• No results found

From cardiogenesis to cardiac regeneration : focus on epicardium-derived cells Winter, E.M.

N/A
N/A
Protected

Academic year: 2021

Share "From cardiogenesis to cardiac regeneration : focus on epicardium-derived cells Winter, E.M."

Copied!
15
0
0

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

Hele tekst

(1)

From cardiogenesis to cardiac regeneration : focus on epicardium-derived cells

Winter, E.M.

Citation

Winter, E. M. (2009, October 15). From cardiogenesis to cardiac regeneration : focus on epicardium-derived cells. Retrieved from https://hdl.handle.net/1887/14054

Version: Corrected Publisher’s Version

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

Downloaded from: https://hdl.handle.net/1887/14054

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

applicable).

(2)

General Introduction

1

(3)
(4)

Background

Embryonic heart development Cardiac regeneration therapy 1. Embryonic versus adult cells

2. Adult cells harvested from extracardiac structures 3. Adult cells derived from the heart

Functional assessment of the infarcted mouse heart Chapter Outline

General

Introduction

(5)

12

|

Chapter 1

General Introduction

Background

The epidemic proportions of cardiovascular disease, with coronary artery occlusions and its consequences ranking first in the mortality list of the Western world

1

, warrant the development of more appropriate treatment modalities. Promising results of recent innovative stem cell studies direct ischemic heart disease research into an entirely novel field. To accurately design such new experiments, and to fully understand the implication of the outcome, knowledge about embryonic development of the heart might be of benefit. Cardiogenesis and cardiac regeneration therapies will be discussed, emphasizing the outer layer of the heart, the epicardium, which might have promising potential in the adult setting.

Embryonic heart development

Early in gestation, two crescent-shaped cardiogenic plates arise from the anterior splanchnic mesoderm and fuse to form the primitive single-chambered heart tube

2,3

(Figure 1a, b). This straight tube undergoes a complex process of looping and segmentation, orchestrated by a cascade of genes that are important for left-right programming and cardiomyocyte differentiation, before a four- chambered heart is formed. During this process various cells from the so-called second heart field (SHF) add to and grow into the primitive heart, which was initially formed solely by derivatives of the first heart-forming field (FHF)

4-10

. Within the cardiac crescent the splanchnic mesoderm destined to form either of these two heart fields can already be distinguished, with the precursors of the SHF located at the centre (Figure 1a). The contribution of the SHF is crucial for proper cardiogenesis. The FHF-derived structure does not contain the entire assembly of necessary elements to build a complete heart.

Initially, before cell populations from the SHF are added, the primitive heart tube consists of only endocardium and myocardium with a cardiac jelly in between (Figure 1c). During the looping of the heart this cardiac jelly becomes unevenly distributed, resulting in large protrusions at the sites of the atrioventricular (AV) junction and outflow tract (OFT). The initially acellular structures become invaded by mesenchymal cells derived from the endocardium as a result of epithelial-mesenchymal transformation (EMT)

11

. These early cushions will give rise to the cardiac valves in the mature heart, but are also instrumental in cardiac septum-formation. At the moment that the cushions become colonized, SHF-derived cells and cardiac neural crest cells (CNCs) start to populate the outflow and inflow portions of the heart (Figure 1d). The CNCs play likewise an important role in cardiac septation and in cushion development

12-15

. Since the majority of the CNCs is destined for apoptosis

12,13

, their contribution is suggested to be mainly instructive, rather than constructive.

The SHF contribution is both instructive as well as constructive. The SHF is divided in an anterior heart field (AHF), which adds to the OFT or arterial pole

4,8,10

, and a posterior field (PHF), from which the derivatives are located at the inflow tract (IFT) or venous pole (Figure 1d)

9

. Besides a considerable contribution to the IFT myocardium

9

, the PHF delivers the epicardial progenitors to the heart by forming the pro-epicardial organ (PEO)

16

.

The PEO consists of mesothelial protrusions and villi in the splanchnic mesoderm located near the venous pole of the heart (Figure 1d). During the looping stages of cardiac development, cells cross the coelomic cavity as free-floating cell aggregates (mammals and fish)

17,18

or guided by tissue bridges (avians)

19,20

, although a combination is also suggested

21

. At first, the epicardial (progenitor) cells attach to the naked heart tube at the dorsal side of the atrioventricular sulcus. From that point, they spread over the heart to cover it with its epicardial layer

22

.

It has only recently been discovered that the role of the outermost layer of the heart, the epicardium,

(6)

is much more extensive than being the ‘cover’ that enables friction-free movement in the pericardial cavity

17,23-26

. The derivatives of the epicardium, the epicardium-derived cells (EPDCs), contribute in both a constructive as well as an instructive way to normal cardiac development. EPDCs are generated soon after the heart has been enveloped entirely by epicardium, as a result of EMT

27

. From the subepicardial space, where the EPDCs are located at first, they migrate into the myocardium. They add to the AV cushions, the cardiac interstitium, and the fibrous heart skeleton by delivering fibroblasts

27-30

. EPDCs also stabilize the coronary arteries as smooth muscle cells and adventitial fibroblasts

26,28-31

. As regulators of cardiogenesis, EPDCs appear to be important for the formation of the compact

myocardium

30,32-34

and for proper ingrowth of coronary arteries into the aorta

35

. Cardiac looping

32,36

and development of the Purkinje fiber system

37

are also dependent on EPDC-generated signaling. It has not yet been investigated whether EPDCs remain situated in the cardiac interstitium or in the wall of coronary arteries once they have migrated there. Besides their ultimate destiny, their function in the adult setting remains to be studied since EPDCs hold great potential for application in cardiac regeneration therapy (discussed further below).

Cardiac regeneration therapy

Compared to the effective, mainly curative treatments of cardiac arrhythmias (catheter ablations, pacemakers and implantable defibrillators), cardiac valve dysfunction (artificial valves), and coronary obstructions (dotter procedures, stents and surgical bypasses), common therapy of the infarcted heart might be considered somewhat old-fashioned. Currently, general treatment aims at limiting the region of injury by reopening the blocked vessel, and at reducing workload of the remaining tissue with pharmaceutics. But real healing or revival of the dead tissue is not yet possible, and homograft transplantation is no first-line therapy because of the small size of donor organ pool, high risk of complications, and costs. Reasonably, the exciting data from the study by Orlic et al

38

which

Figure 1. Schematic picture illustrating consecutive stages of cardiac development. The cardiac crescent, which is derived from the splanchnic mesoderm (a) first forms the primary heart tube (PHT, pink, b) and atrioventricular endocardial cushions (EC, blue, d). The yellow area within the cardiogenic plates (a) and behind the PHT (c) depicts the second heart field (SHF) that in later stages adds myocardium to the developing heart (d). The subset of the SHF contributing to the arterial pole (AP) of the heart is called the anterior heart field (AHF), and the part that adds to the venous pole (VP) the posterior heart field (PHF, d). The pro-epicardial organ (PEO), which is a subpopulation of the SHF that can be distinguished as a lobulated protrusion at the VP of the heart delivers the epicardial cells to the heart (i.e. the epicardium, d). Cardiac neural crest cells (CNCs, dark blue cells) migrate from the neural tube into the heart, thereby passing through the SHF mesoderm (d). G: gut, C: coelomic cavity, Ep: epicardium.

Modified after Gittenberger-de Groot AC et al, Fetal Cardiology 3rd edition, in press.

(7)

14

|

Chapter 1

General Introduction

demonstrated that bone marrow-derived stem cells could regenerate the injured area and improve cardiac function, energized many cardiovascular scientists and instigated even more new research projects in this field.

1. Embryonic versus adult cells

A large variety of cell populations can be applied for cell-based therapy of the infarcted heart, each having its own profile of advantages, limitations and feasibility. Cell types used can be sorted along their developmental potential. The most potent cells in an organism regarding differentiation are the ‘totipotent’ blastomeres. They form the embryo and also the placenta. Later, during the blastocyst stage, the structure giving rise to the placenta (trophoblast) can be distinguished from the source of the three germ layers of the embryo, the inner cell mass. The cells from the inner cell mass are ‘pluripotent’ and are referred to as embryonic stem cells, with the capacity to self-renew, to differentiate into all lineages

3940,41

, and to grow clonogenic

42

. Stem cells that reside within the adult somatic tissue are already committed to a specific lineage, and are only able to differentiate into a more selected variety of differentiation pathways, making them ‘multipotent’ stem or progenitor cells.

In current basic fundamental animal research as well as in clinical experimental treatments mainly adult stem or progenitor cells are used, in comparison to the more scarce application of embryonic stem cells. Arguments for this imbalance could be that i) appropriate handling of embryonic stem cells is intricate, ii) usage of embryonic stem cells is ethically challenging, and iii) includes the risk of teratoma formation, iv) adult stem cells can be derived from the patient itself and thus rejection problems are obviated, v) some adult cells, like bone marrow-derived cells, are easily available in large amounts. Drawbacks of adult cells are, however, that possible genetic defects might impair function of the cells to be applied, that they can not be expanded unlimitedly, and that many types are not immediately available at the moment of e.g. acute infarction. Recently described induced pluripotent cells (iPS) might combine best properties of both. These are reprogrammed adult cells in which pluripotency can be induced in vitro by overexpressing a combination of specific transcription factors

43-46

. However, their feasibility in vivo has not yet been revealed, leaving them to be dreamed of for the

future.

2. Adult cells harvested from extracardiac structures

The most extensively studied adult stem cell populations at the moment are those derived from the bone marrow

47

. This is a rich source of various immature cells that are applied in basic and clinical experimental cardiac regeneration therapy, being hematopoietic stem cells (HSCs)

38,48-50

, a subset of these, the so-called side population cells

51-53

, mesenchymal stem cells (MSCs) or stromal cells

54-56

, multipotent adult progenitor cells (MAPCs)

57,58

, which are a subset of MSCs, and bone marrow-derived multipotent stem cells (BMSCs)

59

. MAPCs and BMSCs are able to differentiate into all three germ layers

57,59

a property which has always been considered to be exclusive for embryonic stem cells.

Commonly, especially in clinical trials, a mixture of these subpopulations is used, which are then referred to as bone marrow-derived mononuclear cells (BMCs)

60

. Bone marrow-derived cells do not necessarily need to be harvested from the bone marrow, some can also be isolated from the blood, like the circulating progenitor cells (CPCs)

61,62

which include the endothelial progenitor cells (EPCs)

63

. Furthermore, potential cardiotherapeutic progenitor cell populations can be derived from adipose tissue

64,65

, skeletal muscle

66-68

, placental cord blood

69

, and probably many other less obvious tissues.

3. Adult cells derived from the heart

It seems logical to expect cell populations derived from the target-organ being the most appropriate cell source for its own tissue regeneration, supposing these cells are present in the organ of interest.

With regard to the heart, the presumed cardiac derived progenitor cells would probably be committed

(8)

to the cardiac lineage, adapted to the specific environment and they might have been programmed to excrete factors of use for the heart. Research of the last decade focused to find progenitor cells in the adult heart revealed indeed several progenitor populations, although the heart has long been considered as an organ without regenerative potential. The heart was demonstrated to possess endogenous regenerative potential

70

and to harbor small multipotent cells that could differentiate into cardiomyocytes. These cells were recognized based on their expression of stem cell markers stem cell antigen-1 (sca-1)

71-76

(for human tissue referred to as cardiomyocyte progenitor cell or CMPC), c-kit

77

, or the primitive SHF marker islet1 (isl1)

78,79

. But they were also phenotyped by their expression profile in combination with adherence properties (cardiospheres)

80

, or their ability to efflux the Hoechst dye

81

. Of special interest are the CMPCs which can be isolated from human fetal and adult cardiac tissue, since they can differentiate into cardiomyocytes in vitro without the necessity of being co-cultured with neonatal cardiomyocytes

74

. The different multipotent residents described by various groups might, however, represent closely related subpopulations of one major cardiac stem cell pool. If these different cell types with cardiomyocyte differentiation potential characterize isolated populations resident in the heart, it is expected that ischemic injury would at least partly be mended by the organ itself. Although adult stem and progenitor cells from non-cardiac tissues are already widely tested for their therapeutic applicability, stem and progenitor cells resident in the heart itself are currently only of modest interest when evaluated for their usage in clinical studies.

Knowing that the adult heart harbors cardiomyocyte-precursors leads to the expectation that other lineage-destined ‘remnants’ of early development are also present in the postnatal heart, including EPDCs. As mentioned before, EPDCs are progenitors of the cardiac interstitial fibroblasts and medial and adventitial cells of the coronary arteries with extensive instructive properties during cardiogenesis. It might be speculated that adult EPDCs could recapitulate their embryonic properties, for example in case of ischemic injury. It is, however, not known whether EPDCs are indeed present and still potent in the adult heart.

In this thesis it is studied whether EPDCs could be derived from the adult epicardium

82

, and if these cells could be of benefit for cardiac repair of the adult ischemic heart. It is hypothesized that the adult EPDCs improve cardiac function after myocardial infarction through paracrine signaling like they demonstrate during embryogenesis. This presumed supportive and instructive effect of the adult EPDCs on the surrounding injured host tissue is also elucidated. It is tested whether their profit could be further increased by co-transplanting complementary CMPCs. Being mainly located in the atrium

83

, CMPCs are suggested to originate from the SHF like EPDCs, designating them as ontogenetically closely related. A further mesenchymal cell type is studied, the bone marrow-derived MSC. It is examined whether their potential is retained if they are derived from ischemic heart disease patients, as opposed to healthy donors.

Functional assessment of the infarcted mouse heart

The extensive interest in the cardiac stem-cell field, with the general aim to cure the adult heart by regenerative cellular therapies, has resulted in the emergence of numerous basic animal studies.

Although small rodents are phylogenetically less related to human than large mammals, mouse and rat are most commonly applied for basic animal studies, since their genetic background is well known and their usage is less complicated and expensive than that of larger animals. To evaluate the effect of the cell type of interest in vivo, i.e. to test whether it can stimulate cardiac performance, functional assessment is needed. This requires highly specialized techniques, in particular for the mouse, considering the extreme small size of the heart (7 mm) and high heart rate (~500 beats per minute).

Nonetheless, magnetic resonance imaging (MRI), echocardiography and invasive pressure-volume (PV)

loop measurements have become available for cardiac phenotyping of small animals.

(9)

16

|

Chapter 1

General Introduction

In this thesis several aspects of MRI, which is the method of choice for longitudinal stem cell

experiments

84

, are described. Additional applications of MRI, being infarct size measurements and

identification of transplanted cells in the living heart by usage of iron labeling, are discussed. Also,

MRI and PV loop measurements, which each address different aspects of left ventricular function, are

compared for their reliability in the infarcted mouse heart.

(10)

Chapter Outline

Chapter 1 provides background information regarding embryonic heart development and stem cell therapy for cardiac regeneration. Technical issues in functional basic experimental stem cell research are shortly introduced.

Chapter 2 describes the contribution of EPDCs to the developing heart. It speculates on the role of these cells in the adult heart, in specific their potential in the infarcted heart.

Chapter 3 translates the knowledge from the embryonic situation to the adult diseased heart, investigating whether EPDCs harvested from the adult heart can contribute to functional improvement after myocardial infarction.

Chapter 4 addresses potential underlying mechanisms that might explain the positive effect of adult EPDCs on the infarcted adult heart. It focuses on histological and functional differences between normal scar development and cardiac healing after adult EPDCs have been transplanted.

Chapter 5 reports on the interaction between CMPCs and EPDCs. Their mutual effect is studied parallel in the in vitro and in vivo situation, from which the latter tests the hypothesis that combined transplantation will be of a synergistic benefit for the infarcted heart.

Chapter 6 concerns the applicability of MSCs from ischemic heart disease patients to cure the infarcted heart. Engraftment and differentiation of transplanted MSCs are addressed, as well as their potential to improve LV function.

Chapter 7 compares two methods used for heart function determination of small laboratory animals.

Reliability of MRI and PV–loop measurements for assessment of cardiac function of the failing mouse heart is analyzed.

Chapter 8 describes methodological issues around iron-oxide labeling for tracing of stem cells in the mouse heart.

Chapter 9 provides a general discussion on the concept of using cardiogenesis as basis for cardiac

regeneration therapy. Application of adult EPDCs to improve cardiac function after myocardial

infarction is addressed, as are potential mechanisms which might explain the detected profit. Other

mesoderm-derived cell types are conferred, and technical issues in functional assessment of the

infarcted heart are placed in perspective.

(11)

Reference List

1.

International Cardiovascular Disease Statistics. American Heart Association.

2009.

2.

DeRuiter MC, Poelmann RE, VanderPlas- de Vries I, Mentink MMT, Gittenberger- de Groot AC. The development of the myocardium and endocardium in mouse embryos. Fusion of two heart tubes? Anat Embryol. 1992; 185:461-473.

3.

Gittenberger-de Groot AC, Bartelings MM, DeRuiter MC, Poelmann RE.

Basics of cardiac development for the understanding of congenital heart malformations. Pediatr Res. 2005;

57:169-176.

4.

Kelly RG, Brown NA, Buckingham ME.

The arterial pole of the mouse heart forms from Fgf10-expressing cells in pharyngeal mesoderm. Dev Cell. 2001;

1:435-440.

5.

Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, Evans S. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell.

2003; 5:877-889.

6.

Buckingham M, Meilhac S, Zaffran S.

Building the mammalian heart from two sources of myocardial cells. Nat Rev Genet. 2005; 6:826-835.

7.

Meilhac SM, Esner M, Kelly RG, Nicolas JF, Buckingham ME. The clonal origin of myocardial cells in different regions of the embryonic mouse heart. Dev Cell.

2004; 6:685-698.

8.

Mjaatvedt CH, Nakaoka T, Moreno- Rodriguez R, Norris RA, Kern MJ, Eisenberg CA, Turner D, Markwald RR. The outflow tract of the heart is recruited from a novel heart-forming field. Dev Biol. 2001; 238:97-109.

9.

Gittenberger-de Groot AC, Mahtab EA, Hahurij ND, Wisse LJ, DeRuiter MC, Wijffels MC, Poelmann RE. Nkx2.5- negative myocardium of the posterior heart field and its correlation with podoplanin expression in cells from the developing cardiac pacemaking and conduction system. Anat Rec (Hoboken ).

2007; 290:115-122.

10.

Kelly RG. Molecular inroads into the anterior heart field. Trends Cardiovasc Med. 2005; 15:51-56.

11.

Markwald RR, Fitzharris TP, Manasek FJ.

Structural development of endocardial cushions. Am J Anat. 1977; 148:85-119.

12.

Poelmann RE, Gittenberger-de Groot AC. A subpopulation of apoptosis-prone cardiac neural crest cells targets to the venous pole: multiple functions in heart development? Dev Biol. 1999;

207:271-286.

13.

Poelmann RE, Mikawa T, Gittenberger-de Groot AC. Neural crest cells in outflow tract septation of the embryonic chicken heart: differentiation and apoptosis. Dev Dyn. 1998; 212:373-384.

14.

Snider P, Olaopa M, Firulli AB, Conway SJ. Cardiovascular development and the colonizing cardiac neural crest lineage. ScientificWorldJournal. 2007;

7:1090-1113.

15.

Kirby ML, Waldo KL. Neural crest and cardiovascular patterning. Circ Res.

1995; 77:211-215.

16.

Mahtab EA, Wijffels MC, Van Den Akker NM, Hahurij ND, Lie-Venema H, Wisse LJ, DeRuiter MC, Uhrin P, Zaujec J, Binder BR, Schalij MJ, Poelmann RE, Gittenberger-de Groot AC. Cardiac malformations and myocardial abnormalities in podoplanin knockout mouse embryos: Correlation with abnormal epicardial development. Dev Dyn. 2008; 237:847-857.

17.

Viragh S, Challice CE. The origin of the epicardium and the embryonic myocardial circulation in the mouse.

Anat Rec. 1981; 201:157-168.

18.

Munoz-Chapuli R, Macias D, Ramos C, Fernandez B., Sans-Coma V.

Development of the epicardium in the dogfish (Scyliorhinus canicula). Acta Zool. 1997; 78:39-46.

19.

Männer J. Experimental study on the formation of the epicardium in chick embryos. Anat Embryol (Berl). 1993;

187:281-289.

20.

Männer J. The development of pericardial villi in the chick embryo.

Anat Embryol (Berl). 1992; 186:379-385.

21.

Nahirney PC, Mikawa T, Fischman DA.

Evidence for an extracellular matrix bridge guiding proepicardial cell migration to the myocardium of chick embryos. Dev Dyn. 2003; 227:511-523.

22.

Vrancken Peeters M-PFM, Mentink MMT, Poelmann RE, Gittenberger-de Groot AC.

Cytokeratins as a marker for epicardial formation in the quail embryo. Anat Embryol. 1995; 191:503-508.

23.

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

188:381-393.

24.

Mikawa T, Fischman DA. Retroviral analysis of cardiac morphogenesis:

discontinuous formation of coronary vessels. Proc Natl Acad Sci U S A. 1992;

89:9504-9508.

25.

Poelmann RE, Gittenberger-de Groot AC, Mentink MM, Bokenkamp R, Hogers B. Development of the cardiac coronary vascular endothelium, studied with antiendothelial antibodies, in chicken-quail chimeras. Circ Res. 1993;

73:559-568.

26.

Mikawa T, Gourdie RG. Pericardial mesoderm generates a population of coronary smooth muscle cells migrating into the heart along with ingrowth of the epicardial organ. Dev Biol. 1996;

174:221-232.

27.

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

(12)

28.

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

29.

Männer J. Does the subepicardial mesenchyme contribute

myocardioblasts to the myocardium of the chick embryo heart? A quail-chick chimera study tracing the fate of the epicardial primordium. Anat Rec. 1999;

255:212-226.

30.

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

31.

Vrancken Peeters MP, Gittenberger-de Groot AC, Mentink MM, Poelmann RE.

Smooth muscle cells and fibroblasts of the coronary arteries derive from epithelial-mesenchymal transformation of the epicardium. Anat Embryol (Berl).

1999; 199:367-378.

32.

Gittenberger-de Groot AC, Vrancken Peeters MP, Bergwerff M, Mentink MM, Poelmann RE. Epicardial outgrowth inhibition leads to compensatory mesothelial outflow tract collar and abnormal cardiac septation and coronary formation. Circ Res. 2000;

87:969-971.

33.

Lie-Venema H, Gittenberger-de Groot AC, van Empel LJ, Boot MJ, Kerkdijk H, de Kant E, DeRuiter MC. Ets-1 and Ets-2 transcription factors are essential for normal coronary and myocardial development in chicken embryos. Circ Res. 2003; 92:749-756.

34.

Kang JO, Sucov HM. Convergent proliferative response and divergent morphogenic pathways induced by epicardial and endocardial signaling in fetal heart development. Mech Dev.

2005; 122:57-65.

Groot AC. Coronary artery and orifice development is associated with proper timing of epicardial outgrowth and correlated Fas-ligand-associated apoptosis patterns. Circ Res. 2005;

96:526-534.

36.

Lie-Venema H, Van Den Akker NM, Bax NA, Winter EM, Maas S, Kekarainen T, Hoeben RC, DeRuiter MC, Poelmann RE, Gittenberger-de Groot AC. Origin, fate, and function of epicardium- derived cells (EPDCs) in normal and abnormal cardiac development.

ScientificWorldJournal. 2007; 7:1777- 1798.

37.

Eralp I, Lie-Venema H, Bax NA, Wijffels MC, van der LA, DeRuiter MC, Bogers AJ, Van Den Akker NM, Gourdie RG, Schalij MJ, Poelmann RE, Gittenberger-de Groot AC. Epicardium-derived cells are important for correct development of the Purkinje fibers in the avian heart.

Anat Rec A Discov Mol Cell Evol Biol.

2006; 288:1272-1280.

38.

Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;

410:701-705.

39.

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

40.

Dawn B, Bolli R. Adult bone marrow- derived cells: Regenerative potential, plasticity, and tissue commitment.

Basic Res Cardiol. 2005; 100:494-503.

41.

Kocher AA, Schlechta B, Gasparovicova A, Wolner E, Bonaros N, Laufer G. Stem cells and cardiac regeneration. Transpl Int. 2007.

42.

Anderson DJ, Gage FH, Weissman IL. Can stem cells cross lineage boundaries? Nat Med. 2001; 7:393-395.

43.

Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures

44.

Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S.

Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007; 131:861-872.

45.

Yu J, Vodyanik MA, Smuga-Otto K, ntosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;

318:1917-1920.

46.

Nishikawa S, Goldstein RA, Nierras CR. The promise of human induced pluripotent stem cells for research and therapy. Nat Rev Mol Cell Biol. 2008;

9:725-729.

47.

Dimmeler S, Zeiher AM, Schneider MD. Unchain my heart: the scientific foundations of cardiac repair. J Clin Invest. 2005; 115:572-583.

48.

Gunsilius E, Gastl G, Petzer AL.

Hematopoietic stem cells. Biomed Pharmacother. 2001; 55:186-194.

49.

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.

50.

Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, 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.

51.

Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med. 1996;

183:1797-1806.

52.

Guo Y, Follo M, Geiger K, Lubbert M, Engelhardt M. Side-population cells from different precursor compartments.

(13)

53.

Jackson KA, Majka SM, Wang H, 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. J Clin Invest. 2001;

107:1395-1402.

54.

Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004; 95:9-20.

55.

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.

56.

Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, Zhang JJ, Chunhua RZ, 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. Am J Cardiol. 2004; 94:92-95.

57.

Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM.

Pluripotency of mesenchymal stem cells derived from adult marrow. Nature.

2002; 418:41-49.

58.

Check E. Stem-cell paper corrected.

Nature. 2007; 447:763.

59.

Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T, Kusano K, Hanley A, Scadova H, Qin G, Cha DH, Johnson KL, Aikawa R, Asahara T, Losordo DW.

Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. J Clin Invest.

2005; 115:326-338.

60.

Dimmeler S, Burchfield J, Zeiher AM.

Cell-based therapy of myocardial infarction. Arterioscler Thromb Vasc Biol. 2008; 28:208-216.

61.

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.

62.

Schachinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND, Vogl TJ, Hofmann WK, Martin H, Dimmeler S, Zeiher AM.

Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J Am Coll Cardiol. 2004; 44:1690-1699.

63.

Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res. 2004;

95:343-353.

64.

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. Circ Res.

2004; 94:223-229.

65.

Yamada Y, Wang XD, Yokoyama SI, Fukuda N, Takakura N. Cardiac progenitor cells in brown adipose tissue repaired damaged myocardium.

Biochem Biophys Res Commun. 2006.

66.

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. Nat Med. 1998; 4:929- 933.

67.

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.

68.

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. J Am Coll Cardiol. 2003;

41:1078-1083.

69.

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 J, Caplan AI, Colletti EJ, meida-Porada G, Muller HW, Zanjani E, Wernet P. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J Exp Med. 2004; 200:123-135.

70.

Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, Bussani R, Nadal-Ginard B, Silvestri F, Leri A, Beltrami CA, Anversa P.

Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med. 2001; 344:1750-1757.

71.

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. Proc Natl Acad Sci U S A. 2003; 100:12313-12318.

72.

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 cardiomyocyte differentiation. Circ Res.

2005; 97:52-61.

73.

Wang X, Hu Q, Nakamura Y, Lee J, Zhang G, From AH, Zhang J. The Role of Sca-1+/

CD31- Cardiac Progenitor Cell Population in Postinfarction LV Remodeling. Stem Cells. 2006.

74.

Goumans MJ, de Boer T, Smits AM, Van Laake LW, van Vliet P, Metz CH, Korfage T, Kats P, Hochstenbach R, Pasterkamp G, Verhaar MC, van der Heyden MS, de Kleijn D, Mummery CL, van Veen TA, Sluijter JP, Doevendans PA. TGFb1 induces efficient differentiation of human cardiomyocyte progenitor cells into functional cardiomyocytes in vitro.

Stem Cell Research. 2008; 1:138-149.

75.

Smits AM, van Vliet P, Metz CH, Korfage T, Sluijter JP, Doevendans PA, Goumans MJ. Human cardiomyocyte progenitor cells differentiate into functional mature cardiomyocytes; an in vitro model for studying human cardiac physiology and pathophysiology. Nat Protoc. 2008; in press.

76.

Matsuura K, Nagai T, Nishigaki N, Oyama T, Nishi J, Wada H, Sano M, Toko H, Akazawa H, Sato T, Nakaya H, Kasanuki H, Komuro I. Adult cardiac

(14)

Sca-1-positive cells differentiate into beating cardiomyocytes. J Biol Chem.

2004; 279:11384-11391.

77.

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.

78.

Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, Lin LZ, Cai CL, Lu MM, Reth M, Platoshyn O, Yuan JX, Evans S, Chien KR. Postnatal isl1+

cardioblasts enter fully differentiated cardiomyocyte lineages. Nature. 2005;

433:647-653.

79.

Laugwitz KL, Moretti A, Caron L, Nakano A, Chien KR. Islet1 cardiovascular progenitors: a single source for heart lineages? Development. 2008; 135:193- 205.

80.

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

95:911-921.

81.

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. Dev Biol. 2004; 265:262-275.

82.

van Tuyn J, Atsma DE, Winter EM, van der Velde-van Dijke I, Pijnappels DA, Bax NAM, Knaan-Shanzer S, Gittenberger- de Groot AC, Poelmann RE, van der Laarse A, van der Wall EE, Schalij MJ, de Vries AAF. Epicardial cells of human adults can undergo an epithelial-to- mesenchymal transition and obtain characteristics of smooth muscle cells in vitro. Stem Cells. 2007; 25:271-278.

83.

van Vliet P, Roccio M, Smits AM, van Oorschot AA, Metz CH, van Veen TA, Sluijter JP, Doevendans PA, Goumans

84.

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.

(15)

Referenties

GERELATEERDE DOCUMENTEN

License: Licence agreement concerning inclusion of doctoral thesis in the Institutional Repository of the University of Leiden Downloaded from: https://hdl.handle.net/1887/14054.

The work presented in this thesis was carried out at the Department of Anatomy and Embryology of the Leiden University Medical Center and was supported by a grant from

OT: outflow tract, SV: sinus venosus, PEO: proepicardial organ, AVC: atrioventricular cushion, EPDC: epicardium-derived cell, Ep: epicardium, V: ventricle, A: atrium,

Key findings of the present study are that in an immune-compromised mouse model of acute MI, intramyocardial injection of hMSCs from patients with IHD resulted in i) a

Figure 5: Mean end-diastolic volume (EDV), end-systolic volume (ESV), and ejection fraction (EF) by magnetic resonance imaging (MRI) and conductance catheter (CC) in mice

PBS phosphate buffered saline PCNA proliferating cell nuclear antigen PDGF platelet derived growth factor PDGFR platelet derived growth factor receptor.. 204 | Abbriviations

Ejectie fractie was toegenomen 2 weken na operatie en transplantatie, en deze verbetering bleef behouden tot het laatste tijdspunt waarop gemeten werd, te weten 6 weken na

The cell-based therapies involve transplantation of human pluripotent stem cell (hPSC)-derived cardiac progenitor cells (CPCs), cardiomyocytes (CMs), or multiple cardiac