Molecular Pathways and Animal Models of Coronary Artery Anomalies



Fig. 45.1
Normal and abnormal coronary anatomy. Cartoons illustrate normal (13) coronary anatomy and relevant cases of coronary congenital anomalies (49). Asterisks indicate the specific location of myocardial bridges (7b), coronary fistulae (8b), and coronary aneurysms (9b)





45.3 Developmental Substrates of Congenital Coronary Anomalies


In principle, any gene required for proper blood vessel development could be a genetic substrate for CAA. However, animal models for these genes (mostly transgenic/mutant mice) often die before coronary blood vessels form, as they often display deficient yolk sac vascularization [10]. Therefore, in this chapter, we will focus on mice, a species displaying defects in coronary blood vessels similar to those observed in humans.


45.3.1 Defective Outflow Tract Septation and Rotation


The majority of the known forms of congenital heart disease affect the outflow tract (arterial pole of the heart), and some of them include coronary anomalies as part of their phenotype. Several cellular mechanisms coordinate outflow tract transformation into the mature aorta and pulmonary arteries and valves. The first one is the septation of the embryonic outflow tract of the heart (common truncus arteriosus), predominantly driven by cardiac neural crest cells (NCC) [11, 12]. Persistent truncus arteriosus (PTA) is a severe congenital heart disease that results from restricted or absent outflow tract septation [13]. This anomaly also affects the structure of arterial semilunar valves and the relative position of the coronary ostia. PTA is characteristic of null mice for genes relevant to NCC development like Pax3 (paired box 3) [14] or Pbx1 (pre-B-cell leukemia homeobox 1) [15].

Abnormal NCC migration defects also arise as a result of mutations of genes in the NOTCH signaling pathway, which has been shown to be required for the proper development of outflow tract, endocardial cushions, and arterial valve primordia [16]. This study emphasizes the importance of tissue interaction during outflow tract development, proving that NCC and secondary heart field progenitor interaction is necessary for endocardial-derived valvuloseptal mesenchyme formation via NOTCH signaling. Accordingly, mutant mice for the NOTCH target gene Hes1 (hes family bHLH transcription factor 1) display outflow defects [17].

The best known mouse model for outflow tract anomalies is that of the Tbx1 (T-box 1) knockout, a candidate gene for DiGeorge or 22q11.2 deletion syndrome. In Tbx1 null mice with PTA both right and left coronary ostia preferentially locate at the right or ventral aortic sinus of Valsalva and present a proximal left coronary artery stem that courses abnormally ventrally rather than laterally [18]. These authors suggest it is the loss of the subpulmonary myocardium, hypothesized to be refractory to blood vessels, that may play a role in this coronary defect.

Proper rotation of the outflow tract is as important as correct outflow tract septation, and both phenomena are developmentally linked by the neural crest [19] and the genetic specification of outflow tract myocardial subpopulations [19, 20]. For example, the loss of the extracellular matrix component PERLECAN (Hspg2 encoding heparan sulfate proteoglycan 2) first affects NCC migration and then outflow tract rotation, resulting in transposition of the great arteries [21] and in multiple defects in left and right coronary stem course [22]. These latter anomalies suggest that it is the location of the aortic root that determines the path followed by coronary blood vessels at the base of the heart and this can be quite variable [1, 23]. As a result of outflow tract malrotation, coronary ostia frequently appear at abnormal anatomical positions [24].

In summary, we suggest that the disruption of outflow tract septation and rotation, especially when combined with mispatterning of the unique antivascular properties of the subpulmonary myocardium [18] and the provascular properties of aortic myocardium (via Vegf-C encoding vascular endothelial growth factor C, [25]), is sufficient to explain an important part of group 1 CAA.


45.3.2 Anomalous Epicardial Development


It has been proposed that the epicardium and embryonic coronary blood vessels are a developmental continuum [26]. The intricate relationship existing between these two structures can explain some CAA (groups 1–3) through the alteration of four basic developmental epicardial mechanisms:


45.3.2.1 Epicardial Formation and EMT


The primitive epicardial epithelium forms after epicardial progenitor cells are transferred from the proepicardium to the myocardial surface [27, 28]. Abnormal primitive epicardium formation is known to affect ventricular myocardium growth and coronary vascular development [29]. Integrins are currently the only type of adhesion molecule that have been shown to be involved in this process [30]. The α4-integrin null mouse displays severe ventricular myocardial thinning and coronary phenotype [31]. Soon after the primitive epicardium forms, epicardial epithelial-to-mesenchymal transition (EMT) is initiated. Epicardial EMT is a finely regulated process which supplies the heart with highly invasive mesenchymal epicardially derived cells (EPDCs) [26]. Multiple molecules participate in triggering EMT. To date, TGFβ (transforming growth factor beta) 1–2 appears to be the strongest epicardial EMT inducer [3234], as shown by the epicardial deletion of the TGFβ type I receptor ALK5 (TGFBR1), which disrupts epicardial EMT and coronary blood vessel development [35].

Epicardial transcription factors Wt1 and Tbx18, together with canonical WNT effectors such as β-CATENIN and retinoic acid, also participate as effectors of the EMT process by modulating cell adhesion and motility [36, 37]. Potentially, genetic defects in any of the genes encoding for all these molecules (or participating in their biosynthesis) could give rise to CCAs, but due to the importance of primitive epicardial formation and EMT, individuals carrying such a defect are likely to die during gestation. Still, experimental studies using the chick embryo as experimental animal model reveal that a simple delay in epicardial formation and EMT activation is sufficient to generate abnormal coronary patterning and arterioventricular connections or fistulae (CAA group 3) [38, 39], anomalies which are associated with survival to adulthood.


45.3.2.2 Epicardial and Myocardial Instructive Signaling


The epicardium does not only contribute cells to the developing coronary vascular system, but it also acts as a signaling center by secreting various morphogens. These epicardial-secreted molecules include retinoic acid, FGF (fibroblast growth factor) 9,16,20, and IGF2 (insulin-like growth factor 2) [26, 40]. Retinoic acid seems to also act in an autocrine fashion supporting the secretory activity of the epicardium [41] which in turn affects coronary development. Although the characterization of this secretome has been carried out with the main goal of identifying epicardial-secreted molecules regulating myocardial compact layer growth, many of these molecules are likely to affect coronary blood vessel development. As a matter of fact, the thin myocardium that characterizes mice mutant epicardial molecules frequently carries aberrant coronary blood vessels [31, 36, 42], reflecting abnormal ventricular patterning and ramification of coronary vasculature (group 2 CCA).

The epicardium also is sensitive to widely distributed molecules like erythropoietin (EPO) [43] and, accordingly, erythropoietin receptor knockouts have severe coronary dysmorphogenesis [44]. Interfering with embryonic epicardial transcriptional regulation, as shown by the molecular characterization of Wt1 (Wilms tumor 1) and Tbx18 mouse mutants [36, 45], reveals anomalies in the expression of secreted molecules such as VEGF-A and ANGIOPOIETIN-1, growth factor receptors (e.g., PDGFRα,β), or mediators like smoothened and patched (components of the hedgehog signal transduction machinery), all of which are known to be involved in coronary blood vessel development [5, 4648].

The developing myocardium also contributes to coronary development by providing a material substrate for blood vessel formation, as evidenced by the altered coronary patterning found in mice defective for myocardially expressed Vcam1 (vascular cell adhesion molecule 1) and Vangl2 (VANGL planar cell polarity protein 2). Of these two genes, Vcam-1 encodes for a cell adhesion molecule which can act as ligand for α4 INTEGRIN-containing cell receptors [49], while VANGL-2 is a molecule involved in the planar cell polarity signaling pathway [50]. Genetic deletion of cofactors involved in the transcriptional regulation of the myocardium like FOG-2 (zinc finger protein, FOG family member 2) also gives rise to anomalous coronary development [51].


45.3.2.3 Determination and Differentiation of Coronary Progenitor Cells


Misspecification of coronary cell types and/or poor deployment of mesenchymal EPDCs can alter early coronary vascular formation. Knockout embryos for the gap junction connexin 43 gene, which is expressed by epicardial progenitor (pro-epicardial) cells, have severe defects in coronary vascular patterning [52]. Accordingly, Wt1-null mice present decreased coronary endothelial and ectopic smooth muscle differentiation, eventually impairing coronary formation at midgestation [36]. Abrogation of NOTCH1 signaling (a well-known cell fate determinant) in the epicardium severely impacts coronary morphogenesis [42, 53].

Some other genes such as TCF21 (transcription factor 21) have been reported to be involved in the early specification of fibroblasts from the epicardium, as shown by the epicardial EMT and coronary morphogenesis defects displayed by TCF21 mutants [54]. It is of particular interest to consider here that epicardial progenitor (pro-epicardial) cells seem to carefully balance their developmental fate as a response to local BMP2 (bone morphogenic protein 2) and FGF2 levels [55], suggesting that genetic defects in any component of these signaling pathways may result in defects in coronary development.


45.3.2.4 Muscularization and Stabilization of Coronary Artery Anatomical Pattern


The muscular fate of EPDCs is modulated by the subepicardial microenvironment and requires activation of SRF-dependent pool of characteristic smooth muscle genes [56] and p160 RHO-KINASE activation [57]. Both VEGF-A and retinoic acid have been shown to act synergistically to favor early endothelial differentiation from EPDCs at the expenses of smooth muscle cell differentiation [58]. This study proposes, for the first time, a mechanism that explains the physiological delay in the muscularization of developing blood vessels, which is supposed to allow for the extensive remodeling of the primitive coronary vasculature before its maturation and stabilization is initiated. Therefore, premature muscularization of the developing coronary blood vessels may alter the ramification and complexity of adult coronary vasculature, whereas a delay in the formation of the vascular medial wall could result in the formation of aneurism-like defects (Group 2 CCA).


45.3.3 Outgrowth of Endocardial Cells


Defective endocardial incorporation to the coronary vascular system from both sinus venosus-derived and ventricular endocardium can impair coronary vascular patterning. In order to establish a functional vascular circuitry, independent coronary vascular plexuses have to connect growing through the ventricular chamber wall. Altered transmural vascular growth gives rise to abnormal coronary arteriovenous shunts, arterioventricular connections (fistulae), defects that are compatible with postnatal and adult life [2].

It is logical to think that VEGF is a good candidate gene to explain these developmental alterations, because epicardial-to-endocardial VEGF gradients (sensitive to the transmural changes in oxygen tension) are known to determine compact myocardium colonization by embryonic blood vessels [5], and developing vascular structures are known to be extremely sensitive to VEGF doses [59].


Conclusion

As a summary for this chapter, a list of selected genes involved in CAA is provided in Table 45.1. Please note that the selection of these molecules is based on their known involvement in coronary morphogenesis in mice. The functional properties of the molecules encoded by these genes have been experimentally tested by germ line (systemic mutants) or conditional (tissue specific) gene deletion, but determining whether these genes also participate in human coronary development will require further, systematic genetic analysis of human CCAs.


Table 45.1
Genes required for coronary vascular development































































































Genes deleted or altered

Phenotype

References

Neuropilin overexpression

Hypoplastic ventricular myocardium; aberrant coronary vasculature

[60]

VCAM k.o.

Hypoplastic ventricular myocardium, poor epicardial integrity, and abnormal coronary development

[49]

a-4 integrin k.o.

Poor epicardial integrity; abnormal coronary development

[31]

Erythropoietin receptor k.o.

Hypoplastic ventricular myocardium; epicardial detachment and underdeveloped subepicardium; abnormal coronary morphogenesis

[44]

WT1 k.o.

Hypoplastic ventricular myocardium, poor epicardial integrity reduced EPDCs, and abnormal coronary morphogenesis

[36, 61]

FOG-2 k.o.

Hypoplastic ventricular myocardium; abnormal coronary vessels

[51]

Connexin 43 k.o.

Abnormal coronary patterning and presence of conal “pouches”

[52]

RXRa epicardial deletion

Thin myocardium; abnormal epicardial epithelial-to-mesenchymal transformation; defects in coronary vascular formation

[62]

KCNJ8 k.o.

Defects in coronary vascular development

[63]

Tgf-b type I receptor Alk5 epicardial deletion

Thin myocardium and defective coronary muscularization

[35]

A 2A receptor k.o.

Defects in coronary artery formation

[64]

Vangl2 k.o.

Disrupted the organization of the cardiomyocytes and formation of the coronary vessel

[50]

PDGFR beta k.o.

Thin myocardium; fail to form coronary vessels on the ventral heart surface

[47, 65]

Tgfbr2 smooth muscle cell deletion

Anomalous epicardium, myocardium, and coronary smooth muscle cell formation

[66]

Notch k.o.

Disrupts coronary artery differentiation, reduces myocardium wall thickness and myocyte proliferation; smooth muscle differentiation

[42, 53]

Tgfbr3 k.o.

Poor coronary vessel development

[67]

Nephrin k.o.

Abnormal epicardial cell morphology and a reduced number of coronary vessels

[68]

FAK smooth muscle cell deletion

Defective coronary smooth muscle cell formation

[69]

Hand2

Defective epicardialization and failure to form coronary arteries

[70]

Tcf21 k.o.

Hemorrhaging in the pericardial cavity and thin myocardium

[54]

COUP-TFII endocardial and myocardial deletion

Atrioventricular septal defects; thin-walled myocardium; abnormal coronary morphogenesis

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Nov 21, 2016 | Posted by in CARDIOLOGY | Comments Off on Molecular Pathways and Animal Models of Coronary Artery Anomalies

Full access? Get Clinical Tree

Get Clinical Tree app for offline access