• No results found

Regenerative medicine in cardiovascular disease: from tissue enginering to tissue regeneration Grauss, R.W.

N/A
N/A
Protected

Academic year: 2021

Share "Regenerative medicine in cardiovascular disease: from tissue enginering to tissue regeneration Grauss, R.W."

Copied!
33
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Citation

Grauss, R. W. (2008, January 17). Regenerative medicine in cardiovascular disease: from tissue enginering to tissue regeneration. Retrieved from https://hdl.handle.net/1887/12556

Version: Corrected Publisher’s Version

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden

Downloaded from: https://hdl.handle.net/1887/12556

Note: To cite this publication please use the final published version (if

applicable).

(2)

145

CHAPTER 7

PRESERVATION OF LEFT VENTRICUL AR FUNC TION AND

AT TENUATION OF REMODELING AFTER TR ANSPL ANTATION OF HUMAN EPICARDIUM - DERIVED CELLS INTO THE INFARC TED MOUSE HEART

E.M. Winter R.W. Grauss B. Hogers J. van Tuyn R. van der Geest H. Lie-Venema R. Vicente Steijn S. Maas M.C. DeRuiter A.A.F. deVries P. Steendijk P.A. Doevendans A. van der Laarse R.E. Poelmann M.J. Schalij D.E. Atsma

A.C. Gittenberger-de Groot

Circulation 2007;116:917-27

(3)

146

ABSTR AC T

Background Proper development of compact myocardium, coronary vessels and Purkinje fi bers depends on the presence of epicardium-derived cells (EPDCs) in embryonic myocardium.

We hypothesized that adult human EPDCs might partly reactivate their embryonic program when transplanted into ischemic myocardium, and improve cardiac performance after myo- cardial infarction.

Methods and Results EPDCs were isolated from human adult atrial tissue. Myocardial inf- arction was created in immunodefi cient mice, followed by intramyocardial injection of 4x105 eGFP-labelled EPDCs (survival 2 weeks: n = 22, 6 weeks: n = 15) or culture medium (n = 24 and n = 18 respectively). Left ventricular function was assessed using a 9.4T animal-MRI.

Ejection fraction was similar between groups on day 2, but was signifi cantly higher in the EPDC- injected group at 2 weeks (short term), as well as after long term survival at 6 weeks.

End-systolic and end-diastolic volume were signifi cantly smaller in the EPDC-injected group than in the medium-injected group at all ages evaluated. At 2 weeks, vascularization was sig- nifi cantly increased in the EPDC-treated group, together with an increased wall thickness which might be explained by augmented DNA-damage repair activity in the infarcted area. Immuno- histochemical analysis showed massive engraftment of injected EPDCs at 2 weeks, expressing α-smooth muscle actin, von Willebrand Factor, sarcoplasmic reticulum Ca2+-ATPase and SCN5a.

EPDCs were negative for cardiomyocyte markers. At 6 weeks survival wall thickness was still increased but only a few EPDCs could be detected.

Conclusions After transplantation into ischemic myocardium, adult human EPDCs preserve cardiac function and attenuate ventricular remodeling. Autologous human EPDCs are promis- ing candidates for clinical application in infarcted hearts.

(4)

147

INTRODUC TION

Current therapy aimed at alleviating the sequelae of sustained myocardial infarction (MI) is not able to restore the function of the scarred area. Stem cell therapy poses a promising alternative therapy. As therapeutic use of embryonic stem cells is ethically intricate and technically diffi cult in relation to possible rejection of the cells and tumor formation, use of adult stem cells seems to be a more feasible option. Many diff erent types of adult cells have been demonstrated to improve cardiac function after a MI, although the underlying mechanism has only partly been unraveled (for review see [1]). Most of the cell types used are not known to be of importance during normal cardiogenesis. We chose to transplant epicardium-derived cells (EPDCs), as these cells are known to be crucial for cardiac development, both because of their physical contribu- tion [2] as well as their modulatory role [3,4] During embryogenesis, epicardium migrates from the extracardiac proepicardium to cover the premature heart consisting by that time of only myocardium and endocardium with cardiac jelly in between [5]. A subset of the epicardial cells undergoes epithelial-mesenchymal transformation (EMT). These cells are called EPDCs [2]. EPDCs migrate into the myocardium to diff erentiate into interstitial cardiac fi broblasts, subendocardial and atrioventricular cushion mesenchymal cells, and into coronary smooth muscle cells and adventitial fi broblasts [2,6-8]. Besides this physical contribution of EPDCs to the coronary vessels and the fi brous heart skeleton, EPDCs have a modulatory role in cardiogenesis [9,10]. Formation of the compact myocardium [3,4], coronary vessel formation [11,12,13], and Purkinje fi ber cell diff erentiation [2,14] are dependent on EPDC regulation. If epicardial outgrowth is inhibited completely, the myocardium remains thin and normal septation does not take place, leading to early embryonic death [3]. Besides hampered formation of the compact myocardium, vascular development is severely disturbed in partial epicardial abrogation [11,15].

The role of EPDCs during postnatal cardiac growth has never been elucidated. It is unknown whether new EPDCs are generated continuously through EMT, contributing to the growing structures of the heart, and regulating developmental processes. It has been shown, however, that adult rat epicardial cells are at least still able to undergo EMT and diff erentiate into smooth muscle cells in vitro [16]. We recently demonstrated that adult human cells derived from epi- cardial explants (hEPDCs) can spontaneously undergo EMT, observed by a transformation from cobble stone into spindle shape morphology, while losing their b-catenin expression [17]. In the present study we also used Wilms’ Tumor 1 suppressor protein (WT1) expression to investigate this aspect. During cardiogenesis, expression of WT1 is observed in the proepicardium and the epicardial cells, but is lost in the EPDCs soon after they have undergone EMT [18,19]. Further- more, it has been shown that adult epicardial cells positively modify cardiomyocyte phenotype and function [20], and that WT1 is switched on de novo in adult coronary vessels in ischemic myocardium [21].

(5)

148 Being relatively undiff erentiated cells that can give rise to diff erentiated progeny of at least smooth muscle cells and fi broblasts, embryonic EPDCs have been referred to as stem cells [22].

Based on the fact that adult hEPDCs can still undergo EMT and can give rise to the above men- tioned cell types [17], we consider adult hEPDCs as progenitor cells. We hypothesized that adult hEPDCs could recapitulate part of their embryonic program when transplanted into diseased myocardium, thereby positively modifying the surrounding myocardium.

In this study, we investigated in a mouse model whether adult hEPDCs could improve cardiac performance after MI. Adult spindle shaped hEPDCs were transplanted into ischemic murine myocardium. Evaluation of this population revealed that these cells showed no expression pat- tern indicative of endothelial cells or cardiomyocytes [17].

(6)

149

MATERIALS AND METHODS

See the online data supplement (page 169) for an expanded Materials and Methods section.

Primary Cultured Human Adult EPD Cs

EPDCs were cultured from human adult epicardium which was separated mechanically from atrial appendages. Spindle shaped cells (see online supplement Figure 1) from passage two to four were used for transplantation experiments after transduction with a human vector express- ing the enhanced green fl uorescent protein (eGFP) gene, enabling cell tracing. An adenoviral and a lentiviral vector were used for short term (2 weeks survival) and long term (6 weeks sur- vival) experiments, respectively.

Creation of the M I and Cell Transplantation

To avoid rejection of transplanted human cells, nonobese diabetic-severe combined immuno- defi cient (NOD/scid) mice were used [23]. All animal procedures were approved by the Animal Ethics Committee of the Leiden University and conformed the Guide for the Care and Use of Laboratory Animals (National Institutes of Health publication No. 85-23, Revised 1996). For short term experiments, the main left anterior descending coronary artery (LAD) was permanently ligated. For long term experiments only the branch supplying the ventral wall of the left ventri- cle was ligated, since extended survival until 6 weeks is not possible with the entire left ventricle infarcted.

Immediately after ligation, transplantation of 4 x 105 hEPDCs suspended in M199 (hEPDC-group, survival 2 weeks: n = 22, 6 weeks: n = 15), or cell-free M199 (medium-group, survival 2 weeks:

n = 24, 6 weeks: n = 18) was performed into the ischemic myocardium. Sham-operated animals (survival 2 weeks: n = 16, 6 weeks: n = 3) were operated similarly, but without ligation of the LAD and without fl uid injection. Animals were randomized to treatment.

Short Term Experiments

M R I

Infarct size (2 days after surgery) and cardiac function (2 and 14 days after surgery) were assessed using contrast-enhanced and cine MRI images (9.4 Tesla). Images were analyzed by manual delineation of endocardial and epicardial borders (hEPDC-group n = 17, medium-group n = 14, sham-group n = 13) using dedicated software (see online supplement for detailed information).

Left ventricular (LV) infarcted area, LV end-diastolic volume (EDV), LV end-systolic volume (ESV),

(7)

150 LV stroke volume (SV), and LV ejection fraction (EF) were computed automatically [24]. Infarcted area measurements were used to correct functional parameters for potential diff erences in ini- tial infarct size.

I M M U N O H I S T O C H E M I C A L A S S E S S M E N T

Animals were sacrifi ced 15 days after surgery. Paraffi n sections of the hearts (n = 5 per group) were used for immunohistochemical analysis. To investigate host tissue properties, serial sec- tions were stained for CD31, α/γ muscle actin, α-smooth muscle actin, proliferating cell nuclear antigen (PCNA), phospho-histone H3, and phospho-histone H2AX. Non-fl uorescent anti- eGFP staining was performed to visualize the injected hEPDCs because the strong and irregular autofl uorescence of the heart disturbs assessment of spontaneous eGFP fl uorescence to detect engrafted cells.

To identify which proteins were expressed by the injected hEPDCs, double stainings were per- formed for eGFP and other proteins, being α-smooth muscle actin, von Willebrand Factor (vWF), sarcoplasmic or endoplasmic reticulum Ca2+-ATPase (SERCA2a), voltage-gated sodium channel (α-subunit) (SCN5a) cardiac troponin I, (cTnI), atrial natriuretic peptide (ANP), α/γ muscle actin (clone HHF35), and sarcomeric myosin (clone MF20). A Red Qdot conjugated goat anti-rabbit antibody was used to visualize eGFP staining. An appropriate biotinylated secondary antibody in combination with Yellow Qdot conjugated to streptavidin was used to visualize the other proteins. For details see online supplement.

L V V A S C U L A R P R O F I L E A N D W A L L T H I C K N E S S

To evaluate the angiogenic eff ect of hEPDC transplantation, the cumulative area of CD31-stained vessel lining per total LV area was determined in the hEPDC- and medium-group (n = 5 for each group).

LV wall thickness was measured in the hearts of the hEPDC- and medium-group (n = 5 for each group) by an observer blinded to treatment. Wall thickness was measured in both border zones of the infarcted wall, in the mid-infarcted area in between the border zones, and in the middle of the interventricular septum.

G E N E R A L H E A L T H A N D S U R V I V A L

Body weight was determined just before surgery at day 0 and before sacrifi ce at day 15. Pulmo- nary water content was estimated after sacrifi ce by subtracting dry weight from wet weight of the lungs. To correct for diff erences in body mass, the amount of lung fl uid was expressed relative to body weight. Survival proportions were assessed.

(8)

151 Long Term Experiments

M R I

The eff ect of hEPDC transplantation on the long term was evaluated by cine MRI images 42 days after surgery only (hEPDC-group n = 15, medium-group n = 14, sham-group n = 3), because of the high risk of mortality during repeated imaging procedures.

I M M U N O H I S T O C H E M I C A L A S S E S S M E N T

Animals were sacrifi ced 43 days after surgery. Excised hearts were treated as described for short term experiments, besides 3 hearts of the medium- and hEPDC- group that were frozen. To iden- tify injected cells, non-fl uorescent stainings for eGFP, human specifi c CD31 and human specifi c vWF were performed in paraffi n sections, and for human nucleus in frozen sections.

L V V A S C U L A R P R O F I L E A N D W A L L T H I C K N E S S

Anti-CD31 and Sirius Red staining, and wall thickness measurements were used for host tissue investigation.

S U R V I V A L

Survival proportions were assessed.

Statistical Analysis

Data are presented as mean ± standard error (SEM). Data were analyzed with SPSS software (SPSS Inc, Chicago, IL, USA). For comparisons of more than two groups, a one-way-ANOVA was performed (or the non-parametric Kruskal-Wallis for the dependent variable lung weight). If the omnibus tests among groups were signifi cantly diff erent, post-hoc tests between groups using t-tests (and Mann-Whitney for lung weight) were performed. Infarct size was used as covariate in an analysis of (co-)variance of the functional data to correct for baseline diff erences in infarct size among groups. Diff erences in mortality were evaluated using the Breslow test. A level of p < 0.05 was considered to represent a signifi cant diff erence.

The authors had full access to the data and take responsibility for its integrity. All authors have read and agree to the manuscript as written.

(9)

152

RESULTS

Cultured Adult hEPD Cs

Within a few days hEPDCs migrated from the epicardium dissected from the atrial append- ages. In the fi rst passage, mainly cobble stone and a few spindle shaped cells were observed, while after two passages all cells had adopted a spindle shaped morphology (with loss of WT1 expression, see online supplement Figure 1). Cells could easily be kept in culture, with a constant doubling time between day 10 and day 50, after which they went into senescence (Figure 1). For a more extensive characterization of in vitro cultured hEPDCs see Van Tuyn et al. [17].

Figure 1. Growth curves of adult hEPDCs in culture. Growth velocity is similar for diff erent cultures.

Short Term Experiments

L V F U N C T I O N

LV EF and SV decreased signifi cantly after onset of MI. LV EF and SV were similar in the medium- and hEPDC-treated group 2 days after surgery. However, at 14 days after surgery a signifi cantly higher LV EF and SV were observed in the hEPDC-treated group than in the medium-treated group (Figure 2a, b). LV EDV and LV ESV were signifi cantly smaller in the hEPDC-treated group than in the medium-treated group, both at 2 and 14 days after surgery (Figure 2c, d). Both medium- and hEPDC-treated mice showed heart-function parameters that diff ered signifi cantly from the sham-operated animals. However, with respect to EDV on day 2 and SV on day 14, the hEPDC-treated group resembled the sham-operated group (Figure 2a-d).

(10)

153

Figure 2. Ejection fraction (a), stroke volume (b), end-diastolic volume (c) and end-systolic volume (d) in hEPDC- treated, medium-treated, and sham-operated mice, 2 and 14 days after surgery. Please note that no intermediate time points were measured. Data are mean ± SEM. *: p < 0.05 versus medium-group, †: p < 0.05 versus sham- group.

L V W A L L S T R U C T U R E

Histological evaluation showed that at day 15, transplanted hEPDCs had engrafted mainly in the ischemic LV wall, and formed layers of cells (Figure 3a, b). Transplanted hEPDCs were not found in the lining of the coronary vessel wall (Figure 4a,b).

The infarcted host tissue only showed remaining cardiomyocytes in the subendocardial and, to a lesser extent, in the subepicardial position, which was not diff erent between the hEPDC- and medium-group (Figure 3c, d). The cardiomyocytes and the intermediate fi brous wall tissue were negative for α-smooth muscle actin (Figure 3e, f ). Nuclear PCNA expression was increased in the infarcted left ventricular wall (Figure 3g) and the border zone (Figure 3h) of the hEPDC-group as compared to the medium-group (Figure 3i, j). DNA damage was present in these areas of both the medium- and the hEPDC-group, as was demonstrated by phospho-histone H2AX staining (not shown). Phospho-histone H3 staining revealed that the low number of mitotic fi gures was not diff erent between groups (not shown).

Hearts from the hEPDC-group showed a complex vascular network consisting of capillaries, veins and arterioles with a high vascular density throughout the ischemic area (Figure 4c),

(11)

154 but lacking the distribution of fi ne capillaries as observed in healthy myocardium (Figure 4e).

Medium-injected infarcted hearts were poorly vascularized with small capillaries, unequally dis- persed throughout the ventricular wall (Figure 4d). When determined quantitatively, free wall endothelial density, expressed relative to septal endothelial density, was signifi cantly higher in all parts of the left ventricle of the hEPDC-group as compared to the medium-group (Figure 4f, g).

Wall thickness of the infarcted LV wall, the border zone and the interventricular septum was signifi cantly larger in the hEPDC-treated group than in the medium-treated group (Figure 4h).

Figure 3. Photomicrograph of representative sec- tions 2 weeks after MI demonstrating engrafted eGFP-positive hEPDCs (brown) in a hEPDC-treated heart (a), which are absent in the medium-treated heart (b). Remaining cardiomyocytes (expressing α/γ muscle actin [HHF35]) in the hEPDC- (c) and the medium-group (d) are positioned in the subendo- cardial and subepicardial region of the ischemic left ventricular wall. Consecutive sections show that the cardiomyocytes are negative for α-smooth muscle actin (ASMA) (e, f ). The number of PCNA-positive cells (brown) is increased in the infarcted left ventricular wall (g) and border zone (h) of the hEPDC-group as compared to the infarcted area (i) and border zone (j) of the medium-group. LV: left ventricle, RV: right ventricle, VS: ventricular septum, END: endocardium, EP: epicardium. Scale bars a, b: 300 μm, scale bars c-f: 90 μm.

(12)

155

Figure 4. Vascular profi le and wall thick- ness at 2 weeks survival. Two consecutive sections demonstrate that eGFP-positive hEPDCs (arrows) (a) were not incorporated into the CD31 positive murine (host) vessels (arrowheads) (b). Endothelial CD31 staining in the LV wall shows a high vascular density and a complex vascular network consist- ing of small capillaries (arrowheads) and irregularly shaped large vessels (arrows) in the hEPDC-group (c), which are lacking in the medium-group (d). These profi les are very diff erent from the capillary rich myocardium in the sham-operated group (e). Quantifi cation of endothelial density (expressed relative to septal values [dot- ted areas in f ]) demonstrated a signifi cant higher degree of vascularization in all parts of the left ventricle, represented by num- bers 1-6 (f ), in the hEPDC-group compared to the medium-group (g). Wall thickness of

the border zone, infarcted area and interventricular septum was signifi cantly larger in the hEPDC-group than in the medium-group (h). This diff erence is also visible in the pictures showing the LV wall of the hEPDC-group (c) and medium-group (d). Data are mean ± SEM. *: p < 0.05 versus medium-group. LV: left ventricle, RV: right ventricle, END: endocardium, EP: epicardium. Scale bars a-e: 200 μm, scale bar d: 400 μm.

By double staining techniques we were able to investigate a number of diff erentiation markers in the engrafted hEPDCs. Almost all hEPDCs expressed α-smooth muscle actin (Figure 5a-c) and vWF (Figure 5d-f ). A number of the hEPDCs was positive for SERCA2a and SCN5a (Figure 5g-l). The transplanted cells did not express the (cardio)myocyte markers ANP, cTnI, sarcomeric myosin, and α/γ muscle actin (not shown).

(13)

156

Figure 5. Confocal microscopic images showing diff erentiation markers of engrafted hEPDCs in ischemic myo- cardium 2 weeks after MI. Most eGFP-positive hEPDCs (red: a, d) show colocalization with α smooth muscle actin (ASMA) (yellow: b, merge: c) and vWF (yellow: e, merge: f ). A number of eGFP-positive hEPDCs (red: g, j) express SERCA2a (yellow: h, merge: i), and SCN5a (yellow: k; merge: l). Scale bars: 20 μm.

(14)

157

G E N E R A L H E A L T H A N D S U R V I V A L

Only a minor decrease in body weight between day of surgery and day of sacrifi ce (day 15) was observed in the hEPDC-treated group (-0.5 ± 0.6 g), which was not statistically diff erent from that of the sham-operated group (+0.1 ± 0.3 g). In contrast, decrease in body weight was signifi cantly larger in the medium-treated group (-2.7 ± 1.0 g) than in the sham-operated group and the hEPDC-treated group (Figure 6a). Decrease in body mass relative to the original weight amounted 10 ± 4 % in the medium-group, 2 ± 2 % in the hEPDC-group and 0 ± 1 % in the sham- group.

Figure 6. Decrease in body weight (a) and amount of lung edema (b) in three groups of mice, being medium-, hEPDC-, and sham-group at 2 weeks after surgery. Data are mean ± SEM. ‡: p < 0.05 versus hEPDC-group, †: p < 0.05 versus sham-group.

Similarly, no signifi cant diff erence at day 15 in amount of lung fl uid, corrected for body weight, was observed between the sham-operated and hEPDC-treated group, while lung edema was present in the medium-treated group (Figure 6b). Cumulative survival of medium-treated mice over the 2 week time period was signifi cantly lower than that of the sham-operated mice. Survival of hEPDC-treated mice was not diff erent from survival of sham-operated animals (Figure 7).

Figure 7. Survival proportions of sham-oper- ated, hEPDC-treated and medium-treated mice until day 15, by which time all animals were sacrifi ced. Signifi cantly more animals died in the medium-group than in the sham-group. †:

p < 0.05 versus sham-group.

(15)

158

Long Term Effect of hEPD C Transplantation

The eff ect of hEPDC transplantation on cardiac function 6 weeks after MI was investigated in separate experiments. LV EF and SV were signifi cantly higher 6 weeks after creation of MI in the hEPDC-group compared to the medium-group (Figure 8a, b). Similarly, LV EDV and ESV were signifi cantly smaller in the hEPDC-group than in the medium-group (Figure 8c, d). Sham-oper- ated animals showed values for these functional parameters that were all signifi cantly diff erent from those of the medium- and hEPDC-operated animals, except for SV (Figure 8a-d). No death was observed in the hEPDC- and sham-group, in contrast to four deaths in the medium-group (Figure 8e). Anti-eGFP staining only demonstrated a few engrafted hEPDCs which were not embedded in the vessel lining (Figure 8f, g). No cells positive for human specifi c CD31 or vWF were observed in sections consecutive to the eGFP stained sections. In the frozen sections of the hEPDC-group no cells were detected that stained positive for human nucleus (not shown).

Wall thickness of infarcted area and border zone was increased in the hEPDC-group compared to the medium-group (Figure 8h). Properties of the scar itself were not signifi cantly diff erent between hEPDC- and medium-treated animals. The remaining cardiomyocytes as observed in the scar area at week 2 had disappeared. The infarcted area consisted in both groups of mainly fi brous tissue (Figure 8i), with many vessels situated in the border zone (Figure 8j) and few ves- sels in the scar area (Figure 8k).

(16)

159

DISCUSSION

The main fi ndings of our study are that adult human EPDCs i) can be isolated and cultured, during which they undergo EMT, ii) engraft and survive in ischemic murine myocardium for at least 2 weeks while only few cells could be detected at 6 weeks, and iii) preserve LV function and attenuate post-ischemic remodeling until 6 weeks after MI. Embryonic EPDCs are known to be of crucial importance during cardiogenesis. They are essential for proper myocardial architecture and coronary vessel formation, both through their physical contribution and through regulation of these developmental processes [2,3,10]. Little is known about the role of adult EPDCs in the normal and diseased adult heart. We describe for the fi rst time the use of adult hEPDCs, cells grown from human adult epicardial explants after EMT [17], in cardiac regeneration therapy, applying them to possibly trigger embryological developmental processes which might restore or preserve cardiac function.

I M M U N O L O G I C A L C H A R A C T E R I Z A T I O N O F H E P D C S I N V I V O

After 2 weeks, many hEPDCs were observed in the LV wall. They expressed the smooth muscle cell marker α-smooth muscle actin. This is in line with the fact that embryonic EPDCs express this protein when they diff erentiate into coronary smooth muscle cells, besides interstitial and adventitial coronary fi broblasts [2]. The α-smooth muscle actin-positive hEPDCs were not observed in the vessel wall, but as isolated cells located in the scar tissue, having a shape similar to that of the surrounding fi broblasts. Part of the engrafted hEPDCs also expressed the marker SERCA2a, which is expressed in smooth muscle cells, skeletal muscle cells and cardiomyocytes [25]. Staining for α/γ muscle actin (clone HHF35), normally expressed by almost all muscle cells [26], was negative in transplanted hEPDCs, suggesting that engrafted cells did not fully dif- ferentiate into a smooth muscle cell phenotype.

The injected hEPDCs did not acquire a cardiomyocyte phenotype, as the engrafted hEPDCs did not express any of the cardiomyocyte markers ANP, sarcomeric myosin or cTnI. This is consistent with the fi nding that EPDCs do not diff erentiate into cardiomyocytes during embryonic heart development [2,6]. Remarkably, immunostaining for SCN5a was positive in some engrafted hEPDCs. SCN5a is mainly expressed in cardiomyocytes, and has recently also been described in human gastrointestinal smooth muscle [27]. Therefore, we consider the expression pattern of the transplanted hEPDCs as that of a smooth muscle cell with extraordinary features, such as SCN5a expression.

Although the engrafted cells did not integrate into the vessel wall, a large part of them was positive for the endothelial cell marker vWF at 2 weeks after MI. It is interesting that the engrafted adult hEPDCs expressed vWF, as the possible contribution of embryonic EPDCs to coronary endothelium is still a subject of debate [13,28,29]. Relevant to these fi ndings are reports

(17)

160

(18)

Figure 8. Results of hEPDC transplantation 6 weeks after MI. Ejection fraction (a) and stroke volume (b) were signifi cantly higher in the hEPDC-injected group 6 weeks after MI. hEPDC transplantation resulted in signifi cantly smaller end-diastolic (c) and end-systolic volumes (d) as compared to medium-injection. A better survival was observed in the hEPDC-group compared to the medium-group (p = 0.057) (e). Only a few eGFP-positive hEPDCs (brown) were detected in the hearts 6 weeks after hEPDC transplantation, mainly in the border zone (f, g). The hEPDCs (arrows) did not integrate in the vessel lining (g). Wall thickness of the infarcted area and the border zone was signifi cantly higher in the hEPDC-group than in the medium-group 6 weeks after MI (h). Pictures i, j and k are from hEPDC-treated hearts (i-k), but are also representative for medium-treated hearts. Sirius Red staining demonstrates that the infarcted area consisted of mainly fi brous tissue (i). Hardly any vessels could be observed in the infarcted area (j), while many vessels were found in the border zone (k). Data are mean ± SEM. *: p < 0.05.

LV: left ventricle, RV: right ventricle, END: endocardium, EP: epicardium, a: artery, v: vene. Scale bars f, g: 30 μm, scale bar i: 600 μm, scale bars j, k: 60 μm.

161 describing that endothelial markers can also be expressed by non-endothelial cells, such as

certain skeletal muscle cells [30].

The expression profi le of cultured hEPDCs in vitro is diff erent from that of engrafted hEPDCs in vivo. Whereas engrafted hEPDCs in vivo stained positive for α-smooth muscle actin, vWF, SERCA2a and SCN5a proteins, hEPDC in vitro contain only α-smooth muscle actin mRNA, while mRNA for SCN5a and SERCA2a is not observed. Moreover, vWF staining is negative in cultured hEPDCs in vitro [17]. This implies that the expression pattern of hEPDCs changes in reaction to the surrounding, ischemic tissue, resulting in relatively undiff erentiated engrafted cells with a myo-endothelial phenotype [30]. The engraftment of the hEPDCs is temporary, since only few hEPDCs could be detected at week 6. These cells did not express vWF, indicating that the myo- endothelial phenotype is at least partly transitional.

H I S T O L O G I C A L C H A R A C T E R I S T I C S O F T H E S U R R O U N D I N G H O S T T I S S U E

High vascular densities were observed in all parts of the LV wall of hEPDC- injected hearts 2 weeks after MI. The highly organized vascular network in the host tissue of the hEPDC-group consisted of variably-sized but mainly large-diameter vessels, whereas the medium-group con- tained only few vessels that were spread irregularly throughout the ischemic area. However, the high density of capillaries as observed in healthy myocardium was lacking in the hEPDC-group.

The vessels must be of mouse origin, since no hEPDCs were observed integrated in the vessel lining, a fi nding confi rmed at 6 weeks survival. It remains to be investigated whether more ves- sels had survived, or new murine (host) vessels were formed after hEPDC injection. An indirect paracrine stimulatory eff ect of hEPDCs on vessel survival or angiogenesis is suggested, as (i) hEPDCs were not found in vessel linings, and (ii) vessels were found throughout the entire LV

(19)

162 wall of the hEPDC-group, and not only in areas with a high density of engrafted hEPDCs. At week 6 diff erences in vascular profi ling had disappeared, suggesting a transitional eff ect of hEPDCs on the vessels.

The increased PCNA expression in the infarcted area and border zone of the hEPDC-group com- pared to the medium-group might partly explain the signifi cantly increased wall thickness in these areas of the hEPDC-group, both shortly and long after MI. PCNA is a central protein in both replication and DNA-damage repair [31,32,33]. Since phospho-histone H3 staining revealed only few mitotic fi gures, which was not diff erent between groups, it is likely that the PCNA positive cells represent cells with DNA-damage repair rather than proliferating cells. This was supported by the fact that DNA-damage was indeed present in both groups. Increased cellular survival [34] due to augmented repair might then have contributed to a thicker wall. Further research, however, is needed to unravel the processes underlying the increased PCNA-activity, and to determine whether the PCNA-positive cells are indeed repairing DNA-damage, or whether they represent activated and proliferating cells as well.

On the other hand, diminished ventricular dilatation itself [35], as well as increased proliferation of cardiac stem cells [36] and other host tissue cells immediately after MI (before day 14) might have aff ected wall thickness [34,37-39]. It seems unlikely that new cardiomyocyte formation [40] contributed to the increment in wall thickness in the hEPDC-group, since the cardiomyocytes observed in the subendocardial and subepicardial region of the infarcted area of the hEPDC- and medium-group were negative for α-smooth muscle actin, which is normally expressed by primitive but not by adult cardiomyocytes [41]. Moreover, while wall thickness at week 6 was still increased in the hEPDC-treated hearts, hardly any cardiomyocytes were observed in the ischemic area.

F U N C T I O N A L I M P R O V E M E N T

LAD occlusion results in MIs that vary in size with associated variability in ventricular volumes [42]. In order to discern possible treatment eff ects, it seems mandatory to determine initial infarct size and correct functional data for any diff erences in infarct size. We performed this by assessing infarct size with contrast-enhanced MRI images and subsequent covariance analysis of functional parameters [43]. Functional data were acquired using MRI, which is considered to be the gold standard for ventricular function assessment in small animals [44], creating high resolution images especially at 9.4 Tesla. In contrast to one- or two- dimensional echocardi- ography and conductance catheter measurements, computation of ventricular volumes from MRI images is not based on specifi c geometric assumptions but on real data, which makes it a reliable method for determination of infarcted, abnormally shaped hearts.

We showed that hEPDC transplantation in the acutely infarcted myocardium improved cardiac function 2 weeks after induction of MI. This improvement was represented by a higher EF, larger

(20)

163 SV, and less lung edema in the hEPDC-group compared to medium-treated animals. Moreover,

a smaller EDV in the hEPDC-group demonstrated that ventricular remodeling was reduced by hEPDC transplantation. However, EDV was still 2-3 fold higher in the hEPDC group than in the sham-group, illustrating the fact hEPDC transplantation does result in less deterioration or pres- ervation of cardiac function, not in restoration of normal function. An early protective eff ect of the hEPDCs was indicated by the fact that a smaller LV EDV and ESV were observed already 2 days after onset of MI. It can not be determined whether this early eff ect is responsible for the observed positive infl uence of hEPDC transplantation after several weeks. Studies in which hEPDCs are injected a few days after MI might reveal this contribution.

The increased survival proportions in the hEPDC-group might be explained by the improved cardiac function and reduced ventricular remodeling, since left ventricular dysfunction and mor- tality are highly correlated [45]. The animals of the hEPDC-group did not show cardiac cachexia 2 weeks after MI, defi ned as weight loss >7.5 % of the original weight [46], and known as a severe complication of chronic heart failure associated with a poor prognosis. Cardiac cachexia, however, was observed in the medium-treated group, which had a relative weight loss of 10 %.

Mice in the hEPDC-group showed less lung edema than mice in the medium-group, illustrating that the absence of weight loss in the hEPDC-group was not obscured by edema.

To investigate whether the benefi cial eff ect of hEPDC transplantation on the infarcted heart remained until a defi nitive scar had been formed, an additional set of experiments was per- formed with analysis 6 weeks after MI. We demonstrated that EF and SV were still signifi cantly higher in the hEPDC-group than in the medium-group. Moreover, ESV and EDV were again sig- nifi cantly smaller in the hEPDC-injected group, demonstrating decreased remodeling 6 weeks after MI. The survival benefi t for the hEPDC-treated group after 6 weeks confi rmed the benefi cial infl uence of hEPDC transplantation on the long term. These data suggest that the eff ect of hEPDCs on cardiac function is stable, although we still can not exclude an early paracrine eff ect, as has been described for other stem cells [34,47].

The exact mechanism underlying the improvement in cardiac function caused by hEPDC transplantation remains to be investigated. We suggest from our data that the injected hEPDCs protect host tissue cells through augmented DNA-damage repair, resulting in augmented cel- lular survival [34] and subsequent prevention of extreme wall thinning, which will contribute to preservation of left ventricular function and reduced functional remodeling both on the short and on the long term. A paracrine eff ect of the transplanted cells on the host tissue is indicated since functional data demonstrate an eff ect of the hEPDCs already at day 2, and since histologi- cal data show diff erences between groups in host tissue properties rather than newly formed donor-derived tissue in the hEPDC-group.

(21)

164 C L I N I C A L R E L E V A N C E

We showed that adult hEPDCs grow easily in culture during several passages and acquire spindle shape morphology, similar to embryonic EPDCs that have undergone epithelial mesen- chymal transformation enabling migration into the myocardium [2]. As they preserved cardiac function and reduced remodeling after MI, both shortly and long after onset of MI, autologous EPDCs seem promising for use in cardiac regeneration therapy. Preferably, these cells should be injected intramyocardially, using catheter-based methods described previously [48]. In the current study, atrial appendages were harvested during CABG procedures. For broader clinical applications, a minimally invasive cardiac surgical technique is to be preferred, such as endo- scopic surgery. Atrial appendages are removed using this technique as therapy in atrial fi bril- lation [49]. Transplantation of autologous EPDCs will not give ethical problems, and there will be little risk of rejection. This makes them ideal candidates for cell therapy. Spontaneous tumor formation is no issue, as we demonstrated that adult hEPDCs do not divide indefi nitely, nor did we observe any tumor formation in the ischemic mouse heart until 6 weeks after transplanta- tion. In a clinical setting, EPDCs can probably only be transplanted in the chronically infarcted, reperfused heart. We are currently studying this aspect.

A C K N O W L E D G M E N T S

The authors are indebted to Pieter Voigt and Robert Klautz for kindly supplying the leftover surgical atrial appendages, to Jan Lens for excellent preparation of the fi gures, and to the animal facility.

F U N D I N G S O U R C E S

This research was funded by grant 53.345 from the Interuniversity Cardiology Institute of the Netherlands.

(22)

165

REFERENCES

[1] Murry CE, Reinecke H, Pabon LM. Regeneration gaps: observations on stem cells and cardiac repair. J Am Coll Cardiol. 2006; 47:1777-1785.

[2] Gittenberger-de Groot AC, Vrancken Peeters MP, Mentink MM, Gourdie RG, Poelmann RE. Epicardium- derived cells contribute a novel population to the myocardial wall and the atrioventricular cushions. Circ Res. 1998; 82:1043-1052.

[3] Gittenberger-de Groot AC, Vrancken Peeters MP, Bergwerff M, Mentink MM, Poelmann RE. Epicardial out- growth inhibition leads to compensatory mesothelial outfl ow tract collar and abnormal cardiac septation and coronary formation. Circ Res. 2000; 87:969-971.

[4] Kang JO, Sucov HM. Convergent proliferative response and divergent morphogenic pathways induced by epicardial and endocardial signaling in fetal heart development. Mech Dev. 2005; 122:57-65.

[5] Viragh S, Gittenberger-de Groot AC, Poelmann RE, Kalman F. Early development of quail heart epicardium and associated vascular and glandular structures. Anat Embryol (Berl). 1993; 188:381-393.

[6] Männer J. Does the subepicardial mesenchyme contribute myocardioblasts to the myocardium of the chick embryo heart? A quail-chick chimera study tracing the fate of the epicardial primordium. Anat Rec. 1999;

255:212-226.

[7] Vrancken Peeters MP, Gittenberger-de Groot AC, Mentink MM, Poelmann RE. Smooth muscle cells and fi broblasts of the coronary arteries derive from epithelial-mesenchymal transformation of the epicardium.

Anat Embryol (Berl). 1999; 199:367-378.

[8] Dettman RW, Denetclaw WJ, Ordahl CP, Bristow J. Common epicardial origin of coronary vascular smooth muscle, perivascular fi broblasts, and intermyocardial fi broblasts in the avian heart. Dev Biol. 1998; 193:169-181.

[9] Männer J, Perez-Pomares JM, Macias D, Munoz-Chapuli R. The origin, formation and developmental signifi - cance of the epicardium: a review. Cells Tissues Organs. 2001; 169:89-103.

[10] Winter EM, Gittenberger-de Groot AC. Cardiovascular development: towards biomedical applicability : Epicardium-derived cells in cardiogenesis and cardiac regeneration. Cell Mol Life Sci. 2007; 64:692-703.

[11] Eralp I, Lie-Venema H, DeRuiter MC, Van Den Akker NM, Bogers AJ, Mentink MM, Poelmann RE, Gitten- berger-de Groot AC. Coronary artery and orifi ce development is associated with proper timing of epicar- dial outgrowth and correlated Fas-ligand-associated apoptosis patterns. Circ Res. 2005; 96:526-534.

[12] Perez-Pomares JM, Phelps A, Munoz-Chapuli R, Wessels A. The contribution of the proepicardium to avian cardiovascular development. Int J Dev Biol. 2001; 45:S155-S156.

[13] Guadix JA, Carmona R, Munoz-Chapuli R, Perez-Pomares JM. In vivo and in vitro analysis of the vasculo- genic potential of avian proepicardial and epicardial cells. Dev Dyn. 2006; 235:1014-1026.

[14] Eralp I, Lie-Venema H, Bax NA, Wijff els MC, van der LA, DeRuiter MC, Bogers AJ, Van Den Akker NM, Gourdie RG, Schalij MJ, Poelmann RE, Gittenberger-de Groot AC. Epicardium-derived cells are important for correct development of the Purkinje fi bers in the avian heart. Anat Rec A Discov Mol Cell Evol Biol. 2006; 288:1272- 1280.

(23)

166 [15] Lie-Venema H, Gittenberger-de Groot AC, van Empel LJ, Boot MJ, Kerkdijk H, de Kant E, DeRuiter MC.

Ets-1 and Ets-2 transcription factors are essential for normal coronary and myocardial development in chicken embryos. Circ Res. 2003; 92:749-756.

[16] Wada AM, Smith TK, Osler ME, Reese DE, Bader DM. Epicardial/mesothelial cell line retains vasculogenic potential of embryonic epicardium. Circ Res. 2003; 92:525-531.

[17] Van Tuyn J, Atsma DE, Winter EM, van der Velde-van Dijke I, Pijnappels DA, Bax NAM, Knaan-Shanzer S, Gittenberger-de Groot AC, Poelmann RE, van der Laarse A, van der Wall EE, Schalij MJ, de Vries AAF.

Epicardial cells of human adults can undergo an epithelial-to-mesenchymal transition and obtain characteristics of smooth muscle cells in vitro. Stem Cells. 2007; 25:271-278.

[18] Moore AW, McInnes L, Kreidberg J, Hastie ND, Schedl A. YAC complementation shows a requirement for Wt1 in the development of epicardium, adrenal gland and throughout nephrogenesis. Development.

1999; 126:1845-1857.

[19] Perez-Pomares JM, Phelps A, Sedmerova M, Carmona R, Gonzalez-Iriarte M, Munoz-Chapuli R, Wessels A. Experimental studies on the spatiotemporal expression of WT1 and RALDH2 in the embryonic avian heart: a model for the regulation of myocardial and valvuloseptal development by epicardially derived cells (EPDCs). Dev Biol. 2002; 247:307-326.

[20] Eid H, Larson DM, Springhorn JP, Attawia MA, Nayak RC, Smith TW, Kelly RA. Role of epicardial mesothe- lial cells in the modifi cation of phenotype and function of adult rat ventricular myocytes in primary coculture. Circ Res. 1992; 71:40-50.

[21] Wagner KD, Wagner N, Bondke A, Nafz B, Flemming B, Theres H, Scholz H. The Wilms’ tumor suppressor Wt1 is expressed in the coronary vasculature after myocardial infarction. FASEB J. 2002; 16:1117-1119.

[22] Wessels A, Perez-Pomares JM. The epicardium and epicardially derived cells (EPDCs) as cardiac stem cells. Anat Rec A Discov Mol Cell Evol Biol. 2004; 276:43-57.

[23] Meyerrose TE, Herrbrich P, Hess DA, Nolta JA. Immune-defi cient mouse models for analysis of human stem cells. Biotechniques. 2003; 35:1262-1272.

[24] van der Geest RJ, Buller VG, Jansen E, Lamb HJ, Baur LH, van der Wall EE, de Roos A, Reiber JH. Com- parison between manual and semiautomated analysis of left ventricular volume parameters from short-axis MR images. J Comput Assist Tomogr. 1997; 21:756-765.

[25] Le Jemtel TH, Lambert F, Levitsky DO, Clergue M, Anger M, Gabbiani G, Lompre AM. Age-related changes in sarcoplasmic reticulum Ca(2+)-ATPase and alpha-smooth muscle actin gene expression in aortas of normotensive and spontaneously hypertensive rats. Circ Res. 1993; 72:341-348.

[26] Tsukada T, Tippens D, Gordon D, Ross R, Gown AM. HHF35, a muscle-actin-specifi c monoclonal anti- body. I. Immunocytochemical and biochemical characterization. Am J Pathol. 1987; 126:51-60.

[27] Ou Y, Gibbons SJ, Miller SM, Strege PR, Rich A, Distad MA, Ackerman MJ, Rae JL, Szurszewski JH, Farrugia G. SCN5A is expressed in human jejunal circular smooth muscle cells. Neurogastroenterol Motil. 2002;

14:477-486.

[28] Poelmann RE, Gittenberger-de Groot AC, Mentink MM, Bokenkamp R, Hogers B. Development of the

(24)

167

cardiac coronary vascular endothelium, studied with antiendothelial antibodies, in chicken-quail chimeras.

Circ Res. 1993; 73:559-568.

[29] Merki E, Zamora M, Raya A, Kawakami Y, Wang J, Zhang X, Burch J, Kubalak SW, Kaliman P, Belmonte JC, Chien KR, Ruiz-Lozano P. Epicardial retinoid X receptor alpha is required for myocardial growth and coro- nary artery formation. Proc Natl Acad Sci U S A. 2005; 102:18455-18460.

[30] Tavian M, Zheng B, Oberlin E, Crisan M, Sun B, Huard J, Peault B. The vascular wall as a source of stem cells.

Ann N Y Acad Sci. 2005; 1044:41-50.

[31] Jonsson ZO, Hubscher U. Proliferating cell nuclear antigen: more than a clamp for DNA polymerases.

Bioessays. 1997; 19:967-975.

[32] Holmes AM, Haber JE. Double-strand break repair in yeast requires both leading and lagging strand DNA polymerases. Cell. 1999; 96:415-424.

33] Essers J, Theil AF, Baldeyron C, van Cappellen WA, Houtsmuller AB, Kanaar R, Vermeulen W. Nuclear dynam- ics of PCNA in DNA replication and repair. Mol Cell Biol. 2005; 25:9350-9359.

[34] Uemura R, Xu M, Ahmad N, Ashraf M. Bone Marrow Stem Cells Prevent Left Ventricular Remodeling of Ischemic Heart Through Paracrine Signaling. Circ Res. 2006.

[35] Gerdes AM, Capasso JM. Structural remodeling and mechanical dysfunction of cardiac myocytes in heart failure. J Mol Cell Cardiol. 1995; 27:849-856.

[36] Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocar- dial regeneration. Cell. 2003; 114:763-776.

[37] Fazel S, Cimini M, Chen L, Li S, Angoulvant D, Fedak P, Verma S, Weisel RD, Keating A, Li RK. Cardioprotective c-kit+ cells are from the bone marrow and regulate the myocardial balance of angiogenic cytokines. J Clin Invest. 2006; 116:1865-1877.

[38] Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, Scadova H, Qin G, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW. Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. J Clin Invest. 2005; 115:326-338.

[39] Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, Salio M, Battaglia M, Latronico MV, Coletta M, Vivarelli E, Frati L, Cossu G, Giacomello A. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ Res. 2004; 95:911-921.

[40] Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, Bussani R, Nadal-Ginard B, Silvestri F, Leri A, Beltrami CA, Anversa P. Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med.

2001; 344:1750-1757.

[41] Leor J, Patterson M, Quinones MJ, Kedes LH, Kloner RA. Transplantation of fetal myocardial tissue into the infarcted myocardium of rat. A potential method for repair of infarcted myocardium? Circulation. 1996; 94:

II332-II336.

[42] Degabriele NM, Griesenbach U, Sato K, Post MJ, Zhu J, Williams J, Jeff ery PK, Geddes DM, Alton EW. Critical appraisal of the mouse model of myocardial infarction. Exp Physiol. 2004; 89:497-505.

(25)

168 [43] Yang Z, Berr SS, Gilson WD, Toufektsian MC, French BA. Simultaneous evaluation of infarct size and cardiac function in intact mice by contrast-enhanced cardiac magnetic resonance imaging reveals contractile dysfunction in noninfarcted regions early after myocardial infarction. Circulation. 2004; 109:1161-1167.

[44] Jacoby C, Molojavyi A, Flogel U, Merx MW, Ding Z, Schrader J. Direct comparison of magnetic resonance imaging and conductance microcatheter in the evaluation of left ventricular function in mice. Basic Res Cardiol. 2006; 101:87-95.

[45] Gottlieb SH, Ouyang P, Gottlieb SO. Death after acute myocardial infarction: interrelation between left ventricular dysfunction, arrhythmias and ischemia. Am J Cardiol. 1988; 61:7B-12B.

[46] Anker SD, Sharma R. The syndrome of cardiac cachexia. Int J Cardiol. 2002; 85:51-66.

[47] Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, Morello F, Mu H, Melo LG, Pratt RE, Ingwall JS, Dzau VJ. Evidence supporting paracrine hypothesis for Akt-modifi ed mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J. 2006; 20:661-669.

[48] Beeres SL, Bax JJ, Kaandorp TA, Zeppenfeld K, Lamb HJ, Dibbets-Schneider P, Stokkel MP, Fibbe WE, de Roos A, van der Wall EE, Schalij MJ, Atsma DE. Usefulness of intramyocardial injection of autologous bone marrow-derived mononuclear cells in patients with severe angina pectoris and stress-induced myocardial ischemia. Am J Cardiol. 2006; 97:1326-1331.

[49] Mack MJ. Minimally invasive cardiac surgery. Surg Endosc. 2006; 20 Suppl 2:S488-S492.

(26)

169

ONLINE DATA SUPPLEMENT

Materials and M ethods

P R I M A R Y C U L T U R E D H U M A N A D U L T E P D C S

EPDCs were cultured from human adult epicardium. Epicardium was separated mechanically from human atrial myocardium, which was obtained as leftover material after coronary artery bypass graft (CABG) procedures. Epicardial tissue was cut into small pieces, placed into two 9.5 cm2 culture dishes (Primaria, BD Biosciences, San Jose, CA, USA) and covered with a coverslip to prevent fl oating. The epicardial graft was removed immediately after the fi rst human EPDCs (hEPDCs) migrated from the explant. Cells were cultured in medium consisting of 45 % DMEM, 45 % M199, and 10 % FCS, to which 100 U/ml penicillin, 100 μg/ml streptomycin (all from Invitro- gen, Paisley, UK) and 2 ng/mL bFGF (BD Biosciences) was added, at 37 °C in a humidifi ed air-5 % CO2 atmosphere incubator. When cultures reached confl uency, cells were trypsinized, diluted three times and reseeded. Growth curves were determined (n = 4).

Cells from passage 1 (cobble stone shape) and passage 2-4 (spindle shape) were tested for their Wilms’ Tumor 1 suppressor protein (WT1) expression. Cells were fi xed with methanol 100 % for 15 min. After antigen retrieval with 0.1 % Triton x-100 for 30 minutes (1086031000, Merck KGaA, Darmstadt, Germany) endogenous peroxidase activity was blocked by incubation with 3 % hydrogen peroxide for 15 min. Cells were incubated overnight with WT1 antibody (sc-192, Santa Cruz, Santa Cruz, CA, USA), combined with biotinylated goat anti-rabbit IgG (BA-1000, Vector Labs, Burlingame, CA, USA). The signal was amplifi cated with the ABC staining kit (PK-6100, Vec- tor Labs), using 3,3’-diamino-benzidine tetrahydrochloride (D5637, Sigma-Aldrich) as substrate for horseradish peroxidase. Sections were briefl y counterstained with Mayer’s hematoxilin.

V I R U S - M E D I A T E D E G F P T R A N S D U C T I O N O F H E P D C S

Cells from passage two to four (spindle shape) were used for transplantation experiments. To enable cell tracing, cultured adult hEPDCs were transduced with the enhanced green fl uores- cent protein (eGFP) gene 72h before transplantation. For the short term experiments (2 weeks survival), cells were transduced with the adenoviral vector hAd5/F50.CMV.eGFP [1]. Cells were incubated with 50 infectious units (IU) of hAd5/F50.CMV.eGFP per cell. Because adenoviral expression is lost within a few weeks, hEPDCs used for long term experiments (6 weeks survival) were transduced with the lentiviral vector Lv.hPgk.eGFP (12,5 HeLa transducing units/ml per cell) instead of the adenoviral vector.

(27)

170 C R E A T I O N O F T H E M I A N D C E L L T R A N S P L A N T A T I O N

To avoid rejection of transplanted human cells, 8-12 weeks old male nonobese diabetic-severe combined immunodefi cient (NOD/scid) mice (Charles River, Wilmington, MA, USA) with a body weight of 25-30 g were used [2]. All animal procedures were approved by the Animal Ethics Committee of the Leiden University and conformed the Guide for the Care and Use of Labora- tory Animals (National Institutes of Health publication No. 85-23, Revised 1996). Animals were anesthetized with isofl urane 5 % for induction and 1.5-2 % for maintenance in a gas mixture of oxygen and N2O (1:1). Mice were placed in a supine position on a controlled heating pad, intubated and ventilated mechanically (Harvard Ventilator) with a tidal volume of 240 μL and a frequency of 200/min.

The thoracic cavity was opened in the fi fth intercostal space. After opening of the pericardial sac, the left anterior descending coronary artery (LAD) was visualized and permanently ligated using 7.0 suture (Prolene, Johnson and Johnson, New Brunswick, NJ, USA). For short (2 weeks) survival experiments, the location of ligation was 1 mm caudally from the tip of the left auricle.

For long (6 weeks) survival experiments only the frontal branch of the LAD was ligated, since extended survival until 6 weeks is not possible with total LAD ligation. Transplantation of 4 x 105 hEPDCs suspended in a total volume of 20 μL M199 (hEPDC-group, survival 2 weeks: n = 22, 6 weeks: n = 15), or 20 μL cell-free M199 (medium-group, survival 2 weeks: n = 24, 6 weeks: n = 18) was performed by 5 injections using a beveled needle into the ischemic myocardium of the left ventricle and the border zone, immediately after ligation of the LAD. Sham-operated animals (survival 2 weeks: n = 16, 6 weeks: n = 3) were operated similarly, but without ligation of the LAD and without fl uid injection. After recovery, animals received food and water with antibiotics (Ciproxin and Polymixin B, 10 mg/mL) and an antimycotic (Fungizone 10 mg/mL) ad libitum.

Animals were randomized to treatment.

S H O R T T E R M E X P E R I M E N T S

MRI

Infarct size (2 days after surgery) and cardiac function (2 and 14 days after surgery) were assessed using a vertical 9.4 Tesla Wide Bore magnet (AVANCE console) (Bruker, Etlingen, Germany) with a 30-mm birdcage resonator and an actively shielded gradient set, which had a maximum gra- dient strength of 1 T/m and a rise time of 110 μs. Bruker ParaVision 3.02 software was used for image acquisition. Animals were anesthetized with 5 % isofl urane for induction and 1.5-2 % iso- fl urane for maintenance in a mixture of oxygen and air (1:1) with a fl ow of 0.6 L/min, and placed head-up into an animal holder. Electrocardiogram (ECG) electrodes (3M, Red DotTM), attached to the right forelimb and the tail, and a respiration detection cushion placed under the thorax, were used to monitor ECG and respiratory rate (Bruker BioTrig). Gadolinium-DOTA (Dotarem,

(28)

171 Guerbet) was injected (37.5 μmol in 150 μL) via the tail vein 40 ± 15 min before contrast-enhanced

images were made [3].

Contrast-enhanced images for infarct size measurement

To visualize and measure infarct size, 18 contiguous 0.5 mm short-axis contrast-enhanced images were obtained on day 2, covering the entire heart, using an ECG- and respiratory-triggered fast gradient echo (FLASH) sequence with a fl ip angle of 60° to null the signal from the myocardium.

A 45 ms repetition time and an echo time of 1.9 ms were used. The signal was averaged 6 times.

The fi eld of view was 25.6 x 25.6 mm, projected on a 256 x 256 matrix, resulting in a pixel size of 100 x 100 μm.

Cine images for cardiac function

An ECG- and respiration-triggered cine FLASH sequence with a fl ip angle of 15°, a repetition time of 7 ms, and an echo time of 1.9 ms was used to assess cardiac function on day 2 and 14. During one cardiac cycle 18-30 frames were acquired, dependent on heart rate. The signal was averaged 4 times. The fi eld of view was as described above. To cover the entire left ventricle, six to nine contiguous short axis slices of 1 mm thickness were made.

Determination of infarct size and cardiac function

The MRI images were converted to DICOM format for analysis with the MASS for Mice software package (Leiden, the Netherlands). The endocardial and epicardial borders were delineated manually by an observer blinded to the experimental groups (hEPDC-group n = 17, medium- group n = 14, sham-group n = 13). Subsequently, the infarcted area of the left ventricle was computed automatically (the contrast-enhanced left ventricular (LV) area divided by the total LV area), as were LV end-diastolic volume (EDV), LV end-systolic volume (ESV), LV stroke volume (SV), and LV ejection fraction (EF) [4]. Infarcted area measurements were used to correct func- tional parameters for potential diff erences in initial infarct size.

I M M U N O H I S T O C H E M I C A L A S S E S S M E N T

Mice were sacrifi ced 15 days after surgery. Hearts were excised and fi xed by perfusion, followed by immersion during 48 h at 4 °C with 4 % paraformaldehyde in phosphate-buff ered saline (0.1 M, pH 7.4). Subsequently, hearts (n = 5 per group) were embedded in paraffi n, and sectioned at 5 μm. To block endogenous peroxidase, sections were incubated with 3 % hydrogen peroxide for 15 min; antigen retrieval was accomplished by microwaving for 12 min at 98 °C for all sec- tions, except for those that were stained for α-smooth muscle actin and proliferating cell nuclear antigen (no antigen retrieval) or the endothelial marker CD31 (6-min treatment with pronase at room temperature). Sections were incubated at room temperature with primary antibodies

(29)

172 overnight, and with secondary antibodies for 60 min. To investigate host tissue properties, serial sections were stained for CD31 (clone MEC13.3, 550274, Pharmingen, San Diego, CA, USA), α/γ muscle actin (clone HHF35, M0635, DAKO, Glostrup, Denmark), α-smooth muscle actin (clone 1A4, A2547, Sigma-Aldrich, St. Louis, MO, USA), proliferating cell nuclear antigen (PCNA, clone PC10, M0879, Dako), phospho-histone H3 (06-570, Upstate, Billerica, MA, USA), and phospho- histone H2A.X (clone JBW301, Upstate) combined with an appropriate secondary antibody. Anti- eGFP (A11122, Molecular Probes, Paisley, UK) staining was performed to visualize the injected hEPDCs because the strong and irregular autofl uorescence of the heart disturbs assessment of spontaneous eGFP fl uorescence to detect engrafted cells. The following secondary antibodies were used: biotinylated goat anti-rat IgG (559286, Pharmingen), biotinylated goat anti-rabbit IgG (BA-1000), biotinylated horse anti-mouse IgG (BA-2000, Vector Labs), and rabbit anti-mouse IgG conjugated to horseradish peroxidase (P0260, DAKO). For visualization of eGFP, α/γ muscle actin, PCNA, phospho-histone H3, and phospo-histone H2AX staining, the signal was ampli- fi ed with the ABC staining kit (PK-6100, Vector Labs), and for CD31 staining, visualization was enforced by the CSA system (K1500, DAKO), using 3,3’-diamino-benzidine tetrahydrochloride (D5637, Sigma-Aldrich) as substrate for horseradish peroxidase. Sections were briefl y counter- stained with Mayer’s hematoxilin.

To identify which proteins were expressed by the injected hEPDCs, double stainings were per- formed for eGFP and other proteins, being α-smooth muscle actin (clone 1A4), von Willebrand Factor (vWF, 4400-5884, Biogenesis, Poole, UK), sarcoplasmic or endoplasmic reticulum Ca2+- ATPase (SERCA2a, clone 2A7-A1, MA3-919, Affi nity BioReagents, Golden, CO, USA), voltage-gated sodium channel (α-subunit) (SCN5a, sc-23174, Santa Cruz), cardiac troponin I, (cTnI, clone 19C7, 4T21, HyTest, Turku, Finland), atrial natriuretic peptide (ANP, clone 23/1, CBL66, Chemicon, Temec- ula, CA, USA), α/γ muscle actin (clone HHF35), and sarcomeric myosin (clone MF20, Hybridoma Bank, Iowa City, IA, USA). A Red Qdot conjugated goat anti-rabbit antibody (Q11421MP, Invitro- gen) was used to visualize eGFP staining. An appropriate biotinylated secondary antibody (don- key anti-goat [605-706-002, BioTrend, Cologne, Germany] or horse anti-mouse [BA-2000, Vector Labs]) in combination with Yellow Qdot conjugated to streptavidin (Q10111MP, Invitrogen) was used to visualize the other proteins. Stained sections were analyzed by light microscopy or con- focal microscopy (Leica CTR 6000, Leica Confocal Software v2.6.1, excitation laser of 488 nm).

L V V A S C U L A R P R O F I L E A N D W A L L T H I C K N E S S

To evaluate the angiogenic eff ect of hEPDC transplantation, the cumulative area of CD31-stained vessel lining per total LV area was determined in the hEPDC- and medium-group (n = 5 for each group). Measurements were performed in fi ve diff erent sections, which were chosen in a uni- form manner, using apex and ligature for orientation. The most apical section was located 50 μm cranially from the apex, the most basal section was situated 50 μm caudally from the ligature.

(30)

173 Three other sections were equally distributed between the apical and basal sections. Endocardial

staining was excluded from quantitative analysis. Per section, two areas in the interventricular septum, and six equally displayed areas in the free wall were analyzed (Figure 4f ). To normalize for diff erences in staining intensity among sections, values determined in the free wall areas were expressed relative to the values of the interventricular septal areas of the corresponding section. Measurements were performed by an observer blinded to treatment, using Image Pro software package (Media Cybernetics, Silver Spring, MD, USA).

LV wall thickness was measured in the hearts of the hEPDC- and medium-group (n = 5 for each group) by an observer blinded to treatment. Average LV wall thickness was quantifi ed from microscopic images of fi ve sections per heart (same sections as described for endothelial den- sity measurements). Wall thickness was measured in both border zones of the infarcted wall, in the mid-infarcted area in between the border zones, and in the middle of the interventricular septum. Measurements were performed perpendicular to the ventricular or septal wall.

G E N E R A L H E A L T H A N D S U R V I V A L

Body weight was determined just before surgery at day 0 and before sacrifi ce at day 15. Wet and dry weights (acquired after freeze-drying) of the lungs were measured after sacrifi ce. Pulmonary water content was estimated by subtracting the dry weight from the wet weight. To correct for diff erences in body mass, the amount of lung fl uid was expressed relative to body weight.

Survival proportions were assessed. Death was defi ned as spontaneous death before sacrifi ce or death during anesthesia throughout MRI experiments.

L O N G T E R M E X P E R I M E N T S

Experiments with evaluation 6 weeks after myocardial infarction were performed to investigate the eff ect of hEPDC transplantation after a defi nitive scar had been formed.

MRI

Because of the high risk of mortality during repeated imaging procedures, cine MRI images were performed (methods were similar to procedures described for short term experiments) only 42 days after surgery (sham-group n = 3, medium-group n = 14, hEPDC-group n = 15), not on day 2.

Immunohistochemical assessment

Animals were sacrifi ced 43 days after MI. Excised hearts were treated as described above. Per group 3 paraffi n embedded hearts were used for immunohistochemical evaluation. To identify injected cells, single non-fl uorescent antibody stainings against eGFP, human specifi c CD31 (clone CLB-HEC/75, M1536, Sanquin, Amsterdam, the Netherlands) and human specifi c vWF (4400-5884, Biogenesis) were performed. Staining methods were similar to the above described

(31)

174 protocols, using an appropriate biotinylated secondary antibody (also biotinylated horse anti- goat IgG [BA-9500, Vector Labs]) together with the ABC kit.

To perform staining for human nucleus (clone 235-1, MAB1281, Chemicon), 3 hearts (without fi xa- tion) of the hEPDC-group and the medium-group were embedded in Tissue-Tek (OCT compound, 4583, Sakura Finetek, Zoeterwoude, the Netherlands), frozen at -80 °C for 48 h, and stored at -20

°C before sectioning at 8 μm. Inhibition of endogenous peroxidase was performed as described above. Sections were incubated overnight with the primary antibody against human nucleus, subsequently incubated with biotinylated horse anti-mouse IgG (BA-2000, Vector Labs), blocked with normal Horse serum, and incubated with ABC staining kit (PK-6100, Vector Labs).

LV vascular profile and wall thickness

Sirius Red staining and antibody staining against CD31 (clone MEC13.3, Pharmingen), together with wall thickness measurements were used for host tissue investigation.

Survival

Survival proportions until sacrifi ce at day 43 were assessed according to the methods described for short term experiments.

S T A T I S T I C A L A N A L Y S I S

Data are presented as mean ± standard error (SEM). Data were analyzed with SPSS software (SPSS Inc, Chicago, IL, USA). For comparisons of more than two groups, a one-way-ANOVA was performed (or the non-parametric Kruskal-Wallis for the dependent variable lung weight). If the omnibus tests among groups were signifi cantly diff erent, post-hoc tests between groups using t-tests (and Mann-Whitney for lung weight) were performed. Infarct size was used as covariate in an analysis of (co-)variance of the functional data to correct for baseline diff erences in infarct size among groups. Diff erences in mortality were evaluated using the Breslow test. A level of p < 0.05 was considered to represent a signifi cant diff erence.

(32)

175 Results

Cells from the fi rst passage, mainly cobble stone shaped, demonstrated a high expression of WT1 (Supplemental Figure 1a). Transformation of these cobble stone cells into spindle shaped cells was accompanied by reduction in WT1 expression (Supplemental Figure 1b).

Supplemental Figure 1. Expression of Wilms’ Tumor 1 suppressor protein (WT1) in adult hEPDCs. High expression of WT1 is observed in the cobble stone shaped cells grown from the epicardial explant (a). Spindle shaped cells from the second passage demonstrate a decreased WT1 expression (b). Scale bars: 60 μm.

(33)

176

REFERENCES

[1] Knaan-Shanzer S, van der Velde I, Havenga MJ, Lemckert AA, de Vries AA, Valerio D. Highly effi cient targeted transduction of undiff erentiated human hematopoietic cells by adenoviral vectors displaying fi ber knobs of subgroup B. Hum Gene Ther. 2001; 12:1989-2005.

[2] Meyerrose TE, Herrbrich P, Hess DA, Nolta JA. Immune-defi cient mouse models for analysis of human stem cells. Biotechniques. 2003; 35:1262-1272.

[3] Yang Z, Berr SS, Gilson WD, Toufektsian MC, French BA. Simultaneous evaluation of infarct size and cardiac function in intact mice by contrast-enhanced cardiac magnetic resonance imaging reveals contractile dysfunction in noninfarcted regions early after myocardial infarction. Circulation. 2004; 109:1161-1167.

[4] van der Geest RJ, Buller VG, Jansen E, Lamb HJ, Baur LH, van der Wall EE, de Roos A, Reiber JH. Comparison between manual and semiautomated analysis of left ventricular volume parameters from short-axis MR images. J Comput Assist Tomogr. 1997; 21:756-765.

Referenties

GERELATEERDE DOCUMENTEN

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden Downloaded.

The studies described in this thesis were performed at the departments of Antomy and Embryology, and Cardiology of the Leiden University Medical Center, Leiden, the

Cell isolation procedures matter: a comparison of diff erent isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial

The aim of the present study was to address histological changes in porcine ECM constitu- tion induced by two diff erent cell extraction methods; a non-ionic detergent Triton X-100 and

The authors of “Decellularization of rat aortic valve allografts reduces leafl et destruction and extracellular matrix remodeling”, have elegantly evaluated various methods

In a rat aortic valve transplantation model [7] transplantation of decellularised AVA between MHC-mismatched rats, valves were still morphologically intact after 3 weeks, whereas

Key fi ndings of the present study are that in an immune-compromised mouse model of acute MI, intra-myocardial injection of hMSCs from patients with IHD resulted in (i) a signifi

Key fi ndings of this study are: (1) Injection of hMSC and hMSC myoc into infarcted myocardium results in a relatively preserved LV function compared to injection of medium alone;