You are on page 1of 9

Stem Cell Research (2011) 6, 206–214

available at www.sciencedirect.com

www.elsevier.com/locate/scr

REGULAR ARTICLE

Human mesenchymal stem cell-conditioned


medium improves cardiac function following
myocardial infarction
Leo Timmers a , Sai Kiang Lim b,c,⁎, Imo E. Hoefer a , Fatih Arslan a ,
Ruenn Chai Lai b,d , Angelique A.M. van Oorschot e , Marie Jose Goumans e ,
Chaylendra Strijder a , Sui Kwan Sze f , Andree Choo g , Jan J. Piek h ,
Pieter A. Doevendans a , Gerard Pasterkamp a , Dominique P.V. de Kleijn a,i,⁎⁎
a
Department of Cardiology, University Medical Center Utrecht, Netherlands
b
Institute of Medical Biology, A*STAR, Singapore
c
Department of Surgery, YLL School of Medicine, NUS, Singapore
d
NUS Graduate School for Integrative Sciences & Engineering, NUS, Singapore
e
Department of Molecular Cell Biology, Leiden University Medical Center, Netherlands
f
Nanyang Technological University, Singapore
g
Biotechnology Institute, Singapore
h
Department of Cardiology, Academic Medical Center, Amsterdam, Netherlands
i
Interuniversity Cardiology Institute of the Netherlands, Utrecht, Netherlands

Received 21 August 2010; received in revised form 29 December 2010; accepted 3 January 2011
Available online 28 January 2011

Abstract Recent studies suggest that the therapeutic effects of stem cell transplantation following myocardial infarction (MI)
are mediated by paracrine factors. One of the main goals in the treatment of ischemic heart disease is to stimulate vascular
repair mechanisms. Here, we sought to explore the therapeutic angiogenic potential of mesenchymal stem cell (MSC)
secretions. Human MSC secretions were collected as conditioned medium (MSC-CM) using a clinically compliant protocol. Based
on proteomic and pathway analysis of MSC-CM, an in vitro assay of HUVEC spheroids was performed identifying the angiogenic
properties of MSC-CM. Subsequently, pigs were subjected to surgical left circumflex coronary artery ligation and randomized to
intravenous MSC-CM treatment or non-CM (NCM) treatment for 7 days. Three weeks after MI, myocardial capillary density was
higher in pigs treated with MSC-CM (645 ± 114 vs 981 ± 55 capillaries/mm2; P = 0.021), which was accompanied by reduced
myocardial infarct size and preserved systolic and diastolic performance. Intravenous MSC-CM treatment after myocardial
infarction increases capillary density and preserves cardiac function, probably by increasing myocardial perfusion.
© 2010 Elsevier B.V. All rights reserved.

⁎ Correspondence to: S.K. Lim, Institute of Medical Biology, A*STAR, Department of Surgery, YLL School of Medicine, NUS, 8A Biomedical
Grove, No. 05–505 Immunos, Singapore 136648. Fax: +65 6464 2048.
⁎⁎ Correspondence to: D.P.V. de Kleijn, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Room G02.523,
Heidelberglaan 100, 3584 CX Utrecht, The Netherlands. Fax: +31 30 2522693.
E-mail addresses: saikiang.lim@imb.a-star.edu.sg (S.K. Lim), d.dekleijn@umcutrecht.nl (D.P.V. de Kleijn).

1873-5061/$ - see front matter © 2010 Elsevier B.V. All rights reserved.
doi:10.1016/j.scr.2011.01.001
207

Introduction development of blood vessels, angiogenesis, proliferation of


endothelial cells, neovascularization, and vascularization
Despite significant advances in myocardial revascularization, are the processes with the highest overrepresentation
coronary artery disease and subsequent myocardial infarc- (supplementary data, Table 1).
tion (MI) are leading causes of morbidity and mortality Having identified the potential involvement of MSC-CM in
worldwide. One of the main goals in the treatment of blood vessel formation, we used an in vitro assay of
ischemic heart disease is the development of effective angiogenesis to further investigate this potential. In an
strategies to stimulate vascular repair mechanisms in order angiogenesis assay based on formation of capillary sprouts by
to achieve adequate tissue perfusion. HUVEC spheroids, MSC-CM induced significantly more capil-
Although the potential of stem cell therapy for MI is still lary sprouting compared with NCM (Fig. 1A). The average
heavily debated as a result of studies that failed to show length of the sprouts did not increase following MSC-CM
improved outcome (Moelker et al., 2006; de Silva et al., 2008; treatment compared with NCM (Fig. 1B). The total length of
Janssens et al., 2006; Lunde et al., 2006; Meyer et al., 2009; all sprouts, however, significantly increased in spheroids
Menasche et al., 2008), there are several preclinical and incubated with MSC-CM when compared with NCM (Fig. 1C).
clinical studies that demonstrated the therapeutic effects of In addition, a HUVEC/Matrigel angiogenesis assay was
stem cell transplantation (Hashemi et al., 2008; Wolf et al., performed, which revealed a longer average tube length with
2009; Assmus et al., 2006; Schachinger et al., 2006; Strauer CM compared with NCM (1.07 ± 0.046 vs 0.99 ± 0.041 mm;
et al., 2002; Yousef et al., 2009). While robust evidence for P = 0.003).
cardiomyocyte replacement is still limited, more evidence is
accumulating for vascular repair mechanisms following stem MSC-CM treatment after infarction increases
cell transplantation (van Laake et al., 2009; Losordo & capillary density and decreases collagen density
Dimmeler, 2004; Erbs et al., 2007). Among the stem cells that
have been tested for cardiac repair, mesenchymal stem cells Capillary density was higher in the border areas following
(MSCs) derived from adult bone marrow have emerged as a MSC-CM treatment compared with NCM treatment (Figs. 2A
promising stem cell type for treating cardiovascular disease and B). In the remote area, capillary density did not differ
(Pittenger & Martin, 2004). Recent studies suggest that at between MSC-CM treatment and NCM treatment (Fig. 2C).
least some of the therapeutic effects of MSCs are mediated by We were unable to reliably quantify capillary density in the
paracrine factors secreted by the cells (Caplan & Dennis, infarct area due to the unorganized structure of the infarct
2006; Gnecchi et al., 2005). These paracrine factors could be granulation tissue 3 weeks following MI.
exploited to extend the therapeutic possibilities of MSCs for Picrosirius staining revealed that collagen density in the
the treatment of a variety of diseases, including MI. border area and in the remote myocardium was lower in pigs
Application of MSC secretions rather than MSCs themselves treated with MSC-CM compared with pigs treated with NCM.
in order to restore tissue perfusion could enable us to avoid In the infarct area, no difference was detected between the
some of the limiting factors associated with cell therapy, such treatment groups (Fig. 3).
as immune incompatibility, tumorigenicity, costs, and wait-
ing time for ex vivo expansion.
MSC-CM preserves cardiac function following MI
We previously described how human MSC lines can be
reproducibly generated from human embryonic stem cell
(hESC) lines, which constitute an invariable source of One hour after coronary artery ligation, cardiac function
consistently uniform batches of MSCs (Lian et al., 2007). In decreased similarly in both groups. Fractional area shorten-
addition, we established for the first time the collection of ing decreased from 45.8 ± 1.9 to 32.3 ± 2.0% in the NCM group
these MSC secretions as conditioned medium (MSC-CM) in a and from 46.9 ± 4.6 to 29.7 ± 3.4% in the MSC-CM group
clinically compliant manner, circumventing exposure to (P = 0.670). Also cardiac output (2.80 ± 0.10 L/min (NCM) vs
virus, mouse cells, or serum (Sze et al., 2007). Recently, we 2.67 ± 0.22 L/min (MSC-CM); P = 0.596), mean aortic pressure
demonstrated that MSC-CM treatment following ischemia/ (90 ± 8.3 mm Hg (NCM) vs 84 ± 5.4 mm Hg (MSC-CM); P = 0.544)
reperfusion injury reduces myocardial apoptosis and oxida- and dP/dtMAX (1135 ± 83 mm Hg/s (NCM) vs 1096 ± 83 mm Hg/s
tive stress (Timmers et al., 2007a). In the current paper, we (MSC-CM); P = 0.741) were similar in both groups 1 h after
reveal the angiogenic potential of clinically compatible MSC- coronary artery ligation.
CM and demonstrate in a large animal model that it can be In the 3 weeks following MI, the LV internal areas
used to improve cardiac function following MI. increased in all pigs. However, the increase in LV internal
area tended to be more pronounced in NCM-treated animals,
pointing to reduced remodeling in MSC-CM-treated pigs
Results (Figs. 4A and B). Echocardiographic wall thickness of the
infarct area decreased in pigs treated with NCM, but not in
MSC-CM stimulates angiogenesis in vitro pigs treated with MSC-CM (Fig. 4C). The fractional area
shortening was higher following MSC-CM treatment, which
Ingenuity pathway analysis of the secretory proteome of reflects increased systolic cardiac performance (Fig. 4D).
MSC-CM revealed that within the physiological system Also other systolic functional parameters such as ejection
development and function group, cardiovascular system fraction, dP/dtMAX, stroke volume, and stroke work were
development and function has the highest overrepresenta- higher in animals treated with MSC-CM. Improved cardiac
tion (P b 10-12) with 75 different focus proteins. Within the function translated into improved hemodynamic parameters,
group of cardiovascular system development and function, with the mean arterial pressure and cardiac output being
208 L. Timmers et al.

Figure 1 HUVEC spheroid assay of angiogenesis. The number of capillary sprouts (A), mean length of sprouts (pixels, B), and total
length of sprouts (pixels, C) after incubation of HUVEC spheroids with MSC-CM (n = 12) or NCM (n = 12). Representative pictures of MSC-
CM and NCM are demonstrated in panels D and E, respectively. * P b 0.01 vs NCM.

higher in MSC-CM-treated pigs. In addition, diastolic function infarct size and preserved systolic and diastolic function in a
improved, as assessed with dP/dtMIN and diastolic chamber porcine model of myocardial infarction.
stiffness. All functional parameters are summarized in Table 1. Recently, it was demonstrated that administration of MSC
Myocardial infarct size 3 weeks following coronary artery secretions improved outcome in a murine model of MI
ligation was smaller in MSC-CM-treated pigs compared to NCM- (Gnecchi et al., 2005). Vascular repair mechanisms consti-
treated pigs (11.6 ± 2.0% vs 16.6 ± 1.2% of the LV; P = 0.050). tute an important means to reduce myocardial injury and to
Two pigs died during the surgical procedure due to improve cardiac function following MI. Although the thera-
refractory ventricular fibrillation before treatment with peutic potential of stem cell transplantation is still a subject
MSC-CM or NCM, and were therefore excluded from the of discussion, evidence for vascular repair mechanisms
study. Two pigs (1 treated with MSC-CM and 1 treated with following stem cell transplantation is growing (van Laake
NCM) died in the third week following MI without signs of et al., 2009; Losordo & Dimmeler, 2004; Erbs et al., 2007). If
heart failure or cardiac rupture, probably due to ventricular vascular repair mechanisms are induced by paracrine
tachycardia/fibrillation. factors, application of stem cell secretions may extend the
repertoire of stem cell-based therapies with a different
dimension.
Discussion An in vitro sprouting assay confirmed Ingenuity analysis of
earlier MSC-CM proteomics data and identified the angio-
Here, we demonstrate that human MSC-CM treatment genic potential of MSC-CM. In order to investigate the
enhances capillary density, resulting in reduced myocardial therapeutic potential of MSC-CM to stimulate angiogenesis
209

Figure 2 Capillary density. Representative sections of border areas after lectin staining to visualize endothelium, 3 weeks after MI,
from pigs treated with NCM (A, n = 9) or MSC-CM (B, n = 9). Quantification of capillary density in remote and border area is
demonstrated in panel C. Scale bars, 500 μm.

in vivo, we used a porcine model of myocardial infarction.


During myocardial ischemia following coronary artery liga-
tion, cardiomyocytes in the central infarct perish within
minutes to hours after coronary occlusion. The border zone
of the ischemic area, however, contains injured but viable
cells under hypoperfused and hypoxic conditions which may
either be salvaged or die, depending on the conditions after
ischemia. Restoration of myocardial perfusion by means of
angiogenesis restores the metabolic needs of cardiomyocytes
in the border area and offers the cells a better chance of
survival, which in turn could lead to reduced myocardial
injury. Intracoronary delivery of bone marrow-derived
progenitor cells has been suggested to promote vascular
repair following MI, potentially via paracrine mechanisms
(Erbs et al., 2007). Such effect was also observed in a rodent
model of hind limb ischemia (Kinnaird et al., 2004). As
became evident from our histological analyses, MSC-CM
increased capillary density in the border areas and myocar-
dial infarct size was reduced, which was accompanied by
Figure 3 Collagen density. Quantification of collaged density preserved LV dimensions and cardiac function.
in the infarct, border, and remote areas in pigs treated with NCM Besides stimulation of angiogenesis, it cannot be excluded
and MSC-CM. that also other mechanisms contributed to reduction of
210 L. Timmers et al.

A B

LVES area (cm2)

LVES area (cm2)


*

C D

*
WT infarct (cm)

FAS (%)
E F
*
dP/dtMAX (mmHg/s)

dP/dtMin (mmHg/s)

Figure 4 Cardiac function. Left ventricular (LV) end-diastolic internal area (A), LV end-systolic internal area (B), posterolateral
myocardial wall (infarct) thickness (C), fractional area shortening (FAS, D), dPdtMAX (E), and dPdtMIN (F), before MI (baseline), 1 h post
MI and 3 weeks post MI in pigs treated with MSC-CM (black dots, n = 9) or NCM (open dots, n = 9). * P b 0.05 vs NCM.

myocardial infarct size and preservation of myocardial ments were performed with open chest. In addition, we have
function. With preserved left ventricular dimensions and not been able to investigate the effect of common
infarct thickness, inhibition of post MI cardiac remodeling is a comorbidities such as hypertension, dyslipidemia, and
potential contributing mechanism. Collagen density in the diabetes, which might influence the biologic effect of MSC
border area and remote myocardium was lower in pigs secretions. Due to the permanent coronary artery ligation,
treated with MSC-CM. Cardiac fibrosis is associated with we were not able to assess the area at risk 3 weeks post MI.
heart failure following myocardial infarction and may We were not able to oversee the effects of human protein
contribute to progression of systolic and diastolic dysfunc- injection on the immune system of the porcine host and the
tion (Burlew & Weber, 2002; Cleutjens et al., 1999). We potential influence on study endpoints. Furthermore, the
recently demonstrated that intravenous and intracoronary molecular mechanism for the induction of angiogenesis and/
MSC-CM treatment reduces myocardial apoptosis and oxida- or arteriogenesis and an exact determination of the
tive stress (Timmers et al., 2007a). In that study, cardio- responsible angiogenic factors are still lacking. Several
myocyte death was targeted acutely following ischemia and clinical trials with potent angiogenic factors failed to
reperfusion injury. In the current study, treatment was improve outcome in patients (Henry et al., 2003; Simons
started late after coronary artery ligation, and a model of et al., 2002). It is not clear how paracrine factors in stem cell
permanent coronary artery ligation was used in order to transplants, or the paracrine factors alone, can induce a
investigate the angiogenic potential of MSC-CM and its effect better effect. Most likely, a combination of angiogenic
on cardiac function independent of reduction of reperfusion factors rather than a single protein is responsible. Also
injury. The fact that cardiac function improved using this exosomes could play an important role. We previously
protocol shows that there may be additional benefit from demonstrated exosomes in MSC-CM to contain and deliver
prolonged treatment with MSC secretions. cardioprotective compounds preventing cardiomyocyte
For this study, a permanent coronary artery ligation was death following ischemia and reperfusion injury (Lai et al.,
performed in anesthetized animals and functional measure- 2010). Exosomes constitute a potential delivery tool to
211

Table 1 Hemodynamic and Functional Parameters.


Baseline 3 weeks post MI Change from baseline (%)
Parameter NCM CM NCM CM NCM CM
HR, bpm 66 ± 4 73 ± 6 84 ± 6 86 ± 6 28 ± 12 12 ± 11
MAP, mm Hg 95 ± 5 89 ± 5 66 ± 6 85 ± 5 * -30 ± 5 -5 ± 8 *
CO, L/min 3.1 ± 0.1 3.0 ± 0.2 3.7 ± 0.6 5.3 ± 0.5 23 ± 21 78 ± 20
dP/dt max, mm Hg/s 1213 ± 77 1241 ± 56 1157 ± 162 1650 ± 124 * 4 ± 13 40 ± 9 *
dP/dt min, mm Hg/s -1274 ± 122 -1130 ± 96 -883 ± 98 -1313 ± 95 * 27 ± 9 -19 ± 19 *
EDP, mm Hg 8.0 ± 0.3 7.1 ± 0.6 13.3 ± 1.6 12.1 ± 1.1 68 ± 21 94 ± 42
WT infarct, cm 0.79 ± 0.04 0.83 ± 0.05 0.55 ± 0.02 0.79 ± 0.05 * -27 ± 4 -5 ± 4 *
Lvia (ED), cm2 15.8 ± 0.5 14.2 ± 1.0 26.0 ± 1.8 21.8 ± 1.5 64 ± 12 55 ± 12
Lvia (ES), cm2 8.5 ± 0.4 7.6 ± 0.9 17.7 ± 1.6 12.5 ± 0.9 * 104 ± 14 60 ± 14 *
FAS, % 45.8 ± 1.9 46.9 ± 4.6 32.6 ± 2.2 42.4 ± 3.3 * -28 ± 6 -6 ± 8 *
SV, ml 48 ± 3 42 ± 2 43 ± 4 62 ± 3 * -4 ± 11 52 ± 10 *
EDV, ml 134 ± 16 129 ± 9
ESV, ml 93 ± 15 73 ± 9
EF, % 33.7 ± 4.4 49.1 ± 3.3 *
SW, mm Hg·ml 2497 ± 333 4595 ± 361 *
Stiffness, mm Hg/ml 0.26 ± 0.04 0.14 ± 0.02 *
Summary of cardiac functional parameters of pigs treated with NCM (n = 9) and MSC-CM (n = 9), determined with echocardiography and
conductance catheter-based LV pressure and volume measurements. MI indicates myocardial infarction; HR, heart rate; MAP, mean arterial
pressure; CO, cardiac output; EDP, end-diastolic pressure; WT, wall thickness; LVia, left ventricular internal area; SV, stroke volume; EDV,
end-diastolic volume; ESV, end-systolic volume; EF, ejection fraction; SW, stroke work; Stiffness, end-diastolic chamber stiffness. * P b 0.05
vs NCM.

effectively carry angiogenic factors into the intracellular expression profile, were generated by trypsinization and
space of the target tissue, protecting the factors from propagation of hESCs from either the HuES9 hESC line or the
proteases. This may explain why relatively low doses can H1 hESC line in feeder- and serum-free selection media (Lian
have such profound angiogenic effects. Whether exosomes et al., 2007). One of these cultures, HuES9.E1, can be stably
and their content are also responsible for the stimulation of expanded for at least 80 population doublings. To harvest MSC
angiogenesis following MI remains a subject for further secretions, hESC-derived MSC cultures were transferred to a
investigation. chemically defined, serum-free culture medium to condition
Here, we demonstrate that human MSC secretions the medium for 3 days before the media containing MSC
stimulate angiogenesis. These data support the hypothesis secretions were collected, clarified by centrifugation, con-
that MSC transplantation induces vascular repair mechanisms centrated 25 times using 10-kDa MW cutoff ultrafiltration
via paracrine pathways. Most importantly, human MSC membranes and sterilized by filtration through a 220-nm
secretions harvested as conditioned medium using a clini- filter. The protein concentration after concentration was
cally compliant protocol reduced infarct size and preserved 0.50 mg/ml. As a negative control, the above-noted serum-
cardiac function in a large animal model of myocardial free culture medium was processed equally (nonconditioned
infarction. The study therefore identifies MSC-CM as a novel medium [NCM]).
therapeutic biologic in the improvement of cardiac function
after myocardial infarction. Ingenuity pathway analysis

Methods For this analysis we used the proteomic data obtained


previously (Sze et al., 2007). Bioinformatic analysis was
MSC-CM preparation performed using Ingenuity pathway analysis (Ingenuity IPA-
2002 version 7.1). The 201 proteins were uploaded in
Ingenuity with Ingenuity Knowledge Base as a reference set
The protocols for MSC generation and CM preparation have
and direct and indirect relationships were included. All
been described in previous papers (Lian et al., 2007; Sze et al.,
molecules or relationships were considered for the filter
2007). In short, a chemically defined serum-free culture
summary. In the summary, the focus was on Top Bio functions
medium (DMEM without phenol red, supplemented with insulin,
in which physiological system development and function was
transferrin, and selenium, 5 ng/ml FGF2, 5 ng/ml PDGF AB,
analyzed in depth.
glutamine-penicillin-streptomycin, and ß-mercaptoethanol)
was conditioned by MSCs derived from human embryonic
stem cells (hESCs) using a clinically compliant protocol. Three HUVEC spheroid assay
polyclonal, karyotypically stable, and phenotypically MSC-like
cultures, that do not express pluripotency-associated markers Spheroids were formed from HUVECs to determine the effect
but displayed MSC-like surface antigens (CD29+, CD44+, of conditioned medium on angiogenic sprout formation (Korff
CD49a+/e+, CD105+, CD166+, CD34–, CD45–) and gene & Augustin, 1998). Spheroids were formed overnight in 20%
212 L. Timmers et al.

methocel in culture medium and subsequently embedded in myocardial infarction, the animals were anesthetized once
a collagen–methocel matrix. Sprouting was induced over- more and the sternum was reopened. Echocardiography and
night with MSC-CM (n = 12) or non-CM (NCM) (n = 12) to conductance catheter-based pressure–volume (PV) loop
determine the effect of the secreted chemotactic proteins recordings were obtained to assess cardiac function and
on capillary sprouting. After overnight sprouting, spheroids geometry. After the functional measurements the heart was
were fixed in 10% formaldehyde and pictures were taken and excised for laboratory analysis.
the length and number of sprouts were quantified.
Immunohistochemistry
Animals
Three weeks following MI, myocardial biopsies were obtained
All experiments were performed in accordance with the from the infarct area, border area, and remote area and
“Guide for the Care and Use of Laboratory Animals” prepared fixated in 4% formalin for 24 h before being embedded in
by the Institute of Laboratory Animal Resources and with prior paraffin. To determine capillary density, a lectin staining,
approval by the Animal Experimentation Committee of the delineating the endothelium, was performed following
Faculty of Medicine, Utrecht University, the Netherlands. antigen retrieval by boiling in 10 mM citrate buffer.
Endogenous biotin was blocked using 0.1% avidin solution
(DakoCytomation Biotin Blocking system X0590) for 15 min at
Sample size calculation room temperature (RT) and subsequently with 0.01% biotin
solution (Dako) for 15 min at RT. After blocking with 3.0% BSA
In a previous study, the mean fractional area shortening in PBS, the sections were incubated overnight with biotiny-
(FAS), after LCx ligation was 36.5% (Timmers et al., 2007b). lated BS-1 (Sigma, L3759) in a 1:300 dilution in PBS with 0.1%
With an alpha of 0.05, power of 0.80, SD of 5.8, and BSA and subsequently with streptavidin-HRPO (1:1000 in PBS)
difference of 20% considered relevant, 10 animals per group for 1 h at RT. Finally, the sections were incubated for 30 min
were needed. To allow for 10% mortality, 11 animals per in 3-amino-9-ethylcarbazole (AEC) and stained with hema-
group were enrolled. toxylin before being embedded. Capillary density was
expressed as the number of capillaries per square millimeter
Pig study design tissue. Quantification of collagen density was performed
using picrosirius red staining with circularly polarized light
Myocardial infarction was induced in 22 Dalland Landrace and digital image microscopy after conversion into gray-
pigs (60–70 kg, IDDLO, Lelystad, the Netherlands) by surgical value images, as described before (Timmers et al., 2007b).
left circumflex coronary artery (LCx) ligation. Two pigs died
perioperatively before randomization and were therefore Myocardial infarct size
excluded from the study. The remaining 20 pigs were
randomized after surgery by using sealed envelopes to After excision of the heart, the LV was isolated and cut into 5
intravenous treatment with MSC-CM (2.0 ml MSC-CM, i.e., slices from apex to base. To discriminate infarct tissue from
1.0 mg protein) or 2.0 ml NCM, initiated 4 h after coronary viable myocardium, the slices were incubated in 1%
artery ligation and the treatment was continued for 7 days triphenyltetrazolium chloride (TTC, Sigma-Aldrich Chemi-
twice daily via a catheter inserted into the jugular vein. The cals, Zwijndrecht, Netherlands) in 37 °C Sörensen buffer
pigs were sacrificed 3 weeks after MI. To prevent thrombotic (13.6 g/L KH2PO4 + 17.8 g/L Na2H PO4·2H2O, pH 7.4) for
complications and arrhythmias, all pigs were treated with 15 min. All slices were scanned from both sides and in each
clopidogrel 75 mg/day from 3 days before MI until termina- slide the infarct area was compared to total area using digital
tion and amiodarone 400 mg/day from 10 days before MI planimetry software. After correction for the weight of the
until termination. slices, infarct size was calculated as a percentage of the LV.

Myocardial infarction and operational procedure Cardiac function

Pigs were anesthetized as described previously (Timmers et Midpapillary short axis epicardial echocardiography was
al., 2007b). During the entire operation, electrocardiogram, performed before coronary artery ligation, 1 h after and
arterial pressure, and capnogram were continuously moni- 3 weeks after coronary artery ligation (Prosound SSD-5000,
tored. After median sternotomy, the left ventricular 5 MHz probe UST-5280-5, Aloka Holding Europe AG, Zug,
pressure (LVP) was measured using a pressure tipped Millar Switzerland). Wall thickness (WT) of the infarct area and left
catheter that was inserted through the apex into the left ventricular internal area (LVia) were measured at end-
ventricle. A transonic flow probe (Transonic Systems Inc., diastole (ED) and end-systole (ES). Systolic wall thickening
Ithaca, NY, USA) was placed around the proximal aorta to (SWT) was calculated as (WT(ES) – WT(ED))/WT(ED) * 100 (%)
measure cardiac output. Sutures were then tightened to and fractional area shortening (FAS) as (LVia(ED) – LVia(ES))/
permanently occlude the proximal LCx. Internal defibrilla- LVia(ED) * 100 (%). Left ventricular (LV) pressure–volume
tion with 50 J was used when VF occurred. Prior to and 1 h loops were measured using a conductance catheter as
after induction of the infarct, echocardiography was described previously (Timmers et al., 2009). Diastolic
performed. After stabilization of hemodynamic parameters chamber stiffness was quantified by means of linear
and heart rhythm, the thorax was closed and the animals regression of the end-diastolic pressure–volume relationship
were allowed to recover. Three weeks after induction of (Sagawa, 1981).
213

Data analysis Hashemi, S.M., Ghods, S., Kolodgie, F.D., Parcham-Azad, K., Keane,
M., Hamamdzic, D., Young, R., Rippy, M.K., Virmani, R., Litt, H.,
Wilensky, R.L., 2008. A placebo controlled, dose-ranging, safety
The data were collected and analyzed in a blinded fashion.
study of allogenic mesenchymal stem cells injected by endo-
Data are presented as mean ± SE. Mortality was compared myocardial delivery after an acute myocardial infarction. Eur.
using Fisher's exact test. Capillary sprouting was compared Heart J. 29, 251–259.
using a one-way ANOVA and Bonferroni post hoc tests. A two- Henry, T.D., Annex, B.H., McKendall, G.R., Azrin, M.A., Lopez, J.J.,
way ANOVA with Bonferroni post hoc tests was used to Giordano, F.J., Shah, P.K., Willerson, J.T., Benza, R.L., Berman,
compare capillary density and collagen density in the D.S., Gibson, C.M., Bajamonde, A., Rundle, A.C., Fine, J.,
myocardial tissue. Cardiac functional parameters that were McCluskey, E.R., 2003. The VIVA trial: vascular endothelial
also measured before MI (baseline) were compared using a growth factor in ischemia for vascular angiogenesis. Circulation
two-way ANOVA for repeated measures and Bonferroni post 107, 1359–1365.
hoc tests. For cardiac functional parameters that were Janssens, S., Dubois, C., Bogaert, J., Theunissen, K., Deroose, C.,
Desmet, W., Kalantzi, M., Herbots, L., Sinnaeve, P., Dens, J.,
measured only 3 weeks after MI, Student's t test was used.
Maertens, J., Rademakers, F., Dymarkowski, S., Gheysens, O., Van
Cleemput, J., Bormans, G., Nuyts, J., Belmans, A., Mortelmans, L.,
Boogaerts, M., Van de Werf, F., 2006. Autologous bone marrow-
Acknowledgments derived stem-cell transfer in patients with ST-segment elevation
myocardial infarction: double-blind, randomised controlled trial.
This work was supported by the Netherlands Heart Founda- Lancet 367, 113–121.
tion [Grant 2005T022, L.T.] and an Internationalization grant Kinnaird, T., Stabile, E., Burnett, M.S., Shou, M., Lee, C.W., Barr,
by the University Medical Center Utrecht, the Netherlands S., Fuchs, S., Epstein, S.E., 2004. Local delivery of marrow-
[D.P.V.K.]. derived stromal cells augments collateral perfusion through
paracrine mechanisms. Circulation 109, 1543–1549.
Korff, T., Augustin, H.G., 1998. Integration of endothelial cells in
Appendix A. Supplementary data multicellular spheroids prevents apoptosis and induces differen-
tiation. J. Cell Biol. 143, 1341–1352.
Lai, R.C., Arslan, F., Lee, M.M., Sze, N.S., Choo, A., Chen, T.S.,
Supplementary data to this article can be found online at Salto-Tellez, M., Timmers, L., Lee, C.N., El Oakley, R.M.,
doi:10.1016/j.scr.2011.01.001. Pasterkamp, G., de Kleijn, D.P., Lim, S.K., 2010. Exosome
secreted by MSC reduces myocardial ischemia/reperfusion
injury. Stem Cell Res. 4, 214–222.
References Lian, Q., Lye, E., Suan Yeo, K., Khia Way Tan, E., Salto-Tellez, M., Liu,
T.M., Palanisamy, N., El Oakley, R.M., Lee, E.H., Lim, B., Lim, S.K.,
Assmus, B., Honold, J., Schachinger, V., Britten, M.B., Fischer- 2007. Derivation of clinically compliant MSCs from CD105+, CD24-
Rasokat, U., Lehmann, R., Teupe, C., Pistorius, K., Martin, H., differentiated human ESCs. Stem Cells 25, 425–436.
Abolmaali, N.D., Tonn, T., Dimmeler, S., Zeiher, A.M., 2006. Losordo, D.W., Dimmeler, S., 2004. Therapeutic angiogenesis and
Transcoronary transplantation of progenitor cells after myocardial vasculogenesis for ischemic disease, part II, cell-based therapies.
infarction. N. Engl. J. Med. 355, 1222–1232. Circulation 109, 2692–2697.
Burlew, B.S., Weber, K.T., 2002. Cardiac fibrosis as a cause of Lunde, K., Solheim, S., Aakhus, S., Arnesen, H., Abdelnoor, M.,
diastolic dysfunction. Herz 27, 92–98. Egeland, T., Endresen, K., Ilebekk, A., Mangschau, A., Fjeld, J.G.,
Caplan, A.I., Dennis, J.E., 2006. Mesenchymal stem cells as trophic Smith, H.J., Taraldsrud, E., Grogaard, H.K., Bjornerheim, R.,
mediators. J. Cell. Biochem. 98, 1076–1084. Brekke, M., Muller, C., Hopp, E., Ragnarsson, A., Brinchmann, J.E.,
Cleutjens, J.P., Blankesteijn, W.M., Daemen, M.J., Smits, J.F., Forfang, K., 2006. Intracoronary injection of mononuclear bone
1999. The infarcted myocardium: simply dead tissue, or a lively marrow cells in acute myocardial infarction. N. Engl. J. Med. 355,
target for therapeutic interventions. Cardiovasc. Res. 44, 1199–1209.
232–241. Menasche, P., Alfieri, O., Janssens, S., McKenna, W., Reich-
de Silva, R., Raval, A.N., Hadi, M., Gildea, K.M., Bonifacino, A.C., enspurner, H., Trinquart, L., Vilquin, J.T., Marolleau, J.P.,
Yu, Z.X., Yau, Y.Y., Leitman, S.F., Bacharach, S.L., Donahue, Seymour, B., Larghero, J., Lake, S., Chatellier, G., Solomon, S.,
R.E., Read, E.J., Lederman, R.J., 2008. Intracoronary infusion of Desnos, M., Hagege, A.A., 2008. The Myoblast Autologous
autologous mononuclear cells from bone marrow or granulocyte Grafting in Ischemic Cardiomyopathy (MAGIC) trial: first ran-
colony-stimulating factor-mobilized apheresis product may not domized placebo-controlled study of myoblast transplantation.
improve remodelling, contractile function, perfusion, or infarct Circulation 117, 1189–1200.
size in a swine model of large myocardial infarction. Eur. Heart J. Meyer, G.P., Wollert, K.C., Lotz, J., Pirr, J., Rager, U., Lippolt, P.,
29, 1772–1782. Hahn, A., Fichtner, S., Schaefer, A., Arseniev, L., Ganser, A.,
Erbs, S., Linke, A., Schachinger, V., Assmus, B., Thiele, H., Diederich, Drexler, H., 2009. Intracoronary bone marrow cell transfer after
K.W., Hoffmann, C., Dimmeler, S., Tonn, T., Hambrecht, R., myocardial infarction: 5-year follow-up from the randomized-
Zeiher, A.M., Schuler, G., 2007. Restoration of microvascular controlled BOOST trial. Eur. Heart J. 24, 2978–2984.
function in the infarct-related artery by intracoronary transplan- Moelker, A.D., Baks, T., van den Bos, E.J., van Geuns, R.J., de
tation of bone marrow progenitor cells in patients with acute Feyter, P.J., Duncker, D.J., van der Giessen, W.J., 2006.
myocardial infarction: the Doppler Substudy of the Reinfusion of Reduction in infarct size, but no functional improvement after
Enriched Progenitor Cells and Infarct Remodeling in Acute bone marrow cell administration in a porcine model of
Myocardial Infarction (REPAIR-AMI) trial. Circulation 116, 366–374. reperfused myocardial infarction. Eur. Heart J. 27, 3057–3064.
Gnecchi, M., He, H., Liang, O.D., Melo, L.G., Morello, F., Mu, H., Pittenger, M.F., Martin, B.J., 2004. Mesenchymal stem cells and
Noiseux, N., Zhang, L., Pratt, R.E., Ingwall, J.S., Dzau, V.J., their potential as cardiac therapeutics. Circ. Res. 95, 9–20.
2005. Paracrine action accounts for marked protection of Sagawa, K., 1981. The end-systolic pressure-volume relation of the
ischemic heart by Akt-modified mesenchymal stem cells. Nat. ventricle: definition, modifications and clinical use. Circulation
Med. 11, 367–368. 63, 1223–1227.
214 L. Timmers et al.

Schachinger, V., Erbs, S., Elsasser, A., Haberbosch, W., Ham- Timmers, L., Sluijter, J.P., Verlaan, C.W., Steendijk, P., Cramer, M.J.,
brecht, R., Holschermann, H., Yu, J., Corti, R., Mathey, D.G., Emons, M., Strijder, C., Grundeman, P.F., Sze, S.K., Hua, L., Piek,
Hamm, C.W., Suselbeck, T., Assmus, B., Tonn, T., Dimmeler, J.J., Borst, C., Pasterkamp, G., de Kleijn, D.P., 2007b. Cycloox-
S., Zeiher, A.M., 2006. Intracoronary bone marrow-derived ygenase-2 inhibition increases mortality, enhances left ventricular
progenitor cells in acute myocardial infarction. N. Engl. J. Med. remodeling, and impairs systolic function after myocardial
355, 1210–1221. infarction in the pig. Circulation 115, 326–332.
Simons, M., Annex, B.H., Laham, R.J., Kleiman, N., Henry, T., Timmers, L., Henriques, J.P., de Kleijn, D.P., Devries, J.H.,
Dauerman, H., Udelson, J.E., Gervino, E.V., Pike, M., White- Kemperman, H., Steendijk, P., Verlaan, C.W., Kerver, M.,
house, M.J., Moon, T., Chronos, N.A., 2002. Pharmacological Piek, J.J., Doevendans, P.A., Pasterkamp, G., Hoefer, I.E.,
treatment of coronary artery disease with recombinant fibroblast 2009. Exenatide reduces infarct size and improves cardiac
growth factor-2: double-blind, randomized, controlled clinical function in a porcine model of ischemia and reperfusion injury.
trial. Circulation 105, 788–793. J. Am. Coll. Cardiol. 53, 501–510.
Strauer, B.E., Brehm, M., Zeus, T., Kostering, M., Hernandez, A., van Laake, L.W., Passier, R., den Ouden, K., Schreurs, C., Monshouwer-
Sorg, R.V., Kogler, G., Wernet, P., 2002. Repair of infarcted Kloots, J., Ward-van Oostwaard, D., van Echteld, C.J., Doevendans,
myocardium by autologous intracoronary mononuclear bone P.A., Mummery, C.L., 2009. Improvement of mouse cardiac
marrow cell transplantation in humans. Circulation 106, function by hESC-derived cardiomyocytes correlates with vascular-
1913–1918. ity but not graft size. Stem Cell Res. 3, 106–112.
Sze, S.K., de Kleijn, D.P., Lai, R.C., Khia Way, E., 2007. Tan, H. Wolf, D., Reinhard, A., Seckinger, A., Gross, L., Katus, H.A., Hansen,
Zhao, K.S. Yeo, T.Y. Low, Q. Lian, C.N. Lee, W. Mitchell, R.M. El A., 2009. Regenerative capacity of intravenous autologous,
Oakley, S.K. Lim, Elucidating the secretion proteome of human allogeneic and human mesenchymal stem cells in the infarcted
embryonic stem cell-derived mesenchymal stem cells. Mol. Cell. pig myocardium-complicated by myocardial tumor formation.
Proteomics 6, 1680–1689. Scand. Cardiovasc. J. 43, 39–45.
Timmers, L., Lim, S.K., Arslan, F., Armstrong, J.S., Hoefer, I.E., Yousef, M., Schannwell, C.M., Kostering, M., Zeus, T., Brehm, M.,
Doevendans, P.A., Piek, J.J., El Oakley, R.M., Choo, A., Lee, C.N., Strauer, B.E., 2009. The BALANCE Study: clinical benefit and
Pasterkamp, G., de Kleijn, D.P., 2007a. Reduction of myocardial long-term outcome after intracoronary autologous bone marrow
infarct size by human mesenchymal stem cell conditioned medium. cell transplantation in patients with acute myocardial infarction.
Stem Cell Res. 1, 129–137. J. Am. Coll. Cardiol. 53, 2262–2269.

You might also like