CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS CHAPTER 1 GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS CHAPTER 1 GENERAL INTRODUCTION AND OUTLINE OF THE THESIS 9 REGENERATIVE MEDICINE IN CARD...
37 downloads 0 Views 291KB Size
CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

CHAPTER 1 GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

9

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

10

BACKGROUND Diseases of the heart and circulatory system (cardiovascular disease or CVD) are the major cause of death in the Western world, causing over 4.35 million deaths a year in Europe alone [1]. Thereby, CVD causes nearly 50 % off all deaths in Europe, and is estimated to cost the European Union economies €169 billion a year [1]. Furthermore, the prevalence of CVD is increasing because of improved treatment options with higher survival rates and because of the increasing elderly population [2]. Although cardiovascular medicine has achieved many breakthroughs, ischemic heart diseases (IHD), such as myocardial infarction (MI) and heart failure (HF) remain among the most important health challenges worldwide [3]. MI, due to coronary artery disease (CAD), is currently one of the major contributors to the development of HF [4]. In a multicenter heart failure treatment trial, comprising over 20,000 patients, CAD was the underlying etiology in 68 % of cases [5]. Aggressive treatment strategies focusing on early reperfusion of the culprit artery and modifying the risk profile of patients improve mortality after MI [6]. However if reperfusion is not accomplished in time, tissue ischemia is followed by irreversible loss of cardiomyocytes accompanied by scar formation, leading to progressive ventricular remodeling and HF [4]. The annual incidence of HF ranges from 1 to 5 per 1000 people and doubles each decade over the age of 45 [7]. Despite advances in medical and surgical treatment, prognosis remains poor with a median survival after establishing the diagnosis of HF of only 1.7 years in men and 3.2 years in women [8,9]. Although valvular heart disease (VHD) is less frequent than CAD it is an increasing problem often requiring intervention. Rheumatic disease used to be the most frequent etiology of valve disease in previous decades, but nowadays degenerative origin is by far the most frequent cause in the Western world [10,11]. The two most frequent valvular disorders are currently calcified aortic stenosis (AS) and mitral regurgitation (MR), whereas aortic regurgitation (AR) and mitral stenosis (MS) are less frequent [12]. AS is the most common valvular disease in the western world [13]. It is associated with significant morbidity and mortality, and valve replacement is often required [14]. As AS is a typical problem of the elderly patient, the number of patients increases due to the ageing population [14]. Surgical replacement of diseased human heart valves by mechanical and tissue valve substitutes is now commonplace, with approximately 275000 valve replacements carried out worldwide every year [15]. Importantly, it is estimated that the prevalence of heart valve disease, and as a consequence the need for replacement will increase threefold over the next 50 years [16]. However, although valve replacement improves the prognosis of patients with valvular disease, current valve prostheses have serious limitations and inherently lack the sophisticated design and function of the native valve. Both the epidemic proportions of CVD and the growing number of patients with valvular diseases warrant the development of alternative treatment regimens.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

Regen e rative M e d i c i n e Regenerative Medicine (RM) is an emerging field focusing on unraveling the mechanism underlying complex regeneration processes in nature with the goal of providing elements required for in vivo repair, to create replacements interacting with the living body, and to stimulate the body’s own regenerative capacity [17,18]. RM aims at using strategies and methodologies used in curing conditions as opposed to treating them [19]. Current RM has its origin in observations made in nature that certain species have regenerative capabilities. For example, urodele amphibians, display the ability to regenerate a variety of body parts, including limbs, tail, jaw and retina [20]. Probably the best known example of this phenomenon is the tail regeneration in amphibians, where the missing part, including all differentiated tissue types, is re-grown from the site of transection [21] (Figure 1). This phenomenon has also been described in invertebrates, where e.g. transection of a planarian worm results in a regenerated tail structure [20]. Furthermore, examples of complex tissue regeneration are observed in higher species. Examples are the seasonal re-growth of deer antlers [22], and the distal fingertip regeneration in children [23]. Moreover, at the first two trimesters, human skin wound healing is characterized by complete regeneration and the absence of scar formation [24]. Interestingly, this ability to regenerate is lost in the third trimester. Ultimately, RM should provide treatment options for diseases and/or injuries not treatable by conventional medicine and even new biologic drug therapies. The field of RM encompasses three areas of technology, 1) tissue engineering; 2) stem cell treatment; and 3) cloning [25]. Tissue engineering uses the principles of cell transplantation and scaffold design, in order to develop biological substitutes able to restore or maintain normal function.

Figure 1. Regeneration of the forelimb in an amphibian after amputation at distal (mid-radius and ulna; shown at left) or proximal (mid-humerus; shown at right) sites. The original limb is shown at the top and the regenerated limb at the bottom of the image. The photographs were taken at 7, 21, 25, 28, 32, 42 and 70 days after amputation. (adapted from Brockes et al., Science 1997;276:81-7)

11

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

12

REGENERATIVE MEDICINE IN HEART VALVE DISEASE: TISSUE ENGINEERING APPROACHES Although heart valve repair is currently the preferred method of treating patients with severe valvular disease [26,27], a substantial number of valves are (especially in case of aortic valve disease) not suitable for repair and require replacement. Therefore, valve replacement is the most common surgical procedure in patients with advanced valvular disease [28]. Since their clinical implementation in 1965, extensive experience has been gained with mechanical and bioprosthetic valve prostheses [29]. However, although valve prostheses improve survival and quality of life [30], each specific prosthetic valve has limitations (figure 2).

M e chan ical Val ves Mechanical prostheses, such as caged ball, tilting disk, or hinged semicircular rigid flap valves have excellent durability but include a high risk of thromboembolic complications (approximately 4 % per patient a year [31]) necessitating lifelong anticoagulant therapy [32]. Therefore, the use of mechanical valves is relatively contraindicated among patients with an active lifestyle or a wish for pregnancy. Furthermore flow-profiles across the valves are (depending on the design of the valve) often far less than optimal.

Bio p rosth eti c Val ves Currently there are three types of commercially available bioprosthetic valves: (1) porcine xenograft valves, (2) bovine pericardial valves, and (3) allograft or homograft valves [33]. The porcine xenograft valve is fabricated from an intact pig aortic valve preserved in low concentration glutaraldehyde solution and mounted on a prosthetic frame. The bovine pericardial valve consists of 3 separate pieces of glutaraldehyde treated calf pericardium attached to a stent and sewing cuff, and is configured very similar to that of the porcine xenograft. Both types of valves are crosslinked in glutaraldehyde and treated with various other chemical agents to reduce their antigenicity and to minimize their tendency to calcify [34]. Human tissue valve prostheses have been introduced as homograft valves, defined as a valve from an individual or equal species. They are usually cryopreserved as complete aortic or pulmonary roots and trimmed before implantation [35,36]. One of the major limitations of the porcine and bovine xenografts is their progressive structural failure, which is more often seen in younger recipients [37,38,39]. Although, homograft valves include a greater durability and demonstrate better resistance to infective endocarditis [40], their availability is limited due to organ scarcity.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

All clinically available valve prostheses basically represent non-viable structures and lack the ability to grow repair, or to remodel [41]. This imposes potentially severe problems, especially in the pediatric population. Moreover, with the currently available valves, approximately 60 % of the recipients will suffer from a serious valve-related complication within 10 years postoperatively [42].

Figure 2. Different artificial valve prostheses. From left to right; S. Jude Medical® and Starr-Edwards aortic valve mechanical valves, Medtronic Freestyle stentless porcine bioprosthesis, human cryopreserved homograft, and Carpentier-Edwards PERIMOUNT bovine tissue heart valve.

Tissu e E n g i n e e r i n g Tissue engineering of heart valves aims to overcome the above described limitations of currently available heart valve prostheses. Tissue engineering is one of the main components of regenerative medicine, and follows the principles of cell transplantation, materials science, and engineering towards the development of biological substitutes that can restore, maintain, or improve normal function [25,43]. A tissue engineered heart valve potentially has several advantages, like non-thrombogeneicity, resistance to infection and cellular viability. It promises to be a living implant with a potential to grow and last a lifetime, like most native valves do [33]. The initial and crucial step is to obtain an optimal valve matrix. Tissue engineering strategies for heart valves therefore can be divided into two categories: engineering of completely artificial valves from biodegradable synthetic polymers, and decellularization of biological heart valves from allogeneic or xenogeneic donors, seeded with autologous cells either in vitro or in vivo. DIFFERENT APPROACHES TO VALVULAR TISSUE ENGINEERING

Synthetic Scaffolds Biodegradable materials break down as a result of macromolecular degradation with dispersal in vivo, but are not eliminated from the body [44]. A biodegradable valve scaffold can thus provide a temporary matrix until repopulating cells produce their own matrix proteins. The use of biodegradable materials is especially appealing from a developmental standpoint, with the ability to create a variety of sizes and lengths in order to tailor the valve for individual patients.

13

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

14

Several biodegradable polymers have been demonstrated to be useful in cardiovascular tissue engineering. Polyglycolic acid (PGA) and polylactic acid (PLA) and their copolymers have been widely used in TE [45]. However, these materials have been abandoned for heart valve TE because of their stiffness and short degradation times. Furthermore they cause a significant local inflammatory response [46,47]. Recent advances include more compliant materials like polyhydroxyalkanoates (PHAs) [48] or combinations of PGA and poly-4-hydroxybutyrate (P4HB) [49,50]. However, in most animal studies, these scaffolds have been implanted in the pulmonary artery only, because the degrading scaffold cannot sustain the higher LV pressures until the new valve has been regenerated [49,50,51]. Furthermore, none of these scaffolds have been used in a clinical setting.

Decellularized Biological Scaffolds Another approach to heart valve TE involves the (bio)-chemical decellularization of homo- or xenografts. Xenogeneic and allogeneic cellular antigens are capable of inducing an inflammatory response or immune-mediated tissue rejection, whereas components of the extracellular matrix (ECM) are well conserved among species and are well tolerated even by xenogeneic recipients [52]. Decellularized heart valves are particularly attractive because of the natural anatomical geometry, which may serve as a template for cellular attachment and retaining many of the mechanical and structural properties of native valves such as tensile strength and ECM composition [53]. The goal of a decellularization protocol is to remove all cellular and nuclear material while minimizing any adverse effect on the composition, biological activity, and mechanical integrity of the remaining ECM [54]. Various decellularization methodologies have been explored for the purpose of heart valve tissue engineering.

Enzymatic Decellularization Methods Enzymatic techniques for heart valve decellularization include the use of protease digestion and nucleases. The proteolyic enzyme Trypsin is one of the most commonly used enzymes in valvular decellularization protocols. It displays its effect by cleaving the peptide bonds on the carbon side of arginine and lysine if the next residue is not praline [54]. Nucleases, such as endonucleases and exonucleases, catalyze the hydrolysis of the interior and terminal bonds of ribonucleotide or deoxyribonucleotide chains, ultimately leading to the degradation of RNA and DNA [54]. Steinhoff et al, demonstrated that a 48-hour Trypsin/EDTA treatment resulted in an almost complete decellularization of ovine pulmonary valve conduits [55]. This protocol was later also successfully applied to human aortic and pulmonary allografts, resulting in complete decellularization [56]. However, others reported (applying the same protocol) only incomplete decellularization of porcine aortic and pulmonary roots [57,58].

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

Detergent Decellularization Methods Detergents are amphipathic molecules that can solubilize membrane proteins by creating a mimic of the natural lipid bilayer environment normally inhabited by the protein [59], and can therefore disrupt cell membranes and bonds responsible for intercellular and extracellular connections. Detergents are classified according to their structure, and generally fall into three categories: 1)

Non-ionic detergents have a relatively mild effect on the ECM structure, and have therefore

been used extensively in decellularization protocols. Non-ionic detergents disrupt lipid-lipid and lipid-protein interactions, but not protein-protein interactions, leaving proteins in their functional conformation [59]. Non-ionic detergents include Triton X-100 and the Tween family of agents. The group of Bader, was the first to use Triton X-100 for the decellularization of porcine aortic valves [60]. Also Numata et al. showed sufficient cellular removal in porcine pulmonary allografts after Triton X-100 treatment [61]. However, Kim et al. could still detect cells after Triton X-100 treatment in porcine heart valve leaflets [62]. Furthermore, Booth et al. also could not achieve complete decellularization of porcine aortic valves after Triton X-100 or Tween 20 treatment [63]. 2)

Ionic detergents effectively solubilize both the cytoplasmatic and nuclear cellular mem-

branes, but have a propensity to denature proteins by disruption of protein-protein interactions [59]. For heart valve decellularization, the two most commonly used ionic-detergents are sodium dodecyl sulfate (SDS) and sodium deoxycholate. Several authors have demonstrated complete decellularization of porcine pulmonary [63,62,64] and aortic valves [57,58] after SDS treatment. Ueda et al. used a combination of SDS and the non-ionic detergent Triton X-100 with successful decellularization [65]. Sodium deoxycholate treatment has been successfully applied in porcine aortic [63] and pulmonary valve leaflets [66]. 3)

Combination of an ionic and a non-ionic substance has been used to decellularize valve

preparations. For example, sodium deoxycholate with Triton X-100 resulted in complete decellularization of porcine aortic [57,58,67] and pulmonary valve conduits [57,58]. COMMERCIALLY AVAILABLE PATENTED DECELLULARIZATION METHODS

To date only two commercially available decellularized heart valve prostheses are available. CryoLife Inc from Kennesaw Ga, has developed a patented procedure called the SynerGraft technology that includes cell lysis in hypotonic solution, enzymatic digestion of nucleic acids, and washout in isotonic, neutral buffer, which is able to produce acellular porcine [68] and aortic [69] and pulmonary [70] human homografts. In addition, recently also the AutoTissue LTdtm method [71] has become available, which involves the use of a solution of 1 % deoxycholic acid and ethanol 70 %, and is able to produce completely acellular cryopreserved human homografts [72].

15

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

16

EFFECTS OF DECELLULARIZATION ON THE EXTRACELLULAR MATRIX

Although the focus of the different cell extraction protocols is primarily its efficiency in removing all cellular elements, it should be noted that chemically induced cell extraction can be quite detrimental to the valvular matrix [73]. Decllularisation can result in degradation or denaturing of the matrix proteins or leave toxic residues which can detrimentally effect recellularization and mechanical function [74]. In addition, it was recently demonstrated that the remaining extracellular matrix composition could be involved in early calcification and incomplete repopulation of transplanted decellularized valves [75]. Different cell extraction protocols could also all have different effects on the remaining ECM [63,76]. CELL SEEDING OF VALVE SCAFFOLDS

There is variability in potential strategies and sources of cells. They include the repopulation of scaffolds with appropriate cell types in vitro, or the use of synthetic or decellularized scaffolds, without in vitro repopulation prior to implantation, which can attract endogenous cells. In the later approach the repopulation of the valve scaffold takes place in vivo after the implantation.

Pre-seeding of Valve Scaffolds Different cell types have been successfully applied for reseeding of synthetic and biological scaffolds. Venous endothelial cells have been preferred due to the simple access to peripheral venous segments. Porcine and goat jugular [62,64] and human saphenous [60,56,58] endothelial cells have also been used for the in vitro repopulation of biological scaffolds. However, other authors have used porcine femoral artery endothelial cells [61], and myofibroblasts and endothelial cells derived from the wall of carotid arteries have been used in synthetic [77,49] and acellular [55] matrix scaffolds. More recently, also ovine and human mesenchymal stem cells have been investigated for the feasibility of tissue engineering [50,78].

Non-seeded Valve Scaffolds Leyh et al demonstrated that decellularized porcine xenograft valves were repopulated in vivo within 24 weeks with interstitial myofibroblasts and endothelial cells after transplantation in the pulmonary circulation of a sheep model [75]. In Contrast, Goldstein et al. found progressive recellularisation of the matrix with fibroblasts and myofibroblasts without endothelialization in the same model [79]. Others showed partial degeneration of unseeded porcine valves in contrast to valves that were in vitro seeded with myofibroblasts and endothelial, were complete histological reconstitution was observed [55]. Interestingly Kim et al. found in a goat model no histological differences between decellularized porcine valve leaflets with or without seeding of autologous endothelial cells [64]. Therefore, role and necessity of in vitro pre-seeding remains controversial.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

C l i n i cal E x p e r i e n ce w ith Tissu e E n g i n e e re d Val ves Despite promising results in animal experiments, to date there have been only a limited number of clinical studies. Dohmen et al described in 2002 the first successful implantation of a 1 % deoxycholic acid decellularized cryopreserved pulmonary allograft in the right ventricular outflow tract (RVOT) of a 43 year old patient that underwent a Ross procedure, with good clinical outcome at one year of follow-up [80]. These valve grafts were seeded with autologous vascular endothelial cell, and conditioned in a bioreactor [80]. The results of the first 50 consecutive patients receiving these decellularized xenografts also showed positive results, with normal right ventricular-pulmonary artery pressure gradients [81]. This method (AutoTissue Ltdtm) was modified, with the addition of ethanol 70 % to the protocol [71]. In a series of 20 patients receiving decellularized (n = 11) or cryopreserved (n = 9) allografts during a Ross procedure, the decellularized valves showed normal function up to 18 months of follow-up and showed a substantial reduction in immune-response compared to cryopreserved valves [72]. To date more than 171 patients underwent a Ross procedure with reconstruction of the RVOT using decellularized allografts or xenografts [82], and its value has recently also been demonstrated in more complex congenital aortic valve disease [83]. The commercially available SynerGraft (CryoLife Inc) cryopreserved decellularized pulmonary homograft valve has been successfully implanted in the RVOT of 22 patients with evidence for reduction of an immunologic response, but no difference in transvalvular pressure gradients, compared to cryopreserved pulmonary homografts [70]. Interestingly, in one patient who died 5 weeks after SynerGraft pulmonary homograft implantation, histopathology showed integrity of the ECM with only an early nonspecific inflammatory phase of recellularization, with no signs of any specific immune response [84]. However, whether this observation precedes a possible repopulation of the valves remains to be determined. More recently, also acellular aortic valve conduit homografts have been developed with the SynerGraft technology, which after transplantation in an initial 22 patients showed low transvalvular gradients similar to cryopreserved homografts, and almost absent allosensitization as determined by panel reactive antibody measurements [69]. Importantly, Miller et al demonstrated that in an aortic SynerGraft homograft, explanted after 2 years, due to a cardiac transplantation, endothelial and smooth muscle cell differentiation in a subset of cells, with a virtual absence of calcification and inflammation [85]. In contrast, Simon et al reported in the first series of 4 patients receiving SynerGraft decellularized porcine xenograft valves for RVOT reconstruction, early severe valvular complications, resulting in the dead of 3 patients [68]. Histological examination revealed severe inflammation with degeneration of the leaflets and vascular wall, indicating that the xenogeneic collagen matrix is capable of eliciting a strong immune response in humans [68].

17

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

18

REGENERATIVE MEDICINE IN ISCHEMIC HEART DISEASE: CELL BASED THERAPY Myocardial infarction (MI) results in acute ischemia, and is accompanied by an irreversible loss of cardiac muscle with formation of scar tissue, eventually leading to progressive ventricular remodeling and heart failure [4]. None of the current available therapies address the underlying cause of the remodeling process (damage of myocardium and vasculature in the infarcted area). Furthermore, no currently available treatment option is able to restore viability and function of necrotic myocardium. Although cardiac transplantation and the use of mechanical LV assist devices are an option for patients with end-stage heart-failure, they are associated by a high comorbidity or limited effectiveness [86,87]. Recent years, several animal studies and a few clinical trials have been published using stem- or progenitor cells as potential therapeutic options ischemic heart disease.

Stem an d Progenito r Cells The zygote, and the cells derived from the first two divisions are called totipotent because they can form both the embryo and parts of the placenta, and are considered the most primitive cells in an organism [88]. In the embryo, the blastocyst then gives rise to the embryonic stem cells that can form all the cell types derived from the three germ layers, and are called pluripotent. Stem cells are primitive cells that are by definition clonogenic, capable of self-renewal and differentiation into all lineages (pluripotent) [89]. Most stem cells found in adult organs are already committed to one or more specific lineages and are therefore referred to as multipotent progenitor cells. The initial thought was that the progeny of a stem cell undergoes an irreversible change during early embryogenesis, making them committed to a specific lineage of undifferentiated progenitor cells. However, recent evidence suggested that adult progenitor cells can differentiate into mature cell types outside their original lineage, in response to specific cues, a characteristic called plasticity [90].

D iffe re nt Ce l l Ty p es fo r Regen e rat i ve Th e rapy Potentially, a variety of stem and progenitor cell populations can be used for cardiac regeneration, each with its own profile of advantages, limitations, ethical considerations and practical use in the clinical setting.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

EMBRYONIC STEM CELLS

Embryonic stem (ES) cells are totipotent stem cells derived from the inner cellular mass of the blastocyst-stage embryo. In contrast to other progenitor cells, ES can give rise to all cell types of the body, including cardiomyocytes [91]. ES cells are able to restore cardiac function in animal models of MI with evidence for long term benefit [92]. However, multiple issues prevent the therapeutic application of ES cells in patients, including ethical issues centering on the use of human embryos, the observation of teratoma formation in immune compromised animals [93], and the need to use allogeneic cells. Possibly, the application of somatic cell nuclear transfer (SCNT), involving the transfer of the nucleus of a somatic cell into an enucleated donor oocyte (“therapeutic cloning”), may provide a means of producing immune-compatible Es cells for the treatment of IHD [94]. SKELETAL MYOBLASTS

Skeletal myoblasts (SM) or satellite cells are progenitor cells located under the basal membrane of muscle fibers, where they serve as a reservoir of regenerative cells for skeletal tissue [95,96]. SMs can be easily obtained from patients and cultured in vitro where they maintain native morphological and functional characteristics [97]. These cells can engraft as shown in clinical and experimental studies of MI with improvement of systolic and diastolic cardiac function [98,99,100,101,102]. However, SMs fail to electromechanically couple with resident cardiomyocytes when transplanted into ischemic myocardium [103,104]. This may result in the formation of arrythmogenic foci and the occurrence of life threatening arrhythmias [105,106]. Therefore, new options to enhance electromechanical and functional coupling need to be explored for these cells to be used in cardiac cell-therapy. CARDIAC STEM CELLS

The heart has been traditionally viewed as a static organ, without the capability of repairing damage. However, evidence has accumulated suggesting that the heart contains a pool of cardiac stem cells (CPCs) that display an endogenous regenerative potential [107]. Over the past few years these cells have been phenotyped using different antigenic approaches. Beltrami et al. isolated lineage negative c-kit positive (lin-/c-kit+) cells from the adult rat heart that could differentiate into cardiomyocytes, smooth muscle cells and endothelial cells in vitro, and are able to regenerate myocardium after MI [108]. Later, also murine stem cell antigen-1 (Sca-1) positive cells that were either CD31 positive [109] or negative [110] were found, with in vitro and in vivo myocardial differentiation potential. Messina et al reported on the identification of cardiospheres from murine hearts, expressing SCA-1, c-kit, KDR and CD31, and capable of transdifferentiation in vitro, as well as myocardial and vascular regeneration after MI [111]. Also several authors reported on a pool of cardiac cells that were identified by their ability to efflux the Hoechst 33342 dye;

19

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

20

these cells were found to express SCA-1 but not CD31, and had the capability to differentiate into cardiomyocytes in vitro [112,113]. Finally, Laugwitz et al isolated a population of cardiac stem cells from post natal mouse hearts using the isl-1 transcription factor [114]. When co-cultured with neonatal cardiomyocytes these cells showed electrical and contractile properties evocative of neonatal cardiomyocytes [114]. The possibility of isolation of CSC from adult tissue, together with their potent differentiation capacity may contribute to future human autologous cell transplantation studies. However, to date there have been no clinical trials using human cardiac stem cells. HEMATOPOIETIC STEM CELLS

Hematopoietic stem cells are the best characterized stem cell populations in bone marrow [115]. Hematopoietic stem cells are negative for lineage markers (lin-), but express the hematopoietic markers CD34, CD45 and CD133, and the stem cell factor c-kit. In a landmark study in 2001, the group of Anversa first reported that murine lin-/c-kit+ bone marrow derived cells could differentiate into cardiomyocytes after MI [116]. However, other groups demonstrated that HSCs do not readily acquire a cardiac phenotype in the ischemic heart, but adopted hematopoietic fates, despite the observed attenuation in ventricular function [117,118]. More recently Kajstura and colleagues provided further evidence that c-kit+ bone marrow cells, when injected properly, can differentiate into cardiomyocytes and coronary vessels, with no differentiation into hematopoietic lineages [119]. However, cardiac differentiation of HSCs still remains controversial. Moreover, all of the above described studies have been performed with murine HSCs, and currently no studies with the human counterpart have been performed. Furthermore, the low percentage of HSC in BM and the lack of clinically usable expansion methods limit the therapeutic application of HSCs. SIDE POPULATION CELLS

Side population (SP) cells are a subset of HSCs that can give rise to all hematopoietic cells and represent only 0.05 % of the bone marrow cell population [120]. SP cells are characterized by their ability to exclude the Hoechst dye via the ABCG2 transporter on the membrane [121]. Although they are isolated independently of the antigenic markers, they are enriched for CD34-/low, c-kit and Sca-1+ [122]. SP cells can migrate into cardiac myofibers and small vessels two to four weeks after myocardial infarction in a mouse model [123]. Future studies are needed to further elucidate this cell type in cardiac regeneration. ENDOTHELIAL PROGENITOR CELLS

Endothelial progenitor cells (EPCs) share phenotypic and functional characteristics similar to the fetal hemangioblast [124,125]. These bone marrow derived cells express some cell surface

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

markers characteristic of mature endothelium (VEGFR2, Tie-2) and hematopoietic cell surface markers (CD34), and are negative for CD45 [126,127]. In addition, EPCs have also been shown to express CD133, a marker of immature HSC and EPCs [128]. EPCs are released from the BM and circulate in the peripheral blood, home to various tissues into foci of neovascularization [129], and can participate in the regeneration of tissue damaged by ischemia [130]. In rat myocardial infarction models, injection of human CD34+ EPC can induce blood vessel formation in the infarcted scar, and proliferation of the existing vasculature [131,124]. Currently, clinical trials using CD34+ BM derived EPC transplantation for angiogenesis after MI are in progress [132]. Of interest, recent studies showed that EPC function and differentiation capacity is inversely correlated with risk factors for IHD [133]. MESENCHYMAL STEM CELLS

Mesenchymal stem cells (MSCs) are rare population of non-hematopoietic multipotent stem cells that can differentiate into a variety of mesodermal cell types, including muscle, fibroblasts, bone, tendon, ligament and adipose tissue [134]. MSCs are isolated by culture flask adherence and are, unlike their hematopoietic counterparts, negative for CD34 and CD45, but characteristically express CD29, CD44, CD71, CD90, CD105, CD106, CD120a, CD124, SH2, SH3 and SH4 [135]. MSCs can differentiate into cardiomyocyte cells after appropriate stimulation in vitro [136]. Moreover, when MSC were injected into the healthy mouse heart, differentiation into cardiomyocytes was demonstrated [137]. Furthermore direct injection of MSCs in the infarcted heart has been shown to improve left ventricular (LV) function in rats [138,139], dogs [140] and pigs [141,142]. Several possibilities have been proposed in the interpretation of the observed positive effects on the LV function after myocardial infarction. They include the differentiation of hMSCs into cardiomyocytes [141,143] and vascular cells [140,143] or a paracrine effect mediated by the MSCs [138,139]. Chen et al demonstrated in the only clinical MSC study to date that intra-coronary injection after acute MI resulted in a significant improvement in global and regional LV function compared to the medium treated patients [144]. Importantly, because MSC can be expanded rapidly in vitro, and are reported to avoid rejection by lacking MHC-II and B-7 costimulatory molecule expression [145], they might also be used in an allogeneic setting. MULTIPOTENT ADULT PROGENITOR CELLS

A subpopulation of the BM-derived stem cell that co-purify with MSCs, originally described as mesodermal progenitor cells, was because of its with spectrum of differentiation renamed as multipotent adult progenitor cells (MAPCs) [146]. MAPCs are CD34-/133+/Flk1+ and have been postulated to comprise the principle endothelial progenitor in human BM [147]. Furthermore, in vitro MAPCs can be expanded indefinitely and differentiate into all three embryonic germ layers

21

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

22

[148,149]. When injected into the tibialis anterior muscle of NOD/Scid mice, human MAPCs have been demonstrated to differentiate into myocytes[150]. However, to date MAPCs have not been tested in animal models of IHD. BONE MARROW-DERIVED MULTIPOTENT STEM CELLS

Recently multipotent BMCs, which can differentiate into all three germ layers, were clonally isolated from human BM [151]. These cells are currently referred to as human bone marrow-derived multipotent stem cells (BMSCs), and do not express any of the characteristic marker panels (e.g. CD34, CD44, CD90, CD105 and CD117) defining HSC, MSCs and MAPCs [151]. hBMSCs were found to acquire a cardiomyocyte phenotype in the ischemic mouse myocardium with attenuation of ventricular function [151]. Difficulties in culturing techniques observed with hBMSCs have to be overcome before this cell type could be used in a clinical setting. ADIPOSE TISSUE DERIVED STEM CELLS

Adipose tissue contains different progenitor cells, including HSCs SPs and MSCs [152,153,154]. Recently it was demonstrated that adipose tissue derived stromal cells, if cultured under specific conditions, could be differentiated into beating cells with a cardiomyocyte phenotype [155]. Furthermore, adipose lineage cells can function as progenitors for endothelial cells that enhanced the neovascularisation in ischemic tissue [156]. However, the exact molecular hallmarks of these cells remain to be established. The abundance and ease of excess to fat makes adipose tissue a potentially useful cell source in future cell-therapy approaches. UMBILICAL CORD-DERIVED CELLS

Several reports have indicated that the umbilical cord blood (UCB) contains both hematopoietic and mesenchymal progenitor cells [157,158]. Furthermore, MSCs have been isolated from the Wharton’s jelly of the human umbilical cord [159]. Recently a population of cells from human UCB, called unrestricted somatic stem cells, has been described that was negative for c-kit, CD34 and CD45, and was capable of differentiating in a variety of tissues, including cardiomyocytes [160]. Interestingly, after injection in the ischemic heart of immuno-suppressed pigs these human unrestricted somatic stem cells improved regional and global function, improved perfusion, and reduced infarct size [161]. Furthermore, UCB derived CD34+ cells were demonstrated to improve cardiac function in a rat model of acute MI after injection in the peri-infarct area, as compared to medium treated animals [162]. However, to date, no clinical of UCB derived cells have been reported.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

EPICARDIAL-DERIVED CELLS

The epicardium is a structure that comprises the outermost layer of the heart. It consists of mesothelial cells that during embryogenesis originate from the extracardiac pro epicardial organ (PEO) [163]. A subset of these epicardial cells can undergo epithelial-mesenchymal transformation (EMT), and migrate into the subepicardial, myocardial and subendocardial layers of the primitive heart, as well as the atrioventricular cushions [164,165,166,167]. These cells have been named EPDCs, and differentiate into perivascular and cardiac interstial fibroblasts, as well as into coronary smooth muscle cells [164,166,168,169]. Furthermore, EPDCs do not only physically contribute to the developing heart; they also have a modulatory role in cardiogenesis [170,171]. EPDCs are involved in myocardial compaction [172,173], Purkinje fiber development [174] and coronary vessel formation [175,176]. Although EPDCs are crucial for proper cardiogenesis in the developing heart, limited information is available on the role of EPDCs in fetal and postnatal hearts. Recently, an adult epicardial rat cell line was described that could be induced to undergo EMT in vitro, and from which a fraction had the capacity to form SMCs [177]. Furthermore, van Tuyn et al. demonstrated that human adult epicardial cells spontaneously undergo EMT early during ex vivo culture, express the cardiac markers GATA4 and cardiac troponin T, and obtain characteristics of SMCs after proper stimulation [178]. These results indicate that adult EPDCs retain part of their differentiation capacity present during embryonic development, making them progenitor cells. Recently, Winter et al. therefore hypothesized that adult human EPDC might also recapitulate their embryonic capacities in the ischemic myocardium, thereby positively modifying the surrounding myocardium [171]. However, to date no in vivo transplantation studies have been performed.

Card iac Cell Th erapy in Hu mans The initial observed beneficial effects observed after different cell based therapies in small and large animal models generated excitement, and prompted investigators to initiate the first clinical studies. So far, cell-based therapies have been performed in patients with acute myocardial infarction, chronic ischemia and heart failure. In most studies, investigators have chosen the pragmatic approach of using bone-marrow mononuclear cell (BM-MNC) preparations containing different stem and progenitor cell populations. CELL THERAPY IN ACUTE MYOCARDIAL INFARCTION

The use of BM-MNCs has been most extensively investigated in the setting of acute MI. Several small clinical studies, which all included patients with acute MI who had undergone primary PTCA and stent implantation, with infusion of the cells via the intra-coronary route showed

23

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

24

beneficial effects. However, most of these studies lacked an adequate control group and were not randomized in a blinded fashion. Several larger scales randomized, controlled trails evaluating the therapeutic effect of BM-MNC transplantation for acute myocardial infarction have been performed recently, all with mixed results. Schachinger and colleagues reported in the largest BM-MNC therapy study to date (204 patients were randomized) the results of the Reinfusion of Enriched Progenitor Cells and Infarct Remodeling in Acute Myocardial Infarction (REPAIR-AMI) trial, a double-blind and fully controlled multicenter study that assessed the effect of intra-coronary infusion of BM-MNCs after successful PTCA for acute MI [179]. They observed a modest, but significant improvement in LVEF at 4 months in the BM-MNC treated patients as compared to those given placebo [179]. Of interest in the also randomized, but not blinded, Bone Marrow Transfer to Enhance ST-Elevation Infarct Regeneration (BOOST) trial, the relative improvement in LVEF observed 6 months after BM-MNC transplantation was no longer significant at 18 months, due to continuous improvement in the control group [180]. In contrast, in the double-blind randomized controlled trial by Janssens et al [181], no difference between control and treated groups was observed 4 months after BM-MNC transplantation. Furthermore, in the also recently published Autologous Stem Cell Transplantation in Acute Myocardial Infarction (ASTAMI) trial no significant difference in LVEF was found at 6 months after BM-MNC transplantation, as compared with the control group [182]. The observed discrepancies between different studies are at present not clear, but may relate to the methods used for cell preparation [183]. Several studies also assessed the use of selected bone marrow cells in the setting of acute MI. In the TOPCARE-AMI trial, BM-MNCs were also compared with circulating blood-derived progenitor cells (mainly EPCs), with an improvement in LVEF in both the groups [184]. In another clinical trial, CD133+ BM cells that were isolated with a mouse antibody were injected into the infarct related artery [185]. Although an improvement of global contractility was observed, several of the patients developed a significant in-stent restenosis [185,186]. The therapeutic effects of intracoronary MSC transplantation after acute MI have been investigated by Chen et al, who demonstrated improvement of LV function and an attenuation of LV end-diastolic volume, compared with a randomized control group that received saline [187]. CELL THERAPY IN PATIENTS WITH CHRONIC MYOCARDIAL ISCHEMIA

Due to the diffuse nature of their disease, a subset of patients with coronary artery disease and chronic myocardial ischemia are not suitable for revascularization, and therefore experience refractory angina pectoris. Several small studies in patients with CAD have used trans-epicardial [188] and trans-endocardial [189,190,191] injections of BM-MNC in this group of patients, with improvements of anginal symptoms, exercise capacity, regional tissue perfusion and LV systolic function [188,189,190,191]. Beeres et al reported on the sustained effect on anginal symptoms,

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

perfusion and LV function for up to 12 months of follow up [192]. These studies therefore demonstrate the feasibility of BN-MNC injection in patients with chronic myocardial ischemia. However, none of these studies included a randomized control group. To determine the true efficacy of this approach, it should be tested in prospective randomized trails. CELL THERAPY IN PATIENTS WITH CHRONIC HEART FAILURE

Several small clinical studies investigated the safety and feasibility of skeletal myoblast transplantation in LV scar tissue during CABG, all with significant improvements in LV function [105,106,193]. Furthermore, Smits and colleagues used a trans-endocardial approach to inject myoblasts as a stand-alone procedure in five patients with chronic heart failure [101], and observed an improvement in LVEF. However, in all of these initial studies episodes of ventricular tachycardies, that necessitated ICD placement, were observed [193,105,106,101]. It is therefore likely that skeletal myoblast transplantation increase the risk of ventricular tachycardies in patients with chronic heart failure. Another group used CD133+ BMC that were injected transepicardially into the infarcted borderzone of patients undergoing CABG 3-12 weeks after MI[194]. Perfusion and LVEF were improved after 6-8 months, however, in contrast to the myoblast studies, no ventriculair arrhythmias were observed [194].More recently, Assmus et al evaluated in The Transplantation of Progenitor Cells and Recovery of LV Function in Patients with Chronic Ischemic Heart Disease (TOPCARE-CHD) trial the effects of BMC or progenitor cells derived from the circulating blood (CPC) in patients who had a MI on average more than 6 years ago, and observed a moderate but significant improvement in the LVEF after 3 months [195].

M echanisms M ediating the Therap eutic Effects of Stem or Progenitor Cells Although almost all cell-based therapies have provided evidence for functional improvement or amelioration of LV remodeling, there has been much controversy surrounding the underlying mechanism. In the original concept, stem or progenitor cells were proposed to repair the damaged heart via transdifferentiation into cardiomyocytes [116]. Although initial studies using different stem and progenitor cells demonstrated that these cells could differentiate into cardiomyocytes, especially in the case of BM-derived cells, the ability of crossing lineage boundaries is under debate. As mentioned earlier, plasticity indicates the ability of stem cells to differentiate into cell types other than their cell origin[89]. Although the group of Anversa demonstrated transdifferentiation of BM derived stem cells [119,116], this could not be reproduced by others [118,117]. Other groups demonstrated that the observed plasticity and repair of BM cells after transplantation in the ischemic heart is due to fusion with existing myocytes [196]. However, even if stem and progenitor cells could differentiate into functional cardiomyocytes, this alone would not be

25

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

26

Figure 3. Schematic diagram of proposed mechanisms mediating the therapeutic effects of stem or progenitor cell transplantation.

enough to explain their observed beneficial effects. Murry and colleagues recently calculated that the cardiomyocyte deficit in MI induced heart failure is approximately one billion cardiomyocytes, and true regeneration would therefore require this amount of stem cells that are all synchronously contracting via electromechanical junctions with the host cardiomyocytes [197]. Although with current techniques this remains impossible to achieve, beneficial effect after celltherapy are observed. Interestingly, even fibroblasts have been shown to improve the diastolic function of the ischemic rabbit heart [198]. Therefore, several alternative mechanisms of action have been proposed (Figure 3). Skeletal myoblasts have been demonstrated to improve the passive mechanical properties of the infracted region, resulting in the amelioration of ventricular remodeling [199]. Other investigators demonstrated in a rat model by atomic force microscopy that human MSC engraftment attenuates post-infarction remodeling by softening of the border zone area, thereby improving the elastic moduli resulting in a more compliant infarct scar [200]. Furthermore, BM derived EPC have been proposed to structurally enhance tissue perfusion by differentiating into endothelial cells at sites of neovascularization [201,124]. Another possibility is the so-called paracrine effect, referring to the production of local signal-

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

ing molecules by stem cells that act on the host myocardium [202]. It is becoming increasingly clear that cell therapy can also improve cardiac function by promoting angiogenesis [203,204], cardiomyogenesis [205] and reducing host myocardial apoptosis [124,206,207]. Interestingly, Gnecchi and colleagues demonstrated that most of the benefit observed after MSC transplantation could also be obtained after injecting the cell-free supernatant of MSC cell cultures [208]. Finally, other investigators suggest that cell therapy may stimulate endogenous resident stem cell migration and proliferation, facilitating the ability of the heart to regenerate itself [209]. At present, due to differences in animal models, delivery modes and cell types used, the relative contribution of each component is not clear, moreover, the beneficial effects observed after cell therapy could be due to multiple mechanisms.

27

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

28

CARDIAC PHENOT YPING IN REGENERATIVE MEDICINE: DIFFERENT METHODS FOR THE MURINE HEART Recent developments in transgeneic mouse models of cardiac dysfunction have led to an increase in the use of these models to study cardiac function and disease. In addition, surgical techniques used to mimic cardiac pathology are becoming more common in laboratories around the world. Models of coronary artery ligation, resulting in acute myocardial ischemia, with subsequent heart failure, as well transverse aortic constriction to induce a pressure overloaded LV, have been developed. However, the extremely small size of the mouse heart (100 mg and 7 mm long axis) and fast heart rates (~500 beats per minute) under physiological conditions, require miniaturized and technically advanced methods to accurately assess the cardiac phenotype of these animals. Moreover, in the light of the increasing interest in regenerative medicine, especially the effects of cell therapy and its preclinical applications in small animals makes potential methods for murine cardiac phenotyping more important than ever. Several methods have been adapted for murine studies, including invasive pressure-volume loop measurements, magnetic resonance imaging (MRI) and echocardiology.

A ssess m e nt of P ressu re -Vo l u m e Lo o ps The pressure-conductance catheter is a miniature (1.4F) catheter that is introduced into the LV of mice via the right coronary artery (Figure 4.) [210]. This method is capable of simultaneously monitoring LV volumes and pressures, so that LV pressure-volume (PV) loops can be documented continuously. In addition, also the time derivates of end-diastolic (dP/dtmin) and end-systolic pressures (dP/ dtmax) can be obtained with this method. Moreover, pressure-volume relations are relatively independent of loading conditions and are therefore considered superior to quantify intrinsic ventricular function [211]. A disadvantage of this method is the invasive nature, resulting in the dead of the animal and therefore precluding longitudinal studies.

Figure 4. A 1.4F miniature pressure-conductance catheter containing four platinum electrodes and a solid-state pressure transducer used for measearing pressure-volume loops in mice.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

M a g n eti c Res o n an ce I m a g i n g MRI is a non-invasive technique that uses intrinsic contrast and is capable of obtaining true 3-D anatomical and function information. Combined with the high temporal resolution, which enables accurate assessment of cardiac function, MRI is the imaging modality of choice to study the effects of stem cell therapy [212]. In addition, the established clinical MR technique of infarct size determination by delayed contrast enhancement imaging after Gd-DPTA was recently adapted and validated in the mouse MI model [213]. Furthermore, being a non-invasive technique allows for serial investigations. Drawbacks of MR imaging in small animals are, that the experiments are costly and time consuming, making it difficult to use for large series of studies. Furthermore, no information on end-diastolic and end-systolic pressures, as well as several other indices of LV function can not be obtained with MRI. Despite these drawbacks, there is an increasing use of this promising technique in laboratories all over the world (Figure 5.) Figure 5. Example of a currently available vertical 9.4-T (400 MHz), 89-mm bore nuclear magnetic resonance (MR) spectrometer (Bruker BioSpin, Rheinstetten, Germany) suitable for MR imaging in small laboratory animals, and used in the present thesis. (Picture was taken at the Gorlaeus laboratory, Leiden, The Netherlands).

Ech o card i o g rap hy Ultrasound echocardiography is currently the most commonly used method for cardiovascular phenotyping in mouse models. Two-dimensional echocardiography (2D-echo) has been used in noninvasive cardiac phenotyping in normal and pathological mouse hearts [214,215]. However, this method requires geometric assumptions for calculation of LV volumes and mass, making it less reliable when the LV has an altered asymmetric shape (e.g. after MI). Recently, 3D-echo has been shown to be superior to standard 2D-echo, and even approaches the accuracy of MRI for determining LV parameters and infarct size [216].

29

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

30

AIM AND OUTLINE OF THE THESIS The main aim of the present study was to explore two main components of regenerative medicine, namely tissue-engineering and stem cell therapy, for its applications in treating CVD. The focus was placed on ischemic heart disease and aortic valve disease. Furthermore, the effectiveness of different methods that could be used to determine changes in cardiac function after regenerative medicine applications is briefly described. Part I

of the thesis centers on different methods to produce acellular porcine and rat aortic

valve scaffolds for the purpose of tissue-engineering. In Chapter 2 the effects of two different cell extraction techniques on the remaining extracellular matrix of porcine aortic valve leaflets was evaluated. In addition, the potential of reseeded endothelial cell to produce ECM molecules was assessed. Next Chapter 3 describes a novel method to produce acellular rat aortic valve conduits, and its effects on the remaining ECM. Furthermore, the in vivo behavior of these acellular valve conduits was studied in a heterotopic rat aortic valve implantation model. Finally Chapter 4 used this model to characterize the cellular immune response to cellular and acellular allogeneic aortic valves, and tried to correlate it to the histological and biochemical changes in the ECM collagen constitution. Part II

of the thesis focuses on the feasibility, in vivo behavior and functional evaluation of dif-

ferent cell types used for the treatment of acute myocardial infarction. Although several studies demonstrated the therapeutic regenerative potential of human MSCs from healthy volunteers, little information is available on the effects of hMSCs from hMSC derived from patients with ischemic heart disease. Therefore Chapter 5 investigated whether hMSC from IHD patients were able to engraft into the acutely infarcted myocardium of the immune-compromised NOD/scid mouse, differentiate into several cardiac cell types, and if these cells could preserve LV function. A 9.4T small animal MRI was used to assess LV function and remodeling. Chapter 6 uses the same model to describe the effects of forced expression of the cardiac transcription factor myocardin in hMSCs from IHD patients. Besides immuno-histochemical assessment of cellular engraftment and possible differentiation into cardiomyocytes, detailed hemodynamic measurements were performed with the 9.4T MRI and the invasive conductance catheter. Next in Chapter 7 the effects hEPDC transplantation on the acutely infarcted mouse myocardium were evaluated by MRI at two and 6 weeks after MI. Particular emphasis was placed on the possibility that these cells could recapitulate their embryonic capacities when injected in the ischemic myocardium. Part III

of the thesis addresses two methods used in cardiac phenotyping of small laboratory

animals. Chapter 8 describes a head-to-head comparison between MRI and the conductance catheter for the assessment of LV volumes and ejection fraction in normal and failing mouse hearts.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

REFERENCES [1]

British heart foundation, European Cardiovascular disease statistics, 2000, London, UK. 2000

[2]

Longer survival, more cases, boost heart failure population. Harv Heart Lett 2007;7-7

[3]

WHO: World Health Report www.who.int/whr/. 2007

[4]

Pfeffer MA, Braunwald E. Ventricular Remodeling After Myocardial-Infarction - Experimental-Observations and Clinical Implications. Circulation 1990;81:1161-1172

[5]

Gheorghiade M, Bonow RO. Chronic heart failure in the United States - A manifestation of coronary artery

[6]

Antman EM, Anbe DT, Armstrong PW, Bates ER, Green LA, Hand M, Hochman JS, Krumholz HM, Kushner

disease. Circulation 1998;97:282-289

FG, Lamas GA, Mullany CJ, Ornato JP, Pearle DL, Sloan MA, Smith SC, Antman EM, Smith SC, Alpert JS, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Gregoratos G, Halperin JL, Hiratzka LF, Hunt SA, Jacobs AK, Ornato JP. ACC/AHA guidelines for the management of patients with ST-elevation myocardial infarction - Executive summary - A report of the American College of Cardiology American Heart Association task force on practice guidelines - (Writing committee to revise the 1999 guidelines for the management of patients with acute myocardial infarction). Canadian Journal of Cardiology 2004;20:977-1025 [7]

Tavazzi L. Epidemiological burden of heart failure. Heart 1998;79:S6-S9

[8]

Miller LW MED. Epidemiology of heart failure. Cardiol Clin 2007;4:547-550

[9]

AHA, Heart and Stroke Statistical Update, AHA, TX, USA (2002). 2007

[10]

Dare AJ, Veinot JP, Edwards WD, Tazelaar HD, Schaff HV. New Observations on the Etiology of Aortic-Valve Disease - A Surgical Pathological-Study of 236 Cases from 1990. Human Pathology 1993;24:1330-1338

[11]

Dare AJ, Harrity PJ, Tazelaar HD, Edwards WD, Mullany CJ. Evaluation of Surgically Excised Mitral-Valves - Revised Recommendations Based on Changing Operative Procedures in the 1990S. Human Pathology 1993; 24: 1286-1293

[12]

Iung B, Baron G, Butchart EG, Delahaye F, Gohlke-Barwolf C, Levang OW, Tornos P, Vanoverschelde JL, Vermeer F, Boersma E, Ravaud P, Vahanian A. A prospective survey of patients with valvular heart disease in Europe: The Euro Heart Survey on Valvular Heart Disease. European Heart Journal 2003;24:1231-1243

[13]

Davies MJ, Treasure T, Parker DJ. Demographic characteristics of patients undergoing aortic valve replacement for stenosis: Relation to valve morphology. Heart 1996;75:174-178

[14]

Lindroos M, Kupari M, Heikkila J, Tilvis R. Prevalence of Aortic-Valve Abnormalities in the Elderly - An Echocardiographic Study of A Random-Population Sample. Journal of the American College of Cardiology 1993; 21:1220-1225

[15]

Schoen FJ. Pathology of heart valve substitution with mechanical and tissue prostheses.;629-677

[16]

Yacoub N, Takkenberg J. Will heart valve tissue engineering change the world? Nature Clinical Practice

[17]

Greenwood HL, Singer PA, Downey GP, Martin DK, Thorsteinsdottir H, Daar AS. Regenerative medicine and

Cardiovascular Medicine 2005;2:60-61

the developing world. Plos Medicine 2006;3:1496-1500

31

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

32

[18]

Gurtner GC, Callaghan MJ, Longaker MT. Progress and potential for regenerative medicine. Annual Review

[19]

Akar AR, Durdu S, Corapcioglu T, Ozyurda U. Regenerative medicine for cardiovascular disorders-new mile-

of Medicine 2007;58:299-312

stones: Adult stem cells. Artificial Organs 2006;30:213-232 [20]

Brockes JP, Kumar A. Appendage regeneration in adult vertebrates and implications for regenerative medicine. Science 2005;310:1919-1923

[21]

Brockes JP. Amphibian limb regeneration: Rebuilding a complex structure. Science 1997;276:81-87

[22]

Price JS, Allen S, Faucheux C, Althnaian T, Mount JG. Deer antlers: a zoological curiosity or the key to under-

[23]

Muneoka K. Regeneration of higher vertebrate limbs: Developing a roadmap. Faseb Journal 2005; 19:A1357-

[24]

Bullard KM, Longaker MT, Lorenz HP. Fetal wound healing: Current biology. World Journal of Surgery 2003;

standing organ regeneration in mammals? Journal of Anatomy 2005;207:603-618

A1357

27:54-61 [25]

Atala A. Tissue engineering and regenerative medicine: Concepts for clinical application. Rejuvenation

[26]

Yacoub MH, Cohn LH. Novel approaches to cardiac valve repair from structure to function: part II. Circula-

[27]

Yacoub MH, Cohn LH. Novel approaches to cardiac valve repair from structure to function: Part I. Circulation

Research 2004;7:15-31

tion 2004;109:1064-1072

2004;109:942-950 [28]

Schoen FJ, Levy RJ. Tissue heart valves: Current challenges and future research perspectives. Journal of

[29]

Bonow RO, Carabello B, de Leon AC, Edmunds LH, Fedderly BJ, Freed MD, Gaasch WH, Mckay CR, Nishimura RA,

Biomedical Materials Research 1999;47:439-465

O’Gara PT, O’Rourke RA, Rahimtoola SH, Ritchie JL, Cheitlin MD, Eagle KA, Gardner TJ, Garson A, Gibbons RJ, Russell RO, Ryan TJ, Smith SC. Guidelines for the management of patients with valvular heart disease - Executive summary - A report of the American College of Cardiology American Heart Association task force on practice guidelines (committee on management of patients with valvular heart disease). Circulation 1998;98:1949-1984 [30]

Rahimtoola SH. Choice of prosthetic heart valve for adult patients. Journal of the American College of Cardiology 2003;41:893-904

[31]

Hammermeister KE, Sethi GK, Henderson WG, Oprian C, Kim T, Rahimtoola S. A Comparison of Outcomes in Men 11 Years After Heart-Valve Replacement with A Mechanical Valve Or Bioprosthesis. New England Journal of Medicine 1993;328:1289-1296

[32]

Vongpatanasin W, Hillis LD, Lange RA. Medical progress - Prosthetic heart valves. New England Journal of Medicine 1996;335:407-416

[33] [34]

Vesely I. Heart valve tissue engineering. Circulation Research 2005;97:743-755 Schoen FJ, Levy RJ. Calcification of tissue heart valve substitutes: Progress toward understanding and prevention. Annals of Thoracic Surgery 2005;79:1072-1080

[35]

Melina G, Al Obaidi M, Yacoub MH. Early and long-term outcome of re-replacement of the aortic valve

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

following homograft full root replacement. A 28-year experience. Circulation 2003;108:620-620 [36]

Gonzalez-Lavin L RD. Homograft aortic valve replacement. A five-year experience at the National Heart Hospital, London. J Thorac Cardiovasc Surg 1970;60:1-12

[37]

Grunkemeier GL, Jamieson WRE, Miller DC, Starr A. Actuarial Versus Actual Risk of Porcine Structural Valve

[38]

Fann JI, Miller DC, Moore KA, Mitchell RS, Oyer PE, Stinson EB, Robbins RC, Reitz BA, Shumway NE. Twenty-

Deterioration. Journal of Thoracic and Cardiovascular Surgery 1994;108:709-718

year clinical experience with porcine bioprostheses. Annals of Thoracic Surgery 1996;62:1301-1311 [39]

Kanter KR, Budde JM, Parks WJ, Tam VKH, Sharma S, Williams WH, Fyfe DA. One hundred pulmonary valve replacements in children after relief of right ventricular outflow tract obstruction. Annals of Thoracic Surgery 2002;73:1801-1806

[40]

Melina G, De Robertis F, Gaer JAR, Amrani M, Khaghani A, Yacoub MH. Mid-term pattern of survival, hemodynamic performance and rate of complications after Medtronic Freestyle versus homograft full aortic root replacement: Results from a prospective randomized trial. Journal of Heart Valve Disease 2004;13:972-975

[41]

Mayer JE Jr. Uses of homograft conduits for right ventricle to pulmonary artery connections in the neonatal period. Semin Thorac Cardiovasc Surg 1995;130-132

[42]

Hammermeister K, Sethi GK, Henderson WG, Grover FL, Oprian C, Rahimtoola SH. Outcomes 15 years after valve replacement with a mechanical versus a bioprosthetic valve: Final report of the Veterans Affairs randomized trial. Journal of the American College of Cardiology 2000;36:1152-1158

[43]

Langer R, Vacanti JP. Tissue Engineering. Science 1993;260:920-926

[44]

Therin M, Christel P, Li SM, Garreau H, Vert M. Invivo Degradation of Massive Poly(Alpha-Hydroxy Acids)

[45]

Shinoka T, Breuer CK, Zund G, Tanel R, Ma P, Langer R, Vacanti JP, Mayer JE. Tissue Engineered Heart-Valve

[46]

vanderGiessen WJ, Lincoff AM, Schwartz RS, vanBeusekom HMM, Serruys PW, Holmes DR, Ellis SG, Topol

- Validation of Invitro Findings. Biomaterials 1992;13:594-600

Leaflets - Valve Leaflet Replacement in A Juvenile Lamb Model. Circulation 1995;92:2108-2108

EJ. Marked inflammatory sequelae to implantation of biodegradable and nonbiodegradable polymers in porcine coronary arteries. Circulation 1996;94:1690-1697 [47]

Hollinger JO. Preliminary report on the osteogenic potential of a biodegradable copolymer of polyactide (PLA) and polyglycolide (PGA). J Biomed Mater Res 1983;17:71-80

[48]

Sodian R, Sperling JS, Martin DP, Egozy A, Stock U, Mayer JE, Vacanti JP. Fabrication of a trileaflet heart valve scaffold from a polyhydroxyalkanoate biopolyester for use in tissue engineering. Tissue Engineering 2000;6:183-188

[49]

Hoerstrup SP, Sodian R, Daebritz S, Wang J, Bacha EA, Martin DP, Moran AM, Guleserian KJ, Sperling JS, Kaushal S, Vacanti JP, Schoen FJ, Mayer JE. Functional living trileaflet heart valves grown in vitro. Circulation 2000; 102:44-49

[50]

Hoerstrup SP, Kadner A, Melnitchouk S, Trojan A, Eid K, Tracy J, Sodian R, Visjager JF, Kolb SA, Grunenfelder J, Zund G, Turina MI. Tissue engineering of functional trileaflet heart valves from human marrow stromal cells. Circulation 2002;106:I143-I150

33

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

34

[51]

Sodian R, Hoerstrup SP, Sperling JS, Daebritz S, Martin DP, Moran AM, Kim BS, Schoen FJ, Vacanti JP, Mayer JE. Early in vivo experience with tissue-engineered trileaflet heart valves. Circulation 2000;102:22-29

[52]

Bernard MP, Myers JC, Chu ML, Ramirez F, Eikenberry EF, Prockop DJ. Structure of A Cdna for the Pro-Alpha2 Chain of Human Type-I Procollagen - Comparison with Chick Cdna for Proalpha2(I) Identifies Structurally Conserved Features of the Protein and the Gene. Biochemistry 1983;22:1139-1145

[53]

Hopkins RA. Tissue engineering of heart valves decellularized valve scaffolds. Circulation 2005;111:27122714

[54] [55]

Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials 2006;27:3675-3683 Steinhoff G, Stock U, Karim N, Mertsching H, Timke A, Meliss RR, Pethig K, Haverich A, Bader A. Tissue engineering of pulmonary heart valves on allogenic acellular matrix conduits - In vivo restoration of valve tissue. Circulation 2000;102:50-55

[56]

Cebotari S, Mertsching H, Kallenbach K, Kostin S, Repin O, Batrinac A, Kleczka C, Ciubotaru A, Haverich A. Construction of autologous human heart valves based on an acellular allograft matrix. Circulation 2002;106: I63-I68

[57]

Kasimir MT, Rieder E, Seebacher G, Silberhumer G, Wolner E, Weigel G, Simon P. Comparison of different decellularization procedures of porcine heart valves. International Journal of Artificial Organs 2003;26:421427

[58]

Rieder E, Kasimir MT, Silberhumer G, Seebacher G, Wolner E, Simon P, Weigel G. Decellularization protocols of porcine heart valves differ importantly in efficiency of cell removal and susceptibility of the matrix to recellularization with human vascular cells. Journal of Thoracic and Cardiovascular Surgery 2004;127:399405

[59]

Seddon AA, Curnow P, Booth PJ. Membrane proteins, lipids and detergents: not just a soap opera. Bio-

[60]

Bader A, Schilling T, Teebken OE, Brandes G, Herden T, Steinhoff G, Haverich A. Tissue engineering of heart

chimica et Biophysica Acta-Biomembranes 2004;1666:105-117

valves - human endothelial cell seeding of detergent acellularized porcine valves. European Journal of Cardio-Thoracic Surgery 1998;14:279-284 [61]

Numata S, Fujisato T, Niwaya K, Ishibashi-Ueda H, Nakatani T, Kitamura S. Immunological and histological evaluation of decellularized allograft in a pig model: Comparison with cryopreserved allograft. Journal of Heart Valve Disease 2004;13:984-990

[62]

Kim WG, Park JK, Lee WY. Tissue-engineered heart valve leaflets: An effective method of obtaining acellular-

[63]

Booth C, Korossis SA, Wilcox HE, Watterson KG, Kearney JN, Fisher J, Ingham E. Tissue engineering of cardiac

ized valve xenografts. International Journal of Artificial Organs 2002;25:791-797

valve prostheses I: Development and histological characterization of an acellular porcine scaffold. Journal of Heart Valve Disease 2002;11:457-462 [64]

Kim WG, Lee WY, Kim JM, Moon HJ. Tissue-engineered acellularized valve xenografts: A comparative animal study between plain acellularized xenografts and autologous endothelial cell seeded acellularized xenografts. International Journal of Artificial Organs 2004;27:501-508

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

[65]

Ueda Y, Torrianni MW, Coppin CM, Iwai S, Sawa Y, Matsuda H. Antigen clearing from porcine heart valves

[66]

Erdbrugger W, Konertz W, Dohmen PM, Posner S, Ellerbrok H, Brodde OE, Robenek H, Modersohn D, Pruss A,

with preservation of structural integrity. International Journal of Artificial Organs 2006;29:781-789

Holinski S, Stein-Konertz M, Pauli G. Decellularized xenogenic heart valves reveal remodeling and growth potential in vivo. Tissue Engineering 2006;12:2059-2068 [67]

Spina M, Ortolani F, El Messlemani A, Gandaglia A, Bujan J, Garcia-Honduvilla N, Vesely I, Gerosa G, Casarotto D, Petrelli L, Marchini M. Isolation of intact aortic valve scaffolds for heart-valve bioprostheses: Extracellular matrix structure, prevention from calcification, and cell repopulation features. Journal of Biomedical Materials Research Part A 2003;67A:1338-1350

[68]

Simon P, Kasimir MT, Seebacher G, Weigel G, Ullrich R, Salzer-Muhar U, Rieder E, Wolner E. Early failure of the tissue engineered porcine heart valve SYNERGRAFT (TM) in pediatric patients. European Journal of Cardio-Thoracic Surgery 2003;23:1002-1006

[69]

Zehr KJ, Yagubyan M, Connolly HM, Nelson SM, Schaff HV. Aortic root replacement with a novel decellularized cryopreserved aortic homograft: Postoperative immunoreactivity and early results. Journal of Thoracic and Cardiovascular Surgery 2005;130:1010-1015

[70]

Bechtel JFM, Muller-Steinhardt M, Schmidtke C, Brunswik A, Stierle U, Sievers HH. Evaluation of the decel-

[71]

Erdbrugger W, Konertz W, Dohmen PM, Posner S, Ellerbrok H, Brodde OE, Robenek H, Modersohn D, Pruss A,

lularized pulmonary valve homograft (SynerGraft (TM)). Journal of Heart Valve Disease 2003;12:734-739

Holinski S, Stein-Konertz M, Pauli G. Decellularized xenogenic heart valves reveal remodeling and growth potential in vivo. Tissue Engineering 2006;12:2059-2068 [72]

da Costa FDA, Dohmen PM, Duarte D, von Glenn C, Lopes SV, Haggi H, da Costa MBA, Konertz W. Immunological and echocardiographic evaluation of decellularized versus cryopreserved allografts. during the Ross operation. European Journal of Cardio-Thoracic Surgery 2005;27:572-577

[73]

Schenke-Layland K, Vasilevski O, Opitz F, Konig K, Riemann I, Halbhuber KJ, Wahlers T, Stock UA. Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. Journal of Structural Biology 2003;143:201-208

[74]

Rieder E, Seebacher G, Kasimir MT, Eichmair E, Winter B, Dekan B, Wolner E, Simon P, Weigel G. Tissue engineering of heart valves - Decellularized porcine and human valve scaffolds differ importantly in residual potential to attract monocytic cells. Circulation 2005;111:2792-2797

[75]

Leyh RG, Wilhelmi M, Rebe P, Fischer S, Kofidis T, Haverich A, Mertsching H. In vivo repopulation of xenogeneic and allogeneic acellular valve matrix conduits in the pulmonary circulation. Annals of Thoracic Surgery 2003;75:1457-1463

[76]

Meyer SR, Chiu B, Churchill TA, Zhu LF, Lakey JRT, Ross DB. Comparison of aortic valve allograft decellularization techniques in the rat. Journal of Biomedical Materials Research Part A 2006;79A:254-262

[77]

Shinoka T, Ma PX, ShumTim D, Breuer CK, Cusick RA, Zund G, Langer R, Vacanti JP, Mayer JE. Tissue-engineered heart valves - Autologous valve leaflet replacement study in a lamb model. Circulation 1996;94:164168

35

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

36

[78]

Sutherland FWH, Perry TE, Yu Y, Sherwood MC, Rabkin E, Masuda Y, Garcia GA, McLellan DL, Engelmayr GC, Sacks MS, Schoen FJ, Mayer JE. From stem cells to viable autologous semilunar heart valve. Circulation 2005;111:2783-2791

[79]

Goldstein S, Clarke DR, Walsh SP, Black KS, O’Brien MF. Transpecies heart valve transplant: Advanced studies of a bioengineered xeno-autograft. Annals of Thoracic Surgery 2000;70:1962-1969

[80]

Dohmen PM, Lembcke A, Hotz H, Kivelitz D, Konertz WF. Ross operation with a tissue-engineered heart valve. Annals of Thoracic Surgery 2002;74:1438-1442

[81]

Konertz W, Dohmen PM, Liu JS, Beholz S, Dushe S, Posner S, Lembcke A, Erdbrugger W. Hemodynamic characteristics of the Matrix P decellularized xenograft for pulmonary valve replacement during the Ross operation. Journal of Heart Valve Disease 2005;14:78-81

[82]

Dohmen PM, Konertz W. Results with decellularized xenografts. Circulation Research 2006;99:E10-E10

[83]

Gabbieri D, Dohmen PM, Linneweber J, Lembcke A, Braun JP, Konertz W. Ross procedure with a tissueengineered heart valve in complex congenital aortic valve disease. Journal of Thoracic and Cardiovascular Surgery 2007;133:1088-1089

[84]

Sayk F, Bos I, Schubert U, Wedel T, Sievers HH. Histopathologic findings in a novel decellularized pulmonary homograft: An autopsy study. Annals of Thoracic Surgery 2005;79:1755-1758

[85]

Miller DV, Edwards WD, Zehr KJ. Endothelial and smooth muscle cell populations in a decellularized cryopreserved aortic homograft (SynerGraft) 2 years after implantation. Journal of Thoracic and Cardiovascular Surgery 2006;132:175-MIL

[86]

Reinlib L, Field L. Cell transplantation as future therapy for cardiovascular disease? A workshop of the

[87]

Reinlib L, Abraham W. Recovery from heart failure with circulatory assist: A working group of the national,

National Heart, Lung, and Blood Institute. Circulation 2000;101:E182-E187

heart, lung, and blood institute. Journal of Cardiac Failure 2003;9:459-463 [88]

Dawn B, Bolli R. Adult bone marrow-derived cells: Regenerative potential, plasticity, and tissue commitment. Basic Research in Cardiology 2005;100:494-503

[89]

Fuchs E, Segre JA. Stem cells: A new lease on life. Cell 2000;100:143-155

[90]

Poulsom R, Alison MR, Forbes SJ, Wright NA. Adult stem cell plasticity. Journal of Pathology 2002;197:441456

[91]

Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. Journal of Clinical Investigation 2001;108:407-414

[92]

Hodgson DM, Behfar A, Zingman LV, Kane GC, Perez-Terzic C, Alekseev AE, Puceat M, Terzic A. Stable benefit of embryonic stem cell therapy in myocardial infarction. American Journal of Physiology-Heart and Circulatory Physiology 2004;287:H471-H479

[93]

Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145-1147

[94]

Lanza R, Moore MAS, Wakayama T, Perry ACF, Shieh JH, Hendrikx J, Leri A, Chimenti S, Monsen A, Nurzynska

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

D, West MD, Kajstura J, Anversa P. Regeneration of the infarcted heart with stem cells derived by nuclear transplantation. Circulation Research 2004;94:820-827 [95]

Koh GY, Klug MG, Soonpaa MH, Field LJ. Differentiation and Long-Term Survival of C2C12 Myoblast Grafts in Heart. Journal of Clinical Investigation 1993;92:1548-1554

[96]

Taylor DA, Atkins BZ, Hungspreugs P, Jones TR, Reedy MC, Hutcheson KA, Glower DD, Kraus WE. Regenerating functional myocardium: Improved performance after skeletal myoblast transplantation. Nature Medicine 1998;4:929-933

[97]

Leobon B, Garcin I, Menasche P, Vilquin JT, Audinat E, Charpak S. Myoblasts transplanted into rat infarcted myocardium are functionally isolated from their host. Proceedings of the National Academy of Sciences of the United States of America 2003;100:7808-7811

[98]

Dowell JD, Rubart M, Pasumarthi KBS, Soonpaa MH, Field LJ. Myocyte and myogenic stem cell transplantation in the heart. Cardiovascular Research 2003;58:336-350

[99]

Menasche P, Hagege AA, Scorsin M, Pouzet B, Desnos M, Duboc D, Schwartz K, Vilquin JT, Marolleau JP. Myoblast transplantation for heart failure. Lancet 2001;357:279-280

[100] Murtuza B, Suzuki K, Bou-Gharios G, Beauchamp JR, Smolenski RT, Partridge TA, Yacoub MH. Transplantation of skeletal myoblasts secreting an IL-1 inhibitor modulates adverse remodeling in infarcted murine myocardium. Proceedings of the National Academy of Sciences of the United States of America 2004;101:4216-4221 [101] Smits PC, van Geuns RJM, Poldermans D, Bountioukos M, Onderwater EEM, Lee CH, Maat APWM, Serruys PW. Catheter-based intramyocardial injection of autologous skeletal myoblasts as a primary treatment of ischemic heart failure - Clinical experience with six-month follow-up. Journal of the American College of Cardiology 2003;42:2063-2069 [102] Toh R, Kawashima S, Kawai M, Sakoda T, Ueyama T, Satomi-Kobayashi S, Hirayama S, Yokoyama M. Transplantation of cardiotrophin-1-expressing myoblasts to the left ventricular wall alleviates the transition from compensatory hypertrophy to congestive heart failure in Dahl salt-sensitive hypertensive rats. Journal of the American College of Cardiology 2004;43:2337-2347 [103] Pagani FD, DerSimonian H, Zawadzka A, Wetzel K, Edge ASB, Jacoby DB, Dinsmore JH, Wright S, Aretz TH, Eisen HJ, Aaronson KD. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans - Histological analysis of cell survival and differentiation. Journal of the American College of Cardiology 2003;41:879-888 [104] Reinecke H, Poppa V, Murry CE. Skeletal muscle stem cells do not transdifferentiate into cardiomyocytes after cardiac grafting. Journal of Molecular and Cellular Cardiology 2002;34:241-249 [105] Menasche P, Hagege AA, Vilquin JT, Desnos M, Abergel E, Pouzet B, Bel A, Sarateanu S, Scorsin M, Schwartz K, Bruneval P, Benbunan M, Marolleau JP, Duboc D. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. Journal of the American College of Cardiology 2003;41:1078-1083 [106] Siminiak T, Kalawski R, Fiszer D, Jerzykowska O, Rzezniczak J, Rozwadowska N, Kurpisz M. Autologous skeletal myoblast transplantation for the treatment of postinfarction myocardial injury: Phase I clinical study with 12 months of follow-up. American Heart Journal 2004;148:531-537

37

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

38

[107] Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, Bussani R, Nadal-Ginard B, Silvestri F, Leri A, Beltrami CA, Anversa P. Evidence that human cardiac myocytes divide after myocardial infarction. New England Journal of Medicine 2001;344:1750-1757 [108] Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003;114:763-776 [109] Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael LH, Behringer RR, Garry DJ, Entman ML, Schneider MD. Cardiac progenitor cells from adult myocardium: Homing, differentiation, and fusion after infarction. Proceedings of the National Academy of Sciences of the United States of America 2003;100:12313-12318 [110] Wang XH, Hu QS, Nakamura Y, Lee J, Zhang G, From AHL, Zhang JY. The role of the Sca-1(+)/CD31(-) cardiac progenitor cell population in postinfarction left ventricular remodeling. Stem Cells 2006;24:1779-1788 [111]

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

[112] Martin CM, Meeson AP, Robertson SM, Hawke TJ, Richardson JA, Bates S, Goetsch SC, Gallardo TD, Garry DJ. Persistent expression of the ATP-binding cassette transporter, Abcg2, identifies cardiac SP cells in the developing and adult heart. Developmental Biology 2004;265:262-275 [113] Pfister O, Mouquet F, Jain M, Summer R, Helmes M, Fine A, Colucci WS, Liao R. CD31(-) but not CD31(+) cardiac side population cells exhibit functional cardiomyogenic differentiation. Circulation Research 2005;97:52-61 [114] Laugwitz KL, Moretti A, Lam J, Gruber P, Chen YH, Woodard S, Lin LZ, Cai CL, Lu MM, Reth M, Platoshyn O, Yuan JXJ, Evans S, Chien KR. Postnatal isl1+cardioblasts enter fully differentiated cardiomyocyte lineages. Nature 2005;433:647-653 [115] Gunsilius E, Gastl G, Petzer AL. Hematopoietic stem cells. Biomedicine & Pharmacotherapy 2001;55:186194 [116] Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li BS, Pickel J, Mckay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature 2001;410:701-705 [117] Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 2004;428:668-673 [118] Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KBS, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 2004;428:664-668 [119] Kajstura J, Rota M, Whang B, Cascapera S, Hosoda T, Bearzi C, Nurzynska D, Kasahara H, Zias E, Bonafe M, Nadal-Ginard B, Torella D, Nascimbene A, Quaini F, Urbanek K, Leri A, Anversa P. Bone marrow cells differentiate in cardiac cell lineages after infarction independently of cell fusion. Circulation Research 2005;96:127-137

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

[120] de Muinck ED, Thompson C, Simons M. Progress and prospects: Cell based regenerative therapy for cardiovascular disease. Gene Therapy 2006;13:659-671 [121] Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. Journal of Experimental Medicine 1996;183:17971806 [122] Collins SD BRWR. Cell therapy in myocardial infarction. Cardiovasc Revasc Med 2007;8:43-51 [123] Jackson KA, Majka SM, Wang HG, Pocius J, Hartley CJ, Majesky MW, Entman ML, Michael LH, Hirschi KK, Goodell MA. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. Journal of Clinical Investigation 2001;107:1395-1402 [124] Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nature Medicine 2001;7:430-436 [125] Urbich C, Dimmeler S. Endothelial progenitor cells - Characterization and role in vascular biology. Circulation Research 2004;95:343-353 [126] Eichmann A, Corbel C, Nataf V, Vaigot P, Breant C, LeDouarin NM. Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic mesodermal cells expressing vascular endothelial growth factor receptor 2. Proceedings of the National Academy of Sciences of the United States of America 1997;94:5141-5146 [127] Sato TN, Qin Y, Kozak CA, Audus KL. Tie-1 and Tie-2 Define Another Class of Putative Receptor Tyrosine Kinase Genes Expressed in Early Embryonic Vascular System. Proceedings of the National Academy of Sciences of the United States of America 1993;90:9355-9358 [128] Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nature Medicine 2003;9:702-712 [129] Asahara T, Takahashi T, Masuda H, Kalka C, Chen DH, Iwaguro H, Inai Y, Silver M, Isner JM. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. Embo Journal 1999;18:3964-3972 [130] Takahashi T, Kalka C, Masuda H, Chen D, Silver M, Kearney M, Magner M, Isner JM, Asahara T. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nature Medicine 1999;5:434-438 [131] Kawamoto A, Tkebuchava T, Yamaguchi JI, Nishimura H, Yoon YS, Milliken C, Uchida S, Masuo O, Iwaguro H, Ma H, Hanley A, Silver 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 [132] www.ClinicalTrials.gov. 2007 [133] Heeschen C, Lehmann R, Honold J, Assmus B, Aicher A, Walter DH, Martin H, Zeiher AM, Dimmeler S. Profoundly reduced neovascularization capacity of bone marrow mononuclear cells derived from patients with chronic ischemic heart disease. Circulation 2004;109:1615-1622

39

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

40

[134] Caplan AI. Mesenchymal Stem-Cells. Journal of Orthopaedic Research 1991;9:641-650 [135] Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circulation Research 2004;95:9-20 [136] Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, Hata J, Umezawa A, Ogawa S. Cardiomyocytes can be generated from marrow stromal cells in vitro. Journal of Clinical Investigation 1999;103:697-705 [137] Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation 2002;105:93-98 [138] Tang YL, Zhao Q, Qin XY, Shen LP, Cheng LL, Ge JB, Phillips MI. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Annals of Thoracic Surgery 2005;80:229-237 [139] Dai WD, Hale SL, Martin BJ, Kuang JQ, Dow JS, Wold LE, Kloner RA. Allogeneic mesenchymal stem cell transplantation in postinfarcted rat myocardium - Short- and long-term effects. Circulation 2005;112:214-223 [140] Silva GV, Litovsky S, Assad JAR, Sousa ALS, Martin BJ, Vela D, Coulter SC, Lin J, Ober J, Vaughn WK, Branco RVC, Oliveira EM, He RM, Geng YJ, Willerson JT, Perin EC. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a canine chronic ischemia model. Circulation 2005;111:150-156 [141] Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J, Redmond JM, Pittenger MF, Martin BJ. Mesenchymal stem cell implantation in a swine myocardial infarct model: Engraftment and functional effects. Annals of Thoracic Surgery 2002;73:1919-1925 [142] Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman AW, Hare JM. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proceedings of the National Academy of Sciences of the United States of America 2005;102:11474-11479 [143] Tomita S, Li RK, Weisel RD, Mickle DAG, Kim EJ, Sakai T, Jia ZQ. Autologous transplantation of bone marrow cells improves damaged heart function. Circulation 1999;100:247-256 [144] Chen SL, Fang W, Ye F, Liu YH, Qian J, Shan S, Zhang J, Zhao RCH, Liao LM, Lin S, Sun JP. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. American Journal of Cardiology 2004;94:92-95 [145] Ryan J, Barry F, Murphy JM, Mahon BP. Human mesenchymal stem cells evade allogeneic immune responses. Immunology 2005;116:102-102 [146] Reyes M, Lund T, Lenvik T, Aguiar D, Koodie L, Verfaillie CM. Purification and ex vivo expansion of postnatal human marrow mesodermal progenitor cells. Blood 2001;98:2615-2625 [147] Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM. Origin of endothelian progenitors in human postnatal bone marrow. Journal of Clinical Investigation 2002;109:337-346 [148] Schwartz RE, Reyes M, Koodie L, Jiang YH, Blackstad M, Lund T, Lenvik T, Johnson S, Hu WS, Verfaillie CM. Multipotent adult progenitor cells from bone marrow differentiate into functional hepatocyte-like cells.

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

Journal of Clinical Investigation 2002;109:1291-1302 [149] Jiang YH, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du JB, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 2002;418:41-49 [150] Muguruma Y, Nakamura Y, Yahata T, Ando K, Kato S, Hotta T. Formation of multinucleated myotubes from human multipotent adult proenitor cells. Blood 2002;100:525A-526A [151] Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, Scadova H, Qin GJ, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW. Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. Journal of Clinical Investigation 2005;115:326-338 [152] Rangappa S, Fen C, Lee EH, Bongso A, Wei ESK. Transformation of adult mesenchymal stem cells isolated from the fatty tissue into cardiomyocytes. Annals of Thoracic Surgery 2003;75:775-779 [153] Mizuno H, Zuk PA, Zhu M, Lorenz HP, Benhaim P, Hedrick MH. Myogenic differentiation by human processed lipoaspirate cells. Plastic and Reconstructive Surgery 2002;109:199-209 [154] Miranville A, Heeschen C, Sengenes C, Curat CA, Busse R, Bouloumie A. Improvement of postnatal neovascularization by human adipose tissue-derived stem cells. Circulation 2004;110:349-355 [155] Planat-Benard V, Menard C, Andre M, Puceat M, Perez A, Garcia-Verdugo JM, Penicaud L, Casteilla L. Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circulation Research 2004;94:223229 [156] Planat-Benard V, Silvestre JS, Cousin B, Andre M, Nibbelink M, Tamarat R, Clergue M, Manneville C, Saillan-Barreau C, Duriez M, Tedgui A, Levy B, Penicaud L, Casteilla L. Plasticity of human adipose lineage cells toward endothelial cells - Physiological and therapeutic perspectives. Circulation 2004;109:656-663 [157] Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. British Journal of Haematology 2000;109:235-242 [158] Lee OK, Kuo TK, Chen WM, Lee KD, Hsieh SL, Chen TH. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood 2004;103:1669-1675 [159] Wang HS, Hung SC, Peng ST, Huang CC, Wei HM, Guo YJ, Fu YS, Lai MC, Chen CC. Mesenchymal stem cells in the Wharton’s jelly of the human umbilical cord. Stem Cells 2004;22:1330-1337 [160] Kogler G, Sensken S, Airey JA, Trapp T, Muschen M, Feldhahn N, Liedtke S, Sorg RV, Fischer J, Rosenbaum C, Greschat S, Knipper A, Bender J, Degistirici O, Gao JZ, Caplan AI, Colletti E, Almeida-Porada G, Muller HW, Zanjani E, Wernet P. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. Journal of Experimental Medicine 2004;200:123-135 [161] Kim BO, Tian H, Prasongsukarn K, Wu J, Angoulvant D, Wnendt S, Muhs A, Spitkovsky D, Li RK. Cell transplantation improves ventricular function after a myocardial infarction - A preclinical study of human unrestricted somatic stem cells in a porcine model. Circulation 2005;112:I96-I104 [162] Hirata Y, Sata M, Motomura N, Takanashi M, Suematsu Y, Ono M, Takamoto S. Human umbilical cord blood cells improve cardiac function after myocardial infarction. Biochemical and Biophysical Research Communications 2005;327:609-614

41

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

42

[163] Wessels A, Perez-Pomares JM. The epicardium and epicardially derived cells (EPDCs) as cardiac stem cells. Anatomical Record Part A-Discoveries in Molecular Cellular and Evolutionary Biology 2004;276A:43-57 [164] Perez-Pomares JM, Carmona R, Gonzalez-Iriarte M, Atencia G, Wessels A, Munoz-Chapuli R. Origin of coronary endothelial cells from epicardial mesothelium in avian embryos. International Journal of Developmental Biology 2002;46:1005-1013 [165] PerezPomares JM, Macias D, GarciaGarrido L, MunozChapuli R. Contribution of the primitive epicardium to the subepicardial mesenchyme in hamster and chick embryos. Developmental Dynamics 1997;210:96-105 [166] Dettman RW, Denetclaw W, Ordahl CP, Bristow J. Common epicardial origin of coronary vascular smooth muscle, perivascular fibroblasts, and intermyocardial fibroblasts in the avian heart. Developmental Biology 1998;193:169-181 [167] Gittenberger-de Groot AC, Peeters MPFM, Mentink MMT, Gourdie RG, Poelmann RE. Epicardium-derived cells contribute a novel population to the myocardial wall and the atrioventricular cushions. Circulation Research 1998;82:1043-1052 [168] Mikawa T, Fischman DA. Retroviral Analysis of Cardiac Morphogenesis - Discontinuous Formation of Coronary Vessels. Proceedings of the National Academy of Sciences of the United States of America 1992;89:9504-9508 [169] Peeters MPFM, Gittenberger-de Groot AC, Mentink MMT, Poelmann RE. Smooth muscle cells and fibroblasts of the coronary arteries derive from epithelial-mesenchymal transformation of the epicardium. Anatomy and Embryology 1999;199:367-378 [170] Manner J, Perez-Pomares JM, Macias D, Munoz-Chapuli R. The origin, formation and developmental significance of the epicardium: A review. Cells Tissues Organs 2001;169:89-103 [171] Winter EM, Gittenberger-de Groot AC. Epicardium-derived cells in cardiogenesis and cardiac regeneration. Cellular and Molecular Life Sciences 2007;64:692-703 [172] Gittenberger-de Groot AC, Peeters MPFM, Bergwerff M, Mentink MMT, Poelmann RE. Epicardial outgrowth inhibition leads to compensatory mesothelial outflow tract collar and abnormal cardiac septation and coronary formation. Circulation Research 2000;87:969-971 [173] Kang JO, Sucov HM. Convergent proliferative response and divergent morphogenic pathways induced by epicardial and endocardial signaling in fetal heart development. Mechanisms of Development 2005;122:5765 [174] Eralp I, Lie-Venema H, Bax NAM, Wijffels MCEF, van der Laarse A, Deruiter MC, Bogers AJJC, Van den Akker NMS, Gourdie RG, Schalij MJ, Poelmann RE, Gittenberger-de Groot AC. Epicardium-derived cells are important for correct development of the Purkinje fibers in the avian heart. Anatomical Record Part A-Discoveries in Molecular Cellular and Evolutionary Biology 2006;288A:1272-1280 [175] Guadix JA, Carmona R, Munoz-Chapuli R, Perez-Pomares JM. In vivo and in vitro analysis of the vasculogenic potential of avian proepicardial and epicardial cells. Developmental Dynamics 2006;235:1014-1026 [176] Perez-Pomares JM, Phelps A, Munoz-Chapuli R, Wessels A. The contribution of the proepicardium to avian cardiovascular development. International Journal of Developmental Biology 2001;45:S155-S156

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

[177] Wada AM, Smith TK, Osler ME, Reese DE, Bader DM. Epicardial/mesothelial cell line retains vasculogenic potential of embryonic epicardium. Circulation Research 2003;92:525-531 [178] van Tuyn J, Atsma DE, Winter EM, van der Velde-Van Dijke, Pijnappels DA, Bax NAM, Knaan-Shanzer S, Gittenberger-de Groot AC, Poelmann RE, van der Laarse A, van der Wall EE, Schalij MJ, de Vries AAF. Epicardial cells of human adults can undergo an epithelial-to-mesenchymal transition and obtain characteristics of smooth muscle cells in vitro. Stem Cells 2007;25:271-278 [179] Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, Yu JT, Corti R, Mathey DG, Hamm CW, Suselbeck T, Assmus B, Tonn T, Dimmeler S, Zeiher AM. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. New England Journal of Medicine 2006;355:1210-1221 [180] Meyer GP, Wollert KC, Lotz J, Steffens J, Lippolt P, Fichtner S, Hecker H, Schaefer A, Arseniev L, Hertenstein B, Ganser A, Drexler 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 [181] Janssens S, Dubois C, Boyaert J, Theunissen K, Deroose C, Desmet W, Kolantzi M, Herbots L, Sinnaeve P, Dens J, Maertens J, Rademakers F, Dymarkowski S, Gheysens O, Van Cleemput J, Bormans G, Nuyts J, Belmans A, Mortelmans L, Boogaerts M, Van de Werf F. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet 2006;367:113-121 [182] Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, Endresen K, Ilebekk A, Mangschau A, Fjeld JG, Smith HJ, Taraldsrud E, Grogaard HK, Bjornerheim R, Brekke M, Muller C, Hopp E, Ragnarsson A, Brinchmann JE, Forfang K. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. New England Journal of Medicine 2006;355:1199-1209 [183] Seeger FH, Tonn T, Krzossok N, Zeiher AM, Dimmeler S. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. European Heart Journal 2007;28:766-772 [184] Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher AM. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction - (TOPCARE-AMI). Circulation 2002;106:3009-3017 [185] Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P, Van Haute I, Lootens N, Heyndrickx G, Wijns W. Intracoronary injection of CD133-positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction - Feasibility and safety. Circulation 2005;112: I178-I183 [186] Mansour S, Vanderheyden M, De Bruyne B, Vandekerckhove B, Delrue L, Van Haute I, Heyndrickx G, Carlier S, Rodriguez-Granillo G, Wijns W, Bartunek J. Intracoronary delivery of hematopoietic bone marrow stem cells and luminal loss of the infarct-related artery in patients with recent myocardial infarction. Journal of the American College of Cardiology 2006;47:1727-1730 [187] Chen SL, Fang W, Ye F, Liu YH, Qian J, Shan S, Zhang J, Zhao RCH, Liao LM, Lin S, Sun JP. Effect on left

43

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

44

ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. American Journal of Cardiology 2004;94:92-95 [188] Hamano K, Nishida M, Hirata K, Mikamo A, Li TS, Harada M, Miura T, Matsuzaki M, Esato K. Local implantation of autologous bone marrow cells for therapeutic angiogenesis in patients with ischemic heart disease - Clinical trial and preliminary results. Japanese Circulation Journal-English Edition 2001;65:845-847 [189] Tse HF, Kwong YL, Chan JKF, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003;361:47-49 [190] Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, Baffour R, Waksman R, Weissman NJ, Cerqueira M, Leon MB, Epstein SE. Catheter-based autologous bone marrow myocardial injection in no-option patients with advanced coronary artery disease - A feasibility study. Journal of the American College of Cardiology 2003;41:1721-1724 [191] Perin EC, Dohmann HFR, Borojevic R, Silva SA, Sousa ALS, Mesquita CT, Rossi MID, Carvalho AC, Dutra HS, Dohmann HJF, Silva GV, Belem L, Vivacqua R, Rangel FOD, Esporcatte R, Geng YJ, Vaughn WK, Assad JAR, Mesquita ET, Willerson JT. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107:2294-2302 [192] Beeres SLMA, Bax JJ, Dibbets-Schneider P, Stokkel MPM, Fibbe WE, van der Wall EE, Schalij MJ, Atsma DE. Sustained effect of autologous bone marrow mononuclear cell injection in patients with refractory angina pectoris and chronic myocardial ischemia: Twelve-month follow-up results. American Heart Journal 2006;152: [193] Herreros J, Prosper F, Perez A, Gavira JJ, Garcia-Velloso MJ, Barba J, Sanchez PL, Caniz C, Rabago G, MartiCliment JM, Hernandez M, Lopez-Holgado N, Gonzalez-Santos JM, Martin-Luengo C, Alegria E. Autologous intramyocardial injection of cultured skeletal muscle-derived stem cells in patients with non-acute myocardial infarction. European Heart Journal 2003;24:2012-2020 [194] Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, Klinge H, Schumichen C, Nienaber CA, Freund M, Steinhoff G. Autologous bone-marrow stem-cell transplantation for myocardial regeneration. Lancet 2003;361:45-46 [195] Assmus B, Honold J, Schachinger V, Britten MB, Fischer-Rasokat U, Lehmann R, Teupe C, Pistorius K, Martin H, Abolmaali ND, Tonn T, Dimmeler S, Zeiher AM. Transcoronary transplantation of progenitor cells after myocardial infarction. New England Journal of Medicine 2006;355:1222-1232 [196] Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J, Taneera J, Fleischmann BK, Jacobsen SEW. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nature Medicine 2004;10:494-501 [197] Murry CE, Reinecke H, Pabon LM. Regeneration gaps - Observations on stem cells and cardiac repair. Journal of the American College of Cardiology 2006;47:1777-1785 [198] Hutcheson KA, Atkins BZ, Hueman MT, Hopkins MB, Glower DD, Taylor DA. Comparison of benefits on myocardial performance of cellular cardiomyoplasty with skeletal myoblasts and fibroblasts. Cell Transplantation 2000;9:359-368

CHAPTER 1: GENERAL INTRODUCTION AND OUTLINE OF THE THESIS

[199] Jain M, DerSimonian H, Brenner DA, Ngoy S, Teller P, Edge ASB, Zawadzka A, Wetzel K, Sawyer DB, Colucci WS, Apstein CS, Liao RL. Cell therapy attenuates deleterious ventricular remodeling and improves cardiac performance after myocardial infarction. Circulation 2001;103:1920-1927 [200] Berry MF, Engler AJ, Woo YJ, Pirolli TJ, Bish LT, Jayasankar V, Morine KJ, Gardner TJ, Discher DE, Sweeney HL. Mesenchymal stem cell injection after myocardial infarction improves myocardial compliance. American Journal of Physiology-Heart and Circulatory Physiology 2006;290:H2196-H2203 [201] Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M, Kearney M, Li T, Isner JM, Asahara T. Transplantation of ex vivo expanded endothelial progenitor cells for therapeutic neovascularization. Proceedings of the National Academy of Sciences of the United States of America 2000;97:3422-3427 [202] Caplan AI, Dennis JE. Mesenchymal stem cells as trophic mediators. Journal of Cellular Biochemistry 2006;98:1076-1084 [203] Tang JM, Xie QY, Pan GD, Wang JN, Wang MJ. Mesenchymal stem cells participate in angiogenesis and improve heart function in rat model of myocardial ischemia with reperfusion. European Journal of CardioThoracic Surgery 2006;30:353-361 [204] Kinnaird T, Stabile E, Burnett MS, Lee CW, Barr S, Fuchs S, Epstein SE. Marrow-derived stromal cells express genes encoding a broad spectrum of arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through paracrine mechanisms. Circulation Research 2004;94:678-685 [205] Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, Scadova H, Qin GJ, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW. Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. Journal of Clinical Investigation 2005;115:326-338 [206] Mangi AA, Noiseux N, Kong DL, He HM, Rezvani M, Ingwall JS, Dzau VJ. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nature Medicine 2003;9:11951201 [207] Uemura R, Xu M, Ahmad N, Ashraf M. Bone marrow stem cells prevent left ventricular remodeling of ischemic heart through paracrine signaling. Circulation Research 2006;98:1414-1421 [208] Gnecchi M, He HM, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang LN, Pratt RE, Ingwall JS, Dzau VJ. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nature Medicine 2005;11:367-368 [209] Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM, Dimmeler S. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. Journal of Molecular and Cellular Cardiology 2005;39:733-742 [210] Georgakopoulos D, Mitzner WA, Chen CH, Byrne BJ, Millar HD, Hare JM, Kass DA. In vivo murine left ventricular pressure-volume relations by miniaturized conductance micromanometry. American Journal of Physiology-Heart and Circulatory Physiology 1998;43:H1416-H1422 [211] Kass DA, Maughan WL, Guo ZM, Kono A, Sunagawa K, Sagawa K. Comparative Influence of Load Versus Inotropic States on Indexes of Ventricular Contractility - Experimental and Theoretical-Analysis Based on Pressure-Volume Relationships. Circulation 1987;76:1422-1436

45

REGENERATIVE MEDICINE IN CARDIOVASCULAR DISEASE: FROM TISSUE ENGINEERING TO TISSUE REGENERATION

46

[212] Fuster V, Sanz J, Viles-Gonzalez JF, Rajagopalan S. The utility of magnetic resonance imaging in cardiac tissue regeneration trials. Nat Clin Pract Cardiovasc Med 2006;3 Suppl 1:S2-S7 [213] Yang Z, Berr SS, Gilson WD, Toufektsian MC, French BA. Simultaneous evaluation of infarct size and cardiac function in intact mice by contrast-enhanced cardiac magnetic resonance imaging reveals contractile dysfunction in noninfarcted regions early after myocardial infarction. Circulation 2004;109:1161-1167 [214] Gardin JM, Siri FM, Kitsis RN, Edwards JG, Leinwand LA. Echocardiographic Assessment of Left-Ventricular Mass and Systolic Function in Mice. Circulation Research 1995;76:907-914 [215] Tanaka N, Dalton N, Mao L, Rockman HA, Peterson KL, Gottshall KR, Hunter JJ, Chien KR, Ross J. Transthoracic echocardiography in models of cardiac disease in the mouse. Circulation 1996;94:1109-1117 [216] Dawson D, Lygate CA, Saunders J, Schneider JE, Ye XJ, Hulbert K, Noble JA, Neubauer S. Quantitative 3dimensional echocardiography for accurate and rapid cardiac phenotype characterization in mice. Circulation 2004;110:1632-1637

Suggest Documents