contents chapter 05 · chapter 5 82 endothelial progenitor cells give rise to smooth muscle progeny...

20
CHAPTER 05 ENDOTHELIAL PROGENITOR CELLS GIVE RISE TO SMOOTH MUSCLE PROGENY: IMPLICATIONS FOR A COMMON PROGENITOR STATUS Jan-Renier AJ Moonen * , Guido Krenning * , Marja GL Brinker Marja JA van Luyn and Martin C Harmsen Stem Cell & Tissue Engineering Research Group, Dept. Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands * Authors contributed equally Manuscript Submitted

Transcript of contents chapter 05 · chapter 5 82 endothelial progenitor cells give rise to smooth muscle progeny...

contents

81

chapter05

endothelial progenitor cells give rise to

smooth muscle progeny:

implications for a common progenitor status

Jan-Renier AJ Moonen*, Guido Krenning*, Marja GL BrinkerMarja JA van Luyn and Martin C Harmsen

Stem Cell & Tissue Engineering Research Group, Dept. Pathology & Medical Biology, University Medical Center Groningen, University of Groningen, The Netherlands

* Authors contributed equally

Manuscript Submitted

chapter 5

82

endothelial progenitor cells give rise to smooth muscle progeny

83

5

abstract

Endothelial cells and smooth muscle cells have long been thought to have dinstinct progenitors. However, embryonic vascular progenitor cells with endothelial and smooth muscle progeny have been described and transdifferentiation between both cell types has been shown. Given the plasticity between both lineages we hypothesized that endothelial progenitor cells, known for their angiogenic properties, could transdifferentiate to smooth muscle cells and thereby gain a common vascular progenitor status. Indeed, umbilical cord blood-derived endothelial outgrowth cells (EOC) can give rise to smooth muscle cells through a process termed endothelial-to-mesenchymal transdifferentiation (EnMT). This EnMT was induced by TGF-β1 and completely inhibited by ALK5 kinase inhibition. After EnMT, expression of endothelial markers was lost, as was endothelial anti-thrombogenic function. Transdifferentiated EOC gained mesenchymal marker expression and acquired a contractile phenotype. Furthermore, transdifferentiated EOC showed bFGF and angiopoietin-1-mediated pro-angiogenic paracrine effects on untransdifferentiated EOC. Our study demonstrates that EOC are capable of TGF-β1-mediated EnMT, and thereby give rise to smooth muscle progeny. This implies a common vascular progenitor status for EOC, which clearly has implications for their therapeutical use.

introduction

During embryogenesis, endothelial cells arise from hemangioblasts [156]. Smooth muscle cells arise from local mesenchyme and the neural crest [157;158]. As both vascular cell types originate from different sources, it has long been thought that these cells have dinstinct progenitors. However, in 2000, Yamashita and collegues described embryonic vascular progenitor cells, that can differentiate into endothelial and smooth muscle cells [159]. These results were later confirmed by Ferreira and co-workers, who showed functional integration of vascular progenitor cell-derived endothelial cells and smooth muscle cells in preexisting vasculature [160].

In addition to endothelial- and smooth muscle maturation of embryonic vascular progenitor cells, transdifferentiation between endothelial and smooth muscle cells has also been described. During embryogenesis, endothelial-to-mesenchymal transdifferentiation (EnMT), takes place during the formation of the heart valves [143;161]. In vitro, EnMT of embryonic endothelial cells to smooth muscle actin expressing mesenchymal cells was also shown [151]. Furthermore EnMT is described for mature endothelial cells [144;155;162-165], and recent evidence supports a role for this process in cardiovascular fibrosis [166]. Although less well documented, mesenchymal-to-endothelial transdifferentiation has also been described [167;168]. Furthermore, endothelial cells and vascular smooth muscle cells have certain markers in common such as VEGFR-2 and the type 3 TGF-β receptor endoglin (CD105). These data indicate a potential plasticity of endothelial and smooth muscle cell differentiation.

Numerous studies have shown that endothelial progenitor cells (EPC) contribute to neovascularization, either by differentiation into cells of the endothelial lineage and local engraftment into to vasculature [61;63] or by secretion of pro-angiogenic factors [102;113;169]. In vitro EPC, termed endothelial outgrowth cells (EOC), can be derived from mononuclear cells from umbilical cord and adult blood [170]. These cells, with phenotypical and functional characteristics of endothelial cells, show stem cell-like properties, such as the ability of clonal expansion and telomerase activity [47].

Given the plasticity between endothelial- and mesenchymal lineages, common marker expression, and the stem-cell like properties of EOC, we hypothesized that EOC can (trans)differentiate into smooth muscle cells. This would imply a common vascular progenitor cell status for the archetype in vitro EPC.

materials & methods.

isolation and culture of umbilical cord blood-derived endothelial outgrowth cells (uc-eoc)

Umbilical cord blood was collected at the department of Gynecology, Medical Center Leeuwarden, the Netherlands. Cord blood was isolated directly after normal term delivery, with informed consent from the parents and according to institutional guidelines and the declaration of Helsinki. Cord blood mononuclear cells were separated by density gradient centrifugation on Lymphoprep (Nycomed Pharma, Norway) and cultured on fibronectin-coated (2 μg/cm2; Harbor Bio-Products, MA) culture flasks at a

chapter 5

84

endothelial progenitor cells give rise to smooth muscle progeny

85

5

from Cell Signaling, MA), phosphorylated SMAD2 (pSMAD2, 2 µg/mL; Millipore, MA) and SMAD1/5/8 (4 µg/mL, Santa Cruz Biotechnology, CA) overnight and incubated the next day using fluorescein-conjugated donkey antibody fragments to rabbit IgG. Labeled samples were visualized using a Leica LMRXA fluorescent microscope and Leica software.

functional characterizations

To assess the uptake of acetylated low density lipoprotein (acLDL), 10 μg/mL DiI-labeled acLDL was added to the culture medium and incubated overnight. Subsequently, cells were fixed using 2% paraformaldehyde (Polysciences, PA), rehydrated and incubated with 3 μM DAPI for 10 minutes.

Binding of lectins from Ulex europaeus (UEA-1) was assessed by incubating 2% paraformaldehyde-fixed cells with fluorescein-conjugated UEA-1 lectins (5 μg/mL) dissolved in 3 μM DAPI for 1 hour. Subsequently, samples were analyzed on a Leica DMRXA fluorescent microscope at 40x lens magnification.

Capillary sprout formation by UC-EOC was assessed on MatriGel™ (BD Biosciences, CA). In short, 10 μL of MatriGel™ was solidified in µ-Slide Angiogenesis plates (Ibidi GmbH, Germany). Next, 15 000 UC-EOC were placed on the MatriGel™ and cultured in endothelial outgrowth medium overnight. Sprout formation was analyzed by microscopic analysis.

Thrombin generation was assessed using a Thrombin Generation Assay (HeamoScan, The Netherlands) as described previously [113;165]. In short, cells were incubated with fibrinogen-depleted plasma under normal culture conditions. A mixture of 30mM CaCl2 and phospholipids was added to induce the formation of thrombin. At regular intervals, samples were drawn, diluted in ice-cold TrisHCL and incubated with 3mM chromophore-releasing Thrombin substrate S2238. Change in color was assessed in a microtiter plate reader (BioRad, VA) and plotted against a calibration curve of known thrombin concentrations. Human umbilical cord endothelial cells and tissue culture-treated plastic served as negative and positive controls, respectively.

Gel contraction experiments were performed as described previously [165]. Briefly, a solution of collagen type I (8 mg/mL; BD Biosciences, MA) was supplemented with 10 mM NaOH, 2.5 mM HEPES (GIBCO/Invitrogen, CA) and transdifferentiated cells (1•106/mL). Aliquots of 50 μL (containing 50 000 cells and 0.2 mg collagen type I) were plated on plastic culture dishes and allowed to solidity for 30 minutes. Thereafter 1.5 mL culture medium was added, gels were released with a spatula, and imaged using a common flatbed-scanner (ScanJet 5370C; HP, CA). Thereafter, gels were allowed to contract for 48 hours. After 20 hours of spontaneous contraction, TGF-β1 was added to the culture medium.

immunoblot analysis

Whole cell lysates were prepared in RIPA buffer supplemented with 1% proteinase inhibitor cocktail and 1% phosphatase inhibitor cocktail (both Sigma-Aldrich, MA). Cell lysates (20 µg/lane) were separated by gel electrophoresis on a 10% non-denaturing polyacrylamide gel and subsequently blotted onto nitrocellulose according to

density of 1-2•106 cells/cm2 in endothelial outgrowth medium containing Medium-199 (Cambrex BioScience, ME), 20% fetal bovine serum (FBS; Invitrogen/GIBCO, CA), 1% Penicillin/Streptomycin, 2 mM L-glutamine (both Sigma Aldrich, MA), 5 U/mL heparin (Leo Pharma, Denmark), 10 ng/mL bFGF, 20 ng/mL HGF, 10 ng/mL IGF-1 and 10 ng/mL VEGFa (all Prepotech, NJ). Cord blood mononuclear cells were cultured for 14-21 days before passaging and culture medium was refreshed every third day.

For endothelial-to-mesenchymal transdifferentiation (EnMT), UC-EOC cultured up to passage 4 were used. EnMT was initiated by replacing the endothelial outgrowth medium to mesenchymal differentiation medium consisting of RPMI 1640 supplemented with 20% FBS, 1% Penicillin/Streptomycin, 2 mM L-glutamine, 5 U/mL heparin, with addition of 5 ng/mL TGF-β1 and 15 ng/mL PDGF-BB (both from Peprotech, NJ). Transdifferentiated cells (UC-EnMT) were subsequently cultured in basal medium consisting of RPMI 1640 supplemented with 20% FBS, 1% Penicillin/Streptomycin, 2 mM L-glutamine and 5 U/mL heparin, for up to 7 additional passages. When applicable, ALK5-kinase activity was inhibited by addition of 20µM SB431542 (Sigma, MA) to the culture media.

phenotypic characterization

Cells were detached using accutase (PAA Laboratories, Austria) and subsequently stained using fluorophore-conjugated antibodies to human CD31 (5 µg/mL; IQ Products, The Netherlands), CD34 (5 µg/mL; BD Biosciences, CA), CD105 (5 μg/mL), CD144 (2.5 μg/mL), VEGF-R2 (5 μg/mL), Tie-2 (2.5 μg/mL), αSMA (2.5 μg/mL), TGFβ-R2 (5 μg/mL), and PDGF-Rβ (5 μg/mL; all R&D Systems, MN), or unconjugated antibodies to human eNOS (2.5 μg/mL; BD Biosciences, CA), von Willebrand Factor (3 μg/mL; DakoCytomation, Denmark), thrombomodulin (10 μg/mL), SM22α (5 μg/mL), SM-MHC2 (5 μg/mL), Calponin (5μg/mL; all Abcam, UK), ALK1 (5 µg/mL; R&D Systems, MN) and ALK5 (2 μg/mL; Santa Cruz Biotechnology, CA). Unlabeled primary antibodies were subsequently stained using either fluorescein-conjugated donkey antibody fragments to rabbit IgG (5 μg/mL; Jackson ImmunoResearch, PA) or fluoroscein-conjugated rabbit antibodies to mouse IgG (5 μg/mL; DakoCytomation, Denmark).

For detection of intracellular proteins, cells were fixed using 2% paraformaldehyde and permeabilized using 0.1% Saponin (Sigma-Aldrich, MA) prior to staining procedures. Flow cytometric analysis was performed on a FACSCalibur™ (BD Biosciences, CA). Fluorophore-conjugated, isotype-matched nonsense antibodies, as well as fluorescein-conjugated donkey antibody fragments to rabbit IgG and fluorescein-conjugated rabbit antibodies to mouse IgG served as controls.

For visualization of cytoskeletal organizations, cells were fixed in ice-cold methanol:acetone (1:1), rehydrated and incubated with 0.1 μM fluorescein-conjugated phallotoxins (Molecular Probes/Invitrogen, OR) in phosphate buffered saline (PBS) containing 3 μM 4’,6-diamidino-2-phenylindole (DAPI; Sigma-Aldrich, MA) for 30 minutes.

For detection of (phosphorylated) SMAD proteins, cells were fixed using 2% paraformaldehyde and permeabilized using 0.1% Saponin (Sigma-Aldrich, MA) prior to staining procedures. Samples were subsequently incubated with antibodies against SMAD2/3 (2 µg/mL) and phosphorylated SMAD1/5/8 (pSMAD1/5/8, 2 µg/mL; both

chapter 5

86

endothelial progenitor cells give rise to smooth muscle progeny

87

5

Activity of telomerase was determined using the TeloTAGGG Assay (Roche Diagnostics, the Netherlands) following manufacturers protocol. In short, whole cell extracts of 2 000 cells were incubated with reaction mixture containing telomerase substrate at 25°C for 60 minutes. After telomerase-mediated substrate elongation, PCR amplification was performed at 30 cycles at 94°C for 30s, 50°C for 30s and 72°C for 90s. Hybridization and enzyme-linked immunosorbent assay (ELISA) of amplicons were performed according to manufacturer’s instructions.

Amplicons of telomeric repeats were visualized using gel electrophoresis on a 12% non-denaturing acrylamide gel following standard protocols. Subsequently, amplicons were transferred to a positively charged nylon membrane (Hybond-N+, Amersham, UK) by standard blotting techniques. Blotting membranes were blocked in 5% BSA for 30 minutes and incubated with alkaline phosphatase-conjugated streptavidins (0.1 µg/mL; Southern Biotech, AL) for 1 hour. NBT/BCIP (Bio-Rad, VA) was used as substrate for the detection.

paracrine effects of uc-enmt

UC-EnMT at passage 5 were cultured in basal medium for 72h. The conditioned medium (CM) was filtered using 0.2 µm filters to remove cell debris. Capillary sprout formation was performed as described above. CM was used with- or without 10 µg/mL bFGF neutralizing antibodies or 10 µg/mL of recombinant human Tie-2/Fc (both from R&D systems, UK). Unconditioned basal medium or basal medium supplemented with 200 ng/mL bFGF and 20 ng/mL angiopoietin-1 (both from Peprotech, NJ), were used as controls. The number of branching points were manually scored and the cumulative length of sprouts was analyzed using ImageJ version 1.41.

statistical analysis

All data are expressed as mean ± standard error of mean (SEM). For multiple comparisons testing, one-way ANOVA followed by Tukey post hoc analyses were performed. Probabilities of P < 0.05 were considered to be statistically significant.

results

characterization of endothelial outgrowth cells (uc-eoc)

The first UC-EOC colonies were observed 9-21 days after plating the initial MNC fraction (Figure 1A) and the culture plates reached confluence within the next 5-8 days (Figure 1B). Cells were cultured up to an additional three passages after which EC characteristics were determined by binding of fluorescein-conjugated lectin from Ulex europaeus and uptake of DiI-acetylated low density lipoprotein (DiI-acLDL) (Figures 1C and D, respectively). The ability of UC-EOC to form capillary-like sprouts on Matrigel was also confirmed (Figure 1E). An important functional parameter of EC is their ability to prevent thrombin formation. Indeed, UC-EOC showed anti-thrombogenic function

standard protocols. Blots were blocked for one hour in Tris-buffered saline (TBS) containing 5% bovine serum albumin (BSA) and incubated at room temperature in TBS-Tween with antibodies to human pSMAD2 (0.3 µg/mL; Millipore, MA), SMAD2/3 (0.3 µg/mL), pSMAD1/5/8 (0.2 µg/mL; both Cell Signaling, MA), SMAD1/5/8 (1 µg/mL), Id2 (1 µg/mL), Id3 (1 µg/mL; all Santa Cruz Biotechnology, CA), or GAPDH (1 µg/mL; AbD Serotec, UK) overnight. Subsequently, blots were washed and incubated with alkaline phosphatase-conjugated antibodies to mouse or rabbit IgG (0.1 µg/mL). NBT/BCIP (Bio-Rad, VA) was used as substrate for detection. Densitometric analysis was performed using ImageJ version 1.41 (Research Services Branch, National Institute of Mental Health, Bethesda, MD).

gene transcript analysis

RNA isolation was performed using the RNeasy Mini Kit (Qiagen Inc., CA) according to manufacturer’s protocol. Subsequently, 1 μg of total RNA was reverse transcribed using the FirstStrand cDNA synthesis kit (Fermentas UAB, Lithuania) according to manufacturer’s instructions. The cDNA-equivalent of 5 ng RNA was used for amplification in 384-well microtiter plates in a TaqMan ABI7900HT cycler (Applied Biosystems, CA) in a final reaction volume of 10 μL containing 5 μL TaqMan universal PCR Master Mix (Applied Biosystems, CA) and 0.5 μL primer/probe mix. Applied Biosystems ‘assay on demand’ primer/probe sets were used to detect amplimers of β-2-Microglobulin (β2M; Hs99999907_m1), SM22α (Hs00162558_m1), Calponin (Hs00154543_m1), ALK1 (Hs00163543_m1), ALK5 (Hs00610319_m1), Collagen type I (Hs00164004_m1), Collagen type 3 (Hs00164103_m1), angiopoietin-1 (Hs00181613_m1), CCL2 (Hs00234140_m1), EGF (Hs00153181_m1), BFGF (Hs00266645_m1), HGF (Hs00300159_m1), IGF-1 (Hs00153126_m1) and VEGFa (Hs00173626_m1). Cycle threshold (CT) values for individual reactions were determined using ABI Prism SDS 2.2 data processing software (Applied Biosystems, CA). To determine differences in expression, CT-values were normalized against β2M-expression using the ΔCT-method (ΔCT(gene) = CT(gene) – CT(β2M)). To correct for interassay variance, ΔCT values were normalized against expression levels of an external calibrator (ΔΔCT(gene) = ΔCT(gene) – ΔCT(calibrator)). Relative expression levels were calculated as 2-(ΔΔCT). All cDNA samples were amplified in triplicate. Differences of a two-fold higher or greater were considered to be biologically relevant.

telomerase expression & activity

Gene expression of Telomerase Reverse Transcriptase (hTERT) was analyzed by RT-PCR. Total RNA was isolated and reverse transcribed as described above. RT-PCR for hTERT (sense 5’-TGGATGATTTCTTGTTGGTG-3’; antisense 5’-TCTTCCAAACT TGCTGATGA-3’) and GAPDH (sense 5’-CTGCCGTCTAGAAAAACCTG-3’; antisense 5’-GTCC AGGGGTCTTACTCCTT-3’) were performed in a final reaction volume of 25 μL under the following reaction conditions of 0.25 mM dNTPs, 1.5 mM MgCl2, 1μM primer-mix, and 1U Taq DNA Polymerase. Amplification was performed on a MyCycler (Bio-Rad, VA) in 96-well plates for 30 cycles. Amplimers were separated by gel electrophoresis in a 2% agarose gel.

chapter 5

88

endothelial progenitor cells give rise to smooth muscle progeny

89

5

comparable to human umbilical vein endothelial cells (HUVEC) in an in vitro Thrombin Generation Assay (Figure 1F).

UC-EOC phenotype was further characterized by flow cytometric analysis. Marker proteins of the endothelial cell lineage, namely CD31 (99.68 ± 0.25%), CD144 (94.84 ± 1.31%), VEGFR-2 (97.26 ± 2.06%), eNOS (99.08 ± 0.28%), vWF (99.21 ± 0.75%), thrombomodulin (83.73 ± 9.50%) and Tie-2 (90.77 ± 0.85%) were abundantly expressed by UC-EOC. In contrast, marker proteins of the mesenchymal cell lineage, namely αSMA (0,82 ± 0,70%), SM22α (2.78 ± 2.00%), SM-MHC2 (3.36 ± 2.13%) and calponin (0.05 ± 0.03%), were virtually not expressed by UC-EOC. UC-EOC were further analyzed for the expression CD34 (18.84 ± 1.63%) and for the expression of TGFβ-related growth factor receptors. Abundant expression of ALK1 (99.77 ± 0.06%), CD105 (99.90 ± 0.05%) and TGFβ-R2 (78.12 ± 1.42%) was observed on UC-EOC. In contrast, expression levels of ALK5 (2.50 ± 0.05%) and PDGF-Rb (4.86 ± 1.92%) were virtually absent (Figure 1).

transdifferentiation of uc-eoc to smooth muscle cells

UC-EOC were cultured up to passage 4 in endothelial medium, after which they were cultured in mesenchymal differentiation medium containing TGF-β1 and PDGF-BB, for an additional 21 days. Transdifferentiated cells (UC-EnMT) had strong proliferative capacity and classical hill and valley morphology, combined with typical cytoskeletal organization, as shown by staining with phallotoxin (Figures 2A & B). EnMT also resulted in changes in functional properties, i.e. gain of contractile phenotype and loss of anti-thrombogenicity (Figure 1F). Cells were embedded in collagen type I gels and cultured for 24 hours after which a spontaneous contraction of 30,1 ± 5,9% could be observed by the collagen-embedded cells. In contrast, cell-free controls and the gels loaded with UC-EOC did not show contraction. Addition of TGF-β1 (5 ng/ml) after 24 hours showed additional contraction of the transdifferentiatied cell-embedded collagen gels to a total of 72.0 ± 2.3% of control, likely due to TGF-β1 induced Rho activation [171], whereas this had no effect on control gels (Figure 2C).

The expression of endothelial lineage markers CD144 (4.06 ± 1.83%), eNOS (0.10 ± 0.10%), vWF (0.50 ± 0.45%), thrombomodulin (4.79 ± 0.35%) and Tie-2 (3.54 ± 1.96%) had diminished after transdifferentiation (Figure 2C). About 32% of transdifferentiated cells (UC-EnMT) still showed CD31 expression, although the molecular density had

TCP

HUVEC

UC-EOCEnMT

0

100

200

300*

Thro

mbi

n Co

ncen

trat

ion

(mU

/mL)

2.78 ± 2.00%SM22

3.36 ± 2.13%SM-MHC2

0.05 ± 0.03%Calponin

0.82 ± 0.70%SMA

99.68 ± 0.25%CD31

18.84 ± 1.63%CD34

94.84 ± 1.31%CD144

97.26 ± 2.06%VEGFR-2

99.08 ± 0.28%eNOS

99.21 ± 0.75%von Willebrand Factor

83.73 ± 9.50%Thrombomodulin

90.77 ± 0.85%Tie-2

99.90 ± 0.05%CD105

99.77 ± 0.06%ALK1

2.50 ± 0.05%ALK5

78.12 ± 1.42%TGFß-R2

4.86 ± 1.92%PDGF-Rb

Cell

Coun

ts

Fluorescence Intensity (log)

A B C D

E F

G

Figure 1. Characterization of umbilical cord blood-derived endothelial outgrowth cells (UC-EOC). A, Cord blood mononuclear cells were cultured in endothelial outgrowth medium. After 9-21 days the first outgrowth colonies could be observed. B, UC-EOC cultures reached confluency within the next 5-8 days. C, Binding of fluorescein-conjugated Ulex europaeus lectin by UC-EOC was confirmed. D, UC-EOC took up DiI-acetylated low density lipoprotein. E, UC-EOC formed capillary-like sprouts on Matrigel. F, Anti-thrombogenic function of UC-EOC was assessed by measuring their inhibition of thrombin generation. Anti-thrombogenicity of UC-EOC was comparable to that of human umbilical vein endothelial cell (HUVEC) controls. Transdifferentiated cells showed loss of anti-thrombogenic properties. G, Flow cytometric analysis of UC-EOC phenotype. Endothelial markers (CD31 = platelet endothelial cell adhesion molecule-1, CD144 = vascular endothelial cadherin, VEGFR-2 = vascular endothelial growth factor receptor-2, eNOS = endothelial nitric oxide synthase, von Willebrand Factor, Thrombomodulin and TEK tyrosine kinase Tie-2) were abundantly expressed. Mesenchymal markers (αSMA = alpha smooth muscle actin, SM22α = smooth muscle protein 22 alpha, SM-MHC2 = smooth muscle myosin heavy chain 2 and calponin) were virtually absent. High expression levels were found for transforming growth factor beta (TGFβ) related growth factor receptors, ALK1 = activin like kinase 1, CD105 = endoglin and TGFβ-R2 = TGFβ receptor type 2. In contrast to expression of ALK5 = activin like kinase 5 and PDGF-Rb = platelet derived growth factor receptor b. TCP = Tissue culture plate, EnMT = Transdifferentiated UC-EOC, * P < 0,05.

chapter 5

90

endothelial progenitor cells give rise to smooth muscle progeny

91

5

decreased by 50-fold. The expression of mesenchymal lineage markers αSMA (96.77 ± 1.43%), SM22α (98.78 ± 1.01%), SM-MHC2 (96.30 ± 2.18%) and calponin (90.27 ± 7.09%) was strongly induced during EnMT (Figure 2C). After 21 days of EnMT, TGF-β1 and PDGF-BB, were omitted from the culture media. UC-EnMT were cultured up to an additional 7 passages and maintained their mesenchymal phenotype and proliferative behavior. The expression of endothelial lineage marker CD31 was fully lost after this additional culture period (data not shown). The expression of ALK1 (87.63 ± 1.20%) was reduced after EnMT, while the expression of ALK5 (89.08 ± 6.08%) had increased, resulting in an shift in ALK1/ALK5 balance (Figure 2C). Furthermore, EnMT resulted in increased expression of PDGF-Rb (38,59 ± 6,42%).

transdifferentiation of uc-eoc is mediated by tgf-β1.

EnMT was induced by stimulating UC-EOC with TGF-β1 for 96h. SMAD signaling through type 1 TGF-β receptors, ALK1 and ALK5, was analyzed by immunoblotting of the receptor regulated SMADs, SMAD1/5/8 and SMAD2/3. Relative gene expression levels of ALK1 and ALK5, and of SMAD-target genes were determined.

UC-EnMT had lower gene transcript levels of ALK1, compared to uninduced UC-EOC, after 96h of TGF-β1 stimulation (Figure 3A). ALK1 is involved in activation of the SMAD1/5/8 route through phosphorylation of SMAD1/5/8. In contrast to signaling via ALK5, another type 1 TGF-β-receptor, which leads activation of the SMAD2/3 route [172]. ALK5 expression increased after TGF-β1 stimulation and completely diminished with addition of its kinase activity inhibitor, SB431542, indicative for a positive feedback system (Figure 3A).

UC-EOC cultured in MDM for 96 hours showed reduced levels of SMAD2/3 and SMAD1/5/8 protein compared to unstimulated controls. The addition of the ALK5 kinase inhibitor SB431542 had no effect on basal levels of SMAD2/3 and SMAD1/5/8, but resulted in lower levels of pSMAD2 (Figures 3B & C). Expression levels of pSMAD1/5/8 decreased after TGF-β1 stimulation and expression levels were comparable with UC-EnMT. Supplementing MDM with SB431542, had no effect on pSMAD1/5/8 levels, which implies an ALK5-independent effect. (Figures 3B & C).

Inhibitor-of-DNA-binding-proteins (Id proteins) are dominant negative regulators of basic helix-loop-helix transcriptional regulators which play a role in lineage commitment, cell cycle control and cell differentiation [173]. Expression of Id genes is mediated via phosphorylation of SMAD1/5/8 which bind to Smad responsive elements (SREs) within the Id promotors, thereby activating transcription. TGF-β1, via SMAD2/3, is known to inhibit expression of Id genes via by activation of transcriptional repressor ATF3, which binds to ATF/CREB site on the Id promotors and represses transcription [174]. Id2 and Id3 are known to antagonize SMAD2/3 signaling by repressing pSMAD2 mediated gene transcription [175]. Ectopic expression of Id2 and Id3 has been shown to inhibit transdifferentiation of epithelial cells [176].

Analysis of Id2 and Id3 protein expression showed stable levels of Id2, in contrast to Id3, which showed strongly reduced levels after stimulation with TGF-β1. This effect was not inhibited by addition of SB431542 (Figures 3B & C).

Gene transcript levels of SM22α and calponin increased after 96h of culture in MDM and reached levels similar to UC-EnMT (Figure 3C). SM22α and calponin both are target

Cell

Coun

ts

Fluorescence Intensity (log)

0 12 24 36 480

20

40

60

80

No CellsUC-EOCUC-EnMT

TGF-ß

Time (Hours)

Gel

Are

a (m

m2 )

S

I

UC-EOC

T= 0h T= 48h

UC-EnMTA B C D

E

SM22 SM-MHC2 CalponinSMA96.77 ± 1.44% 98.78 ± 1.01% 96.30 ± 2.18% 90.27 ± 7.09%

CD31 CD34 CD144 VEGFR-231.65 ± 5.57% 5.25 ± 0.39% 4.06 ± 1.83% 77.47 ± 2.86%

eNOS von Willebrand Factor Thrombomodulin Tie-20.10 ± 0.05% 0.50 ± 0.45% 4.79 ± 0.35% 3.54 ± 1.96%

CD105ALK1 ALK5 TGFß-R2 PDGF-Rb97.31 ± 0.53%87.62 ± 1.20% 89.08 ± 6.08% 79.46 ± 5.29% 38.60 ± 6.42%

Figure 2. Characterization of transdifferentiated cells (UC-EnMT). UC-EOC were cultured in mesenchymal differentiation medium (MDM) containing TGF-β1 and PDGF-BB, for 21 days after which TGF-β1 and PDGF-BB were ommitted from the culture media and cells were cultured up to an additional 7 passages. A, UC-EnMT showed classical hill and valley morphology, characteristical of vascular smooth muscle cells. B, Staining with fluorescein-conjugated phallotoxin, showed typical cytoskeletal organization. C&D, UC-EnMT were embedded in collagen type 1 gels and cultured for 24h after which spontaneous contraction (30.1 ± 5.9%) of the UC-EnMT embedded gels could be seen. In contrast to the UC-EOC embedded gels or the cell-free controls which did not show contraction. After 24h, TGF-β1 (5 ng/ml) was added to the cultures. This induced additional contraction of the UC-EnMT-embedded collagen gels (to a total of 72.0 ± 2.3%). E, Flow cytometric analysis of UC-EnMT after 21 days of culture in MDM. Expression of endothelial markers (upper row) had diminished after transdifferentiation. Mesenchymal markers (αSMA, SM22α, SM-MHC2 and calponin) were strongly induced after endothelial-to-mesenchymal transdifferentiation. Expression of ALK1 was reduced after EnMT, while the expression levels of ALK5 had increased compared to UC-EOC controls (Figure 1G). Endoglin and TGFβ-R2 expression levels were virtually unchanged after transdifferentiation, whereas expression of PDGF-Rb had increased.

chapter 5

92

endothelial progenitor cells give rise to smooth muscle progeny

93

5

genes of pSMAD2. However their increased expression is more likely due to reduced levels of Id3, which, as stated above, acts as a repressor of pSMAD2 mediated gene transcription. Inhibition of ALK5 kinase activity with SB431542 decreased pSMAD2 expression and completely abolished SM22α and calponin gene transcript expression (Figure 3C). Gene expression levels of collagen type I were low in cells stimulated with TGF-β1 for 96 hours compared to expression levels in UC-EnMT. Contradictory, gene transcription of collagen type III increased after 96h of culture with TGF-β1. Transcripts of collagen type I and III genes were low compared to UC-EnMT, and diminished by the addition of SB431542.

uc-enmt show increased responsiveness to tgf-β1

UC-EOC and UC-EnMT were stimulated with 50 ng/ml of TGF-β1 for 1 hour with or without addition of SB431542. Immunoblotting and immunofluorescent imaging was used to study the expression of inactive, and phosphorylated SMAD2/3 and SMAD1/5/8. Stimulation of UC-EOC and UC-EnMT with TGF-β1 had no effect on basal SMAD2/3 and SMAD1/5/8 levels (figures 4A & B), while pSMAD1/5/8 levels were clearly reduced in UC-EnMT compared to UC-EOC. Expression of pSMAD2 increased in TGF-β1-stimulated UC-EnMT, and completely diminished with addition of SB431542. UC-EOC did not show increased pSMAD2 levels after stimulation with TGF-β1 (Figures 4A & B). Differential response to TGF-β1 by UC-EOC and UC-EnMT is likely due to increased expression of ALK5 in UC-EnMT which leads to increased ALK5 kinase activity and thereby increased phosphorylation of SMAD2. Another possible explanation may be found in the observed high molecular weight SMAD2/3 complexes present in transdifferentiated cells (Figure 4A). Pretreatment of the protein extracts with 2M NaCl or 8M urea failed to dissociate these complexes (data not shown).

loss of telomerase activity of uc-eoc with increasing population doublings and after trans-differentiation.

Gene expression of Telomerase Reverse Transcriptase (hTERT) was analyzed by RT-PCR. UC-EOC at low population doublings showed a readily detectable gene expression of hTERT, indicating that these cells are capable of self-renewal (Figure 5A). Subsequently, activity of telomerase was determined in UC-EOC and UC-EnMT. Telomerase activity of UC-EOC at low population doubling numbers was similar to that of hTERT-transfected HUVEC controls (Figures 5B & C). Telomerase activity reduced with increased population doublings, which indicates that UC-EOC give rise to terminally differentiated progeny. Also after EnMT, telomerase activity was lost (data not shown).

pro-angiogenic effects of uc-enmt by paracrine signaling.

To study potential paracrine effects of UC-EnMT on UC-EOC, we analyzed gene transcript levels of several pro-angiogenic factors. Expression levels of HGF, IGF-1 and EGF were low in both UC-EOC and UC-EnMT. Expression levels of VEGFa were similar in

Calponin

0.00

0.05

0.10

0.15

0.20

0.25Collagen type I

0

2

4

6

8

ß2M

Collagen type III

0.0

0.5

1.5

ß2M

A ALK1

0.0

0.1

0.2

0.3

0.4

Rela

tive

Gen

e Ex

pres

sion

(nor

mal

ized

to ß

2M)

ALK5

0.00

0.01

0.02

0.03

0.04

ND ND ND

SB431542TGFß1 +

+-- - -

-+ ++

-- - -

-+

0

2

4

6

8

ß2M

Rela

tive

Gen

e Ex

pres

sion

(nor

mal

ized

to ß

2M)

SM22α

SB431542TGFß1 +

+-- - -

-+ ++

-- - -

-+ ++

-- - -

-+ ++

-- - -

-+

UC-EOC UC-EnMT

ND ND ND

D***

******

****

*

**

C

Arbi

trar

y un

its

SMAD1/5/8 0

20

40

60

80

100pSMAD2/3 pSMAD1/5/8

SMAD2/3 0

20

40

60

80

100

0

20

40

60

80

100

0

20

40

60

80

100

Arbi

trar

y un

its

*

*

0

20

40

60

80

100 Id2

Arbi

trar

y un

its

0

20

40

60

80

100Id3

*

pSMAD1/5/8

SMAD1/5/8

GAPDH

pSMAD2

SMAD2/3complex

SMAD2SMAD3

UC-EOC UC-EnMT

SB431542TGFß1 +

+-- - -

-+

Id2

Id3

B

Figure 3. Transdifferentiation of UC-ECFC is mediated by TGF-β1. Endothelial-to-mesenchymal transdifferentiation was induced by stimulating UC-ECFC with TGF-β1 for 96h. Activin like kinase 5 (ALK5) kinase inhibitor SB431542 was added to block ALK5 mediated TGFβ signaling. A, Real-time RT-PCR showed increased gene transcript expression levels of ALK5 after stimulation with TGF-β1. Addition of SB431542 completely inhibited this effect. B, Representative images of immunoblotting of TGFβ signaling components SMAD1/5/8, SMAD2/3, their phosphorylated forms, pSMAD1/5/8 and pSMAD2/3 and pSMAD1/5/8 regulated Id2 and Id3. C, Densitometric analysis of SMAD immunoblotting. Basal levels of SMAD1/5/8 were reduced after stimulation with TGF-β1 to levels comparable to those of transdifferentiated cells (EnMT). Phosphorylation of SMAD1/5/8 also decreased after TGF-β1 stimulation. SB431542 did not inhibit these effects. Id2 and Id3 are known to antagonize SMAD2/3 signaling by repressing pSMAD2 mediated gene transcription. Id2 levels were stable, in contrast to Id3, which showed strongly reduced levels after stimulation with TGF-β1. D, Quantitative RT-PCR showed upregulation of SM22α, calponin and collagen type III. This was completely blocked by SB431542. Expression of collagen type I remained low after 96h of TGF-β1 stimulation compared to levels of transdifferentiated cells, ***P<0.001, *P<0.05.

chapter 5

94

endothelial progenitor cells give rise to smooth muscle progeny

95

5UC-EOC and UC-EnMT. Interestingly, gene transcript levels of bFGF and angiopoietin-1 were strongly increased in UC-EnMT compared to UC-EOC. In contrast to angiopoietin-1 expression, the increase of bFGF expression was induced within 96 hours of TGF-β1 stimulation (Figure 6A).

SDF-1 and MCP-1, are angiogenic factors which play an important role in recruitment and differentiation of EPC [102;177-179]. UC-EnMT expressed high levels of SDF-1, which were virtually absent in UC-EOC, despite TGF-β1 stimulation for 96 hours. MCP-1 expression, however, was induced by short term TGF-β1 stimulation and almost absent in (quiescent) UC-EnMT.

UC-EnMT conditioned medium (CM) was used to study the paracrine effects of UC-EnMT on capillary sprouting of UC-EOC. Given the increased gene transcript levels of bFGF and angiopoietin-1 found in UC-EnMT, neutralizing antibodies to bFGF and soluble Tie-2 (sTie-2) were added to CM. The total number of branching points was comparable between basal medium (BM), BM with bFGF and angiopoietin-1, and CM (46 ± 8, 52 ± 7 and 57 ± 3 respectively). Neutralization of bFGF or addition of sTie-2 to the culture media, resulted in strongly reduced numbers of branching points (Figures 6B & C). The cumulative sprout length was also determined. CM showed increased cumulative length (45.63 ± 1.18%) compared to unconditioned basal medium. BM supplemented with bFGF and angiopoietin-1 showed a similar increase in cumulative sprout length (42.66 ± 4.04%). Addition of bFGF neutralizing antibodies or sTie-2 completely diminished the observed increase, indicating a bFGF and angiopoietin-1 mediated effect (Figures 6B & C).

discussion

In the present study we show the intrinsic capacity of umbilical cord blood-derived endothelial outgrowth cells (UC-EOC) to transdifferentiate into smooth muscle cells; (1) Endothelial-to-mesenchymal transdifferentiation (EnMT) of EOC resulted in waning

pSMAD1/5/8

pSMAD1/5/8

SMAD1/5/8

GAPDH

pSMAD2

SMAD2/3 SMAD1/5/8

pSMAD2

SMAD2/3 complex

SMAD2SMAD3

0

Arbi

trar

y un

its

20

40

60

80

100

0

Arbi

trar

y un

its20

40

60

80

100

SB431542TGFß1 -

--- - -

+++

++

+ --

-- - -

+++

++

+SB431542TGFß1 -

--- - -

+++

++

+

UC-EOCUC-EnMT

--

+- + +

-++

+-

-

A B***

***

pSM

AD

2SM

AD

2/3

pSM

AD

1/5/

8SM

AD

1/5/

8

ECFC EnMT ECFC EnMTC

Figure 4. Transdifferentiated cells (UC-EnMT) show increased responsiveness to TGF-β1. UC-EOC and UC-EnMT were stimulated with a pulse of 50 ng/ml TGF-β1 for 1h. ALK5 kinase inhibitor SB431542 was added to discriminate ALK5 mediated effects of TGF-β1. Immunoblotting was used to study the expression of basal and phosphorylated SMAD1/5/8 and SMAD2/3. A, Representative images of immunoblotting of TGF-β-signaling components SMAD1/5/8, SMAD2/3 and their phosphorylated forms, pSMAD1/5/8 and pSMAD2/3. Note the formation of high molecular weight (110 kDa) SMAD2/3 complexes, specifically in the transdifferentiated cells. B, Densitometric analysis of SMAD immunoblotting. Stimulation of UC-EOC and UC-EnMT with TGF-β1 had no effect on basal SMAD2/3 and SMAD1/5/8 expression. Levels of pSMAD1/5/8 were reduced in UC-EnMT compared to UC-EOC, irrespective of TGF-β1 stimulation or ALK5 kinase inhibition. TGF-β1 stimulation induced increased expression of pSMAD2 in transdifferentiated cells, which was completely blocked by SB431542. UC-EOC did not show increased pSMAD2 levels after stimulation with TGF-β1. C, Immunofluorescent images showing typical cytoskeletal organization in UC-EnMT by staining with Fluorescein-conjugated phallotoxins (green). Unphosphorylated SMAD2/3 and SMAD1/5/8 show cytoplasmic staining patterns (red) while their phosphorylated forms (red) show typical nuclear (blue) localization. , *** p < 0.001.

CMV-hTERT T

PD < 5

PD 10-15

PD > 200

25

50

75

100

Telo

mer

ase

Activ

ity(%

of C

MV-

hTER

T)

UC-EOC

CMV-hTERT T

UC-EOC T

CMV-hTERT

PD < 5

HUVEC

PD > 20

UC-EOC

GAPDH

hTERTA

B

C

Figure 5. Loss of telomerase activity of UC-ECFC with increasing population doublings. A, Representative image of Telomerase Reverse Transcriptase (hTERT) gene transcript analysis. UC-ECFC at low populations doublings show hTERT gene expression, in contrast to UC-ECFC at higher population doublings. B, Telomerase activity analysis clearly shows high telomerase activity in UC-ECFC at low population doublings, comparable to activity levels of hTERT-transfected human umbilical vein endothelial cell (HUVEC) controls (CMV-hTERT). Telomerase activity decreased with increasing population doublings.

chapter 5

96

endothelial progenitor cells give rise to smooth muscle progeny

97

5

of endothelial markers and loss of endothelial functionality. (2) Mesenchymal marker expression and contractile function were gained. (3) EnMT is mediated by TGF-β1 and PDGF-BB and inhibition of ALK5 kinase activity completely abolished EnMT by the inhibition of SMAD2 phosphorylation. (4) Transdifferentiated cells (UC-EnMT) were phenotypically stable, self-sustaining and showed pro-angiogenic paracrine properties. To our knowledge, this study is the first to describe the plasticity of UC-EOC towards smooth muscle cell differentiation.

Since their first description [46], endothelial progenitor cells (EPC) have been extensively studied and their availablity and angiogenic properties have advocated them an interesting and promising cell source for therapeutic neovascularization [180]. Despite this large research effort, plasticity of EPC has received little attention.

Embryonic vascular progenitor cells have been described to differentiate to endothelial and smooth muscle cells [151]. Besides, EnMT plays a physiological role during development in formation of the heart valves [143;161], and has been described postnatally, both in vivo and in vitro [144;155;162-165]. Furthermore, mesenchymal-to-endothelial transdifferentiation has been reported [167;168]. This plasticity between endothelial and mesenchymal lineages is suggestive for a common vascular progenitor.

In vivo, bone marrow-derived smooth muscle progenitor cells (SMPC) have been shown to play a role in cardiovascular pathology [141;181]. SMPC have been cultured from umbilical cord and adult blood, although their origin remains largely unclear [139;182]. Our results indicate that EOC, the archetype in vitro EPC [170], can give rise to smooth muscle progeny.

Since our data concern in vitro EPC, extrapolation to the in vivo situation is somewhat speculative. One could however hypothesize that EPC fulfill a role in replenishing pools of progenitor cells present in the vascular wall as described by Zengin et al.[183]. These cells would require the capacity to give rise to endothelial and smooth muscle progeny. The fact that bone marrow-derived EPC have been described to be recruited to the vessel wall, rather than to the endothelium itself [184] adds to the hypothesis. Also, Purhonen and co-workers recently confirmed these findings by describing that EPC did not contribute to the vascular endothelium, but were found perivascularly [185].

The capacity of EOC to transdifferentiate to smooth muscle cells certainly has implications for their therapeutic use in cardiovascular disease. The majority of patients eligible for cardiovascular cell therapy commonly share an inflammatory vascular profile [186]. TGF-β and it’s downstream effector pSMAD2 have been shown to be highly expressed at atherosclerotic lesion sites [187]. As our data show, increased TGFβ signaling results in mesenchymal differentiation of EPC. This potentially has beneficial effects by limiting plaque formation and increasing plaque stability [188], but it could also contribute to atherogenesis [181]. Co-administration of smooth muscle progenitor cells and EPC has recently been shown to have synergetic angiogenic effects by increased paracrine signaling, mainly through the angiopoietin/Tie2 signaling pathway [189]. Our results also show angiopoietin 1 mediated pro-angiogenic effects of transdifferentiated EOC, in addition to bFGF mediated effects. EOC EnMT thereby provide a novel therapeutic strategy for treating ischemic disease. Ex vivo priming of EOC has recently been described to enhance their angiogenic potential [179]. Our

A

UC-EOC UC-EnMT

Rela

tive

Gen

e Ex

pres

sion

(nor

mal

ized

to ß

2M)

0.0000

0.0005

0.0010

0.0015

0.00

0.01

0.02

0.03

0.04

0.05

VEGFa

0.00

0.02

0.04

0.06

0.08

bFGF

0.00000

0.00005

0.00010

0.00015

0.00020

0.00025

HGF IGF-1

0.0000

0.0001

0.0002

0.0003

0.0004

0.0005EGF

0.00

0.02

0.04

0.06Angiopoietin-1

0.00

0.05

0.10

0.15

0.20

0.25MCP-1

Rela

tive

Gen

e Ex

pres

sion

(nor

mal

ized

to ß

2M)

SB431542TGFß1 +

+-- - -

-+ ++

-- - -

-+0.00

0.02

0.04

0.06SDF-1

++

-- - -

-+ ++

-- - -

-+

B

C

CM CM +sTie2 CM +bFGF NAbs BM++

BM CM

CM +sTie2

CM +bFGF NAbs

BM++0

6

12

18

24

30

0

15

30

45

60

75

BM CM

CM +sTie2

CM +bFGF NAbs

BM++

Bran

chin

g po

ints

Spro

ut le

ngth

(mm

)

**** *

** * ** *

**** *

** *** *

Figure 6. Pro-angiogenic effects of UC-EnMT by paracrine signaling. A, Quantitative RT-PCR analysis of gene transcript levels of pro-angiogenic factors normalized to β2M expression. Gene transcript levels of basic fibroblast growth factor (bFGF) and angiopoietin-1 were strongly increased in UC-EnMT compared to UC-EOC. The increase of bFGF expression was induced within 96 hours of TGF-β1 stimulation. Capillary sprouting capacity of UC-EOC was studied using UC-EnMT conditioned medium (CM) with or without bFGF neutralizing antibodies (bFGF NAbs) or recombinant human Tie-2/Fc (sTie2). Unconditioned basal medium (BM) or BM supplemented with 200 ng/mL bFGF and 20ng/mL angiopoietin-1 (BM++ )were used as controls. The total numbers of branching points were determined and the cumulative lengths of spouts were analyzed. B, Representative images of capillary sprout formation on Matrigel. Note the reduced capillary formation in sTie2 and bFGF NAbs treated cultures. C, The total number of branching points was similar between BM, BM++, and CM. Numbers were reduced with addition of bFGF NAbs or sTie2 to CM. The cumulative sprout length was increased with CM compared to unconditioned BM, similar to that observed with BM++. Addition of bFGF NAbs or sTie2 completely diminished these effects.

chapter 5

98

results illustrate how ex vivo stimulation can indeed direct EOC differentiation, thereby changing phenotypical and functional properties of the cells. This phenomenon can be used in tuning EOC, creating tailor-made therapy for use in a specific patient with specific underlying pathology and co-morbidity.

conclusion

In conclusion, our study demonstrates that EOC are capable of TGF-β1-mediated EnMT, thereby giving rise to smooth muscle progeny. This plasticity of EOC implies a common vascular progenitor status for EOC and clearly has implications for therapeutical use.

appendices

206

references

207 App

references

vaartjes i, peters rJg, van dis s, bots m. hart- en vaatziekten in nederland. in: vaartjes i, peters rJg, van dis s, bots m, editors. hart- 1.

en vaatziekten in nederland, najaar 2006 - cijfers over ziekte en sterfte. den haag: nederlandse hartstichting, 2006. p. 9-18.

eefting f, rensing b, wigman J, pannekoek wJ, liu wm, cramer mJ, lips dJ, doevendans pa. role of apoptosis in reperfusion injury. 2.

cardiovasc res 2004; 61: 414-426.

boyle em, Jr., pohlman th, Johnson mc, verrier ed. endothelial cell injury in cardiovascular surgery: the systemic inflammatory 3.

response. ann thorac surg 1997; 63: 277-284.

costa c, soares r, schmitt f. angiogenesis: now and then. apmis 2004; 112: 402-412.4.

carmeliet p. mechanisms of angiogenesis and arteriogenesis. nat med 2000; 6: 389-395.5.

harkness ml, harkness rd, mcdonald da. the collagen and elastin content of the arterial wall. J physiol 1955; 127: 33-4p.6.

deyl z, Jelinek J, macek K, chaldakov g, vankov vn. collagen and elastin synthesis in the aorta of spontaneously hypertensive rats. 7.

blood vessels 1987; 24: 313-320.

gudiene d, valanciute a, velavicius J. collagen network changes in basilar artery in aging. medicina (Kaunas) 2007; 43: 964-970.8.

Jenkins c, milsted a, doane K, meszaros g, toot J, ely d. a cell culture model using rat coronary artery adventitial fibroblasts to 9.

measure collagen production. bmc cardiovasc disord 2007; 7: 13.

walker-caprioglio hm, trotter Ja, mercure J, little sa, mcguffee lJ. organization of rat mesenteric artery after removal of cells of 10.

extracellular matrix components. cell tissue res 1991; 264: 63-77.

frid mg, shekhonin bv, Koteliansky ve, glukhova ma. phenotypic changes of human smooth muscle cells during development: late 11.

expression of heavy caldesmon and calponin. dev biol 1992; 153: 185-193.

bennett mr, farnell l, gibson wg. a Quantitative description of the contraction of blood vessels following the release of 12.

noradrenaline from sympathetic varicosities. J theor biol 2005; 234: 107-122.

poliseno l, cecchettini a, mariani l, evangelista m, ricci f, giorgi f, citti l, rainaldi g. resting smooth muscle cells as a model for 13.

studying vascular cell activation. tissue cell 2006; 38: 111-120.

matsuki t, hynes mr, duling br. comparison of conduit vessel and resistance vessel reactivity: influence of intimal permeability. 14.

am J physiol 1993; 264: h1251-h1258.

bardin n, anfosso f, masse J, cramer e, sabatier f, bivic a, sampol J, dignat-george f. identification of cd146 as a component of the 15.

endothelial Junction involved in the control of cell-cell cohesion. blood 2001; 98: 3677-3684.

Komarova ya, mehta d, malik ab. dual regulation of endothelial Junctional permeability. sci stKe 2007; 2007: re8.16.

orlova vv, chavakis t. regulation of vascular endothelial permeability by Junctional adhesion molecules (Jam). thromb haemost 17.

2007; 98: 327-332.

bogatcheva nv, verin ad. the role of cytoskeleton in the regulation of vascular endothelial barrier function. microvasc res 2008; 18.

76: 202-207.

rao rm, yang l, garcia-cardena g, luscinskas fw. endothelial-dependent mechanisms of leukocyte recruitment to the vascular 19.

wall. circ res 2007; 101: 234-247.

salmi m, Koskinen K, henttinen t, elima K, Jalkanen s. clever-1 mediates lymphocyte transmigration through vascular and 20.

lymphatic endothelium. blood 2004; 104: 3849-3857.

lee tJ, shirasaki y, nickols ga. altered endothelial modulation of vascular tone in aging and hypertension. blood vessels 1987; 24: 21.

132-136.

busse r, trogisch g, bassenge e. the role of endothelium in the control of vascular tone. basic res cardiol 1985; 80: 475-490.22.

highsmith rf, pang dc, rapoport rm. endothelial cell-derived vasoconstrictors: mechanisms of action in vascular smooth muscle. 23.

J cardiovasc pharmacol 1989; 13 suppl 5: s36-s44.

stalcup sa, davidson d, mellins rb. endothelial cell functions in the hemodynamic responses to stress. ann ny acad sci 1982; 401: 24.

117-131.

minami t, sugiyama a, wu sQ, abid r, Kodama t, aird wc. thrombin and phenotypic modulation of the endothelium. arterioscler 25.

thromb vasc biol 2004; 24: 41-53.

weintraub ws. the pathophysiology and burden of restenosis. am J cardiol 2007; 100: s3-s9.26.

abbott wm, megerman J, hasson Je, l’italien g, warnock df. effect of compliance mismatch on vascular graft patency. J vasc surg 27.

1987; 5: 376-382.

salacinski hJ, goldner s, giudiceandrea a, hamilton g, seifalian a, edwards a, carson r. the mechanical behavior of vascular grafts: 28.

a review. J biomater appl 2001; 15: 241-278.

Kon zn, white c, Kwon mh, Judy J, brown en, gu J, burris ns, laird pc, brown t, brazio ps, gammie J, brown J, griffith bp, poston 29.

rs. the role of preexisting pathology in the development of neointimal hyperplasia in coronary artery bypass grafts. J surg res

2007; 142: 351-356.

panetta tf, marin ml, veith fJ, goldsmith J, gordon re, Jones am, schwartz ml, gupta sK, wengerter Kr. unsuspected preexisting 30.

saphenous vein disease: an unrecognized cause of vein bypass failure. J vasc surg 1992; 15: 102-110.

autieri mv. allograft-induced proliferation of vascular smooth muscle cells: potential targets for treating transplant vasculopathy. 31.

curr vasc pharmacol 2003; 1: 1-9.

herring m, gardner a, glover J. a single-staged technique for seeding vascular grafts with autogenous endothelium. surgery 1978; 32.

84: 498-504.

herring mb, dilley r, Jersild ra, Jr., boxer l, gardner a, glover J. seeding arterial prostheses with vascular endothelium. the nature 33.

of the lining. ann surg 1979; 190: 84-90.

weinberg cb, bell e. a blood vessel model constructed from collagen and cultured vascular cells. science 1986; 231: 397-400.34.

weinberg cb, bell e. regulation of proliferation of bovine aortic endothelial cells, smooth muscle cells, and adventitial fibroblasts 35.

in collagen lattices. J cell physiol 1985; 122: 410-414.

niklason le, gao J, abbott wm, hirschi KK, houser s, marini r, langer r. functional arteries grown 36. in Vitro. science 1999; 284:

489-493.

solan a, mitchell s, moses ma, niklason le. effect of pulse rate on collagen deposition in the tissue-engineered blood vessel. tissue 37.

eng 2003; 9: 579-586.

l’heureux n, paquet s, labbe r, germain l, auger f. a completely biological tissue-engineered human blood vessel. faseb J 1998; 38.

12: 47-56.

l’heureux n, stoclet J, auger fa, lagaud g, germain l, andriantsitohaina r. a human tissue-engineered vascular media: a new 39.

model for pharmacological studies of contractile responses. faseb J 2001; 15: 515-524.

hersel u, dahmen c, Kessler h. rgd modified polymers: biomaterials for stimulated cell adhesion and beyond. biomaterials 2003; 40.

24: 4385-4415.

petrie t, capadona J, reyes c, garcia a. integrin specificity and enhanced cellular activities associated with surfaces presenting a 41.

recombinant fibronectin fragment compared to rgd supports. biomaterials 2006; 27: 5459-5470.

laflamme K, roberge cJ, grenier g, rémy-zolghadri m, pouliot s, baker K, labbe r, orleans-Juste p, auger fa, germain l. adventitia 42.

contribution in vascular tone: insights from adventitia-derived cells in a tissue-engineered human blood vessel. faseb J 2006;

20: 1245-1247.

huynh t, abraham g, murray J, brockbank K, hagen po, sullivan s. remodeling of an acellular collagen graft into a physiologically 43.

responsive neovessel. nat biotech 1999; 17: 1083-1086.

bouïs d, hospers ga, meijer c, molema g, mulder nh. endothelium 44. in Vitro: a review of human vascular endothelial cell lines for

blood vessel-related research. angiogenesis 2001; 4: 91-102.

unterluggauer h, hütter e, voglauer r, grillari J, vöth m, bereiter-hahn J, Jansen-dürr p, Jendrach m. identification of cultivation-45.

independent markers of human endothelial cell senescence in Vitro. biogerontology 2007; 8: 383-397.

asahara t, murohara t, sullivan a, silver m, vanderzee r, li t, witzenbichler b, schatteman g, isner Jm. isolation of putative 46.

progenitor endothelial cells for angiogenesis. science 1997; 275: 964-967.

ingram da, mead le, tanaka h, meade v, fenoglio a, mortell K, pollok K, ferkowicz m, gilley d, yoder m. identification of a novel 47.

hierarchy of endothelial progenitor cells utilizing human peripheral and umbilical cord blood. blood 2004; 104: 2752-2760.

colton cK. implantable biohybrid artificial organs. cell transplant 1995; 4: 415-436.48.

appendices

208

references

209 App

Kawamoto a, asahara t, losordo dw. transplantation of endothelial progenitor cells for therapeutic neovascularization. cardiovasc 71.

radiat med 2002; 3: 221-225.

tamaki t, uchiyama y, okada y, ishikawa t, sato m, akatsuka a, asahara t. functional recovery of damaged skeletal muscle through 72.

synchronized vasculogenesis, myogenesis, and neurogenesis by muscle-derived stem cells. circulation 2005; 112: 2857-2866.

rehman J, li Jl, orschell cm, march Kl. peripheral blood “endothelial progenitor cells” are derived from monocyte/macrophages 73.

and secrete angiogenic growth factors. circulation 2003; 107: 1164-1169.

rohde e, malischnik c, thaler d, maierhofer t, linkesch w, lanzer g, guelly c, strunk d. blood monocytes mimic endothelial 74.

progenitor cells. stem cells 2006; 24: 357-367.

romagnani p, annunziato f, liotta f, lazzeri e, mazzinghi b, frosali f, cosmi l, maggi l, lasagni l, scheffold a, Kruger m, dimmeler 75.

s, marra f, gensini g, maggi e, romagnani s. cd14+cd34low cells with stem cell phenotypic and functional features are the major

source of circulating endothelial progenitors. circ res 2005; 97: 314-322.

schmeisser a, garlichs cd, zhang h, eskafi s, graffy c, ludwig J, strasser rh, daniel wg. monocytes coexpress endothelial and 76.

macrophagocytic lineage markers and form cord-like structures in matrigel (r) under angiogenic conditions. cardiovasc res 2001;

49: 671-680.

dankers pyw, van beek dJm, ten cate at, sijbesma rp, meijer ew. novel biocompatible supramolecular materials for tissue 77.

engineering. polymeric mater 2003; 88: 52-53.

dankers pyw, harmsen mc, brouwer la, van luyn mJa, meijer ew. a modular and supramolecular approach to bioactive scaffolds 78.

for tissue engineering. nat mater 2005; 4: 568-574.

heijkants rg, van calck rv, de groot Jh, pennings aJ, schouten aJ, van tienen tg, ramrattan n, buma p, veth rp. design, synthesis 79.

and properties of a degradable polyurethane scaffold for meniscus regeneration. J mater sci mater med 2004; 15: 423-427.

hynes ro. a reevaluation of integrins as regulators of angiogenesis. nat med 2002; 8: 918-921.80.

de fougerolles ar, Koteliansky ve. regulation of monocyte gene expression by the extracellular matrix and its functional 81.

implications. immunol rev 2002; 186: 208-220.

folmer bJb, sijbesma rp, versteegen rm, van der rijt JaJ, meijer ew. supramolecular polymer materials: chain extension of telechelic 82.

polymers using a reactive hydrogen-bonding synthon. adv mater 2000; 12: 874-878.

davis lg, dibner md, battey Jf. optical density analytical measurements. in: davis lg, dibner md, battey Jf, editors. basic methods 83.

in molecular biology. new york: elsevier, 1986. p. 327-328.

heijkants r. ph.d. thesis: polyurethane scaffold as meniscus reconstruction materials. rijksuniversiteit groningen, the netherlands, 84.

2004.

li t, hamano K, nishida m, hayashi m, ito h, mikamo a, matsuzaki m. cd11785. + stem cells play a Key role in therapeutic angiogenesis

induced by bone marrow cell implantation. am J physiol heart circ physiol 2003; 285: h931-h937.

Quirici n, soligo d, caneva l, servida f, bossolasco p, deliliers g. differentiation and expansion of endothelial cells from human bone 86.

marrow cd133+ cells. br J haematol 2001; 115: 186-194.

fujiyama s, amano K, uehira K, yoshida m, nishiwaki y, nozawa y, Jin d, takai s, miyazaki m, egashira K, imada t, iwasaka t, 87.

matsubara h. bone marrow monocyte lineage cells adhere on injured endothelium in a monocyte chemoattractant protein-1-

dependent manner and accelerate reendothelialization as endothelial progenitor cells. circ res 2003; 93: 980-989.

fernandez pujol b, lucibello fc, zuzarte m, lutjens p, muller r, havemann K. dendritic cells derived from peripheral monocytes 88.

express endothelial markers and in the presence of angiogenic growth factors differentiate into endothelial-like cells. eur J cell

biol 2001; 80: 99-110.

fernandez pujol b, lucibello fc, gehling um, lindemann K, weidner n, zuzarte ml, adamkiewicz J, elsasser hp, muller r, havemann 89.

K. endothelial-like cells derived from human cd14 positive monocytes. differentiation 2000; 65: 287-300.

zhao y, glesne d, huberman e. a human peripheral blood monocyte-derived subset acts as pluripotent stem cells. proc natl acad 90.

sci u s a 2003; 100: 2426-2431.

zhao y, mazzone t. human umbilical cord blood-derived f-macrophages retain pluripotentiality after thrombopoietin expansion. 91.

exp cell res 2005; 310: 311-318.

zumstein a, mathieu o, howald h, hoppeler h. morphometric analysis of the capillary supply in skeletal muscles of trained and 49.

untrained subjects - its limitations in muscle biopsies. pflügers archiv european Journal of physiology 1983; 397: 277-283.

lopez JJ, laham rJ, stamler a, pearlman Jd, bunting s, Kaplan a, carrozza Jp, sellke fw, simons m. vegf administration in chronic 50.

myocardial ischemia in pigs. cardiovasc res 1998; 40: 272-281.

sato K, wu t, laham rJ, Johnson rb, douglas p, li J, sellke fw, bunting s, simons m, post mJ. efficacy of intracoronary or intravenous 51.

vegf165 in a pig model of chronic myocardial ischemia. J am coll cardiol 2001; 37: 616-623.

hariawala md, horowitz Jr, esakof d, sheriff dd, walter dh, Keyt b, isner Jm, symes Jf. vegf improves myocardial blood flow but 52.

produces edrf-mediated hypotension in porcine hearts. J surg res 1996; 63: 77-82.

rabinovsky e, draghia-akli r. insulin-like growth factor i plasmid therapy promotes 53. in Vivo angiogenesis. mol ther 2004; 9:

46-55.

mack ca, magovern cJ, budenbender Kt, patel sr, schwarz ea, zanzonico p, ferris b, sanborn t, isom p, ferris b, sanborn t, isom 54.

ow, crystal rg, rosengart tK. salvage angiogenesis induced by adenovirus-mediated gene transfer of vascular endothelial growth

factor protects against ischemic vascular occlusion. J vasc surg 1998; 27: 699-709.

choi J, hur J, yoon c, Kim J, lee c, youn s, oh i, skurk c, murohara t, park y, walsh K, Kim h. augmentation of therapeutic angiogenesis 55.

using genetically-modified human endothelial progenitor cells with altered glycogen synthase Kinase-3beta activity. J biol chem

2004; 279: 49430-49438.

lu y, shansky J, del tatto m, ferland p, wang x, vandenburgh h. recombinant vascular endothelial growth factor secreted from 56.

tissue-engineered bioartificial muscles promotes localized angiogenesis. circulation 2001; 104: 594-599.

sakamoto t, spee c, scuric z, gordon em, hinton dr, anderson wf, ryan sJ. ability of retroviral transduction to modify the 57.

angiogenic characteristics of rpe cells. graefes arch clin exp ophthalmol 1998; 236: 220-229.

lee rJ, springer ml, blanco-bose we, shaw r, ursell pc, blau hm. vegf gene delivery to myocardium: deleterious effects of 58.

unregulated expression. circulation 2000; 102: 898-901.

sheridan mh, shea ld, peters mc, mooney dJ. bioabsorbable polymer scaffolds for tissue engineering capable of sustained growth 59.

factor delivery. J control release 2000; 64: 91-102.

wu x, rabkin-aikawa e, guleserian K, perry t, masuda y, sutherland f, schoen f, mayer Je, Jr., bischoff J. tissue-engineered 60.

microvessels on three-dimensional biodegradable scaffolds using human endothelial progenitor cells. am J physiol heart circ

physiol 2004; 287: h480-h487.

Kalka c, masuda h, takahashi t, Kalka-moll w, silver m, Kearney m, li t, isner Jm, asahara t. transplantation of ex vivo expanded 61.

endothelial progenitor cells for therapeutic neovascularization. proc natl acad sci u s a 2000; 97: 3422-3427.

hur J, yoon c, Kim h, choi J, Kang h, hwang K, oh bh, lee m, park y. characterization of two types of endothelial progenitor cells and 62.

their different contributions to neovasculogenesis. arterioscler thromb vasc biol 2004; 24: 288-293.

yoder m, mead le, prater d, Krier t, mroueh K, li f, Krasich r, temm c, prchal J, ingram da. redefining endothelial progenitor cells 63.

via clonal analysis and hematopoietic stem/progenitor cell principals. blood 2007; 109: 1801-1809.

awad o, dedkov e, Jiao c, bloomer s, tomanek r, schatteman g. differential healing activities of cd3464. + and cd14+ endothelial cell

progenitors. arterioscler thromb vasc biol 2006; 26: 758-764.

heil m, ziegelhoeffer t, mees b, schaper w. a different outlook on the role of bone marrow stem cells in vascular growth - bone 65.

marrow delivers software not hardware. circ res 2004; 94: 573-574.

sayers rd, raptis s, berce m, miller Jh. long-term results of femorotibial bypass with vein or polytetrafluoroethylene. br J surg 66.

1998; 85: 934-938.

meinhart Jg, schense Jc, schima h, gorlitzer m, hubbell Ja, deutsch m, zilla p. enhanced endothelial cell retention on shear-stressed 67.

synthetic vascular grafts precoated with rgd-cross-linked fibrin. tissue eng 2005; 11: 887-895.

unger re, peters K, wolf m, motta a, migliaresi c, Kirkpatrick cJ. endothelialization of a non-woven silk fibroin net for use in tissue 68.

engineering: growth and gene regulation of human endothelial cells. biomaterials 2004; 25: 5137-5146.

conklin bs, wu h, lin ph, lumsden ab, chen c. basic fibroblast growth factor coating and endothelial cell seeding of a decellularized 69.

heparin-coated vascular graft. artif organs 2004; 28: 668-675.

asahara t, Kawamoto a. endothelial progenitor cells for postnatal vasculogenesis. am J physiol cell physiol 2004; 287: c572-c579.70.

appendices

210

references

211 App

national Kidney foundation. K/doQi clinical practice guidelines for chronic Kidney disease: evaluation, classification, and 112.

stratification. am J Kidney dis 2002; 39: s1-266.

Krenning g, van der strate bwa, schipper m, gallego y van seijen xJ, fernandes b, van luyn mJa, harmsen mc. cd34113. + cells augment

endothelial cell differentiation of cd14+ endothelial progenitor cells in Vitro. J cell mol med 2008; in press.

sturiale a, coppolino g, loddo s, criseo m, campo s, crasci e, bolignano d, nostro l, teti d, buemi m. effects of haemodialysis on 114.

circulating endothelial progenitor cell count. blood purif 2007; 25: 242-251.

freedman mh, cattran dc, saunders ef. anemia of chronic renal failure: inhibition of erythropoiesis by uremic serum. nephron 115.

1983; 35: 15-19.

hotta t, maeda h, suzuki i, chung tg, saito a. selective inhibition of erythropoiesis by sera from patients with chronic renal failure. 116.

proc soc exp biol med 1987; 186: 47-51.

westerweel pe, hoefer ie, blankestijn pJ, de bree p, groeneveld d, van oostrom o, braam b, Koomans ha, verhaar mc. end-stage 117.

renal disease causes an imbalance between endothelial and smooth muscle progenitor cells. am J physiol renal physiol 2007; 292:

f1132-f1140.

de groot K, hermann bahlmann f, sowa J, Koenig J, menne J, haller h, fliser d. uremia causes endothelial progenitor cell deficiency. 118.

Kidney int 2004; 66: 641-646.

coppolino g, bolignano d, campo s, loddo s, teti d, buemi m. circulating progenitor cells after cold pressor test in hypertensive and 119.

uremic patients. hypertension research 2008; 31: 717-724.

stinghen ae, goncalves sm, martines eg, nakao ls, riella mc, aita ca, pecoits-filho r. increased plasma and endothelial cell 120.

expression of chemokines and adhesion molecules in chronic Kidney disease. nephron clin pract 2009; 111: c117-c126.

Jacobson sh, egberg n, hylander b, lundahl J. correlation between soluble markers of endothelial dysfunction in patients with 121.

renal failure. am J nephrol 2002; 22: 42-47.

umland o, heine h, miehe m, marienfeld K, staubach K, ulmer a. induction of various immune modulatory molecules in cd34122. +

hematopoietic cells. J leukoc biol 2004; 75: 671-679.

vasa m, fichtlscherer s, adler K, aicher a, martin h, zeiher a, dimmeler s. increase in circulating endothelial progenitor cells by 123.

statin therapy in patients with stable coronary artery disease. circulation 2001; 103: 2885-2890.

walter d, rittig K, bahlmann fh, Kirchmair r, silver m, murayama t, nishimura h, losordo dw, asahara t, isner Jm. statin therapy 124.

accelerates reendothelialization: a novel effect involving mobilization and incorporation of bone marrow-derived endothelial

progenitor cells. circulation 2002; 105: 3017-3024.

bahlmann fh, degroot K, duckert t, niemczyk e, bahlmann e, boehm sm, haller h, fliser d. endothelial progenitor cell proliferation 125.

and differentiation is regulated by erythropoietin. Kidney int 2003; 64: 1648-1652.

shepherd J, Kastelein JJp, bittner v, deedwania p, breazna a, dobson s, wilson dJ, zuckerman a, wenger nK, for the treating to 126.

new targets investigators. effect of intensive lipid lowering with atorvastatin on renal function in patients with coronary heart

disease: the treating to new targets (tnt) study. clin J am soc nephrol 2007; 2: 1131-1139.

shepherd J, Kastelein JJp, bittner v, deedwania p, breazna a, dobson s, wilson dJ, zuckerman a, wenger nK. intensive lipid lowering 127.

with atorvastatin in patients with coronary heart disease and chronic Kidney disease: the tnt (treating to new targets) study. J

am coll cardiol 2008; 51: 1448-1454.

shepherd J, barter p, carmena r, deedwania p, fruchart Jc, haffner s, hsia J, breazna a, larosa J, grundy s, waters d, for the treating 128.

to new targets investigators. effect of lowering ldl cholesterol substantially below currently recommended levels in patients

with coronary heart disease and diabetes: the treating to new targets (tnt) study. diabetes care 2006; 29: 1220-1226.

wattanakit K, cushman m, stehman-breen c, heckbert sr, folsom ar. chronic Kidney disease increases risk for venous 129.

thromboembolism. J am soc nephrol 2008; 19: 135-140.

mahmoodi bK, ten Kate mK, waanders f, veeger nJ, brouwer Jl, vogt l, navis g, van der mJ. high absolute risks and predictors of 130.

venous and arterial thromboembolic events in patients with nephrotic syndrome: results from a large retrospective cohort study.

circulation 2008; 117: 224-230.

stenvinkel p, ekstrom tJ. epigenetics and the uremic phenotype: a matter of balance. contrib nephrol 2008; 161: 55-62.131.

stenvinkel p, ekstrom tJ. does the uremic milieu affect the epigenotype? J ren nutr 2009; 19: 82-85.132.

christenson em, dadsetan m, wiggins m, anderson Jm, hiltner a. poly(carbonate urethane) and poly(ether urethane) biodegradation: 92.

in Vivo studies. J biomed mater res a 2004; 69a: 407-416.

matheson la, santerre Jp, labow rs. changes in macrophage function and morphology due to biomedical polyurethane surfaces 93.

undergoing biodegradation. J cell physiol 2004; 199: 8-19.

danen ehJ, sonneveld p, brakebusch c, fassler r, sonnenberg a. the fibronectin-binding integrins a5b1 and avb3 differentially 94.

modulate rhoa-gtp loading, organization of cell matrix adhesions, and fibronectin fibrillogenesis. J cell biol 2002; 159:

1071-1086.

eliceiri b, cheresh d. the role of av integrins during angiogenesis: insights into potential mechanisms of action and clinical 95.

development. J clin invest 1999; 103: 1227-1230.

sijbesma rp, meijer ew. Quadruple hydrogen bonded systems. chem commun 2003; 5-16.96.

dankers pyw, van leeuwen enm, van gemert gml, spiering aJh, harmsen mc, brouwer la, Janssen hm, bosman aw, van luyn 97.

mJa, meijer ew. chemical and biological properties of supramolecular polymer systems based on oligocaprolactones. biomaterials

2006; 27: 5490-5501.

werner n, nickenig g. endothelial progenitor cells in health and atherosclerotic disease. ann med 2007; 39: 82-90.98.

dimmeler s, zeiher am. vascular repair by circulating endothelial progenitor cells: the missing link in atherosclerosis? J mol med 99.

2004; 82: 671-677.

popa er, harmsen mc, tio ra, van der strate bwa, brouwer la, schipper m, Koerts J, de Jongste mJl, hazenberg a, hendriks m, 100.

van luyn mJa. circulating cd34+ progenitor cells modulate host angiogenesis and inflammation in Vivo. J mol cell cardiol 2006;

41: 86-96.

Krenning g, dankers pyw, Jovanovic d, van luyn mJa, harmsen mc. efficient differentiation of cd14101. + monocytic cells into endothelial

cells on degradable biomaterials. biomaterials 2007; 28: 1470-1479.

Krenning g, van luyn mJa, harmsen mc. endothelial progenitor cell-based neovascularization: implications for therapy. trends 102.

mol med 2009; in press.

mall J, philipp aw, rademacher a, paulitschke m, buttemeyer r. re-endothelialization of punctured eptfe graft: an 103. in Vitro study

under pulsed perfusion conditions. nephrol dial transplant 2004; 19: 61-67.

Kaushal s, amiel ge, guleserian KJ, shapira om, perry t, sutherland fw, rabkin e, moran am, schoen fJ, atala a, soker s, bischoff 104.

J, mayer Je, Jr. functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. nat med 2001;

7: 1035-1040.

Krenning g, moonen JraJ, van luyn mJa, harmsen mc. generating new blood flow: integrating developmental biology and tissue 105.

engineering. trends cardiovasc med 2009; in press.

sarnak mJ, levey as, schoolwerth ac, coresh J, culleton b, hamm ll, mccullough pa, Kasiske bl, Kelepouris e, Klag mJ, parfrey p, 106.

pfeffer m, raij l, spinosa dJ, wilson pw. Kidney disease as a risk factor for development of cardiovascular disease: a statement from

the american heart association councils on Kidney in cardiovascular disease, high blood pressure research, clinical cardiology, and

epidemiology and prevention. circulation 2003; 108: 2154-2169.

shishehbor mh, oliveira lpJ, lauer ms, sprecher dl, wolski K, cho l, hoogwerf bJ, hazen sl. emerging cardiovascular risk factors 107.

that account for a significant portion of attributable mortality risk in chronic Kidney disease. am J cardiol 2008; 101: 1741-1746.

moonen JraJ, de leeuw K, van seijen x, Kallenberg c, van luyn mJa, bijl m, harmsen mc. reduced number and impaired function of 108.

circulating progenitor cells in patients with systemic lupus erythematosus. arthritis res ther 2007; 9: r84.

valgimigli m, rigolin gm, fucili a, porta md, soukhomovskaia o, malagutti p, bugli am, bragotti lz, francolini g, mauro e, castoldi 109.

g, ferrari r. cd34+ and endothelial progenitor cells in patients with various degrees of congestive heart failure. circulation 2004;

110: 1209-1212.

choi Jh, Kim Kl, huh w, Kim b, byun J, suh w, sung J, Jeon es, oh hy, Kim dK. decreased number and impaired angiogenic function 110.

of endothelial progenitor cells in patients with chronic renal failure. arterioscler thromb vasc biol 2004; 24: 1246-1252.

rodriguez-ayala e, yao Q, holmen c, lindholm b, sumitran-holgersson s, stenvinkel p. imbalance between detached circulating 111.

endothelial cells and endothelial progenitor cells in chronic Kidney disease. blood purif 2006; 24: 196-202.

appendices

212

references

213 App

ishisaki a, hayashi h, li aJ, imamura t. human umbilical vein endothelium-derived cells retain potential to differentiate into 155.

smooth muscle-like cells. J biol chem 2003; 278: 1303-1309.

lancrin c, sroczynska p, stephenson c, allen t, Kouskoff v, lacaud g. the haemangioblast generates haematopoietic cells through 156.

a haemogenic endothelium stage. nature 2009; 457: 892-895.

hellstrom m, Kalen m, lindahl p, abramsson a, betsholtz c. role of pdgf-b and pdgfr-b in recruitment of vascular smooth muscle 157.

cells and pericytes during embryonic blood vessel formation in the mouse. development 1999; 126: 3047-3055.

hirschi KK, rohovsky sa, d’amore pa. pdgf, tgf-b, and heterotypic cell-cell interactions mediate endothelial cell-induced 158.

recruitment of 10t1/2 cells and their differentiation to a smooth muscle fate. J cell biol 1998; 141: 805-814.

yamashita J, itoh h, hirashima m, ogawa m, nishikawa si, yurugi t, naito m, nakao K, nishikawa si. flk1-positive cells derived from 159.

embryonic stem cells serve as vascular progenitors. nature 2000; 408: 92-96.

ferreira ls, gerecht s, shieh h, watson n, rupnick m, dallabrida s, vunjak-novakovic g, langer r. vascular progenitor cells isolated 160.

from human embryonic stem cells give rise to endothelial and smooth muscle like cells and form vascular networks In Vivo. circ

res 2007; 101: 286-294.

markwald rr, fitzharris tp, manasek fJ. structural development of endocardial cushions. am J anat 1977; 148: 85-119.161.

arciniegas e, sutton ab, allen td, schor am. transforming growth factor-b1 promotes the differentiation of endothelial cells into 162.

smooth muscle-like cells in Vitro. J cell sci 1992; 103 ( pt 2): 521-529.

deissler h, deissler h, lang gK, lang ge. tgfb induces transdifferentiation of ibrec to asma-expressing cells. int J mol med 2006; 163.

18: 577-582.

frid mg, Kale v, stenmark K. mature vascular endothelium can give rise to smooth muscle cells via endothelial-mesenchymal 164.

transdifferentiation: In Vitro analysis. circ res 2002; 90: 1189-1196.

Krenning g, moonen JraJ, van luyn mJa, harmsen mc. vascular smooth muscle cells for use in vascular tissue engineering obtained 165.

by endothelial-to-mesenchymal transdifferentiation (enmt) on collagen matrices. biomaterials 2008; 29: 3703-3711.

zeisberg em, tarnavski o, zeisberg m, dorfman al, mcmullen Jr, gustafsson e, chandraker a, yuan x, pu wt, roberts ab, neilson eg, 166.

sayegh mh, izumo s, Kalluri r. endothelial-to-mesenchymal transition contributes to cardiac fibrosis. nat med 2007; 13: 952-961.

oswald J, boxberger s, Jorgensen b, feldmann s, ehninger g, bornhauser m, werner c. mesenchymal stem cells can be differentiated 167.

into endothelial cells In Vitro. stem cells 2004; 22: 377-384.

wang h, riha gm, yan s, li m, chai h, yang h, yao Q, chen c. shear stress induces endothelial differentiation from a murine embryonic 168.

mesenchymal progenitor cell line. arterioscler thromb vasc biol 2005; 25: 1817-1823.

popa er, van der strate bwa, brouwer la, tadema h, schipper m, fernandes b, hendriks m, van luyn mJa, harmsen mc. dependence 169.

of neovascularization mechanisms on the molecular microenvironment. tissue eng 2007; 13: 2913-2921.

hirschi KK, ingram da, yoder mc. assessing identity, phenotype, and fate of endothelial progenitor cells. arterioscler thromb vasc 170.

biol 2008; 28: 1584-1595.

fukata y, amano m, Kaibuchi K. rho-rho-Kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-171.

muscle cells. trends pharmacol sci 2001; 22: 32-39.

goumans mJ, valdimarsdottir g, itoh s, lebrin f, larsson J, mummery cl, Karlsson s, ten dp. activin receptor-like Kinase (alK)1 is 172.

an antagonistic mediator of lateral tgfb/alK5 signaling. mol cell 2003; 12: 817-828.

norton Jd. id helix-loop-helix proteins in cell growth, differentiation and tumorigenesis. J cell sci 2000; 113 ( pt 22): 3897-3905.173.

ruzinova mb, benezra r. id proteins in development, cell cycle and cancer. trends cell biol 2003; 13: 410-418.174.

saika s, ikeda K, yamanaka o, flanders Kc, ohnishi y, nakajima y, muragaki y, ooshima a. adenoviral gene transfer of bmp-7, id2, 175.

or id3 suppresses injury-induced epithelial-to-mesenchymal transition of lens epithelium in mice. am J physiol cell physiol 2006;

290: c282-c289.

Kowanetz m, valcourt u, bergstrom r, heldin ch, moustakas a. id2 and id3 define the potency of cell proliferation and differentiation 176.

responses to transforming growth factor beta and bone morphogenetic protein. mol cell biol 2004; 24: 4241-4254.

arras m, ito wd, scholz d, winkler b, schaper J, schaper w. monocyte activation in angiogenesis and collateral growth in the rabbit 177.

hindlimb. J clin invest 1998; 101: 40-50.

dahl sl, Koh J, prabhakar v, niklason le. decellularized native and engineered arterial scaffolds for transplantation. cell transplant 133.

2003; 12: 659-666.

wu h, wang t, Kang p, tsuang y, sun J, lin f. coculture of endothelial and smooth muscle cells on a collagen membrane in the 134.

development of a small-diameter vascular graft. biomaterials 2007; 28: 1385-1392.

matsuda t. recent progress of vascular graft engineering in Japan. artif organs 2004; 28: 64-71.135.

rezai n, podor tJ, mcmanus bm. bone marrow cells in the repair and modulation of heart and blood vessels: emerging opportunities 136.

in native and engineered tissue and biomechanical materials. artif organs 2004; 28: 142-151.

melero-martin J, Khan z, picard a, wu x, paruchuri s, bischoff J. 137. In Vivo vasculogenic potential of human blood-derived endothelial

progenitor cells. blood 2007; 109: 4761-4768.

schmidt d, mol a, neuenschwander s, breymann c, gossi m, zund g, turina m, hoerstrup sp. living patches engineered from human 138.

umbilical cord derived fibroblasts and endothelial progenitor cells. eur J cardiothorac surg 2005; 27: 795-799.

simper d, stalboerger p, panetta c, wang s, caplice n. smooth muscle progenitor cells in human blood. circulation 2002; 106: 139.

1199-1204.

sugiyama s, Kugiyama K, nakamura s, Kataoka K, aikawa m, shimizu K, Koide s, mitchell r, ogawa h, libby p. characterization of 140.

smooth muscle-like cells in circulating human peripheral blood. atherosclerosis 2006; 187: 351-362.

liu c, nath K, Katusic z, caplice n. smooth muscle progenitor cells in vascular disease. trends cardiovasc med 2004; 14: 288-293.141.

Kashiwakura y, Katoh y, tamayose K, Konishi h, takaya n, yuhara s, yamada m, sugimoto K, daida h. isolation of bone marrow stromal 142.

cell-derived smooth muscle cells by a human sm22a promoter: In Vitro differentiation of putative smooth muscle progenitor cells of

bone marrow. circulation 2003; 107: 2078-2081.

mercado-pimentel me, hubbard ad, runyan rb. endoglin and alk5 regulate epithelial-mesenchymal transformation during cardiac 143.

valve formation. dev biol 2007; 304: 420-432.

paranya g, vineberg s, dvorin e, Kaushal s, roth s, rabkin e, schoen fJ, bischoff J. aortic valve endothelial cells undergo transforming 144.

growth factor-b-mediated and non-transforming growth factor-b-mediated transdifferentiation in Vitro. am J pathol 2001; 159:

1335-1343.

noseda m, mclean g, niessen K, chang l, pollet i, montpetit r, shahidi r, dorovini-zis K, li l, beckstead b, durand re, hoodless 145.

pa, Karsan a. notch activation results in phenotypic and functional changes consistent with endothelial-to-mesenchymal

transformation. circ res 2004; 94: 910-917.

burgess wh, mehlman t, friesel r, Johnson wv, maciag t. multiple forms of endothelial cell growth factor. rapid isolation and 146.

biological and chemical characterization. J biol chem 1985; 260: 11389-11392.

van oeveren w, haan J, lagerman p, schoen t. comparison of coagulation activity tests 147. in Vitro for selected biomaterials. artif

organs 2002; 26: 506-511.

Kakisis J, liapis c, breuer c, sumpio b. artificial blood vessel: the holy grail of peripheral vascular surgery. J vasc surg 2005; 41: 148.

349-354.

Koike n, fukumura d, gralla o, au p, schechner J, Jain rK. tissue engineering: creation of long-lasting blood vessels. nature 2004; 149.

428: 138-139.

zeisberg em, potenta s, xie l, zeisberg m, Kalluri r. discovery of endothelial to mesenchymal transition as a source for carcinoma-150.

associated fibroblasts. cancer res 2007; 67: 10123-10128.

deruiter mc, poelmann re, vanmunsteren Jc, mironov v, markwald rr, gittenberger-de groot ac. embryonic endothelial cells 151.

transdifferentiate into mesenchymal cells expressing smooth muscle actins in Vivo and in Vitro. circ res 1997; 80: 444-451.

fernandes dJ, ravenhall ce, harris t, tran t, vlahos r, stewart ag. contribution of the p38mapK signalling pathway to proliferation 152.

in human cultured airway smooth muscle cells is mitogen-specific. br J pharmacol 2004; 142: 1182-1190.

ferrari g, pintucci g, seghezzi g, hyman Km, galloway ac, mignatti p. vegf, a prosurvival factor, acts in concert with tgf-b1 to 153.

induce endothelial cell apoptosis. proc natl acad sci u s a 2006; 103: 17260-17265.

hyman Km, seghezzi g, pintucci g, stellari g, Kim Jh, grossi ea, galloway ac, mignatti p. transforming growth factor-β1 induces 154.

apoptosis in vascular endothelial cells by activation of mitogen-activated protein Kinase. surgery 2002; 132: 173-179.

appendices

214

references

215 App

igreja c, fragoso r, caiado f, clode n, henriques a, camargo l, reis em, dias s. detailed molecular characterization of cord blood-198.

derived endothelial progenitors. exp hematol 2008; 36: 193.

zentilin l, taturo s, zacchigna s, arsic n, pattarini l, sinigaglia m, giacca m. bone marrow mononuclear cells are recruited to the 199.

sites of vegf-induced neovascularization but are not incorporated into the newly formed vessels. blood 2006; 107: 3546-3554.

botta r, gao e, stassi g, bonci d, pelosi e, zwas d, patti m, colonna l, baiocchi m, coppola s, ma x, condorelli g, peschle c. heart 200.

infarct in nod-scid mice: therapeutic vasculogenesis by transplantation of human cd34+ cells and low dose cd34+Kdr+ cells.

faseb J 2004; 18: 1392-1394.

tei K, matsumoto t, mifune y, ishida K, sasaki K, shoji t, Kubo s, Kawamoto a, asahara t, Kurosaka m, Kuroda r. administrations 201.

of peripheral blood cd34-positive cells contribute to medial collateral ligament healing via vasculogenesis. stem cells 2008;

2007-0671.

yoon c, hur J, park K, Kim J, lee c, oh i, Kim t, cho h, Kang h, chae i, yang h, oh bh, park y, Kim h. synergistic neovascularization by 202.

mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial cells: the role of angiogenic cytokines

and matrix metalloproteinases. circulation 2005; 112: 1618-1627.

o’riordan e, mendelev n, patschan s, patschan d, eskander J, cohen-gould l, chander p, goligorsky ms. chronic nos inhibition 203.

actuates endothelial-mesenchymal transformation. am J physiol heart circ physiol 2007; 292: h285-h294.

arciniegas e, frid mg, douglas is, stenmark Kr. perspectives on endothelial-to-mesenchymal transition: potential contribution to 204.

vascular remodeling in chronic pulmonary hypertension. am J physiol lung cell mol physiol 2007; 293: l1-l8.

sakao s, taraseviciene-stewart l, cool c, tada y, Kasahara y, Kurosu K, tanabe n, takiguchi y, tatsumi K, Kuriyama t, voelkel n. 205.

vegf-r blockade causes endothelial cell apoptosis, expansion of surviving cd34+ precursor cells and transdifferentiation to smooth

muscle-like and neuronal-like cells. faseb J 2007; 21: 3640-3652.

Krug el, mjaatvedt ch, markwald rr. extracellular matrix from embryonic myocardium elicits an early morphogenetic event in 206.

cardiac endothelial differentiation. dev biol 1987; 120: 348-355.

taipale J, Keski-oja J. growth factors in the extracellular matrix. faseb J 1997; 11: 51-59.207.

duong td, erickson ca. mmp-2 plays an essential role in producing epithelial-mesenchymal transformations in the avian embryo. 208.

dev dyn 2004; 229: 42-53.

song w, Jackson K, mcguire pg. degradation of type iv collagen by matrix metalloproteinases is an important step in the epithelial-209.

mesenchymal transformation of the endocardial cushions. dev biol 2000; 227: 606-617.

ishihara h, Kubota h, lindberg rl, leppert d, gloor sm, errede m, virgintino d, fontana a, yonekawa y, frei K. endothelial cell barrier 210.

impairment induced by glioblastomas and transforming growth factor β2 involves matrix metalloproteinases and tight Junction

proteins. J neuropathol exp neurol 2008; 67: 435-448.

reddy K, mangale ss. integrin receptors: the dynamic modulators of endometrial function. tissue cell 2003; 35: 260-273.211.

frank r, adelmann-grill bc, herrmann K, haustein uf, petri Jb, heckmann m. transforming growth factor-b controls cell-matrix 212.

interaction of microvascular dermal endothelial cells by downregulation of integrin expression. J invest dermatol 1996; 106:

36-41.

lee yh, Kayyali us, sousa am, rajan t, lechleider rJ, day rm. transforming growth factor-β1 effects on endothelial monolayer 213.

permeability involve focal adhesion Kinase/src. am J respir cell mol biol 2007; 37: 485-493.

olsson aK, dimberg a, Kreuger J, claesson-welsh l. vegf receptor signalling - in control of vascular function. nat rev mol cell biol 214.

2006; 7: 359-371.

lampugnani m, orsenigo f, gagliani mc, tacchetti c, dejana e. vascular endothelial cadherin controls vegfr-2 internalization and 215.

signaling from intracellular compartments. J cell biol 2006; 174: 593-604.

calera mr, venkatakrishnan a, Kazlauskas a. ve-cadherin increases the half-life of vegf receptor 2. exp cell res 2004; 300: 216.

248-256.

ha ch, bennett am, Jin zg. a novel role of vascular endothelial cadherin in modulating c-src activation and downstream signaling 217.

of vascular endothelial growth factor. J biol chem 2008; 283: 7261-7270.

zhang xf, groopman Je, wang Jf. extracellular matrix regulates endothelial functions through interaction of vegfr-3 and integrin 218.

a5b1. J cell physiol 2005; 202: 205-214.

de falco e, porcelli d, torella ar, straino s, iachininoto mg, orlandi a, truffa s, biglioli p, napolitano m, capogrossi mc, pesce m. 178.

sdf-1 involvement in endothelial phenotype and ischemia-induced recruitment of bone marrow progenitor cells. blood 2004; 104:

3472-3482.

zemani f, silvestre Js, fauvel-lafeve f, bruel a, vilar J, bieche i, laurendeau i, galy-fauroux i, fischer am, boisson-vidal c. ex vivo 179.

priming of endothelial progenitor cells with sdf-1 before transplantation could increase their proangiogenic potential. arterioscler

thromb vasc biol 2008; 28: 644-650.

rafii s, lyden d. therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. nat med 2003; 180.

9: 702-712.

caplice n, bunch tJ, stalboerger p, wang s, simper d, miller d, russell s, litzow m, edwards w. smooth muscle cells in human 181.

coronary atherosclerosis can originate from cells administered at marrow transplantation. proc natl acad sci u s a 2003; 100:

4754-4759.

le ricousse-roussanne s, barateau v, contreres J, boval b, Kraus-berthier l, tobelem g. ex vivo differentiated endothelial and 182.

smooth muscle cells from human cord blood progenitors home to the angiogenic tumor vasculature. cardiovasc res 2004; 62:

176-184.

zengin e, chalajour f, gehling um, ito wd, treede h, lauke h, weil J, reichenspurner h, Kilic n, ergun s. vascular wall resident 183.

progenitor cells: a source for postnatal vasculogenesis. development 2006; 133: 1543-1551.

rajantie i, ilmonen m, alminaite a, ozerdem u, alitalo K, salven p. adult bone marrow-derived cells recruited during angiogenesis 184.

comprise precursors for periendothelial vascular mural cells. blood 2004; 104: 2084-2086.

purhonen s, palm J, rossi dJ, Kaskenpaa n, rajantie i, yla-herttuala s, alitalo K, weissman il, salven p. bone marrow-derived 185.

circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. proc natl acad

sci u s a 2008; 105: 6620-6625.

libby p, ridker pm, maseri a. inflammation and atherosclerosis. circulation 2002; 105: 1135-1143.186.

volger ol, fledderus Jo, Kisters n, fontijn rd, moerland pd, Kuiper J, van berkel tJ, bijnens ap, daemen mJ, pannekoek h, horrevoets 187.

aJ. distinctive expression of chemokines and transforming growth factor-b signaling in human arterial endothelium during

atherosclerosis. am J pathol 2007; 171: 326-337.

zoll J, fontaine v, gourdy p, barateau v, vilar J, leroyer a, lopes-Kam i, mallat z, arnal Jf, henry p, tobelem g, tedgui a. role of human 188.

smooth muscle cell progenitors in atherosclerotic plaque development and composition. cardiovasc res 2008; 77: 471-480.

foubert p, matrone g, souttou b, lere-dean c, barateau v, plouet J, le ricousse-roussanne s, levy bi, silvestre Js, tobelem g. 189.

coadministration of endothelial and smooth muscle progenitor cells enhances the efficiency of proangiogenic cell-based therapy.

circ res 2008; 103: 751-760.

Kawamoto a, losordo dw. endothelial progenitor cells for cardiovascular regeneration. trends cardiovasc med 2008; 18: 33-37.190.

liebner s, cattelino a, gallini r, rudini n, iurlaro m, piccolo s, dejana e. b-catenin is required for endothelial-mesenchymal 191.

transformation during heart cushion development in the mouse. J cell biol 2004; 166: 359-367.

arciniegas e, ponce l, hartt y, graterol a, carlini rg. intimal thickening involves transdifferentiation of embryonic endothelial cells. 192.

anat rec 2000; 258: 47-57.

hall sm, hislop aa, haworth sg. origin, differentiation, and maturation of human pulmonary veins. am J respir cell mol biol 2002; 193.

26: 333-340.

Kawamoto a, asahara t. role of progenitor endothelial cells in cardiovascular disease and upcoming therapies. catheter cardiovasc 194.

interv 2007; 70: 477-484.

hill J, zalos g, halcox J, schenke w, waclawiw m, Quyyumi aa, finkel t. circulating endothelial progenitor cells, vascular function, 195.

and cardiovascular risk. n engl J med 2003; 348: 593-600.

zhang sJ, zhang h, wei yJ, su wJ, liao zK, hou m, zhou Jy, hu ss. adult endothelial progenitor cells from human peripheral blood 196.

maintain monocyte/macrophage function throughout in Vitro culture. cell res 2006; 16: 577-584.

smadja dm, basire a, amelot a, conte a, bieche i, le bonniec bf, aiach m, gaussem p. thrombin bound to a fibrin clot confers 197.

angiogenic and hemostatic properties on endothelial progenitor cells. J cell mol med 2008; 12: 975-986.

appendices

216

references

217 App

sarkar s, lee gy, wong Jy, desai ta. development and characterization of a porous micro-patterned scaffold for vascular tissue 240.

engineering applications. biomaterials 2006; 27: 4775-4782.

isenberg bc, tsuda y, williams c, shimizu t, yamato m, okano t, wong Jy. a thermoresponsive, microtextured substrate for cell 241.

sheet engineering with defined structural organization. biomaterials 2008; 29: 2565-2572.

choi Js, lee sJ, christ gJ, atala a, yoo JJ. the influence of electrospun aligned poly(epsilon-caprolactone)/collagen nanofiber 242.

meshes on the formation of self-aligned skeletal muscle myotubes. biomaterials 2008; 29: 2899-2906.

thomas v, Jose mv, chowdhury s, sullivan Jf, dean dr, vohra yK. mechano-morphological studies of aligned nanofibrous scaffolds 243.

of polycaprolactone fabricated by electrospinning. J biomater sci polym ed 2006; 17: 969-984.

vasita r, shanmugam iK, Katt ds. improved biomaterials for tissue engineering applications: surface modification of polymers. curr 244.

top med chem 2008; 8: 341-353.

feng y, mrksich m. the synergy peptide phsrn and the adhesion peptide rgd mediate cell adhesion through a common mechanism. 245.

biochemistry 2004; 43: 15811-15821.

zamora po, eshima d, graham d, shattuck l, rhodes ba. biological distribution of 99mtc-labeled yigsr and iKvav laminin peptides 246.

in rodents: 99mtc-iKvav peptide localizes to the lung. biochim biophys acta 1993; 1182: 197-204.

yamamoto m, yamamoto K, noumura t. type i collagen promotes modulation of cultured rabbit arterial smooth muscle cells from 247.

a contractile to a synthetic phenotype. exp cell res 1993; 204: 121-129.

tsilibary ec, reger la, vogel am, Koliakos gg, anderson ss, charonis as, alegre Jn, furcht lt. identification of a multifunctional, 248.

cell-binding peptide sequence from the a1(nc1) of type iv collagen. J cell biol 1990; 111: 1583-1591.

hocking dc, sottile J, mcKeown-longo pJ. activation of distinct a5b1-mediated signaling pathways by fibronectin’s cell adhesion 249.

and matrix assembly domains. J cell biol 1998; 141: 241-253.

alobaid n, salacinski hJ, sales Km, ramesh b, Kannan ry, hamilton g, seifalian am. nanocomposite containing bioactive peptides 250.

promote endothelialisation by circulating progenitor cells: an in Vitro evaluation. eur J vasc endovasc surg 2006; 32: 76-83.

ferreira ls, gerecht s, fuller J, shieh hf, vunjak-novakovic g, langer r. bioactive hydrogel scaffolds for controllable vascular 251.

differentiation of human embryonic stem cells. biomaterials 2007; 28: 2706-2717.

grant ds, tashiro Ki, segui-real b, yamada y, martin gr, Kleinman hK. two different laminin domains mediate the differentiation 252.

of human endothelial cells into capillary-like structures in Vitro. cell 1989; 58: 933-943.

pollman mJ, naumovski l, gibbons gh. vascular cell apoptosis: cell type specific modulation by transforming growth factor-β1 in 253.

endothelial cells Versus smooth muscle cells. circulation 1999; 99: 2019-2026.

zisch a, lutolf mp, hubbell Ja. biopolymeric delivery matrices for angiogenic growth factors. cardiovasc pathol 2003; 12: 295-310.254.

pike d, cai s, pomraning K, firpo m, fisher r, shu x, prestwich g, peattie r. heparin-regulated release of growth factors 255. in Vitro and

angiogenic response in Vivo to implanted hyaluronan hydrogels containing vegf and bfgf. biomaterials 2006; 27: 5242-5251.

nillesen st, geutjes pJ, wismans r, schalkwijk J, daamen wf, van Kuppevelt th. increased angiogenesis and blood vessel maturation 256.

in acellular collagen-heparin scaffolds containing both fgf2 and vegf. biomaterials 2007; 28: 1123-1131.

gong y, he l, li J, zhou Q, ma z, gao c, shen J. hydrogel-filled polylactide porous scaffolds for cartilage tissue engineering. J biomed 257.

mater res b appl biomater 2007; 82: 192-204.

zisch ah, lutolf mp, ehrbar m, raeber gp, rizzi sc, davies n, schmokel h, bezuidenhout d, djonov vg, zilla p, hubbell Ja. cell-258.

demanded release of vegf from synthetic, biointeractive cell-ingrowth matrices for vascularized tissue growth. the faseb Journal

2003; 17: 2260-2262.

seliktar d, zisch ah, lutolf mp, wrana Jl, hubbell Ja. mmp-2 sensitive, vegf-bearing bioactive hydrogels for promotion of vascular 259.

healing. J biomed mater res a 2004; 68: 704-716.

herbrig K, pistrosch f, oelschlaegel u, wichmann g, wagner a, foerster s, richter s, gross p, passauer J. increased total number but 260.

impaired migratory activity and adhesion of endothelial progenitor cells in patients on long-term hemodialysis. am J Kidney dis

2004; 44: 840-849.

vasa m, fichtlscherer s, aicher a, adler K, urbich c, martin h, zeiher a, dimmeler s. number and migratory activity of circulating 261.

endothelial progenitor cells inversely correlate with risk factors for coronary artery disease. circ res 2001; 89: e1-7.

Koshida r, rocic p, saito s, Kiyooka t, zhang c, chilian wm. role of focal adhesion Kinase in flow-induced dilation of coronary 219.

arterioles. arterioscler thromb vasc biol 2005; 25: 2548-2553.

malmstrom J, lindberg h, lindberg c, bratt c, wieslander e, delander el, sarnstrand b, burns Js, mose-larsen p, fey s, marko-220.

varga g. transforming growth factor-b1 specifically induce proteins involved in the myofibroblast contractile apparatus. mol cell

proteomics 2004; 3: 466-477.

sinha s, hoofnagle mh, Kingston pa, mccanna me, owens gK. transforming growth factor-b1 signaling contributes to development 221.

of smooth muscle cells from embryonic stem cells. am J physiol cell physiol 2004; 287: c1560-c1568.

hautmann mb, madsen cs, owens gK. a transforming growth factor beta (tgfb) control element drives tgfb-induced stimulation 222.

of smooth muscle a-actin gene expression in concert with two carg elements. J biol chem 1997; 272: 10948-10956.

li l, miano Jm, mercer b, olson en. expression of the sm22a promoter in transgenic mice provides evidence for distinct transcriptional 223.

regulatory programs in vascular and visceral smooth muscle cells. J cell biol 1996; 132: 849-859.

zilberman a, dave v, miano Jm, olson en, periasamy m. evolutionarily conserved promoter region containing carg-like elements is 224.

crucial for smooth muscle myosin heavy chain gene expression. circ res 1998; 82: 566-575.

olson en, perry m, schulz ra. regulation of muscle differentiation by the mef2 family of mads box transcription factors. dev biol 225.

1995; 172: 2-14.

benchabane h, wrana Jl. gata- and smad1-dependent enhancers in the smad7 gene differentially interpret bone morphogenetic 226.

protein concentrations. mol cell biol 2003; 23: 6646-6661.

camoretti-mercado b, fernandes dJ, dewundara s, churchill J, ma l, Kogut pc, mcconville Jf, parmacek ms, solway J. inhibition 227.

of transforming growth factor β-enhanced serum response factor-dependent transcription by smad7. J biol chem 2006; 281:

20383-20392.

nagai r, suzuki t, aizawa K, miyamoto s, amaki t, Kawai-Kowase K, sekiguchi Ki, Kurabayashi m. phenotypic modulation of vascular 228.

smooth muscle cells: dissection of transcriptional regulatory mechanisms. ann n y acad sci 2001; 947: 56-66.

rensen ss, doevendans pa, van eys gJ. regulation and characteristics of vascular smooth muscle cell phenotypic diversity. neth 229.

heart J 2007; 15: 100-108.

chen cn, li ys, yeh yt, lee pl, usami s, chien s, chiu JJ. synergistic roles of platelet-derived growth factor-bb and interleukin-1b in 230.

phenotypic modulation of human aortic smooth muscle cells. proc natl acad sci u s a 2006; 103: 2665-2670.

li x, van putten v, zarinetchi f, nicks me, thaler s, heasley le, nemenoff ra. suppression of smooth-muscle alpha-actin expression 231.

by platelet-derived growth factor in vascular smooth-muscle cells involves ras and cytosolic phospholipase a2. biochem J 1997;

327: 709-716.

hao h, ropraz p, verin v, camenzind e, geinoz a, pepper ms, gabbiani g, bochaton-piallat ml. heterogeneity of smooth muscle cell 232.

populations cultured from pig coronary artery. arterioscler thromb vasc biol 2002; 22: 1093-1099.

deguchi J, namba t, hamada h, nakaoka t, abe J, sato o, miyata t, makuuchi m, Kurokawa K, takuwa y. targeting endogenous 233.

platelet-derived growth factor b-chain by adenovirus-mediated gene transfer potently inhibits in Vivo smooth muscle proliferation

after arterial injury. gene ther 1999; 6: 956-965.

miyata t, iizasa h, sai y, fujii J, terasaki t, nakashima e. platelet-derived growth factor-bb (pdgf-bb) induces differentiation of 234.

bone marrow endothelial progenitor cell-derived cell line tr-bme2 into mural cells, and changes the phenotype. J cell physiol

2005; 204: 948-955.

couet f, rajan n, mantovani d. macromolecular biomaterials for scaffold-based vascular tissue engineering. macromol biosci 2007; 235.

7: 701-718.

Kannan ry, salacinski hJ, butler pe, hamilton g, seifalian am. current status of prosthetic bypass grafts: a review. J biomed mater 236.

res b appl biomater 2005; 74: 570-581.

brody s, anilkumar t, liliensiek s, last Ja, murphy cJ, pandit a. characterizing nanoscale topography of the aortic heart valve 237.

basement membrane for tissue engineering heart valve scaffold design. tissue eng 2006; 12: 413-421.

lin yK, liu dc. studies of novel hyaluronic acid-collagen sponge materials composed of two different species of type i collagen. J 238.

biomater appl 2007; 21: 265-281.

shadwick re. mechanical design in arteries. J exp biol 1999; 202: 3305-3313.239.

appendices

218

references

219 App

duan hx, cheng lm, Jian w, hu ls, lu gx. angiogenic potential difference between two types of endothelial progenitor cells from 283.

human umbilical cord blood. cell biol int 2006; 30: 1018-1027.

zhang r, yang h, li m, yao Q, chen c. acceleration of endothelial-like cell differentiation from cd14284. + monocytes in Vitro. exp

hematol 2005; 33: 1554-1563.

elsheikh e, uzunel m, he z, holgersson J, nowak g, sumitran-holgersson s. only a specific subset of human peripheral-blood 285.

monocytes has endothelial-like functional capacity. blood 2005; 106: 2347-2355.

sparmann a, bar-sagi d. ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. cancer cell 286.

2004; 6: 447-458.

anghelina m, moldovan l, zabuawala t, ostrowski mc, moldovan ni. a subpopulation of peritoneal macrophages form capillarylike 287.

lumens and branching patterns in Vitro. J cell mol med 2006; 10: 708-715.

charo i, taubman m. chemokines in the pathogenesis of vascular disease. circ res 2004; 95: 858-866.288.

Kuroda t, Kitadai y, tanaka s, yang x, mukaida n, yoshihara m, chayama K. monocyte chemoattractant protein-1 transfection 289.

induces angiogenesis and tumorigenesis of gastric carcinoma in nude mice via macrophage recruitment. clin cancer res 2005;

11: 7629-7636.

hong K, ryu J, han K. monocyte chemoattractant protein-1-induced angiogenesis is mediated by vascular endothelial growth 290.

factor-a. blood 2005; 105: 1405-1407.

niiyama h, Kai h, yamamoto t, shimada t, sasaki K, murohara t, egashira K, imaizumi t. roles of endogenous monocyte 291.

chemoattractant protein-1 in ischemia-induced neovascularization. J am coll cardiol 2004; 44: 661-666.

yamada m, Kim s, egashira K, takeya m, ikeda t, mimura o, iwao h. molecular mechanism and role of endothelial monocyte 292.

chemoattractant protein-1 induction by vascular endothelial growth factor. arterioscler thromb vasc biol 2003; 23: 1996-2001.

li a, varney m, valasek J, godfrey m, dave b, singh r. autocrine role of interleukin-8 in induction of endothelial cell proliferation, 293.

survival, migration and mmp-2 production and angiogenesis. angiogenesis 2005; 8: 63-71.

cai l, Johnstone bh, cook tg, liang z, traktuev d, cornetta K, ingram da, rosen ed, march Kl. suppression of hepatocyte growth 294.

factor production impairs the ability of adipose-derived stem cells to promote ischemic tissue revascularization. stem cells 2007;

25: 3234-3243.

bordoni v, alonzi t, zanetta l, Khouri d, conti a, corazzari m, bertolini f, antoniotti p, pisani g, tognoli f, dejana e, tripodi m. 295.

hepatocyte-conditioned medium sustains endothelial differentiation of human hematopoietic-endothelial progenitors.

hepatology 2007; 45: 1218-1228.

asahara t, masuda h, takahashi t, Kalka c, pastore c, silver m, Kearne m, magner m, isner Jm. bone marrow origin of endothelial 296.

progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. circ res 1999; 85:

221-228.

losordo dw, schatz ra, white cJ, udelson Je, veereshwarayya v, durgin m, poh KK, weinstein r, Kearney m, chaudhry m, burg 297.

a, eaton l, heyd l, thorne t, shturman l, hoffmeister p, story K, zak v, dowling d, traverse Jh, olson re, flanagan J, sodano d,

murayama t, Kawamoto a, Kusano Kf, wollins J, welt f, shah p, soukas p, asahara t, henry td. intramyocardial transplantation of

autologous cd34+ stem cells for intractable angina: a phase i/iia double-blind, randomized controlled trial. circulation 2007; 115:

3165-3172.

schachinger v, assmus b, britten mb, honold J, lehmann r, teupe c, abolmaali nd, vogl tJ, hofmann wK, martin h, dimmeler 298.

s, zeiher am. transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year

results of the topcare-ami trial. J am coll cardiol 2004; 44: 1690-1699.

leor J, rozen l, zuloff-shani a, feinberg ms, amsalem y, barbash im, Kachel e, holbova r, mardor y, daniels d, ocherashvilli a, 299.

orenstein a, danon d. ex vivo activated human macrophages improve healing, remodeling, and function of the infarcted heart.

circulation 2006; 114: i94-100.

urbich c, heeschen c, aicher a, dernbach e. relevance of monocytic features for neovascularization capacity of circulating 300.

endothelial progenitor cells. circulation 2003; 108: 2511-2516.

Kinnaird t, stabile e, burnett ms, lee cw, barr s, fuchs s, epstein se. marrow-derived stromal cells express genes encoding a broad 262.

spectrum of arteriogenic cytokines and promote in Vitro and in Vivo arteriogenesis through paracrine mechanisms. circ res 2004;

94: 678-685.

gnecchi m, zhang z, ni a, dzau vJ. paracrine mechanisms in adult stem cell signaling and therapy. circ res 2008; 103: 1204-1219.263.

takahashi m, li ts, suzuki r, Kobayashi t, ito h, ikeda y, matsuzaki m, hamano K. cytokines produced by bone marrow cells can 264.

contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury. aJp - heart and

circulatory physiology 2006; 291: h886-h893.

hristov m, erl w, weber p. endothelial progenitor cells: mobilization, differentiation, and homing. arterioscler thromb vasc biol 265.

2003; 23: 1185-1189.

Jackson K, majka s, wang h, pocius J, hartley c, majesky mw, entman m, michael lh, hirschi KK, goodell m. regeneration of ischemic 266.

cardiac muscle and vascular endothelium by adult stem cells. J clin invest 2001; 107: 1395-1402.

ma n, stamm c, Kaminski a, li w, Kleine hd, muller-hilke b, zhang l, ladilov y, egger d, steinhoff g. human cord blood cells induce 267.

angiogenesis following myocardial infarction in nod/scid-mice. cardiovasc res 2005; 66: 45-54.

van der strate bwa, popa er, schipper m, brouwer la, hendriks m, harmsen mc, van luyn mJa. circulating human cd34268. + progenitor

cells modulate neovascularization and inflammation in a nude mouse model. J mol cell cardiol 2007; 42: 1086-1097.

van amerongen mJ, bou-gharios g, popa er, van aJ, petersen ah, van dg, van lm, harmsen mc. bone marrow-derived myofibroblasts 269.

contribute functionally to scar formation after myocardial infarction. J pathol 2007; 214: 377-386.

van amerongen mJ, harmsen mc, van rn, petersen ah, van luyn mJa. macrophage depletion impairs wound healing and increases 270.

left ventricular remodeling after myocardial injury in mice. am J pathol 2007; 170: 818-829.

anghelina m, Krishnan p, moldovan l, moldovan n. monocytes and macrophages form branched cell columns in matrigel: 271.

implications for a role in neovascularization. stem cells dev 2004; 13: 665-676.

harraz m, Jiao c, hanlon hd. cd34- blood derived human endothelial cell progenitors. stem cells 2001; 19: 304-312.272.

sharifi bg, zeng z, wang l, song l, chen h, Qin m, sierra-honigmann mr, wachsmann-hogiu s, shah pK. pleiotrophin induces 273.

transdifferentiation of monocytes into functional endothelial cells. arterioscler thromb vasc biol 2006; 26: 1273-1280.

ziegelhoeffer t, fernandez b, Kostin s, heil m, voswinckel r, helisch a, schaper w. bone marrow-derived cells do not incorporate 274.

into the adult growing vasculature. circ res 2004; 94: 230-238.

bautz f, rafii s, Kanz l, mohle r. expression and secretion of vascular endothelial growth factor-a by cytokine-stimulated 275.

hematopoietic progenitor cells: possible role in the hematopoietic microenvironment. exp hematol 2000; 28: 700-706.

majka m, Janowska-wieczorek a, ratajczak J, ehrenman K, pietrzkowski z, Kowalska ma, gewirtz am, emerson sg, ratajczak 276.

mz. numerous growth factors, cytokines, and chemokines are secreted by human cd34+ cells, myeloblasts, erythroblasts, and

megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner. blood 2001; 97: 3075-3085.

bonello l, basire a, sabatier f, paganelli f, dignat-george f. endothelial injury induced by coronary angioplasty triggers mobilization 277.

of endothelial progenitor cells in patients with stable coronary artery disease. J thromb haemost 2006; 4: 979-981.

susen s, sautiere K, mouquet f, cuilleret f, chmait a, mcfadden ep, hennache b, richard f, de groote p, lablanche Jm, dallongeville 278.

J, bauters c, Jude b, van belle e. serum hepatocyte growth factor levels predict long-term clinical outcome after percutaneous

coronary revascularization. eur heart J 2005; 26: 2387-2395.

woywodt a, erdbruegger u, haubitz m. circulating endothelial cells and endothelial progenitor cells after angioplasty: news from 279.

the endothelial rescue squad. J thromb haemost 2006; 4: 976-978.

rohde e, bartmann c, schallmoser K, reinisch a, lanzer g, linkesch w, guelly c, strunk d. immune cells mimic the morphology of 280.

endothelial progenitor colonies In Vitro. stem cells 2007; 25: 1746-1752.

moldovan ni, goldschmidt-clermont pJ, parker-thornburg J, shapiro sd, Kolattukudy pe. contribution of monocytes/macrophages 281.

to compensatory neovascularization - the drilling of metalloelastase-positive tunnels in ischemic myocardium. circ res 2000; 87:

378-384.

anghelina m, Krishnan p, moldovan l, moldovan n. monocytes/macrophages cooperate with progenitor cells during 282.

neovascularization and tissue repair: conversion of cell columns into fibrovascular bundles. am J pathol 2006; 168: 529-541.

appendices

220

references

221 App

Jolicoeur em, granger cb, fakunding Jl, mockrin sc, grant sm, ellis sg, weisel rd, goodell ma. bringing cardiovascular cell-based 323.

therapy to clinical application: perspectives based on a national heart, lung, and blood institute cell therapy working group

meeting. am heart J 2007; 153: 732-742.

perin ec, dohmann hf, borojevic r, silva sa, sousa al, silva gv, mesquita ct, belem l, vaughn wK, rangel fo, assad Ja, carvalho ac, 324.

branco rv, rossi mi, dohmann hJ, willerson Jt. improved exercise capacity and ischemia 6 and 12 months after transendocardial

injection of autologous bone marrow mononuclear cells for ischemic cardiomyopathy. circulation 2004; 110: ii213-ii218.

beeres sl, lamb hJ, roes sd, holman er, Kaandorp ta, fibbe we, de ra, van der wall ee, schalij mJ, bax JJ, atsma de. effect of 325.

intramyocardial bone marrow cell injection on diastolic function in patients with chronic myocardial ischemia. J magn reson

imaging 2008; 27: 992-997.

vandervelde s, van luyn mJa, rosenbaum mh, petersen ah, tio ra, harmsen mc. stem cell-related cardiac gene expression early 326.

after murine myocardial infarction. cardiovasc res 2006.

wiener cm, booth g, semenza gl. 327. In Vivo expression of mrnas encoding hypoxia-inducible factor 1. biochem biophys res commun

1996; 225: 485-488.

huang le, gu J, schau m, bunn hf. regulation of hypoxia-inducible factor 1alpha is mediated by an o2-dependent degradation 328.

domain via the ubiquitin-proteasome pathway. proc natl acad sci u s a 1998; 95: 7987-7992.

asahara t, takahashi t, masuda h, Kalka c, chen d, iwaguro h, inai y, silver m, isner Jm. vegf contributes to postnatal 329.

neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. embo J 1999; 18: 3964-3972.

grunewald m, avraham i, dor y, bachar-lustig e, itin a, yung s, chimenti s, landsman l, abramovitch r, Keshet e. vegf-induced 330.

adult neovascularization: recruitment, retention, and role of accessory cells. cell 2006; 124: 175-189.

morimoto h, takahashi m, shiba y, izawa a, ise h, hongo m, hatake K, motoyoshi K, ikeda u. bone marrow-derived cxcr4331. + cells

mobilized by macrophage colony-stimulating factor participate in the reduction of infarct area and improvement of cardiac

remodeling after myocardial infarction in mice. am J pathol 2007; 171: 755-766.

yamaguchi J, Kusano Kf, masuo o, Kawamoto a, silver m, murasawa s, bosch-marce m, masuda h, losordo dw, isner Jm, asahara t. 332.

stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization.

circulation 2003; 107: 1322-1328.

capoccia bJ, gregory ad, link dc. recruitment of the inflammatory subset of monocytes to sites of ischemia induces angiogenesis 333.

in a monocyte chemoattractant protein-1-dependent fashion. J leukoc biol 2008; 84: 760-768.

hattori K, dias s, heissig b, hackett n, lyden d, tateno m, hicklin d, zhu z, witte l, crystal r, moore ma, rafii s. vascular endothelial 334.

growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem

cells. J exp med 2001; 193: 1005-1014.

heeschen c, aicher a, lehmann r, fichtlscherer s, vasa m, urbich c, mildner-rihm c, martin h, zeiher am, dimmeler s. erythropoietin 335.

is a potent physiologic stimulus for endothelial progenitor cell mobilization. blood 2003; 102: 1340-1346.

wallez y, huber p. endothelial adherens and tight Junctions in vascular homeostasis, inflammation and angiogenesis. biochimica 336.

et biophysica acta (bba) - biomembranes 2008; 1778: 794-809.

gavard J, gutkind Js. vegf controls endothelial-cell permeability by promoting the beta-arrestin-dependent endocytosis of ve-337.

cadherin. nat cell biol 2006; 8: 1223-1234.

hirase t, Kawashima s, wong ey, ueyama t, rikitake y, tsukita s, yokoyama m, staddon Jm. regulation of tight Junction permeability 338.

and occludin phosphorylation by rhoa-p160rocK-dependent and -independent mechanisms. J biol chem 2001; 276: 10423-10431.

stamatovic sm, dimitrijevic ob, Keep rf, andjelkovic av. protein Kinase calpha-rhoa cross-talk in ccl2-induced alterations in brain 339.

endothelial permeability. J biol chem 2006; 281: 8379-8388.

peled a, zipori d, abramsky o, ovadia h, shezen e. expression of alpha-smooth muscle actin in murine bone marrow stromal cells. 340.

blood 1991; 78: 304-309.

peled a, Kollet o, ponomaryov t, petit i, franitza s, grabovsky v, slav mm, nagler a, lider o, alon r, zipori d, lapidot t. the chemokine 341.

sdf-1 activates the integrins lfa-1, vla-4, and vla-5 on immature human cd34+ cells: role in transendothelial/stromal migration

and engraftment of nod/scid mice. blood 2000; 95: 3289-3296.

sengupta s, sellers l, li r, gherardi e, zhao g, watson n, sasisekharan r, fan t. targeting of mitogen-activated protein Kinases 301.

and phosphatidylinositol 3 Kinase inhibits hepatocyte growth factor/scatter factor-induced angiogenesis. circulation 2003; 107:

2955-2961.

tomita n, morishita r, taniyama y, Koike h, aoki m, shimizu h, matsumoto K, nakamura t, Kaneda y, ogihara t. angiogenic property 302.

of hepatocyte growth factor is dependent on upregulation of essential transcription factor for angiogenesis, ets-1. circulation

2003; 107: 1411-1417.

nian m, lee p, Khaper n, liu p. inflammatory cytokines and postmyocardial infarction remodeling. circ res 2004; 94: 1543-1553.303.

vandervelde s, van amerongen mJ, tio ra, petersen ah, van luyn mJa, harmsen mc. increased inflammatory response and 304.

neovascularization in reperfused vs. non-reperfused murine myocardial infarction. cardiovasc pathol 2006; 15: 83-90.

Kocher aa, schuster md, szabolcs mJ, takuma s, burkhoff d, wang J, homma s, edwards nm, itescu s. neovascularization of ischemic 305.

myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves

cardiac function. nat med 2001; 7: 430-436.

shiojima i, sato K, izumiya y, schiekofer s, ito m, liao r, colucci ws, walsh K. disruption of coordinated cardiac hypertrophy and 306.

angiogenesis contributes to the transition to heart failure. J clin invest 2005; 115: 2108-2118.

risau w. mechanisms of angiogenesis. nature 1997; 386: 671-674.307.

distler Jh, hirth a, Kurowska-stolarska m, gay re, gay s, distler o. angiogenic and angiostatic factors in the molecular control of 308.

angiogenesis. Q J nucl med 2003; 47: 149-161.

murohara t, ikeda h, duan J, shintani s, sasaki K, eguchi h, onitsuka i, matsui K, imaizumi t. transplanted cord blood-derived 309.

endothelial precursor cells augment postnatal neovascularization. J clin invest 2000; 105: 1527-1536.

cottler-fox mh, lapidot t, petit i, Kollet o, dipersio Jf, link d, devine s. stem cell mobilization. hematology am soc hematol educ 310.

program 2003; 419-437.

hattori K, heissig b, rafii s. the regulation of hematopoietic stem cell and progenitor mobilization by chemokine sdf-1. leuk 311.

lymphoma 2003; 44: 575-582.

Jujo K, ii m, losordo dw. endothelial progenitor cells in neovascularization of infarcted myocardium. J mol cell cardiol 2008; 45: 312.

530-544.

Jevon m, dorling a, hornick pi. progenitor cells and vascular disease. cell prolif 2008; 41 suppl 1: 146-164.313.

ward mr, stewart dJ, Kutryk mJ. endothelial progenitor cell therapy for the treatment of coronary disease, acute mi, and pulmonary 314.

arterial hypertension: current perspectives. catheter cardiovasc interv 2007; 70: 983-998.

schatteman gc, awad a. 315. In Vivo and in Vitro properties of cd34+ and cd14+ endothelial cell precursors. adv exp med biol 2003; 522:

9-16.

schatteman gc, dunnwald m, Jiao c. biology of bone marrow-derived endothelial cell precursors. aJp - heart and circulatory 316.

physiology 2007; 292: h1-18.

zhang sJ, zhang h, hou m, zheng z, zhou J, su w, wei y, hu ss. is it possible to obtain true endothelial progenitor cells by 317. in Vitro

culture of bone marrow mononuclear cells? stem cells dev 2007; 16: 683-690.

fan cl, li y, gao pJ, liu JJ, zhang xJ, zhu dl. differentiation of endothelial progenitor cells from human umbilical cord blood cd34318. +

cells in Vitro. acta pharmacol sin 2003; 24: 212-218.

nagano m, yamashita t, hamada h, ohneda K, Kimura K, nakagawa t, shibuya m, yoshikawa h, ohneda o. identification of 319.

functional endothelial progenitor cells suitable for the treatment of ischemic tissue using human umbilical cord blood. blood

2007; 110: 151-160.

werner n, Junk s, laufs u, link a, walenta K, bohm m, nickenig g. intravenous transfusion of endothelial progenitor cells reduces 320.

neointima formation after vascular injury. circ res 2003; 93: e17-e24.

zhang w, zhang g, Jin h, hu r. characteristics of bone marrow-derived endothelial progenitor cells in aged mice. biochem biophys 321.

res commun 2006; 348: 1018-1023.

pompilio g, capogrossi mc, pesce m, alamanni f, dicampli c, achilli f, germani a, biglioli p. endothelial progenitor cells and 322.

cardiovascular homeostasis: clinical implications. international Journal of cardiology 2009; 131: 156-167.

appendices

222

references

223 App

sarkar s, sales Km, hamilton g, seifalian am. addressing thrombogenicity in vascular graft construction. J biomed mater res b appl 364.

biomater 2007; 82: 100-108.

plate g, hollier lh, gloviczki p, dewanjee mK, Kaye mp. overcoming failure of venous vascular prostheses. surgery 1984; 96: 365.

503-510.

atluri p, woo yJ. pro-angiogenic cytokines as cardiovascular therapeutics: assessing the potential. biodrugs 2008; 22: 209-222.366.

dardik a, liu a, ballermann bJ. chronic 367. in Vitro shear stress stimulates endothelial cell retention on prosthetic vascular grafts and

reduces subsequent in Vivo neointimal thickness. J vasc surg 1999; 29: 157-167.

urbich c, aicher a, heeschen c, dernbach e, hofmann w, zeiher a, dimmeler s. soluble factors released by endothelial progenitor 368.

cells promote migration of endothelial cells and cardiac resident progenitor cells. J mol cell cardiol 2005; 39: 733-742.

armstrong eJ, bischoff J. heart valve development: endothelial cell signaling and differentiation. circ res 2004; 95: 459-470.369.

tanaka t, saika s, ohnishi y, ooshima a, mcavoy Jw, liu cy, azhar m, doetschman t, Kao ww. fibroblast growth factor 2: roles of 370.

regulation of lens cell proliferation and epithelial-mesenchymal transition in response to injury. mol vis 2004; 10: 462-467.

arciniegas e, neves yc, carrillo lm. potential role for insulin-like growth factor ii and vitronectin in the endothelial-mesenchymal 371.

transition process. differentiation 2006; 74: 277-292.

Kilian o, flesch i, wenisch s, taborski b, Jork a, schnettler r, Jonuleit t. effects of platelet growth factors on human mesenchymal 372.

stem cells and human endothelial cells in Vitro. eur J med res 2004; 9: 337-344.

hyman Km, seghezzi g, pintucci g, stellari g, Kim Jh, grossi ea, galloway ac, mignatti p. transforming growth factor-b1 induces 373.

apoptosis in vascular endothelial cells by activation of mitogen-activated protein Kinase. surgery 2002; 132: 173-179.

ahmadi h, baharvand h, ashtiani sK, soleimani m, sadeghian h, ardekani Jm, mehrjerdi nz, Kouhkan a, namiri m, madani-civi 374.

m, fattahi f, shahverdi a, dizaji av. safety analysis and improved cardiac function following local autologous transplantation of

cd133+ enriched bone marrow cells after myocardial infarction. curr neurovasc res 2007; 4: 153-160.

meyer gp, wollert Kc, lotz J, steffens J, lippolt p, fichtner s, hecker h, schaefer a, arseniev l, hertenstein b, ganser a, drexler 375.

h. intracoronary bone marrow cell transfer after myocardial infarction: eighteen months’ follow-up data from the randomized,

controlled boost (bone marrow transfer to enhance st-elevation infarct regeneration) trial. circulation 2006; 113: 1287-1294.

wollert Kc, meyer gp, lotz J, ringes lichtenberg s, lippolt p, breidenbach c, fichtner s, Korte t, hornig b, messinger d, arseniev l, 376.

hertenstein b, ganser a, drexler h. intracoronary autologous bone-marrow cell transfer after myocardial infarction: the boost

randomised controlled clinical trial. the lancet 2004; 364: 141-148.

hirsch a, nijveldt r, van d, v, biemond bJ, doevendans pa, van rossum ac, tijssen Jg, zijlstra f, piek JJ. intracoronary infusion 377.

of autologous mononuclear bone marrow cells or peripheral mononuclear blood cells after primary percutaneous coronary

intervention: rationale and design of the hebe trial--a prospective, multicenter, randomized trial. am heart J 2006; 152: 434-441.

hirsch a, nijveldt r, van d, v, tio ra, van der giessen wJ, marques Km, doevendans pa, waltenberger J, ten berg Jm, aengevaeren 378.

wr, biemond bJ, tijssen Jg, van rossum ac, piek JJ, zijlstra f. intracoronary infusion of autologous mononuclear bone marrow cells

in patients with acute myocardial infarction treated with primary pci: pilot study of the multicenter hebe trial. catheter cardiovasc

interv 2008; 71: 273-281.

schachinger v, erbs s, elsasser a, haberbosch w, hambrecht r, holschermann h, yu J, corti r, mathey dg, hamm cw, suselbeck 379.

t, werner n, haase J, neuzner J, germing a, mark b, assmus b, tonn t, dimmeler s, zeiher am, for the repair-ami investigators.

improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial

infarction: final 1-year results of the repair-ami trial. eur heart J 2006; 27: 2775-2783.

erbs s, linke a, schachinger v, assmus b, thiele h, diederich Kw, hoffmann c, dimmeler s, tonn t, hambrecht r, zeiher am, schuler 380.

g. restoration of microvascular function in the infarct-related artery by intracoronary transplantation of bone marrow progenitor

cells in patients with acute myocardial infarction: the doppler substudy of the reinfusion of enriched progenitor cells and infarct

remodeling in acute myocardial infarction (repair-ami) trial. circulation 2007; 116: 366-374.

assmus b, schachinger v, teupe c, britten m, lehmann r, dobert n, grunwald f, aicher a, urbich c, martin h, hoelzer d, dimmeler 381.

s, zeiher a. transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (topcare-ami).

circulation 2002; 106: 3009-3017.

voermans c, rood pml, hordijk pl, gerritsen wr, van der schoot ce. adhesion molecules involved in transendothelial migration of 342.

human hematopoietic progenitor cells. stem cells 2000; 18: 435-443.

smadja dm, bieche i, silvestre Js, germain s, cornet a, laurendeau i, duong-van huyen Jp, emmerich J, vidaud m, aiach m, gaussem 343.

p. bone morphogenetic proteins 2 and 4 are selectively expressed by late outgrowth endothelial progenitor cells and promote

neoangiogenesis. arterioscler thromb vasc biol 2008; 28: 2137-2143.

sieveking dp, buckle a, celermajer ds, ng mKc. strikingly different angiogenic properties of endothelial progenitor cell 344.

subpopulations: insights from a novel human angiogenesis assay. J am coll cardiol 2008; 51: 660-668.

van belle e, witzenbichler b, chen d, silver m, chang l, schwall r, isner Jm. potentiated angiogenic effect of scatter factor/345.

hepatocyte growth factor via induction of vascular endothelial growth factor: the case for paracrine amplification of angiogenesis.

circulation 1998; 97: 381-390.

Khoo cp, pozzilli p, alison mr. endothelial progenitor cells and their potential therapeutic applications. regen med 2008; 3: 346.

863-876.

symes Jf, vale pr, schatz ra, losordo dw. clinical trials of myocardial vegf gene transfer. gene therapy and regulation 2001; 1: 347.

311-323.

chen rr, silva ea, yuen ww, brock aa, fischbach c, lin as, guldberg re, mooney dJ. integrated approach to designing growth 348.

factor delivery systems. faseb J 2007; 21: 3896-3903.

Jain rK, duda dg, clark Jw, loeffler Js. lessons from phase iii clinical trials on anti-vegf therapy for cancer. nat clin pract oncol 349.

2006; 3: 24-40.

hao x, silva ea, mansson-broberg a, grinnemo Kh, siddiqui aJ, dellgren g, wardell e, brodin la, mooney dJ, sylven c. angiogenic 350.

effects of sequential release of vegf-a165 and pdgf-bb with alginate hydrogels after myocardial infarction. cardiovasc res 2007;

75: 178-185.

roy r, zhang b, moses ma. making the cut: protease-mediated regulation of angiogenesis. exp cell res 2006; 312: 608-622.351.

richardson tp, peters mc, ennett ab, mooney dJ. polymeric system for dual growth factor delivery. nat biotechnol 2001; 19: 352.

1029-1034.

ehrbar m, djonov vg, schnell c, tschanz sa, martiny-baron g, schenk u, wood J, burri ph, hubbell Ja, zisch ah. cell-demanded 353.

liberation of vegf121 from fibrin implants induces local and controlled blood vessel growth. circ res 2004; 94: 1124-1132.

herbrig K, gebler K, oelschlaegel u, pistrosch f, foerster s, wagner a, gross p, passauer J. Kidney transplantation substantially 354.

improves endothelial progenitor cell dysfunction in patients with end-stage renal disease. am J transplant 2006; 6: 2922-2928.

Kränkel n, Katare rg, siragusa m, barcelos ls, campagnolo p, mangialardi g, fortunato o, spinetti g, tran n, zacharowski K, 355.

wojakowski w, mroz i, herman a, manning fox Je, macdonald pe, schanstra Jp, bascands Jl, ascione r, angelini g, emanueli c,

madeddu p. role of Kinin b2 receptor signaling in the recruitment of circulating progenitor cells with neovascularization potential.

circ res 2008; 103: 1335-1343.

langer r, vacanti Jp. tissue engineering. science 1993; 260: 920-926.356.

hristov m, weber c. endothelial progenitor cells in vascular repair and remodeling. pharmacol res 2008; 58: 148-151.357.

deb a, skelding K, wang s, reeder m, simper d, caplice n. integrin profile and 358. In Vivo homing of human smooth muscle progenitor

cells. circulation 2004; 110: 2673-2677.

sata m, saiura a, Kunisato a, tojo a, okada sy, tokuhisa t, hirai h, makuuchi m, hirata y, nagai r. hematopoietic stem cells 359.

differentiate into vascular cells that participate in the pathogenesis of atherosclerosis. nat med 2002; 8: 403-409.

asahara t. cell therapy and gene therapy using endothelial progenitor cells for vascular regeneration. handb exp pharmacol 2007; 360.

180: 181-194.

Kawamoto a, tkebuchava t, yamaguchi Ji, nishimura h, yoon ys, milliken c, uchida s, masuo o, iwaguro h, ma h, hanley a, silver 361.

m, Kearney m, losordo dw, isner Jm, asahara t. intramyocardial transplantation of autologous endothelial progenitor cells for

therapeutic neovascularization of myocardial ischemia. circulation 2003; 107: 461-468.

gottrup f. oxygen in wound healing and infection. world Journal of surgery 2004; 28: 312-315.362.

moossavi s, tuttle ab, vachharajani tJ, plonk g, bettmann ma, majekodunmi o, russell gb, regan Jd, freedman bi. long-term 363.

outcomes of transposed basilic vein arteriovenous fistulae. hemodial int 2008; 12: 80-84.

appendices

224

tateishi-yuyama e, matsubara h, murohara t, ikeda u, shintani s, masaki h, amano K, Kishimoto y, yoshimoto K, akashi h, shimada 382.

K, iwasaka t, imaizumi t. therapeutic angiogenesis for patients with limb ischaemia by autologous transplantation of bone-

marrow cells: a pilot study and a randomised controlled trial. lancet 2002; 360: 427-435.

van huyen Jp, smadja dm, bruneval p, gaussem p, dal-cortivo l, Julia p, fiessinger Jn, cavazzana-calvo m, aiach m, emmerich J. 383.

bone marrow-derived mononuclear cell therapy induces distal angiogenesis after local injection in critical leg ischemia. mod

pathol 2008; 21: 837-846.

zhang h, zhang n, li m, feng h, Jin w, zhao h, chen x, tian l. therapeutic angiogenesis of bone marrow mononuclear cells (mncs) 384.

and peripheral blood mncs: transplantation for ischemic hindlimb. ann vasc surg 2008; 22: 238-247.

ott i, Keller u, Knoedler m, gotze Ks, doss K, fischer p, urlbauer K, debus g, von bn, rudelius m, schomig a, peschel c, oostendorp 385.

ra. endothelial-like cells expanded from cd34+ blood cells improve left ventricular function after experimental myocardial

infarction. faseb J 2005; 19: 992-994.

sondergaard cs, bonde J, degnæs-hansen f, nielsen Jm, zachar v, holm m, hokland p, pedersen l. minimal engraftment of human 386.

cd34+ cells mobilized from healthy donors in the infarcted heart of athymic nude rats. stem cells dev 2008.

Koponen JK, Kekarainen t, heinonen e, laitinen a, nystedt J, laine J, yla-herttuala s. umbilical cord blood-derived progenitor cells 387.

enhance muscle regeneration in mouse hindlimb ischemia model. mol ther 2007; 15: 2172-2177.