The creation of regenerative stem cell therapies for heart disease requires that we understand the molecular mechanisms that govern the fates and differentiation of the diverse muscle and non-muscle cell lineages of the heart. Recently, different cardiac cell types have been reported to arise from a common, multipotent Islet1 (Isl1)-positive progenitor, suggesting that a clonal model of heart lineage diversification might occur that is analogous to hematopoiesis. The ability to isolate, renew and differentiate Isl1+ precursors from postnatal and embryonic hearts and from embryonic stem cells provides a powerful cell-based system for characterizing the signaling pathways that control cardiovascular progenitor formation, renewal, lineage specification and conversion to specific differentiated progeny.

“Is myogenesis controlled by a single master gene? Possibly,combinatorial schemes and cell lineage are both used to generate positional identity in the embryo for each individual cell.”

Hal Weintraub and colleagues (Davis et al., 1987)

Heart is muscle. Or so it would seem to the outside observer. From this vantage point, the discovery of MyoD almost 20 years ago by Weintraub, Lassar and colleagues (Davis et al.,1987) represents one of the seminal advances in our understanding of myogenesis, not only in skeletal muscle, but also in cardiac muscle. Subsequent studies by many investigators (for a review, see Srivastava and Olson, 2000; Olson, 2006) have underscored this point and have documented the conservation of the pathways and principles of myogenesis across muscle types, spanning the evolutionary spectrum from invertebrates to humans. This beautiful body of work has had major implications for our understanding of both development and disease, and has uncovered a host of genetic regulatory circuits that contribute to the control of cardiogenesis. As such, the molecular paradigm for cardiogenesis has largely been based on studies of skeletal myogenesis, an approach that has yielded spectacular advances in the field of cardiovascular science and medicine.

But is heart more than muscle? And if so, might there be another major paradigm that accounts for the generation of these diverse cell types, which include endothelial, smooth muscle and conduction system cells? Early cell lineage-tracing studies in avian systems indicated that a common muscle cell precursor exists for both working myocardium (atrial and ventricular myocardium) and the conduction system(Mikawa, 1999). At the same time, other studies identified a common progenitor, the hemangioblast, for endothelial and blood cell lineages, a finding which indicated that a common precursor might also exist for endothelial cells in the heart(Choi et al., 1998; Fehling et al., 2003; Kouskoff et al., 2005). The recent discovery of multipotent Islet1-positive (Isl1+) progenitors in several species, including mouse, rat and human, and in different regions of the embryonic and adult heart, implies that there might be a stem cell paradigm for the generation of diverse cell lineages in the heart(Moretti et al., 2006). Unexpectedly, a growing body of evidence from multiple independent laboratories now suggests that, with respect to lineage diversification, the heart could be like blood, an organ in which a single stem/progenitor cell is able to generate all of the major cell types of the system(Kattman et al., 2006; Wu et al., 2006). The concept of progressive lineage restriction is well accepted for hematopoiesis but has not been established in such detail in the development of solid organs such as the heart. In this review, we explore this idea further by focusing on the Isl1 cardiovascular progenitor story, placing it into the context of the generation of diverse cardiovascular lineages, and discussing its implications for cardiovascular development and disease. The reader is also directed to reviews on the potential general role of progenitors in cardiogenesis and disease (see Buckingham et al.,2005; Chien and Karsenty,2005; Srivastava,2006; Black,2007).

The heart is composed of diverse muscle and non-muscle cell lineages:atrial/ventricular cardiac myocytes, conduction system cells, smooth muscle/endothelial cells of the coronary arteries and veins, endocardial cells, valvular components and connective tissue(Fig. 1). During cardiogenesis,the differentiation of these multiple heart lineages is under tight spatial and temporal control, resulting in the coordinated formation of the distinct tissue components of the heart, including the four specialized chambers,diverse structures of the conduction system, the endocardium, the heart valves, the coronary arterial tree and the outflow tract(Harvey, 2002; Brand, 2003). Understanding how this diversity of heart cell lineages arises is a fundamental question that has major implications for understanding and treating both congenital and adult heart diseases, a subset of which have recently been shown to be due to defects in the pathways involved in heart lineage specification(Schott et al., 1998; Benson et al., 1999; Garg et al., 2003; Pashmforoush et al., 2004)(Table 1).

Table 1.

Gene mutations in human non-syndromic congenital cardiovascular malformations

GeneInheritanceCardiac malformationsReferences
NKX2-5 (NK2 transcription factor related, locus 5) Autosomal dominant ASD, conduction system defects (Schott et al., 1998; Benson et al., 1999; Goldmuntz et al., 2001; Elliott et al., 2003; McElhinney et al., 2003
GATA4 (GATA binding protein 4) Autosomal dominant ASD, VSD (Garg et al., 2003
TBX20 (T-box 20) Partial penetrance ASD, VSD, valve abnormalities, cardiomyopathy (Kirk et al., 2007
JAG1 (jagged 1) Partial penetrance TOF (Krantz et al., 1999
CRELD1 (cysteine-rich with EGF-like domains 1) Partial penetrance AVSD (Robinson et al., 2003
NOTCH1 Autosomal dominant BAV, calcification (Garg et al., 2005
MYH6 (myosin heavy chain 6) Autosomal dominant, partial penetrance ASD (Ching et al., 2005
GeneInheritanceCardiac malformationsReferences
NKX2-5 (NK2 transcription factor related, locus 5) Autosomal dominant ASD, conduction system defects (Schott et al., 1998; Benson et al., 1999; Goldmuntz et al., 2001; Elliott et al., 2003; McElhinney et al., 2003
GATA4 (GATA binding protein 4) Autosomal dominant ASD, VSD (Garg et al., 2003
TBX20 (T-box 20) Partial penetrance ASD, VSD, valve abnormalities, cardiomyopathy (Kirk et al., 2007
JAG1 (jagged 1) Partial penetrance TOF (Krantz et al., 1999
CRELD1 (cysteine-rich with EGF-like domains 1) Partial penetrance AVSD (Robinson et al., 2003
NOTCH1 Autosomal dominant BAV, calcification (Garg et al., 2005
MYH6 (myosin heavy chain 6) Autosomal dominant, partial penetrance ASD (Ching et al., 2005

ASD, atrial septal defect; AVSD, atrioventricular septal defect; BAV,bicuspid aortic valve; TOF, tetralogy of Fallot; VSD, ventricular septal defect.

In this regard, three major sources of heart cell precursors have been identified in the embryo: the cardiogenic mesoderm, the cardiac neural crest and the proepicardial organ (see Fig. 1A,B and glossary, Box 1). Each of these represents a spatially and temporally distinct pool of embryonic heart progenitors, which are known to give rise to distinct cardiac structures and cell components. The cardiogenic mesoderm forms the linear heart tube (see Box 1)and ultimately the bulk of the working myocardium in the ventricular and atrial chambers. The cardiac neural crest migrates into the heart at a later stage of cardiac development, and gives rise to the vascular smooth muscle of the aortic arch, ductus arteriosus (see Box 1) and the great vessels; additionally, neural crest contributes to essential components of the cardiac autonomic nervous system(Kirby et al., 1983; Epstein and Buck, 2000). The mesenchyme portion of the developing heart and the majority of epicardial cells are derived from the proepicardium(Dettman et al., 1998; Moore et al., 1999; Manner et al., 2001). The epicardial mantle begins to envelop the myocardium at the same time that coronary precursor cells first appear in the heart tube(Mikawa and Gourdie, 1996) and retroviral genetic labeling experiments have shown that single vasculogenic cells of the proepicardium differentiate into solitary vessel-associated clusters that consist of three cell types: endothelial, smooth muscle and perivascular connective tissue cells(Mikawa and Gourdie, 1996). This evidence suggests that the coronary vasculature might be partially derived from mesodermal cells in the proepicardium, although the contribution of the proepicardium to the endothelial lineage is still controversial(Poelmann et al., 2002). Taken together, these studies on the contribution of the primordial heart field,cardiac neural crest, and of the proepicardium to specialized structures during cardiogenesis indicate that lineage diversification in the heart can arise, in part, via distinct pools of cardiovascular progenitors that are spatially and temporally segregated in the developing embryo (see Fig. 1B).

Fig. 1.

Origin and lineage relationship of cardiac cell types. (A)Contribution of the three populations of embryonic heart progenitors,cardiogenic mesoderm (red), cardiac neural crest (purple) and proepicardial organ (yellow) to different heart compartments during cardiac morphogenesis in the mouse. Progenitors of the cardiogenic mesoderm are first recognizable under the head folds (HFs) of the embryo at E7.5, then move ventrally to the midline (ML) and form initially the linear heart tube and ultimately the four chambers of the heart. After the looping of the heart tube (E8.5), cardiac neural crest progenitors migrate from the dorsal neural tube to engulf the aortic arch arteries and contribute to vascular smooth muscle cells of the outflow tract (OFT) around E10.5. At the same time in mouse development, the proepicardial organ precursors contact the surface of the developing heart,give rise to the epicardial mantle (yellow) and contribute later to the coronary vasculature. In the fetal heart (∼E14), the chambers separate due to septation and are connected to the pulmonary trunk (PT) and aorta (Ao). Cranial (Cr)-caudal (Ca), right (R)-left (L), and dorsal (D)-ventral (V) axes are indicated. (B) Cardiac cell types that arise through the lineage diversification of the three embryonic precursor pools in the mouse heart. Whereas the contribution of the proepicardium to the smooth muscle cells of the coronary system and to the mesenchymal cells of the heart is well accepted, the origin of the endothelial lineage in the coronary vasculature is still controversial. AA, aortic arch; IVS, interventricular septum; LA, left atrium; LV, left ventricle; PhA, pharyngeal arches; PLA, primitive left atrium; PRA, primitive right atrium; RA, right atrium; RV, right ventricle.

Fig. 1.

Origin and lineage relationship of cardiac cell types. (A)Contribution of the three populations of embryonic heart progenitors,cardiogenic mesoderm (red), cardiac neural crest (purple) and proepicardial organ (yellow) to different heart compartments during cardiac morphogenesis in the mouse. Progenitors of the cardiogenic mesoderm are first recognizable under the head folds (HFs) of the embryo at E7.5, then move ventrally to the midline (ML) and form initially the linear heart tube and ultimately the four chambers of the heart. After the looping of the heart tube (E8.5), cardiac neural crest progenitors migrate from the dorsal neural tube to engulf the aortic arch arteries and contribute to vascular smooth muscle cells of the outflow tract (OFT) around E10.5. At the same time in mouse development, the proepicardial organ precursors contact the surface of the developing heart,give rise to the epicardial mantle (yellow) and contribute later to the coronary vasculature. In the fetal heart (∼E14), the chambers separate due to septation and are connected to the pulmonary trunk (PT) and aorta (Ao). Cranial (Cr)-caudal (Ca), right (R)-left (L), and dorsal (D)-ventral (V) axes are indicated. (B) Cardiac cell types that arise through the lineage diversification of the three embryonic precursor pools in the mouse heart. Whereas the contribution of the proepicardium to the smooth muscle cells of the coronary system and to the mesenchymal cells of the heart is well accepted, the origin of the endothelial lineage in the coronary vasculature is still controversial. AA, aortic arch; IVS, interventricular septum; LA, left atrium; LV, left ventricle; PhA, pharyngeal arches; PLA, primitive left atrium; PRA, primitive right atrium; RA, right atrium; RV, right ventricle.

One of the earliest steps in cardiogenesis is the formation of the cardiac crescent (Fig. 2 and see Box 1), which is derived from cells of the mesoderm that become instructed to adopt a cardiac fate in response to signals from adjacent tissues(Harvey, 2002; Srivastava and Olson, 2000). Recent studies have revealed that the cardiogenic mesoderm in fact consists of two populations or fields (see Box 1) of cardiac precursor cells that contribute to different parts of the heart. The earliest population of cardiac progenitors, referred to as the first heart field, originates in the anterior splanchnic mesoderm, gives rise first to the cardiac crescent, later to the linear heart tube, and ultimately contributes to parts of the atrial chambers and the left ventricular region. The second cardiogenic region, known as the second heart field, lies anterior and dorsal to the linear heart tube and is derived from the pharyngeal mesoderm medial to the cardiac crescent (see Fig. 2A and Box 1). Cells from this second heart lineage are added to the developing heart tube and give rise to the outflow tract, the right ventricular region and the main parts of the atrial tissue (reviewed by Kelly and Buckingham,2002; Buckingham et al.,2005). The discovery of the second heart fieldreads like a scientific detective story, in which independent pieces of evidence were brought together to ultimately identify a prime suspect.

Fig. 2.

First and second heart fields and their contributions to the developing heart. (A) The upper drawings show the relative position, movement and contribution of the second heart field progenitors (green) relative to the first heart field cells (red) from the cardiac-crescent through to the looping stages of mouse heart development. The dashed lines indicate the position of the corresponding sections shown in the lower panels. (B) Location(upper) and contribution (beneath) of the second heart field progenitors(blue) to the outflow tract in the chick embryo. Cranial (Cr)-caudal (Ca),right (R)-left (L), and dorsal (D)-ventral (V) axes are indicated. DOFT,distal outflow tract; HFs, head folds; LA, left atrium; LV, left ventricle;ML, midline; PhA, pharyngeal arches; OFT, outflow tract; POFT, proximal outflow tract; RA, right atrium; RV, right ventricle.

Fig. 2.

First and second heart fields and their contributions to the developing heart. (A) The upper drawings show the relative position, movement and contribution of the second heart field progenitors (green) relative to the first heart field cells (red) from the cardiac-crescent through to the looping stages of mouse heart development. The dashed lines indicate the position of the corresponding sections shown in the lower panels. (B) Location(upper) and contribution (beneath) of the second heart field progenitors(blue) to the outflow tract in the chick embryo. Cranial (Cr)-caudal (Ca),right (R)-left (L), and dorsal (D)-ventral (V) axes are indicated. DOFT,distal outflow tract; HFs, head folds; LA, left atrium; LV, left ventricle;ML, midline; PhA, pharyngeal arches; OFT, outflow tract; POFT, proximal outflow tract; RA, right atrium; RV, right ventricle.

Box 1. Glossary of specialized terms

Branchial arches: A series of paired segmental structures composed of ectoderm, mesoderm and neural crest cells that are positioned on either side of the developing pharynx. In mammals, the branchial arches contribute to pharyngeal organs and to the connective, skeletal, neural and vascular tissues of the head.

Cardiac crescent: Crescent-shaped epithelium that represents the first recognizable cardiac progenitors at the cranial and cranial-lateral parts of the embryo. The cardiac crescent fuses at the midline to form the early heart tube.

Cardiogenic mesoderm: Mesoderm is one of the three layers of cells in the early embryo that, together with the endoderm and ectoderm, provides the source of all subsequent cell types that appear during embryogenesis. The cardiogenic mesoderm gives rise to the specialized cells of the heart and includes both first and second heart field precursors.

Ductus arteriosus: A shunt connection between the pulmonary artery and the aortic arch that allows most of the blood from the right ventricle to bypass the embryonic lungs.

Heart field: A defined special area of cells in the mesoderm that is fated to form the cardiac organ.

Linear heart tube: Transient structure of the early developing heart composed of an inner endothelial tube shrouded by a myocardial layer.

Myocardial cell lineage: The term lineage emphasizes the history of a cell and its descendents, and the characterization of their clonal contribution to the heart. The first and second myocardial lineages represent the progenitors of the first and second heart fields and their differentiated progeny.

Neural crest: Progenitor cells present in vertebrates that arise from the dorsal neural tube and migrate to other sites in the embryo and differentiate into various cell types depending on location.

Pharyngeal mesoderm: The mesoderm that is located below the head in the pharynx, which is the part of the embryo where the developing respiratory and digestive systems are situated.

Primitive streak: Transitory embryonic structure, present as a strip of cells that prefigures the anterior-posterior axis of the embryo. Proepicardial organ: A cluster of mesothelial cells located on the right side of the external surface of the sinus venosus. These cells contact the dorsal wall of the tubular heart in the region of the atrioventricular junction and form a monolayer that gradually covers the heart.

Retrospective clonal analysis: A genetic approach for lineage analysis that is based on random labeling of precursor cells. The example cited in this review employed a lacZ carrying a duplication (laacZ)that renders it non-functional. Only upon a rare, spontaneous intragenic recombination which removes the duplication do cells express the lacZreporter and can be clonally analyzed.

The initial evidence that the outflow tract was not present in the linear heart-tube stage came from a series of in vivo lineage-tracing experiments performed in chick embryos by de la Cruz and co-workers in the 1970s, which indicated that the outflow tract myocardium originates from a progenitor population that is different from the cardiac crescent and is situated anterior to the heart tube (reviewed by de la Cruz and Sanchez-Gomez, 2000). Over the past few years, the source of the outflow tract and the right ventricle has been addressed by studies from three different laboratories, two performed in chick embryos and one in mouse embryos (Waldo et al.,2001; Kelly et al.,2001; Mjaatvedt et al.,2001). The lineage studies in chick demonstrated that the splanchnic mesoderm adjacent to the pharyngeal endoderm migrates in through the aortic sac to contribute cells to the outflow tract and the right ventricle of the heart, and that the flow of these progenitors along this path gives rise to distinct outflow tract regions at different times during cardiac development (Mjaatvedt et al.,2001; Waldo et al.,2001) (see Fig. 2B). In mouse embryos, a lacZ reporter gene integrated into the genomic fibroblast growth factor 10 (Fgf10) locus marked the outflow tract and the right ventricle of the developing embryonic heart(Kelly et al., 2001). This transgene was expressed at the cardiac-crescent stage of heart development at embryonic day (E) 7.5, and β-galactosidase (β-gal) activity was observed in splanchnic mesoderm that lies medial to the classical first heart field. As development progressed, β-gal+ cells were found in anterior splanchnic mesoderm adjacent to pharyngeal endoderm and were subsequently observed in branchial arch mesoderm proximal to the heart, in the outflow tract and in the right ventricle. Taken together, these studies demonstrated that cells comprising the earliest fusing myocardium do not contain all of the progenitors of the outflow tract and the right ventricle and that these heart structures derive, wholly or in part, from precursor cells of a second heart field, which are added to and supplement the myocardium that originates from the first heart field progenitors(Kelly and Buckingham, 2002). Furthermore, an elegant retrospective clonal analysis (see Box 1) in the mouse embryo suggested that the first and second lineages (see Box 1) of cardiac progenitors originate from a common precursor population that segregates prior to the cardiac-crescent stage (Meilhac et al.,2004). However, a clear delineation of the existence of the second myocardial lineage as a separate subset of cardiovascular precursors, and the ultimate identification of the heart components that it generates, would await the identification of a suitable genetic marker.

In this regard, recent studies have revealed that the expression of the LIM-homeodomain transcription factor Isl1 is a marker of the second myocardial lineage during mammalian cardiogenesis(Cai et al., 2003), a finding that ultimately allowed the isolation of the Isl1+ cardiovascular progenitors themselves (Laugwitz et al.,2005). LIM/HD proteins are a subset of homeodomain (HD)-containing transcription factors that are defined on the basis of a common LIM domain,which consists of a conserved cysteine- and histidine-rich structure of two tandemly repeated zinc fingers. The acronym of LIM is derived from the first identified members of this family, namely LIN-11 from Caenorhabditis elegans (Frey et al.,1990), Isl1 from rat (Karlsson et al., 1990), and MEC-3 from C. elegans(Way and Chalfie, 1988). Subsequent studies of LIM/HD proteins in embryonic motoneurons have led to the identification of a combinatorial code of several LIM/HD proteins, including the Isl1 gene, that controls various aspects of motoneuron identity(Tsuchida et al., 1994; Thor et al., 1999; Thaler et al., 2004). Aside from its expression in embryonic motoneurons, Isl1 is expressed in a variety of cell lineages of pancreatic endocrine origin during embryogenesis, as well as in normal adult islet cells (Karlsson et al., 1990).

Isl1 knockout mice have been generated and studied for defects both in motoneuron specification and pancreatic development(Pfaff et al., 1996; Ahlgren et al., 1997). Homozygous Isl1 mutants exhibit growth retardation around E9.5-10,and die at approximately E10.5-11. Histological analysis of mutant hearts between E9.0 and 9.5 revealed that homozygous Isl1 mutants have a severe cardiac phenotype. Isl1-null hearts fail to undergo looping morphogenesis and appear to have a common atrium and a uni-ventricular chamber, whereas the right ventricle and the outflow tract are absent(Cai et al., 2003). Genetic marker analysis for expression of Tbx5, Hand1 and Fgf10demonstrated that the remaining ventricular tissue had a left ventricular identity and confirmed the lack of the right ventricle and the outflow tract.

These observations suggested that Isl1 is expressed in cells of the second heart field, which contribute to both the venous and arterial poles of the heart. Lineage tracing of Isl1-expressing cells using the Cre-loxP strategy showed that these cells colonize the outflow tract, the right ventricle, part of the atria and a minor portion of the inner curvature of the left ventricle,confirming that this transcription factor marks cardiac progenitors of the second myocardial lineage (see Box 1). Furthermore, Isl1 seems to be required for the survival,proliferation and migration of these cells into the cardiac tube, and its transcription is turned off as the precursor cells differentiate. Since Isl1 expression delineates undifferentiated and differentiated progenitor states,it represents an excellent lineage tracer for cardiac mesodermal cells during embryogenesis.

Is Isl1 expression restricted to the second heart field of cardiac progenitor cells or is it also transiently expressed in the first? Two lines of evidence support the latter notion. First, recent lineage-tracing experiments that used a highly efficient Isl1-Cre knock-in mouse line showed that the majority of cells in the left ventricle express the genetic marker β-gal (Park et al.,2006). However, it is possible that the inefficiency of the excision of the original Isl1-IRES-Cre allowed cells in which Isl1 was expressed for a longer period of time to be preferentially detected (Srinivas et al.,2001; Cai et al.,2003). The second piece of evidence comes from recent studies in mouse and Xenopus. Harvey and co-workers report that, in contrast to Isl1 mRNA, Isl1 protein is expressed at E7.5 throughout the anterior intra-embryonic coelomic walls and proximal head mesenchyme, regions that encompass both first and second heart fields in mouse(Prall et al., 2007). Similarly, during neurula stages in Xenopus, Isl1 is co-expressed with Nkx2-5 throughout the cardiac crescent, which is the first heart field in amphibians (Brade et al.,2007). These data suggest that Isl1 might be a pan-cardiac progenitor marker, but additional work is needed to clarify this issue. In this regard, the identification of specific markers for the first heart field would be extremely valuable. However, analyses of the cardiovascular phenotype of Isl1 knockout mice suggests that Isl1 does not play such an essential role in the first myocardial precursor lineage as compared with the second. So far, the earliest molecular pan-cardiac markers for both myocardial cell lineages are the transcription factors Mesp1/2 (mesoderm posterior 1/2) and Fgf8, which are expressed transiently in cells of the newly formed mesoderm at the primitive-streak stage (see Box 1), the descendents of which colonize the whole myocardium (Saga et al., 2000; Kitajima et al., 2003; Ilagan et al., 2006).

A comparison of the cardiovascular defects that arise in Isl1-deficient mouse embryos with those of mice with mutations in other cardiac transcription factors has begun to uncover a genetic regulatory network that controls the fate of Isl1+ cardiovascular precursors in the second heart field (Table 2; Fig. 3).

Table 2.

Mutant phenotypes of genes involved in second heart field development

GeneSHF expressionMutant phenotype in the cardiac tubeReferences
Is/1 SHF progenitors; weak in distal OFT Embryonic lethal at E10. Single atrial and ventricular (LV identity)compartments; no RV or OFT; atria at the venous pole are abnormal (Cai et al., 2003
Mef2c Early SHF progenitors Embryonic lethal at E10. OFT reduced; RV does not develop (Hand2 is downregulated); INF abnormalities (Lin et al., 1997; Dodou et al., 2004
Hand2 Both FHF and SHF progenitors at cardiac crescent stage Embryonic lethal at E10.5. Defective looping with RV abnormalities (Srivastava et al., 1997
Hand1/2 Yes No ventricles; only atrial chamber forms (Yamagishi et al., 2001; McFadden et al., 2005
Foxh1 SHF progenitors and pharyngeal mesoderm at E9 Embryonic lethal at E10. OFT reduced or absent; RV does not develop; disrupted expression of Mef2c, Hand2, Fgf8 and Tbx5 (von Both et al., 2004
Foxc1/c2 SHF progenitors at E8 and splanchnic and pharyngeal mesoderm Embryonic lethal at E10.5. Severe defects in RV and OFT formation; abnormal coronary vessels; disrupted expression of Fgf10, Fgf8 and Tbx1 (Kume et al., 2001; Seo and Kume, 2006
Smyd1 Precardiac mesoderm at E7.5 Embryonic lethal. Defective looping and single ventricular chamber (Gottlieb et al., 2002
Nkx2-5 Both FHF and SHF progenitors Single atrial and ventricular compartments; loss of ventricular tissue; no Hand1 expression (Lyons et al., 1995; Tanaka et al., 1999; Yamagishi et al., 2001
Tbx1 Pharyngeal mesoderm Perinatal lethal. Defective OFT and ventricular septation (Xu et al., 2004; Hu et al., 2004
Tbx20 Cardiac crescent stage Embryonic lethal at E11.5. Failed looping; chambers do not develop; no Hand1 expression; hypoplastic RV; OFT disrupted (Takeuchi et al., 2005; Singh et al., 2005; Stennard et al., 2005; Cai et al., 2005
Fgf8 Yes OFT defects (Abu Issa et al., 2002; Brown et al., 2004
Fgf10 Yes No early phenotype detected (Kelly et al., 2001
GeneSHF expressionMutant phenotype in the cardiac tubeReferences
Is/1 SHF progenitors; weak in distal OFT Embryonic lethal at E10. Single atrial and ventricular (LV identity)compartments; no RV or OFT; atria at the venous pole are abnormal (Cai et al., 2003
Mef2c Early SHF progenitors Embryonic lethal at E10. OFT reduced; RV does not develop (Hand2 is downregulated); INF abnormalities (Lin et al., 1997; Dodou et al., 2004
Hand2 Both FHF and SHF progenitors at cardiac crescent stage Embryonic lethal at E10.5. Defective looping with RV abnormalities (Srivastava et al., 1997
Hand1/2 Yes No ventricles; only atrial chamber forms (Yamagishi et al., 2001; McFadden et al., 2005
Foxh1 SHF progenitors and pharyngeal mesoderm at E9 Embryonic lethal at E10. OFT reduced or absent; RV does not develop; disrupted expression of Mef2c, Hand2, Fgf8 and Tbx5 (von Both et al., 2004
Foxc1/c2 SHF progenitors at E8 and splanchnic and pharyngeal mesoderm Embryonic lethal at E10.5. Severe defects in RV and OFT formation; abnormal coronary vessels; disrupted expression of Fgf10, Fgf8 and Tbx1 (Kume et al., 2001; Seo and Kume, 2006
Smyd1 Precardiac mesoderm at E7.5 Embryonic lethal. Defective looping and single ventricular chamber (Gottlieb et al., 2002
Nkx2-5 Both FHF and SHF progenitors Single atrial and ventricular compartments; loss of ventricular tissue; no Hand1 expression (Lyons et al., 1995; Tanaka et al., 1999; Yamagishi et al., 2001
Tbx1 Pharyngeal mesoderm Perinatal lethal. Defective OFT and ventricular septation (Xu et al., 2004; Hu et al., 2004
Tbx20 Cardiac crescent stage Embryonic lethal at E11.5. Failed looping; chambers do not develop; no Hand1 expression; hypoplastic RV; OFT disrupted (Takeuchi et al., 2005; Singh et al., 2005; Stennard et al., 2005; Cai et al., 2005
Fgf8 Yes OFT defects (Abu Issa et al., 2002; Brown et al., 2004
Fgf10 Yes No early phenotype detected (Kelly et al., 2001

FHF, first heart field; INF, inflow region; OFT, outflow tract; RV, right ventricle; SHF, second heart field.

The first and second myocardial lineages seem to be governed by both shared and distinct genetic programs. Myocardial regulatory genes that are activated in both lineages include the zinc-finger-containing transcription factors of the GATA family (Gata4, 5 and 6) (Charron and Nemer, 1999) and NK2 transcription factor related, locus 5(Nkx2-5) (Harvey, 1996). Recent studies have identified Mef2c (myocyte enhancer factor 2C;also known as RSRF, related to serum response factor) as a direct transcriptional target of Isl1 and GATA factors in the second heart field(Dodou et al., 2004). The forkhead box H1 transcription factor Foxh1 is also expressed in the second lineage of cardiac precursors, and Foxh1 mutant mouse embryos, like Isl1- and Mef2c-deficient embryos, display defects in the right ventricle and in outflow tract formation(von Both et al., 2004). Isl1 and Foxh1 directly activate transcription of Mef2c by regulating two independent enhancer regions in collaboration with GATA factors and Nkx2-5(Arceci et al., 1993; Dodou et al., 2004). Thus,these genes appear to act at the top of a cascade of cardiac transcription factors in the second myocardial lineage. The SET-domain protein Smyd1 (Bop)is a direct target of Mef2c and regulates Hand2 (heart and neural crest derivatives expressed transcript 2) expression during second heart field development, implying that Smyd1 is an indirect downstream target of Isl1/GATA factors and Foxh1/Nkx2-5 (Gottlieb et al.,2002; Phan et al.,2005). Recently, it has been shown that Forkhead factors are central components of additional regulatory circuits that intersect and reinforce the Isl1-GATA-Mef2c main pathway in the second heart lineage transcriptional network. Foxa2, Foxc1 and Foxc2 can bind and activate in vitro a Tbx1 enhancer that is sufficient to direct expression to the second heart field (Maeda et al.,2006). Tbx1, in turn, activates Fgf8(Hu et al., 2004), whose loss-of-function in the second heart field results in a reduction of Isl1 expression in the pharyngeal mesoderm and outflow tract(Park et al., 2006).

Taken together, the early segregation of the two lineages, the different time-course of myocytic differentiation and the distinct regional contributions to the embryonic heart support the idea that the two progenitor populations may have discrete properties and distinct transcriptional hierarchies for cardiac development, with Nkx2-5 as the critical transcription factor in the first and second lineages, and Isl1, along with Foxh1 and GATA factors, as the key transcriptional regulators in the second heart progenitor field (Biben and Harvey, 1997)(see Fig. 3).

Alternatively, one might question the existence of two unbridgeable progenitor lineages regulated by distinct transcriptional networks whose descendents form distinct heart compartments, and instead propose the complex patterning of one primordial precursor field(Abu-Issa et al., 2004). Such a view carries the implication that cardiac precursors within this solitary field have the capacity to form different heart structures, depending on positional cues. Temporal microenvironmental stimuli and differing concentrations of diffusing morphogens could create cellular diversity that leads to a progressive restriction of developmental potency within a field that was initially homogenous (Moorman et al., 2007).

The purification, renewal and differentiation of native Isl1+cardiac progenitors provides a means to unravel the steps for both cardiac lineage formation and regeneration, and to identify how these steps are linked to certain forms of congenital and adult cardiac diseases. Taking advantage of the fact that Isl1 expression is downregulated in most cardiac precursor cells as they differentiate, recent studies have utilized inducible Isl1-Cre and knock-in Isl1-nlacZ mice to analyze the timing of Isl1+ progenitor migration into the looping heart and the distinct subdomains of the heart that they colonize during embryonic development (Laugwitz et al.,2005; Sun et al.,2007) (Fig. 4). Interestingly, a subset of Isl1+ undifferentiated progenitors remains embedded in the embryonic heart after its formation and a few cells are still detectable after birth in the compartments that arise from Isl1+ second lineage precursors during cardiac development. Tamoxifen-inducible Cre-lox technology has enabled this novel postnatal Isl1+ cell population and its progeny to be selectively marked at a defined time and purified to relative homogeneity(Laugwitz et al., 2005). The ability of these cells to self-renew in vitro on a cardiac mesenchymal feeder layer and to be stimulated to differentiate into fully mature functional cardiomyocytes indicates that these cells represent native cardiac progenitors, remnants of the embryonic Isl1+ precursors(Laugwitz et al., 2005). Postnatal Isl1-expressing cells can be detected in mouse, rat and human myocardium and appear to be distinct from the previously reported cardiac c-Kit+ and Sca1+ cells (Sca1 is also known as Ly6a),which were reported to activate cardiomyocyte-specific genes in vitro and to differentiate into cardiac muscle cells in vivo(Beltrami et al., 2003; Oh et al., 2003).

Fig. 3.

Regulatory networks within the second heart field lineage. The model presents an Isl1-dependent transcriptional network for the development of the second heart field lineage. The LIM-homeodomain transcription factor Isl1(blue) functions as an early regulator of a core network that determines right ventricle and outflow tract development. The solid lines indicate that direct in vivo activation of regulatory sequences has been demonstrated. Dotted lines indicate genetic data or in vitro activation. Fgf8/10, fibroblast growth factor 8 and 10; Foxa2, forkhead box a2; Foxc1/2, forkhead box c1/2; Foxh1,forkhead box h1; Gata, GATA-binding proteins; Hand2, heart and neural crest derivatives expressed transcript 2; Isl1, insulin gene enhancer protein;Mef2c, myocyte enhancer factor 2C; Nkx2-5, NK2 transcription factor related,locus 5; Smyd1, SET and MYND domain containing 1; Tbx1/20, T-box 1 and 20.

Fig. 3.

Regulatory networks within the second heart field lineage. The model presents an Isl1-dependent transcriptional network for the development of the second heart field lineage. The LIM-homeodomain transcription factor Isl1(blue) functions as an early regulator of a core network that determines right ventricle and outflow tract development. The solid lines indicate that direct in vivo activation of regulatory sequences has been demonstrated. Dotted lines indicate genetic data or in vitro activation. Fgf8/10, fibroblast growth factor 8 and 10; Foxa2, forkhead box a2; Foxc1/2, forkhead box c1/2; Foxh1,forkhead box h1; Gata, GATA-binding proteins; Hand2, heart and neural crest derivatives expressed transcript 2; Isl1, insulin gene enhancer protein;Mef2c, myocyte enhancer factor 2C; Nkx2-5, NK2 transcription factor related,locus 5; Smyd1, SET and MYND domain containing 1; Tbx1/20, T-box 1 and 20.

The Isl1 cell-based in vitro system, which can use both mouse embryos and differentiating ES cells as a source of Isl1-expressing cells, should facilitate the rapid and direct identification of the signaling pathways that guide the formation, renewal and diversification of Isl1+progenitors into distinct heart cell lineages.

In vivo cell lineage tracing using the Cre-loxP strategy in the mouse has been an invaluable tool for precisely delineating the specific cell types that derive from Isl1+ precursors, as well as for defining their specific locations in the components of the heart, vascular and conduction systems. To date, several separate Cre mouse lines have proven to be useful for the in vivo lineage tracing of cardiovascular progenitors from the first and second heart field. The Isl1-IRES-Cre line(Srinivas et al., 2001) has the advantage of driving Cre expression only in undifferentiated cardiac progenitors, but the disadvantage of being widely expressed later on in embryonic development. The inducible Isl1-mER-Cre-mER line offers temporal control over Cre activity(Laugwitz et al., 2005). In this line, the presence of the two mutated oestrogen-receptors (mERs) results in Cre being sequestered in the cytoplasm. In the presence of Tamoxifen, the mER-Cre-mER protein undergoes rapid nuclear translocation, which allows Cre-mediated recombination to then occur in those cells that express Isl1. A transgenic Cre mouse line that allows gene inactivation to be restricted to the second heart field lineage has been recently described in which Cre is expressed under a specific Mef2c promoter/enhancer region (Verzi et al., 2005). Nkx2-5-Cre and Mesp1-Cre deleter lines have also been well characterized, and can be utilized to trace lineages from both the first and second heart fields (Moses et al.,2001; Kitajima et al.,2000). Fate-mapping experiments utilizing these different Cre lines have demonstrated that Mef2c, Nkx2-5 and Mesp1 can mark cell populations that contribute to myocardial cells and to subsets of endocardium(Verzi et al., 2005; Stanley et al., 2002; Kitajima et al., 2000). Furthermore, the Cre-mediated lineage tracing of cells that express Flk1 (also known as Kdr), one of the earliest mesodermal progenitor markers for vascular endothelial and hematopoietic lineages has, interestingly, shown the potential of Flk1+ precursors to give rise to both cardiac and smooth muscle during development (Motoike et al.,2003; Coultas et al.,2005).

Fig. 4.

Isl1+ progenitor cells during cardiovascular development.β-gal expression was analyzed in Isl1-nlacZ knock-in mouse embryos at the indicated days of development and in the postnatal heart at day 3 (PN 3d) by X-Gal staining. Representative sections at each developmental stage from E8.5-14.5 are shown (left), as are the corresponding heart structures that express Isl1 (middle) and the lineage markers used to identify the different cell types (right). Cranial (Cr)-caudal (Ca), right (R)-left(L), and dorsal (D)-ventral (V) axes are indicated. A, atrial cavity; Ao,aorta; AS, atrial septum; AVN, atrioventricular node; CG, cardiac ganglia;Hcn4, hyperpolarization-activated cyclic nucleotide-gated K+ 4; LA,left atrium; LV, left ventricle; MF20, antibody recognizing sarcomeric myosin;OFT, outflow tract; PA, pulmonary artery; RA, right atrium; RV, right ventricle; SAN, sinoatrial node; SM, smooth muscle; sm-actin, smooth muscle actin; V, ventricle cavity. Parts of the figure are reproduced with permission(Sun et al., 2007).

Fig. 4.

Isl1+ progenitor cells during cardiovascular development.β-gal expression was analyzed in Isl1-nlacZ knock-in mouse embryos at the indicated days of development and in the postnatal heart at day 3 (PN 3d) by X-Gal staining. Representative sections at each developmental stage from E8.5-14.5 are shown (left), as are the corresponding heart structures that express Isl1 (middle) and the lineage markers used to identify the different cell types (right). Cranial (Cr)-caudal (Ca), right (R)-left(L), and dorsal (D)-ventral (V) axes are indicated. A, atrial cavity; Ao,aorta; AS, atrial septum; AVN, atrioventricular node; CG, cardiac ganglia;Hcn4, hyperpolarization-activated cyclic nucleotide-gated K+ 4; LA,left atrium; LV, left ventricle; MF20, antibody recognizing sarcomeric myosin;OFT, outflow tract; PA, pulmonary artery; RA, right atrium; RV, right ventricle; SAN, sinoatrial node; SM, smooth muscle; sm-actin, smooth muscle actin; V, ventricle cavity. Parts of the figure are reproduced with permission(Sun et al., 2007).

Fig. 5.

Cell lineage tracing of Isl1+ cardiac progenitors and lineage contribution to the adult heart. Lineage tracing utilizing the Isl1-IRES-Cre mouse crossed with the indicator R26R line to mark all cells that once expressed Isl1 during development. The boxes represent magnifications of different regions of the coronary tree (black boxes) and of the aorta/pulmonary artery (red boxes), where β-gal-positive cells have been detected. (A) Contribution of Isl1+ progenitors (blue)to the coronary vasculature, the valves and the pulmonary artery/aorta. The contribution of Isl1+ cells to the endothelial (purple) and smooth muscle (yellow) cell layers is limited to the proximal area of the great vessels and progressively declines from the proximal to the distal parts of the coronary tree. β-gal+ cells were also detected in connective tissue structures of the aortic and pulmonary leaflets, indicating that components of the conotruncal cushions, which have an endocardial origin,are derived from Isl1+ progenitors. (B) Contribution of Isl1+ progenitors (blue) to the atrial/ventricular myocardium(red), the conduction system (white) and cardiac ganglia (green). In the conduction system, most genetically marked cells were detected in the sino-atrial nodal (SAN) region. AS, atrial septum; AVN, atrioventricular node;CAs, coronary arteries; LA, left atrium; LCA, left coronary artery; LV, left ventricle; RA, right atrium; RCA, right coronary artery; RV, right ventricle;VS, ventricular septum.

Fig. 5.

Cell lineage tracing of Isl1+ cardiac progenitors and lineage contribution to the adult heart. Lineage tracing utilizing the Isl1-IRES-Cre mouse crossed with the indicator R26R line to mark all cells that once expressed Isl1 during development. The boxes represent magnifications of different regions of the coronary tree (black boxes) and of the aorta/pulmonary artery (red boxes), where β-gal-positive cells have been detected. (A) Contribution of Isl1+ progenitors (blue)to the coronary vasculature, the valves and the pulmonary artery/aorta. The contribution of Isl1+ cells to the endothelial (purple) and smooth muscle (yellow) cell layers is limited to the proximal area of the great vessels and progressively declines from the proximal to the distal parts of the coronary tree. β-gal+ cells were also detected in connective tissue structures of the aortic and pulmonary leaflets, indicating that components of the conotruncal cushions, which have an endocardial origin,are derived from Isl1+ progenitors. (B) Contribution of Isl1+ progenitors (blue) to the atrial/ventricular myocardium(red), the conduction system (white) and cardiac ganglia (green). In the conduction system, most genetically marked cells were detected in the sino-atrial nodal (SAN) region. AS, atrial septum; AVN, atrioventricular node;CAs, coronary arteries; LA, left atrium; LCA, left coronary artery; LV, left ventricle; RA, right atrium; RCA, right coronary artery; RV, right ventricle;VS, ventricular septum.

To define the contribution of Isl1+ precursors to cardiac cell lineages in the adult heart, Isl1-IRES-Cre/R26R double heterozygous mice have been generated and analyzed (Fig. 5) (Laugwitz et al.,2005; Moretti et al.,2006). In these mice, the Cre-mediated removal of a stop sequence in the cells that express Isl1 results in the expression/translation of lacZ, which is itself under the control of the endogenous Rosa26(R26) promoter. Thus, this approach marks all the cells that once expressed Isl1 during development, and its use has revealed that a high proportion of the right ventricular myocardium and parts of both atria consist of cells that express lacZ and the myocytic protein sarcomeric α-actinin(Laugwitz et al., 2005). The histochemical analysis of β-gal and acetylcholinesterase expression has revealed that Isl1+ progenitors make a remarkable contribution to the cells of the conduction system, primarily to the sino-atrial node, the pacemaker of the heart. Additionally, β-gal staining was observed throughout the proximal aorta, the trunk of the pulmonary artery and the stems of the main left and right coronary arteries (see Fig. 5). The co-expression of lacZ with endothelial and smooth muscle-specific markers, such as CD31 (Pecam1), VE-cadherin (cadherin 5) and smooth muscle myosin heavy chain(SM-MHC; also known as myosin heavy chain 11), revealed that Isl1+precursors can give rise to vascular lineages. Additional studies in the mouse embryo have confirmed that Isl1+ cardiovascular progenitors contribute to over two-thirds of all the cells in the embryonic heart, and to the major cell types in all of the cardiovascular compartments and conduction system, with the exception of the free left ventricular wall(Cai et al., 2003; Moretti et al., 2006; Sun et al., 2007).

Taken together, these results demonstrate that Isl1 marks cardiac precursors that give rise to working cardiac muscle, to the conduction system and to endothelial/smooth muscle cells in multiple heart tissue compartments during cardiogenesis (see Fig. 5). Furthermore, this evidence raises the question of what role Isl1 plays in the specification of each of these mesodermal lineages. Is Isl1 marking an early anterior mesodermal progenitor not yet committed to a cardiac fate? Or is Isl1 defining a multipotent primordial cardiovascular progenitor,which contributes to distinct cell lineages within heart components known to originate from the second heart field? Or do different lineage-restricted precursors exist, all of which independently express Isl1? The early onset of Isl1 expression is consistent with the first two possibilities, although previous retroviral marking experiments have shown, for the proepicardial progenitors of the coronary vasculature, that clones of precursors can give rise only to a single phenotype (for example, only to epicardial cells,endothelial cells, smooth muscle cells or fibroblasts)(Reese et al., 2002). A clear delineation of the developmental potency of Isl1+ cells during cardiogenesis awaits in vivo clonal differentiation analysis.

Understanding how embryonic precursors generate and control the formation of distinct endothelial, pacemaker, atrial, ventricular and vascular smooth muscle lineages, as well as how these cells become positioned to form the specific chambers, aorta, coronary arteries and conduction system of the heart, is of fundamental importance for understanding the developmental logic and molecular cues that underlie both cardiovascular development and disease. As noted above, the formation of cardiac, smooth muscle and endothelial cell lineages in the heart has largely been ascribed to a set of non-overlapping embryonic precursors that have distinct origins. The discovery of several heart lineage-restricted genes has lead to the suggestion that the generation of different cardiac cell types could be driven by a unique combinatorial subset of transcriptional networks that operate within distinct cardiovascular progenitors (for a review, see Srivastava and Olson, 2000). An alternative possibility exists that diverse muscle and non-muscle lineages arise from multipotent, primordial cardiovascular stem cells, which give rise to a hierarchy of downstream cellular intermediates that represent the tissue-restricted precursors of fully differentiated heart cells (Fig. 6). This clonal model of heart lineage diversification is similar to that of hematopoiesis, which is initiated by a few multipotent hematopoietic stem cells that generate large numbers of differentiated progeny by a process of amplification and progressive lineage restriction(Morrison and Weissman, 1994; Weissman, 2000).

Fig. 6.

Cellular hierarchy of cardiac progenitors and their lineage specification. The expression of the lineage markers shown is based on previous studies (Kattman et al.,2006; Moretti et al.,2006; Wu et al.,2006). The divergence of endothelial and haematopoietic lineages from the myocardial lineage precedes the expression of Nkx2-5 in the precardiac mesoderm (tissue-specific markers are shown in blue). Bry,brachyury (T); MLC2a, atrial myosin light chain 2 (also known as Myl7); MLC2v,ventricular myosin light chain 2 (Myl2); cTnT, cardiac troponin T (Tnnt2);Hcn4, hyperpolarization-activated cyclic nucleotide-gated K+ 4;sm-actin, smooth muscle actin; SM-MHC, smooth muscle myosin heavy chain;VE-Cadh, VE-cadherin.

Fig. 6.

Cellular hierarchy of cardiac progenitors and their lineage specification. The expression of the lineage markers shown is based on previous studies (Kattman et al.,2006; Moretti et al.,2006; Wu et al.,2006). The divergence of endothelial and haematopoietic lineages from the myocardial lineage precedes the expression of Nkx2-5 in the precardiac mesoderm (tissue-specific markers are shown in blue). Bry,brachyury (T); MLC2a, atrial myosin light chain 2 (also known as Myl7); MLC2v,ventricular myosin light chain 2 (Myl2); cTnT, cardiac troponin T (Tnnt2);Hcn4, hyperpolarization-activated cyclic nucleotide-gated K+ 4;sm-actin, smooth muscle actin; SM-MHC, smooth muscle myosin heavy chain;VE-Cadh, VE-cadherin.

Recent work from our laboratory has indeed proven the existence of multipotent Isl1+ cardiovascular progenitors (MICPs), which are marked by the transcriptional signature of Isl1, Nkx2-5 and Flk1. These cells can generate the three major cell types of the heart: cardiac, smooth muscle and endothelial cells (Moretti et al.,2006). MICPs have been cloned from both mouse ES cells and mouse embryos, and can make the decision to enter the muscle or endothelial differentiation pathways at the single-cell level, hinting at a hematopoietic-like system for how the diverse cardiovascular lineages can be generated (Moretti et al.,2006). In support of this concept, a hierarchy of distinct Isl1+ cardiovascular progenitors has been uncovered(Laugwitz et al., 2005; Moretti et al., 2006) (see Fig. 6), including a rare subset of Isl1+ cardioblasts that persists until birth and can develop into fully mature cardiac and smooth muscle cells(Laugwitz et al., 2005; Sun et al., 2007). At the same time, independent in vivo and in vitro studies from other laboratories have also identified other multipotent and bi-potent cardiovascular precursors(Kattman et al., 2006; Moretti et al., 2006; Wu et al., 2006), which might also arise from the early heart fields. However, the multi-lineage potential of MICPs still needs to be confirmed by fate-mapping techniques in vivo. Uncovering the molecular pathways that control the formation, renewal and differentiation of cardiovascular progenitors into specific mature cellular progeny will be crucial for unlocking the potential of stem cell therapy for use in a myriad of cardiovascular degenerative diseases, such as heart failure, conduction system disorders and congenital heart disease (see Table 1).

The microenvironment provided by a stem cell niche plays a vital role in stem cell/progenitor maintenance (for a review, see Scadden, 2006). Previous studies have shown that cardiac mesenchymal cells serve in vitro as an effective microenvironment that allows embryonic-, ES cell- and postnatal-derived Isl1+ progenitors to renew in culture(Laugwitz et al., 2005; Moretti et al., 2006). Utilizing a chemical screen, recent work from our laboratory has identified the Wnt/β-catenin pathway as being a major component of the cardiac mesenchymal microenvironment that controls the pre-specification, renewal, and subsequent differentiation of a hierarchy of Isl1+ cardiovascular precursors from mouse ES cells, embryos and postnatal hearts(Qyang et al., 2007). This study reported that the inhibition of canonical Wnt signaling in the mesenchymal cells of the feeder layer promotes the pre-specification of mesodermal precursors into Isl1+ cardiac progenitors in vitro, while reducing the expansion of already specified Isl1+ precursors. Furthermore, this study and several others demonstrate that the in vivo activation of β-catenin within the Isl1+ progenitors in the second heart field leads to their massive accumulation in the pharyngeal mesoderm, the inhibition of their differentiation, and the onset of outflow tract morphogenetic defects(Cohen et al., 2007; Kwon et al., 2007). Similarly,loss-of-function studies have confirmed the requirement for β-catenin for the expansion and survival of Isl1+ cardiac precursors in the embryo in vivo (Lin et al.,2007). Interestingly, chemical agents that inhibit the activity of glycogen synthase kinase 3 (GSK3), the kinase that phosphorylatesβ-catenin and promotes its degradation, can markedly promote the in vitro proliferation of ISL1+ cells from human neonatal hearts,representing a key advance towards the eventual cloning of human ISL1+ cardiac progenitors(Qyang et al., 2007).

Congenital heart disease (CHD), which is one of the most important and prevalent forms of human birth defect, is present in nearly 1 in 100 live births and is responsible for the vast majority of prenatal losses(Hoffman, 1995). Some congenital cardiovascular malformations occur as syndromes in which multiple organs are affected. Over the past decade, a number of single-gene mutations have been correlated with syndromic CHDs, such as the association of mutations in Tbx1 with DiGeorge syndrome and of Tbx5 with Holt-Oram syndrome (for reviews, see Gruber and Epstein, 2004; Ransom and Srivastava, 2007). Although these syndromes have helped researchers to elucidate some of the mechanisms of CHD, most CHD occurs in the absence of any other organ malformation. In humans, 50% of these non-syndromic CHDs manifest as failed atrial/ventricular septation or outflow tract defects,and necessitate open-heart surgery to restore normal circulation. A subset of non-syndromic CHD is familial, and causative genes have been identified, most of which encode transcription factors that are part of a conserved regulatory network that controls cardiogenesis (see Table 1). Mutations in NKX2-5 have been identified in individuals with atrial/ventricular septal defects and with conduction system abnormalities, whereas mutations in GATA4 and TBX20 are found in patients with septation defects, valve abnormalities and cardiomyopathy (Gruber and Epstein, 2006; Schott et al., 1998; Garg et al., 2003; Kirk et al., 2007). However,most non-syndromic CHD cases are sporadic and multifactorial, with no single gene being wholly responsible. Therefore, a major challenge is to dissect the transcriptional and signaling pathways that regulate cardiogenesis and to better understand at a genetic and mechanistic level how a cardiac progenitor cell can develop into each of the specific cell lineages that form the heart.

The identification of two distinct populations of cardiac precursors, one that exclusively forms the left ventricle and the other that mainly forms the outflow tract, the right ventricle and most of the atria, hints at a new approach to understanding CHDs, not as a defect in a specific gene or transcription factor, but rather as a defect in the lineage decisions of a defined subset of cardiac precursors. In this manner, CHDs might be associated with alterations in the formation, expansion and differentiation of embryonic cardiac progenitor cells, which in turn form essential components of the heart, such as the atria, ventricles, coronary arteries and conduction system(Goldmuntz et al., 2001; Pashmforoush et al., 2004; Prall et al., 2007). The discovery of a unique Isl1+ cardiovascular progenitor that contributes to all of these structures in the heart has important implications for understanding the mechanistic origins of CHD. In this regard, a series of studies of the localization of Isl1+ progenitors in patients with diverse forms of CHD (atresia of the aortic and mitral valve, transposition of the great arteries, ventricular septal defects and an interrupted aortic arch)have added important new insights into the potential role of Isl1+progenitors in the closure of the atrial septum, in remodeling events that occur in the latest stages of cardiac morphogenesis or in the immediate postnatal window when the right ventricle starts to pump blood into the pulmonary circulation (Laugwitz et al.,2005). Moreover, the recent discovery of the role ofβ-catenin pathways in regulating the renewal and differentiation of Isl1+ cardiovascular precursors in vivo(Cohen et al., 2007; Kwon et al., 2007; Lin et al., 2007; Qyang et al., 2007) is likely to have a significant impact on our understanding of several forms of CHDs that involve outflow tract defects, especially those implicating mal-rotations or outflow tract dysplasia.

Whereas the pivotal role of Isl1+ cardiovascular progenitor cells in the formation of the major heart lineages is clear, the importance of these cells in endogenous programs of cardiovascular regeneration is still unknown. Cardiomyocytes rarely seem to enter the cell cycle after birth and,consequently, the heart has a very limited regenerative capacity following injury. Given that the number of Isl1+ cells within the heart is vanishingly small after the postnatal window, it is unlikely that they play a role in the regeneration of the adult working myocardium. However, there is a persistence of the postnatal Isl1+ cells within the cardiac autonomic nervous system in regions that intersect with the cardiac conduction system, raising the question as to their role in the maintenance of normal cardiac conduction (Moretti et al.,2006; Sun et al.,2007). The location of Isl1+ cells in the embryonic/fetal structures that are associated with CHDs (outflow tract,inter-atrial septum, etc) suggests that these cells might participate in regenerative pathways in these tissues in response to genetic or environmentally induced injury in the hearts of newborns. Uncovering the stimuli that might lead to the in vivo mobilization of Isl1+cardiovascular progenitor cells will be of importance for understanding their contribution to endogenous regenerative pathways.

The ability to isolate and clonally expand multipotent Isl1+cardiovascular progenitors from mouse ES cells encourages the hope that the same could be achieved in human ES cells. If so, it could form the basis for the generation of models of human CHD and of adult forms of heart disease. The technology to genetically manipulate human ES cells is moving forward rapidly,as are technologies that will allow the generation of ES cells via somatic cell nuclear transfer (SCNT) without requiring the use of eggs(Hochedlinger and Jaenisch,2002). Moreover, recent work from several independent groups has demonstrated the possibility of reprogramming in vitro, differentiated mouse adult fibroblasts into a pluripotent ES cell-like state through the ectopic expression of only a few defined factors: Oct3/4 (also known as Pou5f1), Sox2,c-Myc and Klf4 (Maherali et al.,2007; Okita et al.,2007; Takahashi and Yamanaka,2006; Wernig et al.,2007). This finding represents an important achievement in controlling pluripotency and might allow patient-specific pluripotent ES-like cells to be created directly from somatic cells, which could be a powerful tool for studying human disease in a `culture dish'. The human ES cell-based ISL1+ cardiovascular progenitor model system could prove to be extremely valuable in designing new human-based assay systems for screening the toxicity of new drugs during the early stages of cardiac development, as well as for identifying and validating new therapeutic targets in specific human cardiovascular cell types, such as in coronary vascular smooth muscle,pulmonary arterial smooth muscle, coronary endothelial cells and conduction system cells.

In the search for master cardiovascular genes, it has become increasingly apparent that the generation of the diverse cell lineages of the heart is likely to be due to the employment of combinatorial codes for specific cell types, akin to the theory proposed by Weintraub and colleagues almost 20 years ago (Davis et al., 1987). At the same time, recent studies of Isl1+ cardiovascular progenitors suggest that a key element of lineage diversification in the heart relates to the presence of a renewable, rare subset of multipotent cells, perhaps a master heart progenitor, that ultimately gives rise to over two-thirds of the heart and to the heart's three major cell types: cardiac muscle, smooth muscle and endothelium. The generation of specific cardiovascular cell types might ultimately relate more to a series of decisions that result in a sequential increase in the restriction of multipotent cardiovascular progenitors to specific cellular intermediates and their differentiated derivatives, than to the functioning of dominantly acting genes that enforce lineage specification. As such, understanding cardiogenesis at the level of specific decisions that are made by discrete heart cell lineages requires that we identify the pathways that govern critical steps in the formation, renewal, specification and differentiation of the hierarchy of Isl1+ progenitors and their derivatives. The identification of specific markers that enable FACS purification of these intermediates, and genetic approaches to reveal key steps in their cell fate decisions, will be essential. In short, if heart is like blood, then approaching cardiogenesis as `cardiopoiesis' might represent the road ahead.

The authors especially thank Sylvia Evans for her continuous support. We also thank members of the laboratories of Karl-Ludwig Laugwitz and Kenneth R. Chien for their helpful discussions and comments. We apologize to colleagues whose work is not mentioned here owing to space limitations. The authors are supported by the Massachusetts General Hospital and the Cardiovascular Disease Program of the Harvard Stem Cell Institute, a Marie Curie Excellence Team Grant from the European Research Council (MEXT-23208), the German Research Foundation (La 1238 3-1/4-1), the National Heart, Lung and Blood Institute,and the Jean Le Ducq Foundation.

Abu-Issa, R., Smyth, G., Smoak, I., Yamamura, K. and Meyers, E. N. (
2002
). Fgf8 is required for pharyngeal arch and cardiovascular development in the mouse.
Development
129
,
4613
-4625.
Abu-Issa, R., Waldo, K. and Kirby, M. L.(
2004
). Heart fields: one, two or more?
Dev. Biol.
272
,
281
-285.
Ahlgren, U., Pfaff, S. L., Jessell, T. M., Edlund, T. and Edlund, H. (
1997
). Independent requirement for ISL1 in formation of pancreatic mesenchyme and islet cells.
Nature
385
,
257
-260.
Arceci, R. J., King, A. A., Simon, M. C., Orkin, S. H. and Wilson, D. B. (
1993
). Mouse GATA-4: a retinoic acid-inducible GATA-binding transcription factor expressed in endodermally derived tissues and heart.
Mol. Cell. Biol.
13
,
2235
-2246.
Beltrami, A. P., Barlucchi, L., Torella, D., Baker, M., Limana,F., Chimenti, S., Kasahara, H., Rota, M., Musso, E., Urbanek, K. et al.(
2003
). Adult cardiac stem cells are multipotent and support myocardial regeneration.
Cell
114
,
763
-776.
Benson, D. W., Silberbach, G. M., Kavanaugh-McHugh, A.,Cottrill, C., Zhang, Y., Riggs, S., Smalls, O., Johnson, M. C., Watson, M. S.,Seidman, J. G. et al. (
1999
). Mutations in the cardiac transcription factor Nkx2.5 affect diverse cardiac developmental pathways.
J. Clin. Invest.
104
,
1567
-1573.
Biben, C. and Harvey, R. P. (
1997
). Homeodomain factor Nkx2.5 controls left/right asymmetric expression of bHLH gene eHand during murine heart development.
Genes Dev.
11
,
1357
-1369.
Black, B. L. (
2007
). Transcriptional pathways in second heart field development.
Cell Dev. Biol.
18
,
67
-76.
Brade, T., Gessert, S., Kühl, M. and Pandur, P.(
2007
). The amphibian second heart field: Xenopus islet-1 is required for cardiovascular development.
Dev. Biol.
doi: .
Brand, T. (
2003
). Heart development: molecular insights into cardiac specification and early morphogenesis.
Dev. Biol.
258
,
1
-19.
Brown, C. B., Wenning, J. M., Lu, M. M., Epstein, D. J., Meyers,E. N. and Epstein, J. A. (
2004
). Cre-mediated excision of Fgf8 in the Tbx1 expression domain reveals a critical role for Fgf8 in cardiovascular development in the mouse.
Dev. Biol.
267
,
190
-202.
Buckingham, M., Meilhac, S. and Zaffran, S.(
2005
). Building the mammalian heart from two sources of myocardial cells.
Nat. Rev. Genet.
6
,
826
-835.
Cai, C. L., Liang, X., Shi, Y., Chu, P. H., Pfaff, S. L., Chen,J. and Evans, S. (
2003
). Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart.
Dev. Cell
5
,
877
-889.
Cai, C. L., Zhou, W., Yang, L., Bu, L., Qyang, Y., Zhang, X.,Li, X., Rosenfeld, M. G., Chen, J. and Evans, S. (
2005
). T-box genes coordinate regional rates of proliferation and regional specification during cardiogenesis.
Development
132
,
2475
-2487.
Charron, F. and Nemer, M. (
1999
). GATA transcription factors and cardiac development.
Semin. Cell Dev. Biol
.
10
,
85
-91.
Chien, K. R. and Karsenty, G. (
2005
). Longevity and lineages: toward the integrative biology of degenerative disease.
Cell
120
,
533
-544.
Ching, Y. H., Ghosh, T. K., Cross, S. J., Packham, E. A.,Honeymon, L., Loughana, S., Robinson, T. E., Dearlove, A. M., Ribos, G,Bonser, A. J. et al. (
2005
). Mutation in myosin heavy chain 6 causes atrial septal defect.
Nat. Genet.
37
,
423
-428.
Choi, K., Kennedy, M., Kazarov, A., Papadimitriou, J. C. and Keller, G. (
1998
). A common precursor for hematopoietic and endothelial cells.
Development
125
,
725
-732.
Cohen, E. D., Wang, Z., Lepore, J. J., Lu, M. M., Taketo, M. M.,Epstein, D. J. and Morrisey, E. E. (
2007
). Wnt/β-catenin signalling promotes expansion of Isl1-positive cardiac progenitor cells through regulation of FGF signalling.
J. Clin. Invest.
117
,
1794
-1804.
Coultas, L., Chawengsaksophak, K. and Rossant, J.(
2005
). Endothelial cells and VEGF in vascular development.
Nature
438
,
937
-945.
de la Cruz, M. V. and Sanchez-Gomez, C. (
2000
). Straight heart tube. Primitive cardiac cavities vs. primitive cardiac segments. In
Living Morphogenesis of the Heart
(ed. M. V. de la Cruz and R. R. Markwald), pp.
85
-99. Boston:Birkhauser.
Davis, R. L., Weintraub, H. and Lassar, A. B.(
1987
). Expression of a single transfected cDNA converts fibroblasts to myoblasts.
Cell
51
,
987
-1000.
Dettman, R. W., Denetclaw, W., Ordahl, C. P. and Bristow, J.(
1998
). Common epicardial origin of coronary vascular smooth muscle, perivascular fibroblasts, and intermyocardial fibroblasts in avian heart.
Dev. Biol.
193
,
169
-181.
Dodou, E., Verzi, M. P., Anderson, J. P., Xu, S.-M. and Black,B. L. (
2004
). Mef2c is a direct transcriptional target of isl1 and GATA factors in the anterior heart field during mouse embryonic development.
Development
131
,
3931
-3942.
Elliott, D. A., Kirk, E. P., Yeoh, T., Chandor, S., McKenzie,F., Taylor, P., Grossfeld, P., Fatkin, D., Jones, O., Hayes, P. et al.(
2003
). Cardiac homeobox gene NKX2.5 mutations and congenital heart disease: associations with atrial septal defect and hypoplastic left heart syndrome.
J. Am. Coll. Cardiol.
41
,
2072
-2076.
Epstein, J. A. and Buck, C. A. (
2000
). Transcriptional regulation of cardiac development: implications for congenital heart disease and DiGeorge syndrome.
Pediatr. Res.
48
,
717
-724.
Fehling, H. J., Lacaud, G., Kubo, A., Kennedy, M., Robertson,S., Keller, G. and Kouskoff, V. (
2003
). Tracking mesoderm induction and its specification to the hemangioblast during embryonic stem cell differentiation.
Development
130
,
4217
-4227.
Frey, G., Kim, S. K. and Horvitz, H. R. (
1990
). Novel cysteine-rich motif and homeodomain in the product of Caenorhabditis elegans cell lineage gene Lin-11.
Nature
344
,
876
-879.
Garg, V., Kathiriya, I. S., Barnes, R., Schluterman, M. K.,King, I. N., Butler, C. A., Rothrock, C. R., Eapen, R. S., Hirayama-Yamada,K., Joo, K. et al. (
2003
). GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5.
Nature
424
,
443
-447.
Garg, V., Muth, A. N., Ransom, J. F., Schluterman, M. K.,Barnes, R., King, I. N., Grossfeld, P. D. and Srivastava, D.(
2005
). Mutations in NOTCH1 cause aortic valve disease.
Nature
437
,
270
-274.
Goldmuntz, E., Geiger, E. and Benson, D. W.(
2001
). Nkx2.5 mutations in patients with tetralogy of Fallot.
Circulation
104
,
2565
-2568.
Gottlieb, P. D., Pierce, S. A., Sims, R. J., Yamagishi, H.,Weihe, E. K., Harriss, J. V., Maika, S. D., Kuziel, W. A., King, H. L., Olson,E. N. et al. (
2002
). Bop encodes a muscle-restricted protein containing MYND and SET domains and is essential for cardiac differentiation and morphogenesis.
Nat. Genet.
31
,
25
-32.
Gruber, P. J. and Epstein, J. A. (
2004
). Development gone awry.
Circ. Res.
94
,
273
-283.
Harvey, R. P. (
1996
). NK-2 homeobox genes and heart development.
Dev. Biol.
178
,
203
-216.
Harvey, R. P. (
2002
). Patterning the vertebrate heart.
Nat. Rev. Genet.
3
,
544
-556.
Hochedlinger, K. and Jaenisch, R. (
2002
). Monoclonal mice generated by nuclear transfer from mature B and T donor cells.
Nature
415
,
1035
-1038.
Hoffman, J. I. E. (
1995
). Incidence of congenital heart disease: I. Postnatal incidence.
Pediatr. Cardiol.
16
,
103
-113.
Hu, T., Yamagishi, H., Maeda, J., McAnally, J., Yamagishi, C. and Srivastava, D. (
2004
). Tbx1 regulates fibroblast growth factors in the anterior heart field through a reinforcing autoregulatory loop involving forkhead transcription factors.
Development
131
,
5491
-5502.
Ilagan, R., Abu-Issa, R., Brown, D., Yang, Y. P., Jiao, K.,Schwartz, R. J., Klingensmith, J. and Meyers, E. N. (
2006
). Fgf8 is required for anterior heart field development.
Development
133
,
2435
-2445.
Karlsson, O., Thor, S., Norberg, T., Ohlsson, H. and Edlund,T. (
1990
). Insulin gene enhancer binding protein isl-1 is a member of a novel class of proteins containing both homeo- and Cys-His domain.
Nature
344
,
879
-882.
Kattman, S. J., Huber, T. L. and Keller, G. M.(
2006
). Multipotent flk-1+ cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages.
Dev. Cell
11
,
723
-732.
Kelly, R. G. and Buckingham, M. E. (
2002
). The anterior-heart forming field:voyage to the arterial pole of the heart.
Trends Genet.
18
,
210
-216.
Kelly, R. G., Brown, N. A. and Buckingham, M. E.(
2001
). The arterial pole of the mouse heart froms from Fgf10-expressing cells in pharyngeal mesoderm.
Dev. Cell
1
,
435
-440.
Kirby, M. L., Gale, T. F. and Stewart, D. E.(
1983
). Neural crest cells contribute to normal aorticopulmonary septation.
Science
220
,
1059
-1061.
Kirk, E. P., Sunde, M., Costa, M. W., Rankin, S. A., Wolstein,O., Castro, M. L., Butler, T. L., Hyun, C., Guo, G., Otway, R. et al.(
2007
). Mutations in cardiac T-box factor gene TBX20 are associated with diverse cardiac pathologies, including defects of septation and valvulogenesis and cardiomyopathy.
Am. J. Hum. Genet.
81
,
280
-291.
Kitajima, S., Takagi, A., Inoue, T. and Saga, Y.(
2000
). MesP1 and MesP2 are essential for the development of cardiac mesoderm.
Development
127
,
3215
-3226.
Kouskoff, V., Lacaud, G., Schwantz, S., Fehling, H. J. and Keller, G. (
2005
). Sequential development of hematopoietic and cardiac mesoderm during embryonic stem cell differentiation.
Proc. Natl. Acad. Sci. USA
102
,
13170
-13175.
Krantz, I. D., Smith, R., Colliton, R. P., Tinkel, H., Zackei,E. H., Piccoli, D. A., Goldmuntz, E. and Spinner, N. B.(
1999
). Jagged1 mutations in patients ascertained with isolated congenital heart defects.
Am. J. Med. Genet.
84
,
56
-60.
Kume, T., Jiang, H., Topczewska, J. M. and Hogan, B. L.(
2001
). The murine winged helix transcription factors, Foxc1 and Foxc2, are both required for cardiovascular development and somitogenesis.
Genes Dev.
15
,
2470
-2482.
Kwon, C., Arnold, J., Hsiao, E. C., Taketo, M. M., Conklin, B. R. and Srivastava, D. (
2007
). Canonical Wnt signaling is a positive regulator of mammalian cardiac progenitors.
Proc. Natl. Acad. Sci. USA
104
,
10894
-10899.
Laugwitz, K.-L., Moretti, A., Lam, J., Gruber, P., Chen, Y.,Woodard, S., Lin, L.-Z., Cai, C.-L., Lu, M. M., Reth, M. et al.(
2005
). Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages.
Nature
433
,
647
-653.
Lin, L., Cui, K., Zhou, W., Dufort, D., Zhang, X., Cai, C.-L.,Bu, L., Yang, L., Martin, J., Kemler, R. et al. (
2007
).β-catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple aspects of cardiogenesis.
Proc. Natl. Acad. Sci. USA
104
,
9313
-9318.
Lin, Q., Schwarz, J., Bucana, C. and Olson, E. N.(
1997
). Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C.
Science
276
,
1404
-1407.
Lyons, I., Parsons, L. M., Hartley, L., Li, R., Andrews, L. E.,Robb, L. and Harvey, R. P. (
1995
). Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeobox gene Nkx2.5.
Genes Dev.
9
,
1654
-1666.
Maeda, J., Yamagishi, H., McAnally, J., Yamagishi, C. and Srivastava, D. (
2006
). Tbx1 is regulated by forkhead proteins in the secondary heart field.
Dev. Dyn.
235
,
701
-710.
Maherali, N., Sridharah, R., Xie, W., Utikal, J., Eminli, S.,Arnold, K., Stadtfeld, M., Yachechko, R., Tchieu, J., Jaenisch, R. et al.(
2007
). Directly reprogrammed fibroblasts show global epigenetic remodeling and widespread tissue contribution.
Cell Stem Cell
1
,
55
-70.
Manner, J., Perez-Pomares, J. M., Macias, D. and Munoz-Chapuli,R. (
2001
). The origin, formation and developmental significance of the epicardium: a review.
Cells Tissues Organs
169
,
2205
-2218.
McElhinney, D. B., Geiger, E., Blinder, J., Benson, D. W. and Goldmuntz, E. (
2003
). NKX2.5 mutations in patients with congenital heart disease.
J. Am. Coll. Cardiol.
42
,
1650
-1655.
McFadden, D. G., Barbosa, A. C., Richardson, J. A., Schneider,M. D., Srivastava, D. and Olson, E. N. (
2005
). The Hand1 and Hand2 transcription factors regulate expansion of the embryonic cardiac ventricles in a gene dosage-dependent manner.
Development
132
,
189
-201.
Meilhac, S. M., Esner, M., Kelly, R. G., Nicolas, J.-F. and Buckingham, M. E. (
2004
). The clonal origin of myocardial cells in different regions of the embryonic mouse heart.
Dev. Cell
6
,
685
-696.
Mikawa, T. (
1999
). Cardiac lineages. In
Heart Development
(ed. R. Harvey and N. Rosenthal),pp.
19
-33. San Diego: Academic Press.
Mikawa, T. and Gourdie, R. G. (
1996
). Pericardial mesoderm generates a population of coronary smooth muscle cells migrating into the heart along with in-growth from the epicardial organ.
Dev. Biol.
173
,
221
-232.
Mjaatvedt, C. H., Nakaoka, T., Moreno-Rodriguez, R., Norris, R. A., Kern, M. J., Eisenberg, C. A., Turner, D. and Markwald, R. R.(
2001
). The outflow tract of the heart is recruited from a novel heart-forming field.
Dev. Biol.
238
,
97
-109.
Moore, A. W., McInnes, L., Kreidberg, J., Hastie, N. D. and Schedl, A. (
1999
). YAC complementation shows a requirement for Wt1 in the development of epicardium, adrenal gland, and throughout nephrogenesis.
Development
126
,
1845
-1857.
Moorman, A. F., Christoffels, V. M., Anderson, R. H. and van den Hoff, M. J. (
2007
). The heart-forming fields: one or multiple?
Philos. Trans. R. Soc. Lond. B Biol. Sci.
362
,
1257
-1265.
Moretti, A., Caron, L., Nakano, A., Lam, J. T., Bernshausen, A.,Chen, Y., Qyang, Y., Bu, L., Sasaki, M., Martin-Puig, S. et al.(
2006
). Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification.
Cell
127
,
1151
-1165.
Morrison, S. J. and Weissman, I. L. (
1994
). The long-term repopulating subset of hematopoietic stem cells is determinitstic and isolatable by phenotype.
Immunity
1
,
661
-673.
Moses, K. A., DeMayo, F., Braun, R. M., Reecy, J. L. and Schwartz, R. J. (
2001
). Embryonic expression of an Nkx2.5/Cre gene using ROSA26 reporter mice.
Genesis
31
,
176
-180.
Motoike, T., Markham, D. W., Rossant, J. and Sato, T. N.(
2003
). Evidence for novel fate of flk1+ progenitors:contribution to muscle lineage.
Genesis
35
,
153
-159.
Oh, H., Bradfute, S. B., Mishina, Y., Pocius, J., Michael, L. H., Behringe, R. R., Schwartz, R. J., Entman, M. L. and Schneider, M. D.(
2003
). Cardiac progenitor cells from adult myocardium: homing,differentiation, and fusion after infarction.
Proc. Natl. Acad. Sci. USA
100
,
12313
-12318.
Okita, K., Ichisaka, T. and Yamanaka, S.(
2007
). Generation of germline-competent induced pluripotent stem cells.
Nature
448
,
313
-317.
Olson, E. N. (
2006
). Gene regulatory networks in the evolution and development of the heart.
Science
313
,
1922
-1927.
Park, E. J., Ogden, L. A., Talbot, A., Evans, S., Cai, C. L.,Black, B. L., Frank, D. U. and Moon, A. M. (
2006
). Required,tissue-specific roles for Fgf8 in outflow tract formation and remodelling.
Development
133
,
2419
-2433.
Pashmforoush, M., Lu, J. T., Chen, H., Armand, T. S., Kondo, R.,Pradervand, S., Evans, S. M., Clark, B., Feramisco, J. R., Giles, W. et al. (
2004
). Nkx2.5 pathways and congenital heart disease:loss of ventricular myocyte lineage specification leads to progressive cardiomyopathy and complete heart block.
Cell
117
,
373
-386.
Pfaff, S. L., Mendelsohn, M., Stewart, C. L., Edlund, T. and Jessell, T. M. (
1996
). Requirement for LIM homeobox gene isl1 in motor neuron generation reveals a motor neuron-dependent step in interneuron differentiation.
Cell
84
,
309
-320.
Phan, D., Rasmussen, T. L., Nakagawa, O., McAnally, J.,Gottlieb, P. D., Tucker, P. W., Richardson, J. A., Bassel-Duby, R. and Olson,E. N. (
2005
). BOP, a regulator of right ventricular heart development, is a direct transcriptional target of MEF2C in the developing heart.
Development
132
,
2669
-2678.
Poelmann, R. E., Lie-Venema, H. and Gittenberger-de Groot, A. C. (
2002
). The role of the epicardium and neural crest.
Tex. Heart Inst. J.
29
,
255
-261.
Prall, O. W. J., Menon, M. K., Solloway, M. J., Watanabe, Y.,Zaffran, S., Bajolle, F., Biben, C., McBride, J. J., Robertson, B. R.,Chauleet, H. et al. (
2007
). An Nkx2-5/BMP2/Smad1 negative feedback loop controls heart progenitor specification and proliferation.
Cell
128
,
947
-959.
Qyang, Y., Martin-Puig, S., Ghiravuri, M., Chen, S., Xu, H., Bu,L., Jiang, X., Granger, A., Moretti, A., Caron, L. et al.(
2007
). The in vivo renewal and differentiation of Isl1+ cardiovascular progenitors is controlled by a Wnt/β-catenin pathway.
Cell Stem Cell
1
,
165
-179.
Ransom, J. and Srivastava, D. (
2007
). The genetics of cardiac birth defects.
Cell Dev. Biol.
18
,
132
-139.
Reese, D. E., Mikawa, T. and Bader, D. M.(
2002
). Development of the coronary vessel system.
Circ. Res.
91
,
751
-768.
Robinson, S. W., Morris, C. D., Goldmuntz, E., Reller, M. D.,Jones, M. A., Steiner, R. D. and Maslen, C. L. (
2003
). Missense mutations in CRELD1 are associated with cardiac atrioventricular septal defects.
Am. J. Hum. Genet.
72
,
1047
-1052.
Saga, Y., Kitajima, S. and Miyagawa-Tomita, S.(
2000
). Mesp1 expression is the earliest sign of cardiovascular development.
Trends Cardiovasc. Med.
10
,
345
-352.
Scadden, D. T. (
2006
). The stem-cell niche as an entity of action.
Nature
441
,
1075
-1079.
Schott, J. J., Benson, D. W., Basson, C. T., Pease, W.,Silberbach, G. M., Moak, J. P., Maron, B. J., Seidman, C. E. and Seidman, J. G. (
1998
). Congenital heart disease caused by mutations in the transcription factor NKX2-5.
Science
281
,
108
-111.
Seo, S. and Kume, T. (
2006
). Forkhead transcription factors, Foxc1 and Foxc2, are required for the morphogenesis of the cardiac outflow tract.
Dev. Biol.
296
,
421
-436.
Singh, M. K., Christoffels, V. M., Dias, J. M., Trowe, M.-O.,Petry, M., Schuster-Gossler, K., Bürger, A., Ericson, J. and Kispert,A. (
2005
). Tbx20 is essential for cardiac chamber differentiation and repression of Tbx2.
Development
132
,
4578
-4588.
Srinivas, S., Watanabe, T., Lin, C. S., William, C. M., Tanabe,Y., Jessell, T. M. and Costantini, F. (
2001
). Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26locus.
BMC Dev. Biol.
1
,
4
.
Srivastava, D. (
2006
). Making or breaking the heart: from lineage determination to morphogenesis.
Cell
126
,
1037
-1048.
Srivastava, D. and Olson, E. N. (
2000
). A genetic blueprint for cardiac development.
Nature
407
,
221
-226.
Srivastava, D., Thomas, T., Lin, Q., Kirby, M. L., Brown, D. and Olson, E. N. (
1997
). Regulation of cardiac mesodermal and neural crest development by the bHLH transcription factor, dHAND.
Nat. Genet.
16
,
154
-160.
Stanley, E. G., Biben, C., Elefanty, A., Barnett, L., Koentgen,F., Robb, L. and Harvey, R. P. (
2002
). Efficient Cre-mediated deletion in cardiac progenitor cells conferred by a 3ÙTR-ires-Cre allele of the homeobox gene Nkx2-5.
Int. J. Dev. Biol.
46
,
431
-439.
Stennard, F. A., Costa, M. W., Lai, D., Biben, C., Furtado, M. B., Solloway, M. J., McCully, D. J., Lumena, C., Preis, J. I., Dunwoodie, S. L. et al. (
2005
). Murine T-box transcription factor Tbx20 as a repressor during heart development, and is essential for adult heart integrity, function and adaptation.
Development
132
,
4567
-4578.
Sun, Y., Liang, X., Najafi, N., Cass, M., Lin, L., Cai, C.-L.,Chen, J. and Evans, S. M. (
2007
). Islet 1 is expressed in distinct cardiovascular lineages, including pacemaker and coronaryvascular cells.
Dev. Biol.
304
,
286
-296.
Takahashi, K. and Yamanaka, S. (
2006
). Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors.
Cell
126
,
663
-676.
Takeuchi, J. K., Mileikovskaia, M., Koshiba-Takeuchi, K., Heidt,A. B., Mori, A. D., Arruda, E. P., Gertsenstein, M., Georges, R., Davidson,L., Mo, R. et al. (
2005
). Tbx20 dose-dependently regulates transcription factor networks required for mouse heart and motoneuron development.
Development
132
,
4534
-4544.
Tanaka, M., Chen, Z., Bartunkova, M., Yamazaki, N. and Izumo,S. (
1999
). The cardiac homeobox gene Csx/Nkx2.5 lies genetically upstream of multiple genes essential for heart development.
Development
126
,
1269
-1280.
Thaler, J. P., Koo, S. J., Konia, A., Lettieri, K., Andrews, S.,Cox, C., Jessell, T. M. and Pfaff, S. L. (
2004
). A postmitotic role for Isl-class LIM homeodomain proteins in the assignment of visceral spinal neuron identity.
Neuron
41
,
337
-350.
Thor, S., Andersson, S. G. E., Tomlinson, A. and Thomas, J. B. (
1999
). A LIM-homeodomain combintaotiral code for motor neuron pathway selection.
Nature
397
,
76
-80.
Tsuchida, T., Ensini, M., Mortin, S. B., Baldassare, M., Edlund,T., Jessell, T. M. and Pfaff, S. L. (
1994
). Topographic organization and embryonic motor neurons defined by expression of LIM homeobox genes.
Cell
79
,
957
-970.
Verzi, M. P., McCulley, D. J., De Val, S., Dodou, E. and Black,B. L. (
2005
). The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field.
Dev. Biol.
287
,
134
-145.
von Both, I., Silvestri, C., Erdemir, T., Lickert, H., Walls, J. R., Henkelma, R. M., Rossant, J., Harvey, R. P., Attisano, L. and Wrana, J. L. (
2004
). Foxh1 is essential for development of the anterior heart field.
Dev. Cell
7
,
331
-345.
Waldo, K. L., Kumiski, D. H., Wallis, K. T., Stadt, H. A.,Hutson, M. R., Platt, D. H. and Kirby, M. L. (
2001
). Conotruncal myocardium arises from a secondary heart field.
Development
128
,
3179
-3188.
Way, J. and Chalfie, C. M. (
1988
). Mec-3, a homeobox-containing gene that specifies differentiation of the touch receptor neurons in C. elegans.
Cell
54
,
5
-16.
Weissman, I. L. (
2000
). Stem cells: units of development, units of regeneration, and units of evolution.
Cell
100
,
157
-168.
Wernig, M., Meissner, A., Foreman, R., Brambrink, T., Ku, M.,Hochedlinger, K., Bernstein, B. E. and Jaenisch, R. (
2007
). In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state.
Nature
448
,
318
-324.
Wu, S. M., Fujiwara, Y., Cibulsky, S. M., Clapham, D. E., Lien,C. L., Schultheiss, T. M. and Orkin, S. H. (
2006
). Developmental origin of a bipotential myocardial and smooth muscle cell precursor in the mammalian heart.
Cell
127
,
1137
-1150.
Xu, H., Morishima, M., Wylie, J. N., Schwartz, R. J., Bruneau,B. G., Lindsay, E. A. and Baldini, A. (
2004
). Tbx1 has a dual role in the morphogenesis of the cardiac outflow tract.
Development
131
,
3217
-3227.
Yamagishi, H., Yamagishi, C., Nakagawa, O., Harvey, R. P.,Olson, E. N. and Srivastava, D. (
2001
). The combinatorial activities of Nkx2.5 and dHAND are essential for cardiac ventricle formation.
Dev. Biol.
239
,
190
-203.