMSCs from
Yao et al., 2015). Neovascularization of the ischemic tissues could be various sources on the proliferation, migration, invasion and vessel-
accomplished by either angiogenesis, which generates new blood forming capacity of EPCs, important steps during the process of
vessels from the pre-existing vessels, or vasculogenesis that gener- vasculogenesis. The expression levels of several angiogenic genes
ates new vessels de novo from endothelial progenitor cells (EPCs). in gestational tissues-derived hMSCs (PL-hMSCs, UC-hMSCs, WJ-
The multi-step neovascularization processes are tightly regulated hMSCs and AM-hMSCs) are different from those of BM-hMSCs.
by many angiogenic factors, such as vascular endothelial growth Moreover, the effect of gestational tissues-derived hMSCs on the
factor (VEGF), basic broblast growth factors (bFGF), angiopoietin migration, invasion capacity and vessel-forming capacity of EPCs
(ANGPT), and transforming growth factor (TGF-), among others were also different from those of BM-hMSCs.
(Carmeliet and Jain, 2011). The BM-hMSCs expressed signicantly higher levels of IGF1,
Although several previous studies showed that BM-MSCs pro- SDF1, and PlGF gene and induced higher level of EPC invasion
duced and secreted several angiogenic factors, including VEGF, compared to gestational tissue-derived hMSCs. IGF1 released from
162 W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163

BM-hMSCs were responsible, at least in part to the enhancing effect References


of BM-hMSCs on EPC invasion. A recent previous study showed
that IGF1 increased the colony formation, migration and vessel- Asahara, T., Masuda, H., Takahashi, T., Kalka, C., Pastore, C., Silver, M., Kearne, M.,
Magner, M., Isner, J.M., 1999. Bone marrow origin of endothelial progenitor
formation of EPCs (Thum et al., 2007). To our knowledge, this is the cells responsible for postnatal vasculogenesis in physiological and pathological
rst report on the effect of IGF1 on EPC invasion. neovascularization. Circ. Res. 85, 221228.
For EPC migration, the PL-hMSCs signicantly induced higher Au, P., Tam, J., Fukumura, D., Jain, R.K., 2008. Bone marrow-derived mesenchymal
stem cells facilitate engineering of long-lasting functional vasculature. Blood
level of EPC migration compare to other sources of hMSCs. The abil- 111, 45514558.
ity of PL-hMSCs to induce EPC migration was mediated, at least in Bunnell, B.A., Flaat, M., Gagliardi, C., Patel, B., Ripoll, C., 2008. Adipose-derived stem
part, through the action of secreted PDGF-BB. The effect of PDGF- cells: isolation, expansion and differentiation. Methods 45, 115120.
Burlacu, A., Grigorescu, G., Rosca, A.M., Preda, M.B., Simionescu, M., 2013. Factors
BB observed in this study is in agreement with a previous study secreted by mesenchymal stem cells and endothelial progenitor cells have
which reported that the overexpression of PDGFR- gene in EPCs complementary effects on angiogenesis in vitro. Stem Cells Dev. 22, 643653.
enhanced their proliferative, migratory and vessel forming capacity Carmeliet, P., Jain, R.K., 2011. Molecular mechanisms and clinical applications of
angiogenesis. Nature 473, 298307.
(Wang et al., 2012). Our ndings suggested that the higher level of
Ceradini, D.J., Kulkarni, A.R., Callaghan, M.J., Tepper, O.M., Bastidas, N., Kleinman,
PDGF- in PL-hMSCs might make this particular source of hMSCs M.E., Capla, J.M., Galiano, R.D., Levine, J.P., Gurtner, G.C., 2004. Progenitor cell
more efcient in inducing EPC migration compared to other hMSC trafcking is regulated by hypoxic gradients through HIF-1 induction of SDF-1.
sources. Nat. Med. 10, 858864.
Davani, S., Marandin, A., Mersin, N., Royer, B., Kantelip, B., Herve, P., Etievent, J.P.,
The addition of PDGF-BB, SDF1, and IGF1 could signicantly Kantelip, J.P., 2003. Mesenchymal progenitor cells differentiate into an
enhance the migration, invasion and vessel-forming capacity of endothelial phenotype, enhance vascular density, and improve heart function
EPCs but the enhancing effects were only 5070% of those with PL- in a rat cellular cardiomyoplasty model. Circulation 108 (Suppl. 1), Ii253Ii258.
Gnecchi, M., Zhang, Z., Ni, A., Dzau, V.J., 2008. Paracrine mechanisms in adult stem
hMSC- and BM-hMSC-secretome. Moreover, the inhibition of SDF-1 cell signaling and therapy. Circ. Res. 103, 12041219.
and IGF-1 failed to abolish the enhancing effects of hMSCs on EPC Gruber, R., Kandler, B., Holzmann, P., Vogele-Kadletz, M., Losert, U., Fischer, M.B.,
functions. Taken together, these results suggest that, apart from Watzek, G., 2005. Bone marrow stromal cells can provide a local environment
that favors migration and formation of tubular structures of endothelial cells.
PDGF-BB, SDF-1 and IGF-1, PL-MSCs and BM-MSCs might secrete Tissue Eng. 11, 896903.
other additional unidentied factors that enhance the migration, Hung, S.C., Pochampally, R.R., Chen, S.C., Hsu, S.C., Prockop, D.J., 2007. Angiogenic
invasion and vessel-forming capacity of EPCs. effects of human multipotent stromal cell conditioned medium activate the
PI3K-Akt pathway in hypoxic endothelial cells to inhibit apoptosis, increase
Our in vitro vessel formation assay showed that SDF1 and IGF1 survival, and stimulate angiogenesis. Stem Cells 25, 23632370.
which were highly expressed in BM-hMSCs could signicantly Int Anker, P.S., Scherjon, S.A., Kleijburg-van der Keur, C., de Groot-Swings, G.M.,
enhance vessel-forming capacity of EPCs either individually or in Claas, F.H., Fibbe, W.E., Kanhai, H.H., 2004. Isolation of mesenchymal stem cells
of fetal or maternal origin from human placenta. Stem Cells 22, 13381345.
combination. Previous studies showed that IGF1 could increase
Jiang, Y., Jahagirdar, B.N., Reinhardt, R.L., Schwartz, R.E., Keene, C.D.,
proliferative, migratory and vessel-forming capacity of EPCs (Thum Ortiz-Gonzalez, X.R., Reyes, M., Lenvik, T., Lund, T., Blackstad, M., Du, J., Aldrich,
et al., 2007) while SDF1 was shown to regulate the mobilization of S., Lisberg, A., Low, W.C., Largaespada, D.A., Verfaillie, C.M., 2002. Pluripotency
EPCs toward ischemic tissue (Ceradini et al., 2004) and enhance of mesenchymal stem cells derived from adult marrow. Nature 418, 4149.
Jiraritthamrong, C., Kheolamai, P., U-Pratya, Y., Chayosumrit, M., Supokawej, A.,
angiogenic potential of EPCs (Zemani et al., 2008). Although a Manochantr, S., Tantrawatpan, C., Sritanaudomchai, H., Issaragrisil, S., 2012.
previous study demonstrated that the combination of VEGF and In vitro vessel-forming capacity of endothelial progenitor cells in high glucose
PlGF could efciently enhance vessel formation in vitro (Li et al., conditions. Ann. Hematol. 91, 311320.
Kamihata, H., Matsubara, H., Nishiue, T., Fujiyama, S., Tsutsumi, Y., Ozono, R.,
2006), our results showed that PlGF, which also highly expressed Masaki, H., Mori, Y., Iba, O., Tateishi, E., Kosaki, A., Shintani, S., Murohara, T.,
in BM-hMSCs, did not increase the vessel-forming capacity of EPCs. Imaizumi, T., Iwasaka, T., 2001. Implantation of bone marrow mononuclear
Therefore, our results suggested that PlGF might exhibit its angio- cells into ischemic myocardium enhances collateral perfusion and regional
function via side supply of angioblasts, angiogenic ligands, and cytokines.
genic potential only in the presence of other angiogenic factors, Circulation 104, 10461052.
such as VEGF. Similar to the previous studies which indicate that Kinnaird, T., Stabile, E., Burnett, M.S., Lee, C.W., Barr, S., Fuchs, S., Epstein, S.E., 2004.
BM-hMSC-derived cytokines had no effect on the proliferation of Marrow-derived stromal cells express genes encoding a broad spectrum of
arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through
endothelial cells (Burlacu et al., 2013; Gruber et al., 2005), all ges- paracrine mechanisms. Circ. Res. 94, 678685.
tational tissue-derived hMSCs generated in the present study did Lee, O.K., Kuo, T.K., Chen, W.M., Lee, K.D., Hsieh, S.L., Chen, T.H., 2004. Isolation of
not promote EPC proliferation. multipotent mesenchymal stem cells from umbilical cord blood. Blood 103,
16691675.
In conclusion, our ndings provide a better understanding of
Li, B., Sharpe, E.E., Maupin, A.B., Teleron, A.A., Pyle, A.L., Carmeliet, P., Young, P.P.,
the angiogenic potential of soluble factors secreted from bone mar- 2006. VEGF and PlGF promote adult vasculogenesis by enhancing EPC
row and gestational tissue-derived hMSCs. We demonstrate for the recruitment and vessel formation at the site of tumor neovascularization.
rst time that hMSCs derived from distinct sources produced and FASEB J. 20, 14951497.
Li, H., Zuo, S., He, Z., Yang, Y., Pasha, Z., Wang, Y., Xu, M., 2010. Paracrine factors
secreted a distinct combination of angiogenic factors which exert released by GATA-4 overexpressed mesenchymal stem cells increase
different biological effects on EPC function. The knowledge gained angiogenesis and cell survival. Am. J. Physiol. Heart Circ. Physiol. 299,
from this study might help to improve the therapeutic applica- H1772H1781.
Meirelles Lda, S., Fontes, A.M., Covas, D.T., Caplan, A.I., 2009. Mechanisms involved
tions of hMSCs as sources of angiogenic cytokines for inducing in the therapeutic properties of mesenchymal stem cells. Cytokine Growth
neovascularization in the injured/ischemic tissues. Furthermore, Factor Rev. 20, 419427.
the administration of hMSC-derived angiogenic factors which play Melero-Martin, J.M., Khan, Z.A., Picard, A., Wu, X., Paruchuri, S., Bischoff, J., 2007.
In vivo vasculogenic potential of human blood-derived endothelial progenitor
important role during the specic stages of vasculogenesis might be cells. Blood 109, 47614768.
used to induce neovascularization in the injured/ischemic tissues Nauta, A.J., Fibbe, W.E., 2007. Immunomodulatory properties of mesenchymal
without the need for cell transplantation. stromal cells. Blood 110, 34993506.
Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., Douglas, R., Mosca, J.D.,
Moorman, M.A., Simonetti, D.W., Craig, S., Marshak, D.R., 1999. Multilineage
potential of adult human mesenchymal stem cells. Science 284, 143147.
Acknowledgements Tepper, O.M., Capla, J.M., Galiano, R.D., Ceradini, D.J., Callaghan, M.J., Kleinman,
M.E., Gurtner, G.C., 2005. Adult vasculogenesis occurs through in situ
recruitment, proliferation, and tubulization of circulating bone
This research project was funded by grants from Thailand marrow-derived cells. Blood 105, 10681077.
Research Fund (grant no. RTA 488-0007) and the Commission on Thum, T., Hoeber, S., Froese, S., Klink, I., Stichtenoth, D.O., Galuppo, P., Jakob, M.,
Higher Education (grant no. CHE-RES-RG-49). Witchayaporn Kam- Tsikas, D., Anker, S.D., Poole-Wilson, P.A., Borlak, J., Ertl, G., Bauersachs, J., 2007.
Age-dependent impairment of endothelial progenitor cells is corrected by
prom was supported by the Royal Golden Jubilee Ph.D. program of growth-hormone-mediated increase of insulin-like growth-factor-1. Circ. Res.
the Thailand Research Fund. 100, 434443.
W. Kamprom et al. / European Journal of Cell Biology 95 (2016) 153163 163

Uccelli, A., Moretta, L., Pistoia, V., 2008. Mesenchymal stem cells in health and Williams, A.R., Hatzistergos, K.E., Addicott, B., McCall, F., Carvalho, D., Suncion, V.,
disease. Nat. Rev. Immunol. 8, 726736. Morales, A.R., Da Silva, J., Sussman, M.A., Heldman, A.W., Hare, J.M., 2013.
Wang, H., Yin, Y., Li, W., Zhao, X., Yu, Y., Zhu, J., Qin, Z., Wang, Q., Wang, K., Lu, W., Enhanced effect of combining human cardiac stem cells and bone marrow
Liu, J., Huang, L., 2012. Over-expression of PDGFR-beta promotes mesenchymal stem cells to reduce infarct size and to restore cardiac function
PDGF-induced proliferation, migration, and angiogenesis of EPCs through after myocardial infarction. Circulation 127, 213223.
PI3K/Akt signaling pathway. PLoS ONE 7, e30503. Wu, Y., Chen, L., Scott, P.G., Tredget, E.E., 2007. Mesenchymal stem cells enhance
Wang, H.S., Hung, S.C., Peng, S.T., Huang, C.C., Wei, H.M., Guo, Y.J., Fu, Y.S., Lai, M.C., wound healing through differentiation and angiogenesis. Stem Cells 25,
Chen, C.C., 2004. Mesenchymal stem cells in the Whartons jelly of the human 26482659.
umbilical cord. Stem Cells 22, 13301337. Yao, Y., Huang, J., Geng, Y., Qian, H., Wang, F., Liu, X., Shang, M., Nie, S., Liu, N.,
Wang, M., Crisostomo, P.R., Herring, C., Meldrum, K.K., Meldrum, D.R., 2006. Du, X., Dong, J., Ma, C., 2015. Paracrine action of mesenchymal stem cells
Human progenitor cells from bone marrow or adipose tissue produce VEGF revealed by single cell gene proling in infarcted murine hearts. PLoS ONE 10,
HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am. e0129164.
J. Physiol. Regul. Integr. Comp. Physiol. 291, R880R884. Zemani, F., Silvestre, J.S., Fauvel-Lafeve, F., Bruel, A., Vilar, J., Bieche, I., Laurendeau,
Wang, Y., Chen, X., Cao, W., Shi, Y., 2014. Plasticity of mesenchymal stem cells in I., Galy-Fauroux, I., Fischer, A.M., Boisson-Vidal, C., 2008. Ex vivo priming of
immunomodulation: pathological and therapeutic implications. Nat. Immunol. endothelial progenitor cells with SDF-1 before transplantation could increase
15, 10091016. their proangiogenic potential. Arterioscler. Thromb. Vasc. Biol. 28, 644650.

Вам также может понравиться