Review

Molecular insights into bicuspid aortic valve development and the associated aortopathy

  • Bicuspid Aortic valve (BAV) is one of the most common congenital cardiac malformations with a prevalence of 1–2% in the general population. Patients with BAV have a 9-fold increased risk of developing serious secondary complications including stenosis, endocarditis, regurgitation, dilation of the aorta, aortic aneurysms and subsequent dissection resulting in a significant increase in morbidity. Progressive decline in valve functionality and associated complications warrants surgical intervention in 27% of the affected individuals. The understanding of genetic and molecular mechanisms underlying disease pathology has been largely confounded by phenotypic heterogeneity, incomplete penetrance and variable expressivity. Additionally, the complex interplay between genetic, epigenetic and haemodynamic factors during and after development along with their dynamic expression depending on tissue type contribute to the elusiveness of the disease. While the exact mechanism of pathogenesis remains unclear, recent advances in genetics, propelled by large scale candidate gene discovery strategies employing next generation sequencing, epigenetics, haemodynamic modelling and imaging have provided insights into the development of BAV and associated aortopathy, thus accelerating advances in clinical management and diagnosis of the disease. This review aims at providing a comprehensive understanding of cardiac valve development and the underlying genetic and molecular mechanisms contributing to BAV associated aortopathy.

    Citation: Nikhita Bolar, Aline Verstraeten, Lut Van Laer, Bart Loeys. Molecular insights into bicuspid aortic valve development and the associated aortopathy[J]. AIMS Molecular Science, 2017, 4(4): 478-508. doi: 10.3934/molsci.2017.4.478

    Related Papers:

    [1] Yu Sato, Hiroyuki Jinnouchi, Atsushi Sakamoto, Anne Cornelissen, Masayuki Mori, Rika Kawakami, Kenji Kawai, Renu Virmani, Aloke V. Finn . Calcification in human vessels and valves: from pathological point of view. AIMS Molecular Science, 2020, 7(3): 183-210. doi: 10.3934/molsci.2020009
    [2] Zhepeng Wang, Aiwu Lu . Cartilage type IIB procollagen NH2-propeptide, PIIBNP, inhibits angiogenesis. AIMS Molecular Science, 2021, 8(4): 291-300. doi: 10.3934/molsci.2021022
    [3] Naba Hasan, Waleem Ahmad, Feroz Alam, Mahboob Hasan . Ferroptosis-molecular mechanisms and newer insights into some diseases. AIMS Molecular Science, 2023, 10(1): 22-36. doi: 10.3934/molsci.2023003
    [4] Tateki Kikuchi . Circular breakdown of neural networks due to loss of deubiquitinating enzyme (UCH-L1) in gracile axonal dystrophy (gad) mouse. AIMS Molecular Science, 2021, 8(4): 311-324. doi: 10.3934/molsci.2021024
    [5] Amal Feiroze Farouk, Areez Shafqat, Shameel Shafqat, Junaid Kashir, Khaled Alkattan, Ahmed Yaqinuddin . COVID-19 associated cardiac disease: Is there a role of neutrophil extracellular traps in pathogenesis?. AIMS Molecular Science, 2021, 8(4): 275-290. doi: 10.3934/molsci.2021021
    [6] Mayuko Ichimura, Akari Minami, Noriko Nakano, Yasuko Kitagishi, Toshiyuki Murai, Satoru Matsuda . Cigarette smoke may be an exacerbation factor in nonalcoholic fatty liver disease via modulation of the PI3K/AKT pathway. AIMS Molecular Science, 2015, 2(4): 427-439. doi: 10.3934/molsci.2015.4.427
    [7] Sreemita Majumdar, Song-Tao Liu . Cell division symmetry control and cancer stem cells. AIMS Molecular Science, 2020, 7(2): 82-101. doi: 10.3934/molsci.2020006
    [8] Tara P. Hurst, Caroline Coleman-Vaughan, Indu Patwal, Justin V. McCarthy . Regulated intramembrane proteolysis, innate immunity and therapeutic targets in Alzheimer’s disease. AIMS Molecular Science, 2016, 3(2): 138-157. doi: 10.3934/molsci.2016.2.138
    [9] Sarmishta Mukhopadhyay, Sayak Ganguli, Santanu Chakrabarti . Shigella pathogenesis: molecular and computational insights. AIMS Molecular Science, 2020, 7(2): 99-121. doi: 10.3934/molsci.2020007
    [10] Seidu A. Richard, Wu Min, Zhaoliang Su, Hua-Xi Xu . Epochal neuroinflammatory role of high mobility group box 1 in central nervous system diseases. AIMS Molecular Science, 2017, 4(2): 185-218. doi: 10.3934/molsci.2017.2.185
  • Bicuspid Aortic valve (BAV) is one of the most common congenital cardiac malformations with a prevalence of 1–2% in the general population. Patients with BAV have a 9-fold increased risk of developing serious secondary complications including stenosis, endocarditis, regurgitation, dilation of the aorta, aortic aneurysms and subsequent dissection resulting in a significant increase in morbidity. Progressive decline in valve functionality and associated complications warrants surgical intervention in 27% of the affected individuals. The understanding of genetic and molecular mechanisms underlying disease pathology has been largely confounded by phenotypic heterogeneity, incomplete penetrance and variable expressivity. Additionally, the complex interplay between genetic, epigenetic and haemodynamic factors during and after development along with their dynamic expression depending on tissue type contribute to the elusiveness of the disease. While the exact mechanism of pathogenesis remains unclear, recent advances in genetics, propelled by large scale candidate gene discovery strategies employing next generation sequencing, epigenetics, haemodynamic modelling and imaging have provided insights into the development of BAV and associated aortopathy, thus accelerating advances in clinical management and diagnosis of the disease. This review aims at providing a comprehensive understanding of cardiac valve development and the underlying genetic and molecular mechanisms contributing to BAV associated aortopathy.


    1. Cardiac development and congenital heart disease

    The heart, in addition to being one of the most important organs and the first to develop, is also one of the organs most susceptible to disease and dysfunction and hence extremely widely studied. Heart development begins with the appearance of two distinct mesodermal heart fields; the First Heart Field (FHF) and the Second Heart Field (SHF) both with a common origin contributing to cells of the developing heart in a spatio-temporal manner. While the SHF precursors contribute to the distal developing outflow tract (OFT) myocardium, the proximal smooth muscle cells that give rise to the tunica media of the great arteries, parts of the inflow, ventricular septum and the right ventricle, cells from the FHF contribute to mainly the left ventricle [1,2,3,4]. Both lineage precursors are regulated by a complex network of signaling pathways involving factors of the bone morphogenic pathway (BMP), sonic hedgehog pathway (Shh), Wnt, NOTCH, fibroblast growth factor (FGF) and the TGFβ pathway, some of which emanate from the adjacent endoderm although the exact role and nature of these signals remain unknown [5]. Cells of the FHF differentiate at the cardiac crescent stage while the SHF exhibits delayed differentiation. This delayed differentiation of the SHF is due to the presence of inhibitory Wnt signals rising from the ventral midline [6]. In addition to the FHF and SHF precursors, there exists another key player; the Cardiac Neural Crest Cells (cNCCs). The cNCCs originate from neural crest cells (NCCs) which are derived from the dorsal aspect of the developing neural tube [7]. These cells possess the ability to differentiate into two populations of cells namely the cranial and truncal neural crest. Cells from both the cranial and truncal neural crest give rise to the peripheral nervous system and melanocytes but only the cranial neural crest gives rise to a unique derivate of cells known as the ectomesenchyme. These ectomesenchyme derived from the preotic cranial neural crest contributes to the skeletal structures of the head, face and neck while the ectomesenchyme derived from the postotic cranial neural crest, also known as the cNCC region, contributes to the development of the cardiovascular structures [8,9].

    The NCCs that populate the pharyngeal arches are responsible for the development of all arch derivatives. A sub population of these NCCs, destined to contribute to cardiac development, continue to migrate via the circumpharyngeal region to pharyngeal arches 3, 4 and 6 and into the outflow tract forming both the aortopulmonary septum and the conotruncal portions of the outflow septation complex [10,11]. Depending on which region of the pharyngeal arches the cNCCs migrate from, they contribute different parts of the septation complex and conotruncus at different times. Along with contributing to septation of the OFT, the cNCCs differentiate into smooth muscle cells (SMCs) of the tunica media layer of the aortic arteries distally while the SHF contributes proximally to the SMC layer of the great arteries, this region-specific contribution is crucial to its structure and function. In mice, the migration of the cNCCs occurs at around E9.5, shortly after cardiac looping. Between E8.5 and E9.5, the first signs of valvulogenesis appear. This is marked by the formation of endocardial cushions or local tissue swellings in the lumen of the atrioventricular canal (AVC) and OFT of the looped and segmented heart. A number of signaling molecules produced by the myocardium inhibit the expression of chamber specific gene in the AVC and proximal OFT and instead trigger an increased synthesis of extracellular matrix (ECM) between the endocardium and myocardium [12]. This increased deposition of ECM, also known as the cardiac jelly, leads to protrusions into the lumen of the AVC and OFT giving rise to valve precursors. As the endocardial cushions move into the lumen, signals emanating from the OFT myocardium or the AVC, for example secretion of VEGF, TGβ or BMP factors, trigger a subset of endocardial cells that line the cushions undergo a highly complex and tightly regulated process termed as endothelial-mesenchymal transition (End-MT) causing the specified cells to break away and migrate into the cardiac jelly where they complete their mesenchymal transition in order to populate the precursor valves with mesenchymal cells [12]. In the lumen of the distal OFT localised tissue swelling, termed as truncal cushions arise. These are subsequently populated by the mesenchymal cells originating from the neural crest and form the aortopulmonary septum [13]. Apart from the fact that the AVC valves develop approximately one day earlier than the OFT valves, another distinguishing factor between the two valves is the relative contribution of cells to these valves. While the AVC valves consist of cells that are primarily of endothelial endocardial origin, the semilunar valves are complicated by the presence of cNC progenitor derivatives [12].

    Given that the SHF contributes to progenitor cells of the OFT, small perturbations in this cell lineage can lead to defects involving not only of the OFT but also the OFT valves. Furthermore, the SHF contributes to the OFT myocardium, responsible for the secretion of a number of signaling molecules that trigger Endo-MT/EMT in the conal endocardial cushions aiding in OFT valve and septum development. Additionally, the SHF derived OFT myocardium expresses chemoattractant molecules like SEMA3C which is responsible for cNCC migration and guidance upon entry into the OFT [14,15,16]. At the base of the aorta and pulmonary trunk, the SHF contributes to the vascular smooth muscle cells which supports segregation of the arteries from their respective left and right ventricular outlets [13,17]. Studies targeting SHF factors like Isl1, Mef2c, Tbx20, Tbx1 revealed disruption of a number of signaling pathways namely, TGFβ, Wnt/B-catenin, BMP, FGF, NOTCH, eNOS, Sonic Hedgehog, Retinoic Acid, Nfatc1 and calcineurin and the recently described Robo-Slit pathway [18,19,20,21,22,23,24,25]. Mutations in components of these pathways can lead to a spectrum of phenotypes affecting cardiac septation and valve development (Table 1) [26,27,28].

    Table 1. Genes associated with syndromic and non-syndromic forms of BAV in humans.
    Gene Identified via Phenotype Mutations observed Reference
    NOTCH1 Familial Sequencing, NGS BAV, calcific aortic valve disease, left side congenital heart disease, left hypoplastic heart disease [83,112,113,114] [108]
    MATR3 CNV analysis and Sequencing LVOTO, BAV, CoA, PDA [85]
    GATA4 Sequencing AR, pulmonary stenosis, BAV [86,87,88,89,115,116]
    GATA5 Sequencing Autosomal dominant pattern of BAV inheritance [90,91,92]
    GATA6 Sequencing Tetralogy of Fallot (TOF), persistent truncus arteriosus and atrial septal defects [95,97]
    NKX2.5 Sequencing BAV, ASD, VSD, AVSD, TOF, SVAS, LVNC, PA, PS, PDA, MV anomalies, AV conduction defects, DORV, PAPVR, TAPVR, heterotaxy, TGA [93,117,118,119,120]
    SMAD6 Sequencing BAV and/or aortic stenosis, coarctation of the aorta and with thoracic aortic aneurysms [106,107]
    FBN1 Direct sequencing along with comprehensive assessment of BAV phenotype and function using Doppler echocardiography Atypical mutations associated with BAV, aortic dilation, aneurysm [98,99]
    AXIN/PDIA2/ENG Gene Network Analysis on BAV probands Associated with BAV [121]
    ACTA2 Sequencing BAV, thoracic aneurysm and dissection (TAAD) (BAV, CoA or PDA observed) [122], [109]
    MAT2A Sequencing Thoracic aortic aneurysms, variable presentation of BAV [123]
    UF1DL Protein analysis Reduced levels of UF1DL in BAV aortas [124]
    EGFR Targeted Sequencing of nine genes Associated with BAV [125]
    TGFBR1, TGFBR2 Sequencing Non syndromic BAV and proximal aortic aneurysm [104]
    TGFBR1/2, TGFB2, TGFB3, SMAD3 Sequencing, NGS Loeys-Dietz Syndrome (LDS) or phenotypes within the LDS spectrum [37,100,101,102,103]
    COL1A2 Sequencing Ehlers Danlos Syndrome (skin and bone abnormalities as well as mitral and aortic valve dysfunction observed) [126]
    KCNJ2 Familial sequencing Andersons Syndrome (Cardiovascular abnormalities: polymorphic ventricular tachycardia and periodic paralysis and semilunar valve abnormalities including BAV) [81]
    HOXA1 Sequencing Bosley-Salih-Alorainy syndrome and Athabascan Brainstem Dysgenesis syndrome (Cardiovascular abnormalities observed: interrupted aortic arch type B, subclavian artery abnormalities, VSD, TOF, and BAV) [127]
    Abbreviations: BAV: Bicuspid Aortic Valve; LVOTO: Left ventricular outflow tract obstruction; CoA: Coarctation; PDA: Patent ductus arteriosus; AR: Aortic regurgitation; ASD: Atrial septal defect; VSD: Ventricular septal defect; AVSD: Atrioventricular septal defect; TOF: Tetralogy of Fallot; SVAS: Supravalvular aortic stenosis; LVNC: Left ventricular non-compaction; PA: Pulmonary atresia; PS: Pulmonary stenosis; PDA: Patent ductus arteriosus; MV: Mitral valve; AV: Atrioventricular; DORV: Double outlet right ventricle; PAPVR: Partial Anomalous Pulmonary Venous Return; TAPVR: Total anomalous pulmonary venous return; TGA: Transposition of the great arteries
     | Show Table
    DownLoad: CSV

    Signals emanating from the myocardium trigger Endo-MT/EMT and constitute a number of pathways and factors like BMP2/4, TGFB2/3 and VEGF. The sensitivity of the endocardium to these factors, in turn initiating Endo-MT/EMT, requires the expression of receptors and effectors of downstream myocardial signaling pathways such as Alk1, Alk3, Alk5, B-catenin, VEGF, NOTCH1, Twist1, Sox9, Tbx20, Gata4, Pitx2 and Hedgehog signaling [29]. Increased ECM production for valve development requires the suppression of chamber specific genes at the myocardium of the cushions. This process is specified and programmed by Tbx2, Nfatc2-4 and Bmp2 among others to suppress chamber-specific genes, produce factors important for EMT and to increase ECM synthesis creating a support for EMT [30,31]. Both the myocardium and endocardium at the cushion is different from that in the cardiac chambers. These tissues at the cushion express genes that are essential for valve and septal development like Nfatc1 [31]. This regional specificity of EMT is defined by the temporal pattern of co-ordinated yet distinct and specific gene programming events and expression of an array of signaling factors and receptors at the endocardium and myocardium cushions of the OFT and AVC.

    Given the complexity and intricacy of the pathways and signaling molecules that orchestrate cardiac development in a spatio-temporal manner, slight dysregulation at any stage can lead to serious consequences on the development of the heart and embryo, sometimes fatal. Defects in gene expression of factors governing development, differentiation, migration and proliferation in either of the two myocardial lineages (FHF and SHF) and cNCC can lead to an array of cardiac phenotypes (Table 1 and 2)

    Table 2. Animal models of valvular defects including BAV and great artery defects.
    Gene Expression Organism Genotype Phenotype Reference
    NOS3 Endocardium, myocardium, SL valves Mouse, Syrian Hamster Nos –/– Congenital heart defects with BAV [45,46,132]
    GATA5 AV and OFT Endocardial cushions, endocardium Mouse Gata5–/–, eGATA5–/–, Tie2Cre; Gata5F/F BAV [131]
    NOTCH1 OFT and AV endocardial cushions Mouse, Zebrafish Notch1–/– Septation defects, valve atrophy [134]
    GATA6 AV and OFT, endocardial cushions, endocardium, myocardium Mouse Targeted deletion in VSMCs and NCCs; Conditional knockout; SMCre+GATA-6F/F; Wnt1Cre+GATA-6F/F DORV, VSD, aortic arch patterning defects [130]
    FGF8 Heart, OFT, endocardial cushions, great arteries, pharyngeal endoderm Mouse Fgf8; hoxa3-IRESCre (Ablation in the 3rd and 4th pharyngeal endoderm and ectoderm) BAV, parathyroid and thymic defects [130]
    Fgf8; AP2α-IRESCre (Ablation in the pharyngeal arch extoderm) Postnatal lethality, Failed formation of the 4th pharyngeal arch artery, aortic arch defects, IAA
    Nkx2.5 SHF, cNCCs, OFT endocardial cushions Mouse Nkx2.5GFPhet, Nkx2.5HDneohet BAV, AV block, ASD [135]
    MATR3 cNCCs, heart, great arteries Mouse Matr3GT-ex13–/– Lethal [85]
    Matr3GT-ex13+/– BAV, PDA, CoA
    ROBO1 and ROBO2 Endothelial cells, endocardium. OFT Endocardial cushions Mouse Robo 1–/– VSD, Overriding Aorta (OA) [21]
    Robo1–/–Robo2–/– VSDs, OA, BAV,
    SLIT2 Endocardium lining Mouse Slit2–/– VSD, thickening of SL valves [21]
    SLIT3 OFT and myocardium adjacent to endocardial cushions Mouse Slit3–/– VSD, hypoplastic aortic valves [21]
    Alk2 (Acvr1) cNCC, OFT endocardial cushions Mouse Targeted deletion in endocardial cushions; Alk2–/–/Gata5-Cre BAV, AS, AI [136,137]
    Targeted deletion in NCCs Wnt-Cre Alk2–/– PTA, abnormal morphogenesis of the aortic arch, impaired migration of cNCCs into the OFT and loss of cNC derived smooth muscle cell differentiation around the aortic arteries.
    ALK6 cNCC, OFT endocardial cushions Mouse Targeted deletion in NCCs Wnt-Cre Bmpr1a–/– shortened outflow tract and septation defects including reduced proliferation of the ventricular myocardium [138]
    SMAD4 cNCCs, myocardium, SHF Mouse Targeted deletion in NCC Wnt-Cre Smad4fl/fl Embryonic lethality, reduced cNCC migration into OFT, PTA, OFT hypoplasia and septation defects [139]
    TBX20 cNCCs, SHF, OFT endocardial mesenchyme Mouse Tbx20–/– Embryonic lethality, OFT defects [140,141]
    Targeted deletion in the endocardial cushions; Nfatc1-Cre Tbx20–/– Dysmorphic and hypoplastic valve, endocardial cushion proliferation defects
    ET-A cNCCs, OFT endocardial cushions, endocardium, Mouse ETA–/– IAA, DA, aortic arch patterning defects, OA, DORV, PTA, TGA [18,142]
    ET-1 Endothelium of great arteries and OFT, cNCCs, OFT endocardial cushions, endocardium, epithelium of pharyngeal arches Mouse ET-1-/- IAA, VSD, tubular hypoplasia of the aortic arch, OFT malformations [143]
    ECE1 OFT endocardial cushions, endocardium Mouse Ece1–/– IAA, PTA, VSD, DORV [144,145]
    Mouse Ece1–/–: Ece2–/– AV and OFT dysplasia, DORV, VSD, hypoplastic endocardial cushion
    ECE2 Endocardial cushion mesenchyme Mouse Ece2–/– No detectable phenotype
    NF1 cNCC, SHF, endocardial OFT endocardial cushions, myocardium, endothelium Mouse NF1–/–, Endothelial specific deletion NF1ec (Tie2-Cre NF1–/–) Enlarged endothelial cushions, DORV, VSD, impaired EMT [146]
    NFATC1 OFT endocardial cushions, endothelium Mouse NFATC1–/– Embryonic lethality, hypoplastic valves, impaired SL valve development [147,148]
    Ltbp1L cNCC, OFT endocardial cushions Mouse Ltbp1L–/– PTA, IAA [149,150]
    SEMA3C cNCC, SHF, endocardial OFT endocardial cushions, myocardium Mouse Sema3c–/– Quadricuspid Aortic valve, CoA, PTA, septal defects abnormal migration of cNCCs [14]
    CXCR4 SDF1 cNCC, SHF, endocardial OFT endocardial cushions, myocardium Chick miRNA-CXCR4 silencing, Dominant negative-CXCR4 (DN-CXCR4) to inhibit SDF1 signaling delayed cNCC migration and increased cNCC death cNCC migration defects, severe conotruncal and cardiac defects [151]
    CXCR7 cNCC, OFT endocardial cushions Mouse CxCr7–/– Severe SL valve stenosis [152]
    Cnb1 SHF, OFT endocardial cushions Mouse Targeted deletion in SHF Mef2c-Cre Cnb1–/– Absent SL valves, reduced conal cushions [26]
    Rac 1 cNCCs Mouse Wnt-Cre Rac1–/– Abnormal patterning of the aortic arteries, absence of VSMC differentiation.OFT septation failure [153]
    Rae 28 cNCC, OFT endocardial cushions Mouse Rae28–/– TOF, DORV and aortic valve stenosis [154,155]
    Ets1 OFT septum Mouse Ets1–/– Septal defects [156]
    HOXA1 NCCs, OFT myocardium, endocardium, SHF Mouse HOXA1–/– BAV and heart defects [157]
     | Show Table
    DownLoad: CSV

    Congenital heart defects involving the OFT, aortic arch and the ductus arteriosus constitute about 20–30% of all congenital heart disease. The presence of a bicuspid aortic valve (BAV), a common congenital cardiac anomaly with a relatively high prevalence in the general population, results in an increased risk of complications later on in life. Due to the significant associated morbidity and mortality, these congenital heart defects prove to be a continuing medical challenge for prognosis and treatment, most of the time demanding invasive approaches of therapy.


    2. Bicuspid aortic valve associated thoracic aortic aneurysm

    The normal aortic valve is tricuspid and comprises three semilunar shaped leaflets or cusps, each of which housed within a small dilation of the proximal aorta called the Sinus of Valsalva or the aortic sinus. Each of these sinuses and the leaflets arising from them are annotated relative to their relationship to the coronary arteries; right, left and non-coronary sinuses/cusps. The outward edges of the semicircular border of each cusp attach it to the wall of the aorta via an attachment point called a commissure. The bicuspid aortic valve is defined as a congenital cardiac abnormality that arises due to perturbations in tightly regulated process involved in valve formation and OFT septation during cardiogenesis. These perturbations result in an aortic valve comprised of only two asymmetrical valve leaflets, as a consequence of fusion of two out of the three valve leaflets, instead of three distinct valve leaflets. The region at which the fusion between two leaflets occurs is marked by a raphe, commonly present with most fusion patterns, although rarely fusion of leaflets without a marked raphe does occur, giving rise to symmetrical valve leaflets. The most common fusion pattern observed in BAV is the fusion of the right and left (R-L) coronary cusps and this occurs in 80% of individuals that present with BAV while the right-noncoronary cusp fusion (R-N) occurs in ~17% followed by the rarest fusion pattern, the left-noncoronary cusp (L-N) fusion in about ~2% of BAV patients (Figure 1) [32,33]. With a prevalence of 1–2% in the general population and a male to 138 female preponderance of ~3:1, BAV is one of the most common congenital malformations as compared to a total of 0.8% for all other forms of congenital cardiac disease combined. This large prevalence is reflective of the immense clinical burden that BAV imposes for the vast majority of the cases that will develop serious complications later on in life, such as aortic dilation and dissection. As a consequence of BAV, some of the serious and common complications like aortic stenosis (AS) and aortic regurgitation (AR) may occur. Dilation involving any or all segments of the proximal aorta right from the aortic root to the arch, termed as BAV related/associated aortopathy is observed in about 50% of the affected individuals [34,35]. Abnormalities of the aortic wall are common in BAV patients and, from a developmental perspective, can be explained by the relative contribution of NCCs to both, the semilunar valves and the tunica media of the distal great arteries. Hence, defects of the NC can result in an abnormal valve in combination with ascending aorta or aortic arch abnormalities. In fact, studies have shown that impaired differentiation and contraction of the cNCC derived smooth muscle cells contribute to aortopathy observed in BAV individuals [36]. Craniofacial abnormalities have also been observed in patients with syndromic aneurysms, with BAV as one of the cardiovascular anomalies, reflective of defects in the neural crest that also contribute to the craniofacial mesenchyme [37,38,39,40,41].

    Figure 1. Modified and referenced from Kang et al.: BAV subtypes with and without a raphe [69].

    BAV individuals present with an 8-fold increased risk of aortic dissection and a 53% risk of valve replacement over a 25-year period. In addition, affected individuals carry a 26% risk for aneurysm formation and a 25% risk for aortic surgery [42]. Almost 75–80% of the stenotic valves in children and at least 50% of aortic valve stenosis in adults is due to the presence of BAV [43]. BAV associated aortopathy can be classified into 3 different subtypes (Figure 2): Aortic root dilation (Type 1), Aortic enlargement involving the tubular portion of the ascending aorta (Type 2) and diffuse involvement of the complete ascending aorta and the transverse aortic arch (Type 3) [44]. A normal non-dilated aorta is classified as Type 0. Studies have hypothesized that while the different valve fusion subtypes have distinct etiologies which are reflective of the defects in molecular and genetic mechanisms involved in the developmental processes of valve development and OFT septation, the subsequent BAV associated aortopathy observed later on in life, may be different depending on the valve fusion subtype observed [44,45,46,47]. BAV associated aortopathy follows a heterogeneous pattern with variable presentation within affected members of families and isolated individuals. This suggests that the underlying pathomechanism of BAV associated aortopathy may consist of a complex interplay of genetic, hemodynamic and epigenetic factors.

    Figure 2. Modified and referenced from Kang et al.: Types of Aortopathy observed in BAV patients [69].

    3. Hemodynamics in relation to valvular fusion

    The altered hemodynamics of blood flow through the aorta as a result of a morphologically or functionally defective aortic valve, has been one of factors, suggested to lead to BAV-associated aortopathy. Several studies have established the presence of a nonaxial transvalvular turbulent jet flow within the aortic root, as a result of the restricted mobility of the fused cusps [48,49,50]. This turbulent jet flow creates altered biomechanics and fluid dynamics within the proximal aorta causing uneven wall stress distribution along the aortic wall, in turn leading to aortic dilation or aneurysm formation and subsequent dissection and rupture. As consequence of this hemodynamic stress, the dilation protrudes asymmetrically towards the right anterior aortic wall due to the aortic convexity [51]. A correlation has been established between the degree of restrictive mobility of the fused cusp and the eccentricity of the transvalvular systolic flow, with the severity and rate of dilation at the root and ascending aorta [50,52]. In addition to determining the rate of progression of aortic dilation due to turbulent flow patterns, the BAV cusp fusion subtypes have been shown to produce unique jet orientations in the proximal aorta [51]. The different BAV subtypes lead to different degrees and orientations of asymmetry in aneurysms that in turn leads to varying rates of dilation progression and altered signatures of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) [53,54]. Comprehensive studies involving MDCT imaging and transthoracic echocardiography of BAV patients with varying BAV subtypes and associated aortopathy, reported that, the R-L fusion subtype was most commonly associated with a normal aorta followed by type 2 aortopathy, while the R-N/L-N fusion subtype was most frequently associated with type 3 aortopathy. In addition, although both subtypes showed hemodynamically significant valvular dysfunction in terms of stenosis and regurgitation, moderate-to-severe aortic stenosis (AS) is predominantly associated with R-N/L-N subtype as compared to moderate-to-severe aortic regurgitation (AR) in R-L subtypes [44]. This study also confirmed the R-L subtype as being the most common valve fusion phenotype among patients. 4D cardiac MRI also showed that the R-L subtype produced a right-anteriorly directed helical systolic jet flow with considerable peripheral skewing towards the ascending aortic convexity, which may explain the asymmetrical dilation of the mid-region of the ascending aorta along with increased aortic root dimensions [48,51,53]. These patients also show severe degeneration of the aortic wall accompanied by higher proteolytic indices. The less frequent R-N subtype is associated with a left-posteriorly directed eccentric flow jet towards the proximal aortic arch which explains the diffuse involvement of the ascending aorta and the transverse arch leaving out the aortic root [48,55]. Interestingly, phase locked particle image velocimetry experiments on non-dilated ascending BAV aortas subjected to different flow patterns observed in TAV, R-L BAV, R-N BAV and N-L BAV revealed significant flow abnormalities measured by degree of jet skewness, velocity, turbulence and shear stress. These substantial flow abnormalities observed in non-dilated aortas, although different from dilated aortas, have been shown to coincide with regions of the aortic wall prone to dilation [56]. Abnormal shear stress created as a result of cusp fusion, can exert an effect on molecular events and modulate genes responsible for ECM remodeling, MMP production and apoptosis of VSMCs hereby increasing the predisposition to aneurysm formation [57]. Levels of global MMP activity including MMP-2, MMP-9 were increased while expression of MMP-7, MMP-8, TIMP-1 and TIMP-4 was decreased in BAV patients [53,58]. MMP and TIMP levels in the R-L patients were consistent with the overall expression observed in BAV patients. In the case of L-N patients, although elevated levels of global MMPs were observed, only MMP-8 and TIMP-4 expression levels showed a decrease. The R-N subgroup showed only a decrease in levels of MMP-7, MMP-8, TIMP-1 and TIMP-4. In addition, the R-L subgroup showed higher proteolytic balance indices for MMP-1, MMP-9 and MMP-12 relative to the R-N and L-N subtypes. This further highlights the unique MMP and TIMP signatures specific to BAV subtypes, and implicating the R-L subtype as associated with the most aggressive deterioration of aortic wall requiring early surgical intervention. Differential profiles of circulating MMPs, TIMPs and recently implicated miRNAs (eg; miRNA-21, miRNA-1, miRNA-29a) may prove to be important biomarkers to predict progression of aortopathy and to distinguish between different BAV associated aortopathy etiologies [59]. Apart from the differential MMP and TIMP expression levels, differential levels of endothelial nitric oxide synthase (eNOS) have also been reported in aortic walls from BAV patients as compared to individuals with TAV [60,61,62,63]. While a reduction in levels of eNOS was observed in the proximal aorta (above the sinotubular junction), eNOS levels in the concavity and distal aorta were increased. This site-dependent variation is probably reflective of aggressive and variable shear stress along the aortic wall [60].

    Although valvular dysfunction along with hemodynamic factors, as a result of BAV, can explain some of the progressive, typically asymmetric aortic enlargement observed in these patients, it cannot entirely describe the full spectrum of BAV-associated aortopathy. Understanding not only the hemodynamic factors but also the developmental mechanisms and the underlying genetics, that govern this congenital abnormality, will be essential to explain the phenotypic heterogeneity and the propensity to develop BAV associated aortopathy seen in these patients.


    4. Genetics

    The "genetic" hypothesis around BAV and its associated aortopathy evolved from the observation that pediatric patients with BAV had larger aortic root and ascending aorta measurements indicating that the inherent structural fragility of the aortic wall was determined early on life [64,65]. Furthermore, progressive aortic dilation is seen in patients both with a normally functioning BAV and following valve replacement indicating an inherent weakness of the aortic wall leading to vascular complications independent of valvular dysfunction [62,66]. Familial studies also demonstrated that within families, BAV can occur independently of any vascular complication or aortopathy and vice versa [67]. Histopathological examination of dilated aortas from BAV individuals revealed localized regions of matrix disruption, VSMC apoptosis, altered collagen content, increased elastic fragmentation and MMP levels and accumulation of mucopolysaccharide ground substance. These findings were strikingly similar to aortas analyzed from Marfan syndrome (MFS) patients caused by a deficiency in fibrillin-1 (FBN1) and was also observed in non-dilated aortas [68,69,70]. Interestingly, loss of aortic elasticity, increased MMPs and fibrillin-1 deficiency has been observed independent of aortic diameter in BAV individuals [71,72]. FBN1 deficiency triggers the activation of MMPs, degrading the structural integrity of the aorta. Notably, decreased fibrillin-1 expression has been observed in the aortic wall of BAV individuals and it is postulated that a microfibrillar gene defect causes increased VSMC detachment from the elastic laminae leading to VSMC apoptosis, subsequently resulting in an increase of MMP and proteolytic factors [72,73,74,75]. This MMP and proteolytic factor activation accelerates the degeneration of the aortic wall and causes loss of structural integrity and elasticity, resulting in progressive dilation prior to aneurysm development even after valve replacement or in the presence of a normally functioning BAV. The FBN-1 deficiency and dilation is also observed in the walls of the pulmonary artery in the absence of any pulmonary valve abnormality in BAV individuals, indicating an underlying developmental abnormality since the great arteries originate from a common embryonic trunk [72]. Interestingly, a recent study observed and upregulation of fibrillin 2 (FBN2) in the ascending aorta of patients with BAV irrespective of dilation [76]. These observations suggest a genetic contribution to the pathogenesis of BAV associated aortopathy. In addition, familial clustering studies and echocardiography screening revealed a high incidence of BAV with 9% prevalence in first degree relatives of BAV patients and about 24% in families with more than 1 affected individual, proposing an autosomal dominant pattern of inheritance with reduced penetrance and variable expressivity [77,78]. A heritability of 89% was estimated indicating that BAV was almost entirely genetically determined with mutations in a number of different genes responsible for the phenotypic heterogeneity observed in BAV patients [43].

    The genetics of BAV and its associated aortopathy remain elusive and largely uncharacterized. Linkage analysis in BAV families using microsatellite markers have identified regions on chromosome 5q, 13q and 18q linked to BAV and cardiovascular malformations, while a region on 15q25-26 has been linked particularly to BAV and ascending aortic aneurysms. The specific genes within these regions have not yet been identified [79,80]. One of the first genes to be associated with BAV was KCNJ2, identified via ascertainment of a 41-member pedigree presenting with polymorphic ventricular tachycardia and periodic paralysis and semilunar valve abnormalities including BAV [81]. However, the strongest association came shortly after with the identification of mutations in NOTCH1 in non-syndromic BAV and calcification in a small number of families and ~4% of sporadic cases [82,83]. Missense and loss of function mutations in NOTCH1 in affected individuals with BAV and/or aneurysms revealed haploinsufficiency as a probable mechanism of aortic valve disease. NOTCH1 is expressed in the OFT mesenchyme and aortic valve leaflets highlighting the role of Notch signaling in aortic valve development and morphogenesis. NOTCH1 has been shown to regulate Sox9 expression; a chondrogenic transcription factor influencing the expression of genes involved in ECM remodeling and valve development. Inhibition of NOTCH1 signaling results in decreased Sox9 expression causing aortic valve calcification while overexpression of Sox9 curbs the calcific process associated with Notch1 inhibition [84]. Hence, NOTCH1 mutations drive the dysregulation of developmental processes involved in aortic valve development and remodeling probably via a Sox9 dependent and Runx2 upregulation mechanism leading to calcification/stenosis of the aortic valve [82,84]. Recently, MATR3 was identified at a translocation breakpoint in a 46, XY, t(1:5)(p36.11;q31.2) dn translocation carrier with pervasive developmental delay and left ventricular outflow tract obstruction (LVOTO) defects, including BAV, coarctation of the aorta and patent ductus arteriosus [85]. The 5q translocation breakpoint resulted in the disruption of the 3' UTR of MATR3, which encodes for the nuclear matrix protein; Matrin 3. The 3'UTR disruption of the MATR3 transcripts resulted in polyadenylation defects affecting transcript and protein expression [85]. The GATA family of transcription factors have long been known to be essential for development and morphogenesis of the cardiac system (Table 1). Mutations in GATA4, an essential transcription factor in OFT development lead to atrial septal defects and valve abnormalities like aortic regurgitation, mitral valve regurgitation and pulmonary stenosis [86,87,88]. Some of the mutations identified have been shown to disrupt GATA4 binding to Smad4 or Tbx5 indicating the conserved function of these Gata complexes in AV and OFT valve development [86,89]. GATA5, has been shown to be essential for aortic valve development. Mutations in GATA5 were identified in families presenting with an autosomal dominant pattern of inheritance of BAV. Few of the identified rare GATA5 variants resulted in significantly decreased transcriptional activity as compared to wildtype when tested via functional assays [90,91,92]. Nkx2.5 maps to region 5q34 which has been linked to BAV and recently, the first mutation in Nkx2.5 was found in a family with BAV, segregating in an autosomal dominant pattern. As a consequence of the mutation, the interaction of Nkx2.5 with GATA5 was abolished rendering Nkx2.5 incapable of regulating downstream effectors in a GATA5 dependent manner [93]. In addition, Nkx2.5 is also known to be a modulator of ECM remodeling through collagen type 1 (COL1A1). The activation of collagen type 1 is dependent on the interaction between Nkx2.5 and GATA6 [94]. Mutations in GATA6, a transcription factor expressed in the developing heart, OFT and VSMC, are associated with Tetralogy of Fallot (TOF), persistent truncus arteriosus and atrial septal defects [95]. Gata6 is involved in OFT development and has been shown to transcriptionally activate SEMA3C and PLXNA2 to aid the migration of NCC into the OFT, which is important for OFT septation [96,97]. Some of the identified mutations in patients with persistent truncus arteriosus have been known to disrupt the binding of GATA6 to SEMA3C and PLXNA2 promoters abrogating its transcriptional activity and nuclear localization [97]. Although, GATA6 maps to chromosome 18q, a region already known to be linked to BAV, no mutations have been reported in BAV individuals so far. Direct sequencing along with comprehensive assessment of BAV phenotype and function using Doppler echocardiography in 10 BAV individuals revealed novel mutations in FBN1 in the absence of Marfan syndrome (MFS) [98,99]. Mutations in genes involved in the TGFβ signaling pathway, TGFBR1, TGFBR2, SMAD3, TGFB2 and more recently TGFB3 are known to cause Loeys-Dietz Syndrome (LDS) or phenotypes within the LDS spectrum [37,100,101,102,103]. A TGFBR2 mutation was identified in a patient with BAV and aneurysm without a LDS presentation [104]. Analysis of global expression levels between BAV and tricuspid aortic valve (TAV) individuals with and without an aneurysm revealed a significant increase of TGFβ binding proteins; particularly LTBP3/4, ADAMTSL1 and an alternatively spliced isoform of fibronectin-1 (FN1) exclusively in BAV patients with aortic dilation [105]. Additionally, non-synonymous variants in SMAD6 have been observed in patients with BAV and/or aortic stenosis, coarctation of the aorta and with thoracic aortic aneurysms [106,107]. Recently, whole exome sequencing of 182 individuals of 51 pedigrees with congenital heart disease including BAV identified mutations in NOTCH1 and other factors of the NOTCH signaling cascade including MAML1, JARID2, SMARCA4 and JAG1. Furthermore, mutations were also identified in ARHGAP31 and MYH6 [108]. Linkage analysis of family members with BAV and/or aortic aneurysms identified the ACTA2 gene [109]. ACTA2 encodes for α-smooth muscle actin and analysis of tissues obtained from the aortas of these patients show proteoglycan accumulation, fragmentation of elastic fibers and loss of VSMCs. Although it is clear that mutations in ACTA2 can lead to thoracic aneurysm and dissection (TAAD) as a result of increased fragility and degeneration of the aortic wall, it is unclear if these mutations can cause BAV [109]. A class of small noncoding RNAs that regulate gene expression, microRNAs (miRNAs) have been shown to be differentially expressed in BAV patients. miR-26a, miR-30b, and miR-195 are downregulated in BAV patients with aortic stenosis as compared to aortic regurgitation [110]. In vitro studies, transfecting each of the above miRNAs into aortic valve interstitial cells activated a number of genes like Bmp2, Runx2, SMAD3, SMAD1 and alkaline phosphatase (ALPL) [110]. Stenotic valves also have a reduced expression of miR-41, a repressor of Bmp2 [111]. The above studies highlight the complexity and the genetic heterogeneity that confound the molecular dissection of this disease.

    Although the studies mentioned above are in humans, the largest contribution to understanding the development and pathogenesis of BAV-TAA comes from animal studies (Table 2). Tissue specific ablation of Notch1 in either cNCCs or the SHF leads to developmental abnormalities of the semilunar valves [28,128,129]. Interestingly, SHF specific inhibition of Notch1 causes a dysregulation of Fgf8 signaling leading to VSMC abnormalities of the great arteries and BAV thus suggesting a common mechanism underlying both valve defects and the associated aortopathy [130]. Gata5 knockout mice showed a downregulation of endothelial nitric oxide synthase (eNOS) expression levels in the OFT endocardial cushions. Of note, the R-N cusp fusion subtype observed in Gata5 knockout mice was identical to the valve phenotype observed in Nos3 knockout mice suggesting that R-N fusion is a result of perturbations in early developmental processes, prior to OFT septation, and may rely on the nitric oxide (NO)-dependent differentiation/migration of endocardial cells during EMT [46,131]. Studies in eNOS knockout mice and inbred syrian hamsters have shown that while the R-N fusion subtype is reflective of an early EMT defect, the R-L fusion subtype is a late defect caused due to abnormal cNCC migration resulting in impaired septation of the proximal OFT [45,46,132]. The abnormal migration of the neural crest could explain the severe aortopathy in the R-L fusion subtype associated with accelerated degeneration of the aortic wall as compared to the R-N fusion subtype. This study for the first time shed light on the different etiological identities of the BAV subtypes, which may play an important role in understanding the genetic and haemodynamic mechanisms at play. These observations re-instate the importance of Nos3 in valve development but also suggest a dose dependent role during valve morphogenesis. Gata4, Gata5 and Gata6 expression has been shown to overlap during cardiac development. Gata4+/– Gata5+/– and Gata5+/– Gata6+/– double knockout mice die perinatally or embryonically due to severe cardiac defects including double outlet right ventricle (DORV) and ventricular septal defects (VSD) [133]. Examination of Gata4+/– Gata5+/– mice revealed an extremely high pressure gradient through the aortic valve along with the presence of BAV. The prevalence of BAV in Gata5+/– Gata6+/– mice was ~25%. Collectively, the above data suggest a dose-dependent sensitivity for GATA factors during cardiogenesis and aortic valve formation.

    The above mentioned findings provide a foundation for the genetic basis of the pathogenesis of BAV and highlights the complex crosstalk between hemodynamic, developmental, environmental and genetic factors.


    5. Epigenetics

    In addition to mutations in the genes themselves, another important heritable factor in regulating or modulating the expression of genes involves the modification of chromatin. The higher order structure containing packaged DNA around histone proteins serves not only a structural purpose but also one that is critical for gene expression controlling the access of certain factors to specific regions in the genome in a temporal manner. Regulation at the chromatin level is mediated by three different processes; DNA methylation and acetylation, ATP dependent chromatin remodeling and covalent histone modifications. Combinations of specific histone modifications coupled with nucleosome structure at defined regions provide a histone code or mark which act as docking sites for transcription factors and other chromatin regulators exerting an array of specificity and complexity on gene expression control.

    One of the important molecules known to play a role in cardiac and OFT development is the Brg1/Brm-associated factor (BAF) complex. The BAF complex consists of 12 subunits and is part of the family of SWI-SNF type of chromatin remodeling complexes. One of two highly homologous genes, Brg1 and Brm encode for the ATPase subunit of the BAF complex. Brg1 aids in the binding of the myocardin family of coactivators to promoters of genes involved in the expression of smooth muscle contractile proteins [158,159]. Targeted deletion of Brg1 in VSMCs in mice results in patent ductus arteriosis causing cyanosis and dilation of the cardiac chambers in 1/3 of the mice [160]. Furthermore, in vitro studies on VSMCs have shown that knockdown of Brg1 or Brm results in decreased expression of SMC contractile proteins which could provide an explanation for the patent ductus arteriosus phenotype. While the patency of the ductus arteriosus is essential for fetal circulation, its closure, mediated by the contraction of SMCs of the DA is essential for adult circulation, hence disorganization of SMCs as a result of Brg1 deficiency can lead to PDA due to absent perinatal closure of the ductus arteriosus. Deletion of Brm, does not recapitulate any of the Brg1 knockout defects and neither does it exacerbate the patent ductus arteriosus phenotype of Brg1 deficient mice. This highlights the fact that Brg1 is the dominant ATPase of the BAF complex and is crucial for smooth muscle cell maturation. These studies have brought to light the importance of factors involved in VSMC maturation and homeostasis, dysregulation of which could possibly contribute to the underlying aortopathy observed in BAV individuals including increased VSMC apoptosis, aberrant ECM remodeling and loss of elasticity and contractile ability resulting in a weakened aortic wall.

    While mutations in NOTCH1 are known to be associated with BAV and calcific valve disease, recently, human disease modelling in induced pluripotent stem cells (iPSCs) derived from endothelial cells, revealed a complex network of transcriptional and epigenetic mechanisms governing NOTCH1 haploinsufficiency [161]. This study showed that NOTCH1 haploinsufficiency caused by heterozygous nonsense mutations in NOTCH1, affected H3K27 acetylation at NOTCH1-bound enhancers leading to disruption of the transcriptional process of almost 1000 genes involved in osteogenesis, inflammation and oxidative stress. This suggests that the altered epigenetic architecture caused an activation of the pro-osteogenic factors exacerbated in the presence of hemodynamic shear stress leading to calcification of the aortic valves [161]. This study highlights the complex interplay between genetic, epigenetic and hemodynamic factors, in addition to transcription factor dosage effect leading to human disease. Acetylation and deacetylation of lysine residues on Notch1 intracellular domain (NICD) act as important modulators of NOTCH signaling and response by regulating NICD protein turnover shuttling between NICD stabilization and degradation respectively. Interestingly, endothelial specific deletion of the lysine deacetylase Sirt1, important for the deacetylation of NICD, caused increased sensitivity to Notch signals and aberrant cell signal response due to the activation of downstream Notch target genes leading to impaired vascular growth and sprout elongation [162]. Further studies are required to determine the exact role of Sirt1 in vascular development. The involvement of the mixed lineage leukemia (MLL) complexes in cardiovascular development has also been suggested. Mutations in MLL2, known to cause Kabuki syndrome, are characterized by an array of birth defects including cardiac defects. Patients with kabuki syndrome show a high incidence of LVOTO defects and aortic dilation at approximately 46% with the most common defects being coarctation of the aorta, ventricular septal defects and atrial septal defects [163]. While de novo and partial deletions of the X chromosome gene, KDM6, which encodes a histone demethylase that interacts with MLL2 have been shown to also cause Kabuki Syndrome, loss of function mutations in MLL2 remain the major cause of Kabuki Syndrome. Recently, a Kabuki patient presented with BAV as the only cardiac defect observed [164]. Impaired histone methylation due to deficiency of MLL2 causing reduced expression of the downstream targets provide a mechanism of pathogenesis in Kabuki Syndrome patients. Another important factor in chromatin remodeling is the histone deacetyltransferase; EP300. P300 proteins act as transcriptional coactivators bridging communication between the transcriptional activators located at enhancer sites and the basal transcription machinery at promoter sites. Crucial to myocardial cell differentiation, EP300 is known to act as a coactivator of GATA5, via physical interaction of the two proteins, to regulate the transcription of the cardiac restricted atrial natriuretic factor (ANF) gene expressed in cells committed to the cardiac lineage early on in embryonic development [165]. Interestingly, mutations in EP300 are known to be associated with Rubenstein-Taybi Syndrome (RSTS) characterized by delayed development, microcephaly and dysmorphic facial features. Several secondary complications are observed in patients with RSTS including cardiovascular malformations like BAV, ASD, VSD, PDA, coarctation of the aorta and hypoplastic left heart [166]. Given the role of the TGFβ superfamily in cardiac development, with mutations in various signaling factors and receptors causing an array of syndromic cardiovascular phenotypes, adding another layer of complexity, studies have shown that alternative TGFβ splicing patterns driven by diverse epigenetic modification mechanisms at specific loci can contribute to the phenotypic heterogeneity of TAA aortopathy observed in BAV patients [167].


    6. TGFβ, BMP and NOTCH-The crosstalk


    6.1. TGFβ pathway

    A few of the first signaling molecules shown to be involved in EMT were the TGFβ signaling factors [168,169,170,171,172]. Studies have shown a number of differentially spliced TGFB isoforms are expressed in the developing endocardial cushions in mice [173]. These isoforms are highly redundant, which could highlight the existence of a number of compensatory mechanisms which may uncover an important role for TGFβ ligands in cardiogenesis. On binding to their corresponding TGFβ type 2 receptors, TGFβ ligands activate them, hereby triggering the transphosphorylation of type 1 receptors within the receptor ligand complex. The phosphorylated type 1 receptor in turn acts as a serine/threonine kinase to phosphorylate and activate the cytosolic smad proteins which act as important intracellular mediators and enhancers of TGFβ signaling. The TGFβ ligands also exert their effects on downstream gene expression and regulation via the non-canonical signaling pathway via ERK, JNK and MEKK proteins (Figure 3). During EMT, the TGFβ ligands are known to signal through the canonical signaling pathway, particularly through SMAD2/3, hereby activating the expression of downstream genes including Snail and Slug. These are two important transcriptional repressors that can tightly modulate EMT in the developing heart valves [174,175,176,177]. Slug/Snail trigger endothelial cell activation, in the endocardial cushions, and invasion. Another important molecule is VE-cadherin. Its expression is reciprocal to that of Slug and Snail. The maintenance of low levels of VE-cadherin is essential for the developing mesenchyme of the heart valves [134]. TGFβ2 acts downstream of Notch, TBX5 and Bmp2 pathways to activate the WNT/β-catenin pathway [30,178,179]. Important for the transduction of signals in the TGFβ pathway are the SMAD proteins. Targeted deletion of SMAD4 in NCCs results in defects of the pharyngeal arch including OFT cushion hypoplasia and patent truncus arteriosus [139]. SMAD4 deficient NCCs exhibit increased apoptosis and reduced presence in OFT along with decreased expression of Bmp4, SEMA3C and PLXNA2; signals that are all required for OFT septation. Another member of the TGFβ family of inhibitory SMADs, SMAD6 is expressed in the endocardial cells of the AVC and OFT. Interestingly, knockdown of Madh6, which encodes for SMAD6 and acts as a repressor of BMP signaling, results in hyperplasia of the atrioventricular canal and OFT cushions leading to hyperplastic valves and reduced endothelial cell mediated vasodilation due to an increase in BMP signaling [180]. In vitro studies have shown BMP signaling may be regulated via a SMAD6 dependent physiological feedback loop and hence might play a role in maintaining the number of endothelial cells that undergo EMT [181]. Knockdown of Erk2/Mapk1, a non-canonical TGFβ signaling factor known to be expressed in the pharyngeal arches, in mice causes cardiovascular defects including double outlet right ventricle, patent truncus arteriosus and ventricular septal defects [182,183].

    Figure 3. Canonical and non-canonical TGFβ signaling pathway including BMPs.

    6.2. BMP pathway

    The TGFβ cytokine superfamily includes the bone morphogenic proteins (BMPs) that signal mainly through the activation of Smad1/5/8. BMPs have a number of roles in OFT septation and valve development. Myocardially expressed Bmp2 activates the expression of hyaluronic acid synthetase 2 (Has2), important for the production of cushion ECM, and other genes (Twist1, Msx2 and Msx1) that are important for EMT [184,185]. In vitro studies on atrioventricular canal endothelial explants from mice, following treatment with Bmp2, indicated an increase in TGFB2 suggesting that Bmp2 triggers an endocardial autocrine TGFβ loop promoting EMT [186]. Bmp4 is strongly expressed in the OFT and is required for septation. Mice deficient in Bmp4 exhibits atrial septal defects, ventricular septal defects and patent truncus arteriosus [187]. BMP6 is expressed in the endocardium, myocardium and the developing valve mesenchyme and shows functional redundancy with BMP7 with a similar pattern of expression. Bmp6–/–/Bmp7–/– double knockout mice exhibit hypocellular OFT cushions with a delay in OFT formation uncovering for the first time the role of BMP signaling in OFT and valve development [188]. Some of the important BMP receptors expressed in the developing heart are Alk2, Alk3 and Bmpr2. Hypomorphic Bmpr2 alleles in mice present with interrupted aortic arch, patent truncus arteriosus and absent semilunar valves [189]. Knockdown of Bmpr2 in endothelial cells results in atrial septal defects, hyperplastic semilunar and atrioventricular valve leaflets and ventricular septal defects while myocardial knockdown or NCC knockdown of Bmpr2 causes double outlet right ventricle or overriding aorta, indicating a cell specific role of Bmpr2 [190]. Conditional deletion of Alk3, a BMP type 1A receptor, resulted in hypoplastic cushions. Further analysis of the endocardial cushions showed a decrease of TGFβ2 and p-Smad1/5/8 suggesting that the hypoplasia observed in the valves might be secondary to TGFβ mediated endocardial cushion EMT [191].


    6.3. Notch pathway

    The Notch signaling pathway (Figure 4) is important for a larger number of developmental processes particularly cell fate determination and differentiation and is highly conserved [192]. As described before, NOTCH signaling has been shown to be involved in BAV and its related aortopathy. Notch signaling is important for EMT. Endocardial and OFT expression of Notch1 is indicative of its role in EMT and valve development. Mutations in NOTCH1 or its effector RBPj-κ results in aberrant EMT. In humans, the NOTCH family comprises 4 type 1 receptors (Notch1-4) and 5 type 1 transmembrane ligands (Jagged 1-2, Delta like (DLL) 1, DLL3 and DLL4) [193]. Ligand binding to the Notch receptors triggers the cleavage of NICD by gamma secretase, followed by the translocation of NICD to the nucleus activating gene expression via binding to RBPj-κ. Initiation of EMT requires the Notch1 induced TGFβ2 activation of Snail1 and Snail2, which in turn represses VE-cadherin disrupting cell-cell contact [134,176]. Endocardial cells exposed to Notch1/TGFβ2 require Bmp2 to trigger EMT. This Bmp2 expression is repressed by the myocardial Notch1 indicating a tissue specific reciprocal effect of myocardial and endocardial Notch1 adding a layer of complexity to the EMT and septation processes involved in early development [179]. In the SHF, Notch1 inhibition has been shown to be associated with a reduction in Fgf8, aberrant EMT in OFT cushions and abnormal migration of NCC consistent with thick, asymmetric semilunar valve leaflets [27,128]. These defects are reversed by the overexpression of Fgf8 indicating that Notch1 acts through Fgf8 to trigger EMT in the OFT [27]. Deletion of Notch in the SHF in mice leads to atrial septal defects, double outlet right ventricle, ventricular septal defects and patent truncus arteriosus [27]. While Notch1 mutations, in humans, are associated with calcified aortic valve disease, mutations in JAG1 or NOTCH2 cause Alagille syndrome comprising heart defects that are recapitulated in mice deficient in Hey2, a downstream target of Notch [194,195,196].

    Figure 4. NOTCH-JAG-DLL signaling pathway. (Referenced from Radtke et al., 2013) [180].

    Given the crosstalk between these large pathways and the multitude of genes and signaling factors involved, dissecting the molecular mechanisms leading to the pathogenesis of BAV associated aortopathy requires gene discovery strategies ranging from candidate gene association and linkage to next generation sequencing.


    7. Future directions and conclusions

    The complexity of BAV, along with the underlying predisposition to secondary cardiac complications coupled with increased morbidity, poses both a research and clinical challenge. While experimental animal models, linkage analysis and more recently next generation sequencing approaches have broadened the existing knowledge of the underlying genetics of BAV associated aortopathy, the exact mechanisms by which these networks of factors interact with each other leading to the development of disease and influencing disease progression remains to be understood. However, data from these large candidate gene identification studies does suggest a complex interplay of genetic factors synergistically involved in OFT and aortic valve development perturbations leading to the development of BAV-associated aortopathy. In addition to current next generation sequencing approaches, identification of rare copy number variants in congenital heart disease may also provide further insight into the genetics and the molecular mechanisms involved in disease pathogenesis. It is important to note that although much of the candidate gene discovery strategies have been applied to mainly familial cases, these constitute only a fraction of the total BAV cases with a large number of cases being isolated or sporadic. Implication of candidate genes in causality requires not only careful functional downstream analysis but also validation in replication cohorts using a case-control approach. Large sample sized case and control cohorts are essential in order to account for low frequency disease causing variants especially given the prevalence and heterogeneity of BAV, both phenotypically and genetically. Furthermore, loci identified in human cohorts can be tested in animal models to determine their causative link to disease. BAV is a risk factor for early onset of serious cardiovascular complications however, the molecular basis and tools (such as biomarkers) to monitor disease progression remain largely undefined. Moving forward, the results of these candidate gene approaches and biomarker studies will be pertinent in influencing clinical practice via translation into better diagnosis, follow up and early intervention of secondary complications in individuals with family history of valve disease.


    Acknowledgements

    The authors are supported by funding from the Fund for Scientific Research, Flanders (FWO, Belgium, G.0221.12), the Fondation Leducq (MIBAVA – Leducq 12CVD03). BL is senior clinical investigator of the Fund for Scientific Research, Flanders and holds a starting grant from the European Research Council (ERC-StG-2012-30972-BRAVE).


    Conflict of Interest

    The authors declare no conflict of interest.


    [1] Kelly RG (2012) The second heart field. Curr Top Dev Biol 100: 33-65. doi: 10.1016/B978-0-12-387786-4.00002-6
    [2] Buckingham M, Meilhac S, Zaffran S (2005) Building the mammalian heart from two sources of myocardial cells. Nat Rev Genet 6: 826-835. doi: 10.1038/nrg1710
    [3] Kelly RG, Buckingham ME, Moorman AF (2014) Heart fields and cardiac morphogenesis. Cold Spring Harb Perspect Med 4.
    [4] Yang YP, Li HR, Cao XM, et al. (2013) Second heart field and the development of the outflow tract in human embryonic heart. Dev Growth Differ 55: 359-367. doi: 10.1111/dgd.12050
    [5] Zaffran S, Frasch M (2002) Early signals in cardiac development. Circ Res 91: 457-469. doi: 10.1161/01.RES.0000034152.74523.A8
    [6] Srivastava D (2006) Making or breaking the heart: from lineage determination to morphogenesis. Cell 126: 1037-1048. doi: 10.1016/j.cell.2006.09.003
    [7] Graham A (2003) The neural crest. Curr Biol 13: R381-384. doi: 10.1016/S0960-9822(03)00315-4
    [8] van den Hoff MJ, Moorman AF (2000) Cardiac neural crest: the holy grail of cardiac abnormalities? Cardiovasc Res 47: 212-216. doi: 10.1016/S0008-6363(00)00127-9
    [9] Kirby ML, Hutson MR (2010) Factors controlling cardiac neural crest cell migration. Cell Adh Migr 4: 609-621. doi: 10.4161/cam.4.4.13489
    [10] Kirby ML, Waldo KL (1995) Neural crest and cardiovascular patterning. Circ Res 77: 211-215. doi: 10.1161/01.RES.77.2.211
    [11] Hutson MR, Kirby ML (2003) Neural crest and cardiovascular development: a 20-year perspective. Birth Defects Res C Embryo Today 69: 2-13. doi: 10.1002/bdrc.10002
    [12] Combs MD, Yutzey KE (2009) Heart valve development: regulatory networks in development and disease. Circ Res 105: 408-421. doi: 10.1161/CIRCRESAHA.109.201566
    [13] Lin CJ, Lin CY, Chen CH, et al. (2012) Partitioning the heart: mechanisms of cardiac septation and valve development. Development 139: 3277-3299. doi: 10.1242/dev.063495
    [14] Feiner L, Webber AL, Brown CB, et al. (2001) Targeted disruption of semaphorin 3C leads to persistent truncus arteriosus and aortic arch interruption. Development 128: 3061-3070.
    [15] Brown CB, Feiner L, Lu MM, et al. (2001) PlexinA2 and semaphorin signaling during cardiac neural crest development. Development 128: 3071-3080.
    [16] Toyofuku T, Yoshida J, Sugimoto T, et al. (2008) Repulsive and attractive semaphorins cooperate to direct the navigation of cardiac neural crest cells. Dev Biol 321: 251-262. doi: 10.1016/j.ydbio.2008.06.028
    [17] Verzi MP, McCulley DJ, De Val S, et al. (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. doi: 10.1016/j.ydbio.2005.08.041
    [18] Clouthier DE, Hosoda K, Richardson JA, et al. (1998) Cranial and cardiac neural crest defects in endothelin-A receptor-deficient mice. Development 125: 813-824.
    [19] Koenig SN, Bosse KM, Nadorlik HA, et al. (2015) Evidence of Aortopathy in Mice with Haploinsufficiency of in -Null Background. J Cardiovasc Dev Dis 2: 17-30. doi: 10.3390/jcdd2010017
    [20] Feng Q, Song W, Lu X, et al. (2002) Development of heart failure and congenital septal defects in mice lacking endothelial nitric oxide synthase. Circulation 106: 873-879. doi: 10.1161/01.CIR.0000024114.82981.EA
    [21] Mommersteeg MT, Yeh ML, Parnavelas JG, et al. (2015) Disrupted Slit-Robo signalling results in membranous ventricular septum defects and bicuspid aortic valves. Cardiovasc Res 106: 55-66. doi: 10.1093/cvr/cvv040
    [22] Li P, Pashmforoush M, Sucov HM (2010) Retinoic acid regulates differentiation of the secondary heart field and TGFbeta-mediated outflow tract septation. Dev Cell 18: 480-485. doi: 10.1016/j.devcel.2009.12.019
    [23] Gruber PJ, Kubalak SW, Pexieder T, et al. (1996) RXR alpha deficiency confers genetic susceptibility for aortic sac, conotruncal, atrioventricular cushion, and ventricular muscle defects in mice. J Clin Invest 98: 1332-1343. doi: 10.1172/JCI118920
    [24] Zhou B, Wu B, Tompkins KL, et al. (2005) Characterization of Nfatc1 regulation identifies an enhancer required for gene expression that is specific to pro-valve endocardial cells in the developing heart. Development 132: 1137-1146. doi: 10.1242/dev.01640
    [25] Bruneau BG (2013) Signaling and transcriptional networks in heart development and regeneration. Cold Spring Harb Perspect Biol 5: a008292.
    [26] Lin CY, Lin CJ, Chen CH, et al. (2012) The secondary heart field is a new site of calcineurin/Nfatc1 signaling for semilunar valve development. J Mol Cell Cardiol 52: 1096-1102. doi: 10.1016/j.yjmcc.2012.01.013
    [27] High FA, Jain R, Stoller JZ, et al. (2009) Murine Jagged1/Notch signaling in the second heart field orchestrates Fgf8 expression and tissue-tissue interactions during outflow tract development. J Clin Invest 119: 1986-1996.
    [28] High FA, Zhang M, Proweller A, et al. (2007) An essential role for Notch in neural crest during cardiovascular development and smooth muscle differentiation. J Clin Invest 117: 353-363. doi: 10.1172/JCI30070
    [29] Chakraborty S, Combs MD, Yutzey KE (2010) Transcriptional regulation of heart valve progenitor cells. Pediatr Cardiol 31: 414-421. doi: 10.1007/s00246-009-9616-x
    [30] Shirai M, Imanaka-Yoshida K, Schneider MD, et al. (2009) T-box 2, a mediator of Bmp-Smad signaling, induced hyaluronan synthase2 and Tgfbeta2 expression and endocardial cushion formation. Proc Natl Acad Sci USA 106: 18604-18609. doi: 10.1073/pnas.0900635106
    [31] Chang CP, Neilson JR, Bayle JH, et al. (2004) A field of myocardial-endocardial NFAT signaling underlies heart valve morphogenesis. Cell 118: 649-663. doi: 10.1016/j.cell.2004.08.010
    [32] Schaefer BM, Lewin MB, Stout KK, et al. (2008) The bicuspid aortic valve: an integrated phenotypic classification of leaflet morphology and aortic root shape. Heart 94: 1634-1638. doi: 10.1136/hrt.2007.132092
    [33] Sievers HH, Schmidtke C (2007) A classification system for the bicuspid aortic valve from 304 surgical specimens. J Thorac Cardiovasc Surg 133: 1226-1233. doi: 10.1016/j.jtcvs.2007.01.039
    [34] Carro A, Teixido-Tura G, Evangelista A (2012) Aortic dilatation in bicuspid aortic valve disease. Rev Esp Cardiol 65: 977-981. doi: 10.1016/j.recesp.2012.04.025
    [35] Girdauskas E, Borger MA, Secknus MA, et al. (2011) Is aortopathy in bicuspid aortic valve disease a congenital defect or a result of abnormal hemodynamics? A critical reappraisal of a one-sided argument. Eur J Cardiothorac Surg 39: 809-814.
    [36] Jiao J, Xiong W, Wang L, et al. (2016) Differentiation defect in neural crest-derived smooth muscle cells in patients with aortopathy associated with bicuspid aortic valves. EBioMedicine 10: 282-290. doi: 10.1016/j.ebiom.2016.06.045
    [37] Loeys BL, Chen J, Neptune ER, et al. (2005) A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in TGFBR1 or TGFBR2. Nat Genet 37: 275-281. doi: 10.1038/ng1511
    [38] Kappetein AP, Gittenberger-de Groot AC, Zwinderman AH, et al. (1991) The neural crest as a possible pathogenetic factor in coarctation of the aorta and bicuspid aortic valve. J Thorac Cardiovasc Surg 102: 830-836.
    [39] Martinez-Vargas J, Ventura J, Machuca A, et al. (2017) Cardiac, mandibular and thymic phenotypical association indicates that cranial neural crest underlies bicuspid aortic valve formation in hamsters. PLoS One 12: e0183556. doi: 10.1371/journal.pone.0183556
    [40] Van Hemelrijk C, Renard M, Loeys B (2010) The Loeys-Dietz syndrome: an update for the clinician. Curr Opin Cardiol 25: 546-551. doi: 10.1097/HCO.0b013e32833f0220
    [41] van de Laar IM, van der Linde D, Oei EH, et al. (2012) Phenotypic spectrum of the SMAD3- related aneurysms-osteoarthritis syndrome. J Med Genet 49: 47-57. doi: 10.1136/jmedgenet-2011-100382
    [42] Michelena HI, Khanna AD, Mahoney D, et al. (2011) Incidence of aortic complications in patients with bicuspid aortic valves. JAMA 306: 1104-1112. doi: 10.1001/jama.2011.1286
    [43] Cripe L, Andelfinger G, Martin LJ, et al. (2004) Bicuspid aortic valve is heritable. J Am Coll Cardiol 44: 138-143. doi: 10.1016/j.jacc.2004.03.050
    [44] Kang JW, Song HG, Yang DH, et al. (2013) Association between bicuspid aortic valve phenotype and patterns of valvular dysfunction and bicuspid aortopathy: comprehensive evaluation using MDCT and echocardiography. JACC Cardiovasc Imaging 6: 150-161. doi: 10.1016/j.jcmg.2012.11.007
    [45] Sans-Coma V, Fernandez B, Duran AC, et al. (1996) Fusion of valve cushions as a key factor in the formation of congenital bicuspid aortic valves in Syrian hamsters. Anat Rec 244: 490-498. doi: 10.1002/(SICI)1097-0185(199604)244:4<490::AID-AR7>3.0.CO;2-Z
    [46] Fernandez B, Duran AC, Fernandez-Gallego T, et al. (2009) Bicuspid aortic valves with different spatial orientations of the leaflets are distinct etiological entities. J Am Coll Cardiol 54: 2312-2318. doi: 10.1016/j.jacc.2009.07.044
    [47] Mahadevia R, Barker AJ, Schnell S, et al. (2014) Bicuspid aortic cusp fusion morphology alters aortic three-dimensional outflow patterns, wall shear stress, and expression of aortopathy. Circulation 129: 673-682. doi: 10.1161/CIRCULATIONAHA.113.003026
    [48] Girdauskas E, Disha K, Borger MA, et al. (2012) Relation of bicuspid aortic valve morphology to the dilatation pattern of the proximal aorta: focus on the transvalvular flow. Cardiol Res Pract 2012: 478259.
    [49] Robicsek F, Thubrikar MJ, Cook JW, et al. (2004) The congenitally bicuspid aortic valve: how does it function? Why does it fail? Ann Thorac Surg 77: 177-185.
    [50] Della Corte A, Bancone C, Conti CA, et al. (2012) Restricted cusp motion in right-left type of bicuspid aortic valves: a new risk marker for aortopathy. J Thorac Cardiovasc Surg 144: 360-369. doi: 10.1016/j.jtcvs.2011.10.014
    [51] Hope MD, Hope TA, Crook SE, et al. (2011) 4D flow CMR in assessment of valve-related ascending aortic disease. JACC Cardiovasc Imaging 4: 781-787. doi: 10.1016/j.jcmg.2011.05.004
    [52] Hope MD, Wrenn J, Sigovan M, et al. (2012) Imaging biomarkers of aortic disease: increased growth rates with eccentric systolic flow. J Am Coll Cardiol 60: 356-357. doi: 10.1016/j.jacc.2012.01.072
    [53] Ikonomidis JS, Ruddy JM, Benton SM, et al. (2012) Aortic dilatation with bicuspid aortic valves: cusp fusion correlates to matrix metalloproteinases and inhibitors. Ann Thorac Surg 93: 457-463. doi: 10.1016/j.athoracsur.2011.09.057
    [54] Russo CF, Cannata A, Lanfranconi M, et al. (2008) Is aortic wall degeneration related to bicuspid aortic valve anatomy in patients with valvular disease? J Thorac Cardiovasc Surg 136: 937-942. doi: 10.1016/j.jtcvs.2007.11.072
    [55] Fazel SS, Mallidi HR, Lee RS, et al. (2008) The aortopathy of bicuspid aortic valve disease has distinctive patterns and usually involves the transverse aortic arch. J Thorac Cardiovasc Surg 135: 901-907. doi: 10.1016/j.jtcvs.2008.01.022
    [56] McNally A, Madan A, Sucosky P (2017) Morphotype-Dependent Flow Characteristics in Bicuspid Aortic Valve Ascending Aortas: A Benchtop Particle Image Velocimetry Study. Front Physiol 8: 44.
    [57] Dolan JM, Meng H, Singh S, et al. (2011) High fluid shear stress and spatial shear stress gradients affect endothelial proliferation, survival, and alignment. Ann Biomed Eng 39: 1620- 1631. doi: 10.1007/s10439-011-0267-8
    [58] Rabkin SW (2014) Differential expression of MMP-2, MMP-9 and TIMP proteins in thoracic aortic aneurysm - comparison with and without bicuspid aortic valve: a meta-analysis. Vasa 43: 433-442. doi: 10.1024/0301-1526/a000390
    [59] Ikonomidis JS, Ivey CR, Wheeler JB, et al. (2013) Plasma biomarkers for distinguishing etiologic subtypes of thoracic aortic aneurysm disease. J Thorac Cardiovasc Surg 145: 1326- 1333. doi: 10.1016/j.jtcvs.2012.12.027
    [60] Mohamed SA, Radtke A, Saraei R, et al. (2012) Locally different endothelial nitric oxide synthase protein levels in ascending aortic aneurysms of bicuspid and tricuspid aortic valve. Cardiol Res Pract 2012: 165957.
    [61] Grewal N, DeRuiter MC, Jongbloed MR, et al. (2014) Normal and abnormal development of the aortic wall and valve: correlation with clinical entities. Neth Heart J 22: 363-369. doi: 10.1007/s12471-014-0576-2
    [62] Aicher D, Urbich C, Zeiher A, et al. (2007) Endothelial nitric oxide synthase in bicuspid aortic valve disease. Ann Thorac Surg 83: 1290-1294. doi: 10.1016/j.athoracsur.2006.11.086
    [63] Grewal N, Gittenberger-de Groot AC, DeRuiter MC, et al. (2014) Bicuspid aortic valve: phosphorylation of c-Kit and downstream targets are prognostic for future aortopathy. Eur J Cardiothorac Surg 46: 831-839. doi: 10.1093/ejcts/ezu319
    [64] Gurvitz M, Chang RK, Drant S, et al. (2004) Frequency of aortic root dilation in children with a bicuspid aortic valve. Am J Cardiol 94: 1337-1340. doi: 10.1016/j.amjcard.2004.07.130
    [65] Fernandes S, Khairy P, Graham DA, et al. (2012) Bicuspid aortic valve and associated aortic dilation in the young. Heart 98: 1014-1019. doi: 10.1136/heartjnl-2012-301773
    [66] Yasuda H, Nakatani S, Stugaard M, et al. (2003) Failure to prevent progressive dilation of ascending aorta by aortic valve replacement in patients with bicuspid aortic valve: comparison with tricuspid aortic valve. Circulation 108 Suppl 1: II291-294.
    [67] Silberbach M (2009) Bicuspid aortic valve and thoracic aortic aneurysm: toward a unified theory. J Am Coll Cardiol 53: 2296-2297. doi: 10.1016/j.jacc.2009.03.028
    [68] Bunton TE, Biery NJ, Myers L, et al. (2001) Phenotypic alteration of vascular smooth muscle cells precedes elastolysis in a mouse model of Marfan syndrome. Circ Res 88: 37-43. doi: 10.1161/01.RES.88.1.37
    [69] Tadros TM, Klein MD, Shapira OM (2009) Ascending aortic dilatation associated with bicuspid aortic valve: pathophysiology, molecular biology, and clinical implications. Circulation 119: 880-890. doi: 10.1161/CIRCULATIONAHA.108.795401
    [70] Bonderman D, Gharehbaghi-Schnell E, Wollenek G, et al. (1999) Mechanisms underlying aortic dilatation in congenital aortic valve malformation. Circulation 99: 2138-2143. doi: 10.1161/01.CIR.99.16.2138
    [71] Nistri S, Grande-Allen J, Noale M, et al. (2008) Aortic elasticity and size in bicuspid aortic valve syndrome. Eur Heart J 29: 472-479. doi: 10.1093/eurheartj/ehm528
    [72] Fedak PW, de Sa MP, Verma S, et al. (2003) Vascular matrix remodeling in patients with bicuspid aortic valve malformations: implications for aortic dilatation. J Thorac Cardiovasc Surg 126: 797-806. doi: 10.1016/S0022-5223(03)00398-2
    [73] Nataatmadja M, West M, West J, et al. (2003) Abnormal extracellular matrix protein transport associated with increased apoptosis of vascular smooth muscle cells in marfan syndrome and bicuspid aortic valve thoracic aortic aneurysm. Circulation 108 Suppl 1: II329-334.
    [74] Grewal N, Franken R, Mulder BJ, et al. (2016) Histopathology of aortic complications in bicuspid aortic valve versus Marfan syndrome: relevance for therapy? Heart Vessels 31: 795- 806. doi: 10.1007/s00380-015-0703-z
    [75] Leme MP, David TE, Butany J, et al. (2003) Molecular evaluation of the great vessels of patients with bicuspid aortic valve disease. Braz J Cardiovasc Surg 18: 148-156.
    [76] Rueda-Martinez C, Lamas O, Mataro MJ, et al. (2017) Fibrillin 2 is upregulated in the ascending aorta of patients with bicuspid aortic valve. Eur J Cardiothorac Surg 51: 104. doi: 10.1093/ejcts/ezw277
    [77] Siu SC, Silversides CK (2010) Bicuspid aortic valve disease. J Am Coll Cardiol 55: 2789-2800. doi: 10.1016/j.jacc.2009.12.068
    [78] Ward C (2000) Clinical significance of the bicuspid aortic valve. Heart 83: 81-85. doi: 10.1136/heart.83.1.81
    [79] Vallely MP, Semsarian C, Bannon PG (2008) Management of the ascending aorta in patients with bicuspid aortic valve disease. Heart Lung Circ 17: 357-363. doi: 10.1016/j.hlc.2008.01.007
    [80] Martin LJ, Ramachandran V, Cripe LH, et al. (2007) Evidence in favor of linkage to human chromosomal regions 18q, 5q and 13q for bicuspid aortic valve and associated cardiovascular malformations. Hum Genet 121: 275-284. doi: 10.1007/s00439-006-0316-9
    [81] Andelfinger G, Tapper AR, Welch RC, et al. (2002) KCNJ2 mutation results in Andersen syndrome with sex-specific cardiac and skeletal muscle phenotypes. Am J Hum Genet 71: 663-668. doi: 10.1086/342360
    [82] Garg V, Muth AN, Ransom JF, et al. (2005) Mutations in NOTCH1 cause aortic valve disease. Nature 437: 270-274. doi: 10.1038/nature03940
    [83] Mohamed SA, Aherrahrou Z, Liptau H, et al. (2006) Novel missense mutations (p.T596M and p.P1797H) in NOTCH1 in patients with bicuspid aortic valve. Biochem Biophys Res Commun 345: 1460-1465.
    [84] Acharya A, Hans CP, Koenig SN, et al. (2011) Inhibitory role of Notch1 in calcific aortic valve disease. PLoS One 6: e27743. doi: 10.1371/journal.pone.0027743
    [85] Quintero-Rivera F, Xi QJ, Keppler-Noreuil KM, et al. (2015) MATR3 disruption in human and mouse associated with bicuspid aortic valve, aortic coarctation and patent ductus arteriosus. Hum Mol Genet 24: 2375-2389. doi: 10.1093/hmg/ddv004
    [86] Moskowitz IP, Wang J, Peterson MA, et al. (2011) Transcription factor genes Smad4 and Gata4 cooperatively regulate cardiac valve development. Proc Natl Acad Sci USA 108: 4006-4011. doi: 10.1073/pnas.1019025108
    [87] Okubo A, Miyoshi O, Baba K, et al. (2004) A novel GATA4 mutation completely segregated with atrial septal defect in a large Japanese family. J Med Genet 41: e97. doi: 10.1136/jmg.2004.018895
    [88] Garg V, Kathiriya IS, Barnes R, et al. (2003) GATA4 mutations cause human congenital heart defects and reveal an interaction with TBX5. Nature 424: 443-447. doi: 10.1038/nature01827
    [89] Maitra M, Schluterman MK, Nichols HA, et al. (2009) Interaction of Gata4 and Gata6 with Tbx5 is critical for normal cardiac development. Dev Biol 326: 368-377. doi: 10.1016/j.ydbio.2008.11.004
    [90] Shi LM, Tao JW, Qiu XB, et al. (2014) GATA5 loss-of-function mutations associated with congenital bicuspid aortic valve. Int J Mol Med 33: 1219-1226. doi: 10.3892/ijmm.2014.1700
    [91] Bonachea EM, Chang SW, Zender G, et al. (2014) Rare GATA5 sequence variants identified in individuals with bicuspid aortic valve. Pediatr Res 76: 211-216. doi: 10.1038/pr.2014.67
    [92] Padang R, Bagnall RD, Richmond DR, et al. (2012) Rare non-synonymous variations in the transcriptional activation domains of GATA5 in bicuspid aortic valve disease. J Mol Cell Cardiol 53: 277-281 doi: 10.1016/j.yjmcc.2012.05.009
    [93] Qu XK, Qiu XB, Yuan F, et al. (2014) A novel NKX2.5 loss-of-function mutation associated with congenital bicuspid aortic valve. Am J Cardiol 114: 1891-1895.
    [94] Ponticos M, Partridge T, Black CM, et al. (2004) Regulation of collagen type I in vascular smooth muscle cells by competition between Nkx2.5 and deltaEF1/ZEB1. Mol Cell Biol 24: 6151-6161. doi: 10.1128/MCB.24.14.6151-6161.2004
    [95] Lin X, Huo Z, Liu X, et al. (2010) A novel GATA6 mutation in patients with tetralogy of Fallot or atrial septal defect. J Hum Genet 55: 662-667. doi: 10.1038/jhg.2010.84
    [96] Lepore JJ, Mericko PA, Cheng L, et al. (2006) GATA-6 regulates semaphorin 3C and is required in cardiac neural crest for cardiovascular morphogenesis. J Clin Invest 116: 929-939. doi: 10.1172/JCI27363
    [97] Kodo K, Nishizawa T, Furutani M, et al. (2009) GATA6 mutations cause human cardiac outflow tract defects by disrupting semaphorin-plexin signaling. Proc Natl Acad Sci USA 106: 13933-13938. doi: 10.1073/pnas.0904744106
    [98] Pepe G, Nistri S, Giusti B, et al. (2014) Identification of fibrillin 1 gene mutations in patients with bicuspid aortic valve (BAV) without Marfan syndrome. BMC Med Genet 15: 23.
    [99] Pereira L, Andrikopoulos K, Tian J, et al. (1997) Targetting of the gene encoding fibrillin-1 recapitulates the vascular aspect of Marfan syndrome. Nat Genet 17: 218-222. doi: 10.1038/ng1097-218
    [100] Loeys BL, Schwarze U, Holm T, et al. (2006) Aneurysm syndromes caused by mutations in the TGF-beta receptor. N Engl J Med 355: 788-798. doi: 10.1056/NEJMoa055695
    [101] van de Laar IM, Oldenburg RA, Pals G, et al. (2011) Mutations in SMAD3 cause a syndromic form of aortic aneurysms and dissections with early-onset osteoarthritis. Nat Genet 43: 121-126. doi: 10.1038/ng.744
    [102] Bertoli-Avella AM, Gillis E, Morisaki H, et al. (2015) Mutations in a TGF-beta ligand, TGFB3, cause syndromic aortic aneurysms and dissections. J Am Coll Cardiol 65: 1324-1336. doi: 10.1016/j.jacc.2015.01.040
    [103] Lindsay ME, Schepers D, Bolar NA, et al. (2012) Loss-of-function mutations in TGFB2 cause a syndromic presentation of thoracic aortic aneurysm. Nat Genet 44: 922-927. doi: 10.1038/ng.2349
    [104] Girdauskas E, Schulz S, Borger MA, et al. (2011) Transforming growth factor-beta receptor type II mutation in a patient with bicuspid aortic valve disease and intraoperative aortic dissection. Ann Thorac Surg 91: e70-71. doi: 10.1016/j.athoracsur.2010.12.060
    [105] Folkersen L, Wagsater D, Paloschi V, et al. (2011) Unraveling divergent gene expression profiles in bicuspid and tricuspid aortic valve patients with thoracic aortic dilatation: the ASAP study. Mol Med 17: 1365-1373.
    [106] Tan HL, Glen E, Topf A, et al. (2012) Nonsynonymous variants in the SMAD6 gene predispose to congenital cardiovascular malformation. Hum Mutat 33: 720-727. doi: 10.1002/humu.22030
    [107] Gillis E, Kumar AA, Luyckx I, et al. (2017) Candidate Gene Resequencing in a Large Bicuspid Aortic Valve-Associated Thoracic Aortic Aneurysm Cohort: SMAD6 as an Important Contributor. Frontiers in Physiology 8: 400. doi: 10.3389/fphys.2017.00400
    [108] Preuss C, Capredon M, Wunnemann F, et al. (2016) Family Based Whole Exome Sequencing Reveals the Multifaceted Role of Notch Signaling in Congenital Heart Disease. PLoS Genet 12: e1006335. doi: 10.1371/journal.pgen.1006335
    [109] Guo DC, Pannu H, Tran-Fadulu V, et al. (2007) Mutations in smooth muscle alpha-actin (ACTA2) lead to thoracic aortic aneurysms and dissections. Nat Genet 39: 1488-1493. doi: 10.1038/ng.2007.6
    [110] Nigam V, Sievers HH, Jensen BC, et al. (2010) Altered microRNAs in bicuspid aortic valve: a comparison between stenotic and insufficient valves. J Heart Valve Dis 19: 459-465.
    [111] Yanagawa B, Lovren F, Pan Y, et al. (2012) miRNA-141 is a novel regulator of BMP-2-mediated calcification in aortic stenosis. J Thorac Cardiovasc Surg 144: 256-262. doi: 10.1016/j.jtcvs.2011.10.097
    [112] McKellar SH, Tester DJ, Yagubyan M, et al. (2007) Novel NOTCH1 mutations in patients with bicuspid aortic valve disease and thoracic aortic aneurysms. J Thorac Cardiovasc Surg 134: 290-296. doi: 10.1016/j.jtcvs.2007.02.041
    [113] Foffa I, Ait Ali L, Panesi P, et al. (2013) Sequencing of NOTCH1, GATA5, TGFBR1 and TGFBR2 genes in familial cases of bicuspid aortic valve. BMC Med Genet 14: 44.
    [114] Kerstjens-Frederikse WS, van de Laar IM, Vos YJ, et al. (2016) Cardiovascular malformations caused by NOTCH1 mutations do not keep left: data on 428 probands with left-sided CHD and their families. Genet Med 18: 914-923. doi: 10.1038/gim.2015.193
    [115] Yang B, Zhou W, Jiao J, et al. (2017) Protein-altering and regulatory genetic variants near GATA4 implicated in bicuspid aortic valve. Nat Commun 8: 15481. doi: 10.1038/ncomms15481
    [116] Misra C, Sachan N, McNally CR, et al. (2012) Congenital heart disease-causing Gata4 mutation displays functional deficits in vivo. PLoS Genet 8: e1002690. doi: 10.1371/journal.pgen.1002690
    [117] Schott JJ, Benson DW, Basson CT, et al. (1998) Congenital heart disease caused by mutations in the transcription factor NKX2-5. Science 281: 108-111.
    [118] Reamon-Buettner SM, Hecker H, Spanel-Borowski K, et al. (2004) Novel NKX2-5 mutations in diseased heart tissues of patients with cardiac malformations. Am J Pathol 164: 2117-2125. doi: 10.1016/S0002-9440(10)63770-4
    [119] De Luca A, Sarkozy A, Consoli F, et al. (2010) Familial transposition of the great arteries caused by multiple mutations in laterality genes. Heart 96: 673-677. doi: 10.1136/hrt.2009.181685
    [120] Draus JM, Hauck MA, Goetsch M, et al. (2009) Investigation of somatic NKX2-5 mutations in congenital heart disease. J Med Genet 46: 115-122.
    [121] Wooten EC, Iyer LK, Montefusco MC, et al. (2010) Application of gene network analysis techniques identifies AXIN1/PDIA2 and endoglin haplotypes associated with bicuspid aortic valve. PLoS One 5: e8830. doi: 10.1371/journal.pone.0008830
    [122] Martin M, Lorca R, Rozado J, et al. (2017) Bicuspid aortic valve syndrome: a multidisciplinary approach for a complex entity. J Thorac Dis 9: S454-S464. doi: 10.21037/jtd.2017.05.11
    [123] Guo DC, Gong L, Regalado ES, et al. (2015) MAT2A mutations predispose individuals to thoracic aortic aneurysms. Am J Hum Genet 96: 170-177. doi: 10.1016/j.ajhg.2014.11.015
    [124] Mohamed SA, Hanke T, Schlueter C, et al. (2005) Ubiquitin fusion degradation 1-like gene dysregulation in bicuspid aortic valve. J Thorac Cardiovasc Surg 130: 1531-1536. doi: 10.1016/j.jtcvs.2005.08.017
    [125] Dargis N, Lamontagne M, Gaudreault N, et al. (2016) Identification of Gender-Specific Genetic Variants in Patients With Bicuspid Aortic Valve. Am J Cardiol 117: 420-426. doi: 10.1016/j.amjcard.2015.10.058
    [126] Malfait F, Symoens S, Coucke P, et al. (2006) Total absence of the alpha2(I) chain of collagen type I causes a rare form of Ehlers-Danlos syndrome with hypermobility and propensity to cardiac valvular problems. J Med Genet 43: e36.
    [127] Tischfield MA, Bosley TM, Salih MA, et al. (2005) Homozygous HOXA1 mutations disrupt human brainstem, inner ear, cardiovascular and cognitive development. Nat Genet 37: 1035-1037. doi: 10.1038/ng1636
    [128] Jain R, Engleka KA, Rentschler SL, et al. (2011) Cardiac neural crest orchestrates remodeling and functional maturation of mouse semilunar valves. J Clin Invest 121: 422-430. doi: 10.1172/JCI44244
    [129] Garg V (2006) Molecular genetics of aortic valve disease. Curr Opin Cardiol 21: 180-184. doi: 10.1097/01.hco.0000221578.18254.70
    [130] Macatee TL, Hammond BP, Arenkiel BR, et al. (2003) Ablation of specific expression domains reveals discrete functions of ectoderm- and endoderm-derived FGF8 during cardiovascular and pharyngeal development. Development 130: 6361-6374. doi: 10.1242/dev.00850
    [131] Laforest B, Andelfinger G, Nemer M (2011) Loss of Gata5 in mice leads to bicuspid aortic valve. J Clin Invest 121: 2876-2887. doi: 10.1172/JCI44555
    [132] Lee TC, Zhao YD, Courtman DW, et al. (2000) Abnormal aortic valve development in mice lacking endothelial nitric oxide synthase. Circulation 101: 2345-2348. doi: 10.1161/01.CIR.101.20.2345
    [133] Laforest B, Nemer M (2011) GATA5 interacts with GATA4 and GATA6 in outflow tract development. Dev Biol 358: 368-378. doi: 10.1016/j.ydbio.2011.07.037
    [134] Timmerman LA, Grego-Bessa J, Raya A, et al. (2004) Notch promotes epithelial-mesenchymal transition during cardiac development and oncogenic transformation. Genes Dev 18: 99-115. doi: 10.1101/gad.276304
    [135] Biben C, Weber R, Kesteven S, et al. (2000) Cardiac septal and valvular dysmorphogenesis in mice heterozygous for mutations in the homeobox gene Nkx2-5. Circ Res 87: 888-895. doi: 10.1161/01.RES.87.10.888
    [136] Kaartinen V, Dudas M, Nagy A, et al. (2004) Cardiac outflow tract defects in mice lacking ALK2 in neural crest cells. Development 131: 3481-3490. doi: 10.1242/dev.01214
    [137] Thomas PS, Sridurongrit S, Ruiz-Lozano P, et al. (2012) Deficient signaling via Alk2 (Acvr1) leads to bicuspid aortic valve development. PLoS One 7: e35539. doi: 10.1371/journal.pone.0035539
    [138] Stottmann RW, Choi M, Mishina Y, et al. (2004) BMP receptor IA is required in mammalian neural crest cells for development of the cardiac outflow tract and ventricular myocardium. Development 131: 2205-2218. doi: 10.1242/dev.01086
    [139] Jia Q, McDill BW, Li SZ, et al. (2007) Smad signaling in the neural crest regulates cardiac outflow tract remodeling through cell autonomous and non-cell autonomous effects. Dev Biol 311: 172-184. doi: 10.1016/j.ydbio.2007.08.044
    [140] Cai X, Zhang W, Hu J, et al. (2013) Tbx20 acts upstream of Wnt signaling to regulate endocardial cushion formation and valve remodeling during mouse cardiogenesis. Development 140: 3176-3187. doi: 10.1242/dev.092502
    [141] Takeuchi JK, Mileikovskaia M, Koshiba-Takeuchi K, et al. (2005) Tbx20 dose-dependently regulates transcription factor networks required for mouse heart and motoneuron development. Development 132: 2463-2474. doi: 10.1242/dev.01827
    [142] Clouthier DE, Williams SC, Hammer RE, et al. (2003) Cell-autonomous and nonautonomous actions of endothelin-A receptor signaling in craniofacial and cardiovascular development. Dev Biol 261: 506-519. doi: 10.1016/S0012-1606(03)00128-3
    [143] Kurihara Y, Kurihara H, Oda H, et al. (1995) Aortic arch malformations and ventricular septal defect in mice deficient in endothelin-1. J Clin Invest 96: 293-300. doi: 10.1172/JCI118033
    [144] Yanagisawa H, Hammer RE, Richardson JA, et al. (2000) Disruption of ECE-1 and ECE-2 reveals a role for endothelin-converting enzyme-2 in murine cardiac development. J Clin Invest 105: 1373-1382. doi: 10.1172/JCI7447
    [145] Yanagisawa H, Yanagisawa M, Kapur RP, et al. (1998) Dual genetic pathways of endothelin- mediated intercellular signaling revealed by targeted disruption of endothelin converting enzyme-1 gene. Development 125: 825-836.
    [146] Gitler AD, Zhu Y, Ismat FA, et al. (2003) Nf1 has an essential role in endothelial cells. Nat Genet 33: 75-79.
    [147] Ranger AM, Grusby MJ, Hodge MR, et al. (1998) The transcription factor NF-ATc is essential for cardiac valve formation. Nature 392: 186-190. doi: 10.1038/32426
    [148] de la Pompa JL, Timmerman LA, Takimoto H, et al. (1998) Role of the NF-ATc transcription factor in morphogenesis of cardiac valves and septum. Nature 392: 182-186. doi: 10.1038/32419
    [149] Todorovic V, Frendewey D, Gutstein DE, et al. (2007) Long form of latent TGF-beta binding protein 1 (Ltbp1L) is essential for cardiac outflow tract septation and remodeling. Development 134: 3723-3732. doi: 10.1242/dev.008599
    [150] Todorovic V, Finnegan E, Freyer L, et al. (2011) Long form of latent TGF-beta binding protein 1 (Ltbp1L) regulates cardiac valve development. Dev Dyn 240: 176-187. doi: 10.1002/dvdy.22521
    [151] Escot S, Blavet C, Hartle S, et al. (2013) Misregulation of SDF1-CXCR4 signaling impairs early cardiac neural crest cell migration leading to conotruncal defects. Circ Res 113: 505-516. doi: 10.1161/CIRCRESAHA.113.301333
    [152] Yu S, Crawford D, Tsuchihashi T, et al. (2011) The chemokine receptor CXCR7 functions to 930 regulate cardiac valve remodeling. Dev Dyn 240: 384-393. doi: 10.1002/dvdy.22549
    [153] Thomas PS, Kim J, Nunez S, et al. (2010) Neural crest cell-specific deletion of Rac1 results in defective cell-matrix interactions and severe craniofacial and cardiovascular malformations. Dev Biol 340: 613-625. doi: 10.1016/j.ydbio.2010.02.021
    [154] Shirai M, Osugi T, Koga H, et al. (2002) The Polycomb-group gene Rae28 sustains Nkx2.5/Csx expression and is essential for cardiac morphogenesis. J Clin Invest 110: 177-184.
    [155] Takihara Y, Tomotsune D, Shirai M, et al. (1997) Targeted disruption of the mouse homologue of the Drosophila polyhomeotic gene leads to altered anteroposterior patterning and neural crest defects. Development 124: 3673-3682.
    [156] Gao Z, Kim GH, Mackinnon AC, et al. (2010) Ets1 is required for proper migration and differentiation of the cardiac neural crest. Development 137: 1543-1551. doi: 10.1242/dev.047696
    [157] Makki N, Capecchi MR (2012) Cardiovascular defects in a mouse model of HOXA1 syndrome. Hum Mol Genet 21: 26-31. doi: 10.1093/hmg/ddr434
    [158] Zhou J, Zhang M, Fang H, et al. (2009) The SWI/SNF chromatin remodeling complex regulates myocardin-induced smooth muscle-specific gene expression. Arterioscler Thromb Vasc Biol 29: 921-928. doi: 10.1161/ATVBAHA.109.187229
    [159] Zhang M, Fang H, Zhou J, et al. (2007) A novel role of Brg1 in the regulation of SRF/MRTFA- dependent smooth muscle-specific gene expression. J Biol Chem 282: 25708-25716. doi: 10.1074/jbc.M701925200
    [160] Zhang M, Chen M, Kim JR, et al. (2011) SWI/SNF complexes containing Brahma or Brahma-related gene 1 play distinct roles in smooth muscle development. Mol Cell Biol 31: 2618-2631. doi: 10.1128/MCB.01338-10
    [161] Theodoris CV, Li M, White MP, et al. (2015) Human disease modeling reveals integrated transcriptional and epigenetic mechanisms of NOTCH1 haploinsufficiency. Cell 160: 1072-1086. doi: 10.1016/j.cell.2015.02.035
    [162] Guarani V, Deflorian G, Franco CA, et al. (2011) Acetylation-dependent regulation of endothelial Notch signalling by the SIRT1 deacetylase. Nature 473: 234-238. doi: 10.1038/nature09917
    [163] Yuan SM (2013) Congenital heart defects in Kabuki syndrome. Cardiol J 20: 121-124.
    [164] Kim SJ, Cho SY, Maeng SH, et al. (2013) A novel MLL2 gene mutation in a Korean patient with Kabuki syndrome. Korean J Pediatr 56: 355-358. doi: 10.3345/kjp.2013.56.8.355
    [165] Kakita T, Hasegawa K, Morimoto T, et al. (1999) p300 protein as a coactivator of GATA-5 in the transcription of cardiac-restricted atrial natriuretic factor gene. J Biol Chem 274: 34096-34102. doi: 10.1074/jbc.274.48.34096
    [166] Stevens CA, Bhakta MG (1995) Cardiac abnormalities in the Rubinstein-Taybi syndrome. Am J Med Genet 59: 346-348. doi: 10.1002/ajmg.1320590313
    [167] Kurtovic S, Paloschi V, Folkersen L, et al. (2011) Diverging alternative splicing fingerprints in the transforming growth factor-beta signaling pathway identified in thoracic aortic aneurysms. Mol Med 17: 665-675.
    [168] Lamouille S, Xu J, Derynck R (2014) Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 15: 178-196. doi: 10.1038/nrm3758
    [169] Brown CB, Boyer AS, Runyan RB, et al. (1996) Antibodies to the Type II TGFbeta receptor block cell activation and migration during atrioventricular cushion transformation in the heart. Dev Biol 174: 248-257. doi: 10.1006/dbio.1996.0070
    [170] Ramsdell AF, Markwald RR (1997) Induction of endocardial cushion tissue in the avian heart is regulated, in part, by TGFbeta-3-mediated autocrine signaling. Dev Biol 188: 64-74. doi: 10.1006/dbio.1997.8637
    [171] Boyer AS, Ayerinskas II, Vincent EB, et al. (1999) TGFbeta2 and TGFbeta3 have separate and sequential activities during epithelial-mesenchymal cell transformation in the embryonic heart. Dev Biol 208: 530-545. doi: 10.1006/dbio.1999.9211
    [172] Brown CB, Boyer AS, Runyan RB, et al. (1999) Requirement of type III TGF-beta receptor for endocardial cell transformation in the heart. Science 283: 2080-2082. doi: 10.1126/science.283.5410.2080
    [173] Millan FA, Denhez F, Kondaiah P, et al. (1991) Embryonic gene expression patterns of TGF beta 1, beta 2 and beta 3 suggest different developmental functions in vivo. Development 111: 131-143.
    [174] Nieto MA (2002) The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3: 155-166. doi: 10.1038/nrm757
    [175] Nieto MA, Sargent MG, Wilkinson DG, et al. (1994) Control of cell behavior during vertebrate development by Slug, a zinc finger gene. Science 264: 835-839. doi: 10.1126/science.7513443
    [176] Romano LA, Runyan RB (2000) Slug is an essential target of TGFbeta2 signaling in the developing chicken heart. Dev Biol 223: 91-102. doi: 10.1006/dbio.2000.9750
    [177] Carver EA, Jiang R, Lan Y, et al. (2001) The mouse snail gene encodes a key regulator of the epithelial-mesenchymal transition. Mol Cell Biol 21: 8184-8188. doi: 10.1128/MCB.21.23.8184-8188.2001
    [178] Liebner S, Cattelino A, Gallini R, et al. (2004) Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J Cell Biol 166: 359-367. doi: 10.1083/jcb.200403050
    [179] Luna-Zurita L, Prados B, Grego-Bessa J, et al. (2010) Integration of a Notch-dependent mesenchymal gene program and Bmp2-driven cell invasiveness regulates murine cardiac valve formation. J Clin Invest 120: 3493-3507. doi: 10.1172/JCI42666
    [180] Galvin KM, Donovan MJ, Lynch CA, et al. (2000) A role for smad6 in development and homeostasis of the cardiovascular system. Nat Genet 24: 171-174. doi: 10.1038/72835
    [181] Takase M, Imamura T, Sampath TK, et al. (1998) Induction of Smad6 mRNA by bone morphogenetic proteins. Biochem Biophys Res Commun 244: 26-29. doi: 10.1006/bbrc.1998.8200
    [182] Corson LB, Yamanaka Y, Lai KM, et al. (2003) Spatial and temporal patterns of ERK signaling during mouse embryogenesis. Development 130: 4527-4537. doi: 10.1242/dev.00669
    [183] Newbern J, Zhong J, Wickramasinghe RS, et al. (2008) Mouse and human phenotypes indicate a critical conserved role for ERK2 signaling in neural crest development. Proc Natl Acad Sci USA 105: 17115-17120. doi: 10.1073/pnas.0805239105
    [184] Rivera-Feliciano J, Tabin CJ (2006) Bmp2 instructs cardiac progenitors to form the heart-1006 valve-inducing field. Dev Biol 295: 580-588. doi: 10.1016/j.ydbio.2006.03.043
    [185] Ma L, Lu MF, Schwartz RJ, et al. (2005) Bmp2 is essential for cardiac cushion epithelial-mesenchymal transition and myocardial patterning. Development 132: 5601-5611. doi: 10.1242/dev.02156
    [186] Sugi Y, Yamamura H, Okagawa H, et al. (2004) Bone morphogenetic protein-2 can mediate myocardial regulation of atrioventricular cushion mesenchymal cell formation in mice. Dev Biol 269: 505-518. doi: 10.1016/j.ydbio.2004.01.045
    [187] McCulley DJ, Kang JO, Martin JF, et al. (2008) BMP4 is required in the anterior heart field and its derivatives for endocardial cushion remodeling, outflow tract septation, and semilunar valve development. Dev Dyn 237: 3200-3209. doi: 10.1002/dvdy.21743
    [188] Kim RY, Robertson EJ, Solloway MJ (2001) Bmp6 and Bmp7 are required for cushion formation and septation in the developing mouse heart. Dev Biol 235: 449-466. doi: 10.1006/dbio.2001.0284
    [189] Delot EC, Bahamonde ME, Zhao M, et al. (2003) BMP signaling is required for septation of the outflow tract of the mammalian heart. Development 130: 209-220. doi: 10.1242/dev.00181
    [190] Beppu H, Malhotra R, Beppu Y, et al. (2009) BMP type II receptor regulates positioning of outflow tract and remodeling of atrioventricular cushion during cardiogenesis. Dev Biol 331: 167-175. doi: 10.1016/j.ydbio.2009.04.032
    [191] Gaussin V, Van de Putte T, Mishina Y, et al. (2002) Endocardial cushion and myocardial defects after cardiac myocyte-specific conditional deletion of the bone morphogenetic protein receptor ALK3. Proc Natl Acad Sci U S A 99: 2878-2883. doi: 10.1073/pnas.042390499
    [192] Radtke F, MacDonald HR, Tacchini-Cottier F (2013) Regulation of innate and adaptive immunity by Notch. Nat Rev Immunol 13: 427-437. doi: 10.1038/nri3445
    [193] Niessen K, Karsan A (2008) Notch signaling in cardiac development. Circ Res 102: 1169-1181. doi: 10.1161/CIRCRESAHA.108.174318
    [194] McDaniell R, Warthen DM, Sanchez-Lara PA, et al. (2006) NOTCH2 mutations cause Alagille syndrome, a heterogeneous disorder of the notch signaling pathway. Am J Hum Genet 79: 169-173. doi: 10.1086/505332
    [195] Warthen DM, Moore EC, Kamath BM, et al. (2006) Jagged1 (JAG1) mutations in Alagille syndrome: increasing the mutation detection rate. Hum Mutat 27: 436-443. doi: 10.1002/humu.20310
    [196] Donovan J, Kordylewska A, Jan YN, et al. (2002) Tetralogy of fallot and other congenital heart defects in Hey2 mutant mice. Curr Biol 12: 1605-1610. doi: 10.1016/S0960-9822(02)01149-1
  • This article has been cited by:

    1. Halil Ibrahim Bulut, Arian Arjomandi Rad, Angeliki-Aikaterini Syrengela, Iakovos Ttofi, Jasmina Djordjevic, Ramanjit Kaur, Amar Keiralla, George Krasopoulos, A Comprehensive Review of Management Strategies for Bicuspid Aortic Valve (BAV): Exploring Epidemiology, Aetiology, Aortopathy, and Interventions in Light of Recent Guidelines, 2023, 10, 2308-3425, 398, 10.3390/jcdd10090398
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(6210) PDF downloads(998) Cited by(1)

Article outline

Figures and Tables

Figures(4)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog