Fact-checked by Grok 2 weeks ago

Heart development


Heart development refers to the embryological processes that transform mesodermal cells into a functional, four-chambered during early , beginning at the end of the second week with and culminating in a septated heart capable of coordinated circulation by the eighth week. Cardiac cells arise from the epiblast lateral to the , migrating to form bilateral heart fields that fuse midline to create a primitive heart tube around day 21, marking the first to exhibit contractile activity in the .
Subsequent morphogenesis involves rightward looping of the heart tube between days 22 and 28, establishing the basic topological arrangement of future chambers, followed by partitioning via and septa to divide the single atrium and ventricle, while outflow and inflow tracts align for efficient blood flow. This tightly regulated cascade, driven by genetic programs including transcription factors like Nkx2.5 and Gata4, integrates , , and hemodynamic forces to yield a mature structure resilient to postnatal demands, with disruptions frequently resulting in congenital anomalies affecting up to 1% of live births. Key milestones include the onset of by day 22 and completion of septation by week 7, underscoring the heart's rapid evolution from a peristaltic tube to a valved pump.

Specification and Initial Formation

Cardiogenic Mesoderm Induction

Cardiogenic mesoderm induction refers to the specification of cells into progenitors committed to cardiac lineages during early in . This process occurs in the anterior region of the , where primitive streak-derived mesodermal cells receive inductive cues to express cardiac-specific transcription factors. In mice, a model for mammalian cardiogenesis, this specification begins around embryonic day 6.5-7.5, with cardiogenic progenitors migrating anteriorly to form bilateral heart fields. In humans, analogous events transpire approximately 16-18 days post-fertilization, preceding overt heart tube formation. Induction primarily arises from interactions between overlying , underlying , and nascent , with anterior visceral endoderm (AVE) in mice or anterior endoderm in other vertebrates secreting diffusible signals. Experiments in and embryos demonstrate that anterior endoderm explants can induce cardiac in non-cardiogenic , indicating its instructive role independent of mesoderm-intrinsic programs. (BMP) signaling, particularly BMP2 and BMP4 from and , is essential for initial mesoderm specification toward cardiogenic fates, activating downstream targets like GATA4 and NKX2.5. (FGF) pathways, including FGF8, cooperate transiently with BMP to promote proliferation and differentiation of these progenitors, as evidenced by loss-of-function studies in and mice where FGF inhibition abolishes cardiac markers. Wnt/β-catenin signaling exhibits biphasic regulation: canonical Wnt promotes primitive formation but must be antagonized anteriorly for cardiogenic commitment, via inhibitors like , Crescent, and Frzb secreted by . Overexpression of Wnt antagonists in posterior induces ectopic cardiac tissue, underscoring their necessity. Nodal/TGF-β signaling upstream integrates with these, driving mesendoderm formation and Eomesodermin (Eomes) expression, which licenses primitive for cardiogenic potential. Mesoderm-specific Mesp1 patterns nascent into cardiac subsets by restricting alternative fates like somitogenesis, with Mesp1-null mice exhibiting severe cardiac . Core cardiac transcription factors emerge post-induction: NKX2.5, GATA4, and TBX5 form regulatory amplified by and FGF, with NKX2.5 mutants in mice showing disrupted despite specification. Id genes, as BMP effectors, inhibit non-cardiac repressors like Tcf3 to facilitate . These mechanisms are conserved across vertebrates, though human pluripotent models reveal nuanced differences, such as enhanced reliance on timed Wnt modulation for efficient cardiogenic yield. Disruptions in these pathways underlie congenital defects, linking failures to conditions like heterotaxy via genes such as SOX17.

Endocardial and Myocardial Tube Fusion

During the third week of , approximately days 19 to 22 post-fertilization, bilateral primordia of the and myocardium converge in the ventral midline to form the primitive heart tube through a driven by embryonic folding. Cephalocaudal and lateral folding of the positions the paired endocardial tubes, originating from angioblastic cells in the splanchnic layer of , adjacent to one another. These endocardial tubes initially appear as paired endothelial-lined channels within the cardiogenic . Fusion initiates in the cranial region near the buccopharyngeal membrane and proceeds caudally, uniting the endocardial tubes into a single conduit by day 22. Concurrently, myocardial precursor cells, also derived from the cardiogenic mesoderm, differentiate into a sleeve of contractile myocardium that envelops the endocardial tube, with an intervening acellular layer of extracellular matrix termed cardiac jelly. This results in a trilayered primitive heart tube: inner endocardium providing endothelial lining, cardiac jelly as a supportive scaffold, and outer myocardium responsible for contractility. The myocardium at this stage exhibits peristaltic contractions, initiating blood flow shortly after fusion, around day 23. In avian models such as the chick embryo, equivalent occurs between the 1- to 13-somite stages, starting with ventricular primordia and extending to outflow and atrial regions by 8 to 9 somites, facilitated by the ventral mesocardium which anchors the heart tube to the . Endocardial cells actively contribute to myocardial alignment and ; experimental disruptions, such as of endocardial progenitors, prevent proper myocardial tube , leading to cardia bifida where unfused bilateral heart primordia persist. Molecular signals, including VEGF and pathways, regulate endocardial-myocardial interactions essential for tube integrity, with defects in these signaling causing failures observed in animal models.

Morphogenetic Remodeling

Heart Tube Looping and Asymmetry

The straight heart tube, formed by the fusion of bilateral endocardial primordia around day 21 of gestation, undergoes looping morphogenesis beginning at approximately day 22-23 ( stage 10). This process transforms the linear tube into a dextral C-shaped loop, with the cranial portion bending ventrally and to the right, while the caudal portion shifts dorsally and leftward, establishing the bulboventricular configuration. Looping is complete by day 28, aligning the future outflow tract cranially, the ventricle caudally and rightward, and the atrium dorsally. Mechanically, looping arises from differential and cytoskeletal rearrangements in the myocardium, particularly at the atrioventricular and bulboventricular junctions, coupled with remodeling via cardiac . Computational models indicate that non-uniform and myocardial trabeculation contribute to the tube's helical twisting, with the rightward bias emerging from asymmetric in the dorsal mesocardium before its resorption. In and models, which recapitulate looping dynamics, this involves ventral bending followed by rightward torsion, driven by left-right differences in shape and motility. Left-right asymmetry in looping is orchestrated by a genetic cascade initiated at the embryonic node, where nodal flow generates asymmetric Nodal signaling on the left (LPM). This activates left-specific expression of the Pitx2, which patterns second heart field progenitors and directs asymmetric of the outflow tract and atria. Pitx2-null mice exhibit randomized or reversed looping, underscoring its role in enforcing dextral , while human heterotaxy syndromes often involve mutations in Nodal pathway components, linking disrupted asymmetry to congenital heart defects like . The integration of biomechanical forces with Pitx2-mediated ensures precise chamber positioning, critical for subsequent septation and valve formation.

Cardiac Jelly and Cushion Formation

The cardiac jelly is an acellular, gelatinous extracellular matrix secreted by myocardial cells into the space between the endocardium and myocardium of the primitive heart tube during early cardiogenesis, typically forming a thick cuff that provides structural support and facilitates morphogenetic processes. This matrix, rich in glycosaminoglycans, fibronectin, collagen types I and III, and proteoglycans, emerges shortly after fusion of the endocardial tubes around embryonic day 8.5 in mice or equivalent stages in humans (approximately 3-4 weeks post-fertilization), acting as a hydraulic skeleton analogous to mesoglea in jellyfish, which maintains tubular integrity against hemodynamic forces. Cushion formation initiates during heart tube looping, with localized expansion of the cardiac jelly in the atrioventricular canal (AVC) and outflow tract (OFT), driven by myocardial synthesis of components and signaling molecules that induce endocardial . In the AVC and OFT, a subset of overlying endocardial cells undergoes epithelial-to-mesenchymal transition (), delaminating, migrating through the jelly, and differentiating into mesenchymal cells that proliferate and populate the swelling cushions; this process is triggered by myocardial-derived signals such as transforming growth factor-β (TGF-β) and bone morphogenetic proteins (BMPs) diffused via the jelly . The cushions initially function as temporary valves to regulate unidirectional blood flow and prevent during looping, with hyaluronan synthesis contributing to jelly expansion and EMT competence around embryonic days 9-10 in chick models, homologous to stages at 5-6 weeks. These , mesenchymalized from approximately 10-20% of local endocardial cells, fuse and remodel to form the primitive atrioventricular valves (mitral and tricuspid) from AVC cushions and contribute to semilunar valve primordia and septation in the OFT, with cell migration into OFT cushions aiding partitioning of the . Disruptions in jelly composition or , such as hyaluronan deficiencies, lead to incomplete cushion formation and congenital defects like atrioventricular septal defects, underscoring the jelly's causal role in enabling mesenchymal invasion and structural partitioning essential for four-chambered heart functionality.

Chamber Specification and Septation

Atrial and Ventricular Patterning

Atrial and ventricular patterning initiates in the nascent linear heart tube following of the endocardial and myocardial layers, establishing distinct myocardial identities along the anterior-posterior axis through graded of transcription factors and signaling cues. This process divides the tube into regions fated for the primitive ventricle anteriorly and atrium posteriorly, with subsequent ballooning of the outer curvature forming mature chambers. In mice, chamber-specific emerges around embryonic day 8.5 (E8.5), while in humans, ventricular chambers are evident by day 32 post-fertilization and a four-chambered by week 7. Key transcription factors drive this specification. Tbx5 exhibits a posterior-to-anterior , with high levels in the future atrium and left ventricle, promoting atrial septation and left ventricular identity through interactions with Nkx2.5 and Gata4; its mutations, as in Holt-Oram syndrome, disrupt septal formation and cause atrial or ventricular septal defects. Nkx2.5, broadly expressed in cardiac progenitors from E8.5 in mice, cooperates with Hand2 and Mef2c to specify ventricular myocardium and trabeculation, with deficiencies impairing ventricular compaction and conduction. Hand1 and Hand2 further delineate ventricular subtypes, with Hand2 directing right ventricular and outflow tract progenitors and Hand1 influencing left ventricular development; disruptions lead to ventricular or double-outlet right ventricle. Gata4 synergizes with Tbx5 and Nkx2.5 to regulate cardiomyocyte proliferation and chamber boundaries, and its absence results in atrioventricular septal defects. Atrial-specific factors like Pitx2 and Coup-TFII (Nr2f1) reinforce posterior identity, while ventricular repressors such as Tbx2, Tbx3, Hey2, and Irx1-6 suppress atrial genes anteriorly. Signaling pathways modulate these networks to enforce chamber fates. Retinoic acid (RA), active in the posterior second heart field (SHF) from E9.5-E12.5 in mice, promotes atrial by activating Nr2f1 and atrial markers like Nppa and Mlc2a, while its inhibition favors ventricular identity marked by Mlc2v and Myh7; excess or deficiency causes atrial or ventricular septal defects. BMP signaling, via Bmp10, drives anterior first heart field (FHF) ventricular trabeculation and upregulates Nkx2.5 through Gata4, with Alk3 receptor mutations yielding thin ventricular walls. Wnt signaling exhibits biphasic regulation, initially promoting SHF proliferation and later inhibiting to enable myocardial and ventricular ballooning; canonical Wnt/β-catenin restricts atrial expansion. Additional inputs like and TGF-β influence atrioventricular canal boundaries to prevent chamber fusion, while left-right asymmetry cues (e.g., Nodal-Pitx2) align atrial appendages. These mechanisms ensure functional heterogeneity, with atrial cardiomyocytes expressing Hcn4 and Cx40 for rapid conduction and ventricular cells featuring Cx43 and thicker myocardium for force generation. Disruptions, often from (e.g., Tbx5-Nkx2.5), yield congenital defects like atrioventricular canal defects or heterotaxy, highlighting the interplay of FHF (left ventricle, parts of atria) and SHF (right ventricle, atria, outflow) progenitors prepatterned in the .

Interatrial and Interventricular Septa

The develops from the end of the fourth gestational week, beginning with the formation of the septum primum as a sickle-shaped crest extending from the roof of the common atrium toward the . This growth leaves an initial opening known as the ostium primum, which permits blood flow between the developing atria. As the septum primum approaches the cushions around the fifth week, the ostium primum closes upon fusion, while perforations in the upper portion of the septum primum coalesce to form the ostium secundum, ensuring continued right-to-left shunting. Subsequently, the arises as a thicker, crescent-shaped of the atrial wall to the right of the septum primum, growing downward but not fully to the cushions, thereby overlapping the ostium secundum and creating the foramen ovale. The persistent lower edge of the septum primum functions as a flap over the foramen ovale, which remains patent for oxygenated blood passage from the right to left atrium. Postnatally, elevated left atrial pressure typically seals the foramen ovale, with morphological occurring in approximately two-thirds of individuals within two years. The forms concurrently starting in the fourth week, primarily through the muscular component, which arises from the apposition and merger of the medial walls of the developing ventricles, growing upward from the toward the . This muscular portion constitutes the bulk of the , leaving a temporary primary at its superior aspect. Closure of the occurs via the membranous portion, derived from of the and contributions from the truncoconal ridges, completing septation by the seventh week. The membranous is thinner and smaller than the part, separating the left ventricle from both the right ventricle and, inferiorly, the right atrium. Full septation divides the single ventricle into right and left chambers, aligning with the completion of atrioventricular and outflow tract partitioning around days 27 to 37 post-fertilization.

Atrioventricular Canal Septation

The atrioventricular () canal, initially a common passageway connecting the primitive atrium and ventricle during early heart tube formation, undergoes septation to establish separate right and left junctions, enabling unidirectional blood flow to the tricuspid and mitral valves. This process primarily occurs between the 27th and 37th days of embryonic through the formation, growth, and fusion of derived from the cardiac jelly matrix secreted by the myocardium. The cushions consist of mesenchymal tissue arising from endothelial-to-mesenchymal transition () of endocardial cells in the canal region, regulated by signaling molecules such as transforming growth factor-beta 2 (TGF-β2) and bone morphogenetic protein 2 (BMP2), which induce and cushion swelling. Septation begins with the development of superior (dorsal or rostral) and inferior (ventral or caudal) endocardial cushions that protrude into the AV canal lumen, followed by the emergence of paired lateral cushions. The superior and inferior cushions elongate toward each other, fusing in the midline to divide the canal into distinct left and right orifices by approximately the 7th week of gestation. This fusion is complemented by contributions from the dorsal mesenchymal protrusion (DMP), a structure originating from the second heart field that provides additional mesenchyme to bridge the primary atrial septum with the AV cushions, ensuring continuity of the atrial and AV septa. Concurrently, the muscular component of the interventricular septum grows upward from the ventricular floor, meeting the fused cushions to complete the membranous interventricular septum. The lateral cushions, forming later, differentiate into the valvular leaflets of the AV valves, with their mesenchymal cells remodeling into fibrous tissue under the influence of transcription factors like Tbx2 and Tbx3, which maintain AV canal myocardial identity distinct from chamber myocardium. Failure in cushion fusion or , often linked to genetic disruptions such as in CRELD1 or GATA4 genes, results in atrioventricular septal defects (AVSD), underscoring the precision of these cellular interactions. Experimental models in mice demonstrate that targeted inactivation of TGF-β signaling impairs cushion mesenchyme formation, halting septation and confirming the pathway's causal role in AV partitioning.

Outflow Tract and Valve Development

Truncus Arteriosus Partitioning

The partitioning of the occurs during the fifth week of , following the completion of heart tube looping, and involves the division of the common outflow vessel into the and pulmonary via formation of the aorticopulmonary septum. This process ensures separate systemic and pulmonary circulations, with the septum's spiral orientation aligning the with the left ventricle and the pulmonary with the right ventricle. Endocardial swellings, or truncal cushions, initially form in the through epithelial-to-mesenchymal transition () of endocardial cells, generating mesenchymal ridges: a right-superior cushion and a left-inferior cushion. These cushions migrate toward each other—the right-superior toward the left and the left-inferior toward the right—while elongating and spiraling due to differential growth and cellular proliferation. Fusion of the cushions midway along the truncus establishes the aorticopulmonary septum, which extends proximally to connect with the conal septum and distally to the aortic sac, completing septation by approximately the seventh week. Cardiac neural crest cells (CNCCs), originating from the postotic (rhombomeres 6–8), play a critical role by migrating through pharyngeal arches 3, 4, and 6 to populate the truncal cushions starting around embryonic day 10 in mammalian models equivalent to early fifth week in humans. These CNCCs contribute mesenchymal cells that reinforce the cushions, promote ridge fusion, and provide structural integrity to the ; experimental of CNCCs in and mouse models results in failure of septation, leading to . CNCC migration and differentiation are guided by semaphorin signaling, such as Sema3C-Plxna2 interactions, with disruptions in genes like Gata6 impairing this process. Additional molecular regulation involves BMP4 and TGFβ signaling for cushion maturation, alongside and Wnt/β-catenin pathways that coordinate CNCC invasion and septal alignment. Tbx1 expression in the secondary heart field supports outflow tract elongation, indirectly facilitating partitioning by providing a scaffold for CNCC integration. This coordinated cellular and molecular interplay ensures precise septation, with the truncal region's mesenchymal ridges distinguishing it from proximal conal septation, which relies more on myocardial contributions.

Semilunar and Atrioventricular Valves

The atrioventricular () valves, comprising the tricuspid and mitral valves, originate from that form in the AV canal around the fifth week of human gestation. These cushions arise as swellings of proteoglycan-rich beneath the , populated by mesenchymal cells generated through epithelial-mesenchymal transition () of endocardial cells, with additional contributions from epicardium-derived cells and minor input from cranial cells. By the seventh to eighth week, superior and inferior cushions fuse to form the septal leaflets, while lateral cushions develop into the mural leaflets, delineating right and left AV orifices and establishing the fibrous annulus that electrically insulates atrial and ventricular myocardium. Morphogenesis of AV valves involves elongation of cushion-derived leaflets, formation of from subendocardial , and papillary muscle attachment from myocardial progenitors, completing basic structure by the ninth week. The nonmuscular portions of leaflets derive exclusively from AV cushions, which remodel via , extracellular matrix stratification, and hemodynamic influences, with processes advancing through the twelfth week and refining postnatally into thin, flexible structures resistant to regurgitation. Semilunar valves, including the aortic and pulmonary valves, develop from endocardial cushions in the outflow tract (OFT) or , initiating around the fifth to sixth week following initial septation by neural crest-derived ridges. These cushions, formed similarly via of endocardial cells with substantial cranial cell invasion, undergo excavation and sculpting post-partitioning of the OFT into and pulmonary , yielding three cusps per valve by the sixth to ninth week. Cellular semilunar valves first appear at embryonic stage 17 (approximately six weeks), with mesenchymal cores covered by that differentiates under —flat on ventricular surfaces facing high flow and cuboidal on arterial sides. Maturation of semilunar valves proceeds through margin growth via localized in low-flow zones, accumulation of and fibers correlating with fluctuations, and overall thinning, achieving fibroelastic maturity near birth. Disruptions in cushion remodeling or neural crest migration can yield fused cusps, as observed in congenital anomalies, underscoring the precision of these temporally coordinated events.

Conduction System Maturation

Pacemaker Node Formation

The (SAN), the primary of the heart, forms from progenitor cells at the embryonic venous pole, specifically within the right , which derives from Tbx18-positive and Isl1-positive mesodermal precursors of the second heart field. These cells originate in the right and contribute to the pro-pacemaking region, distinct from the primary myocardium of the heart tube. In mammalian embryos, including humans, this process coincides with the incorporation of the into the developing right atrium, establishing the SAN at the junction of the and . Morphological development begins with the specification of precursors, followed by and . In mouse models, progenitors emerge around embryonic day 8.5 (E8.5), with the initial Tbx3-positive appearing in Hcn4-positive myocardium by E9.5; the node becomes discernible by E10.5–E11.5 through recruitment of mesenchymal cells and maturation by E13.5. In human embryos, the manifests as a myocardial with a tail-like extension at the right atrium–right common cardinal vein junction by Carnegie stage 13 (approximately 32 days post-fertilization, or 4–5 weeks ), aligning with the onset of coordinated around day 22–23. Early electrical pacemaking precedes overt , initiating in the posterior heart tube before shifting to the region as venous structures remodel. Molecular regulation involves a network of transcription factors that specify and maintain pacemaker identity via mutual repression of chamber myocardial genes. Tbx18 drives initial formation of the SAN head by differentiating mesenchymal precursors into nodal myocardium, while Tbx3 sustains this fate by suppressing atrial markers like Cx40 and Nppa. Shox2 promotes pacemaker gene expression (e.g., Hcn4) by inhibiting Nkx2-5, which otherwise drives working myocardium differentiation; disruption of this balance, as in Shox2-null mice, impairs SAN development. Canonical Wnt signaling initiates precursor specification at the venous pole, with later restriction to the right side influenced by left-right asymmetry cues like Pitx2c, whose absence leads to bilateral SAN formation. These mechanisms ensure localized automaticity, with SAN cells exhibiting slower conduction and distinct ion channel profiles (e.g., elevated HCN4 for hyperpolarization-activated currents) compared to surrounding atrium. ![Embryo at 5 weeks 5 days with heartbeat - annotated.gif][center]
This annotated image depicts an at approximately 5 weeks , when early pacemaker activity initiates visible cardiac contractions, reflecting the functional onset tied to SAN precursor specification.

His-Purkinje Network Development

The His-Purkinje network, comprising the penetrating His bundle, bundle branches, and distal , forms the ventricular component of the , enabling rapid electrical propagation from apex to base for synchronized ventricular contraction. It arises through myogenic differentiation of cardiomyocytes rather than derivatives, with proximal elements (His bundle and branches) originating from a primary myocardial ring and distal emerging from ventricular trabecular myocardium. This differentiation involves recruitment of surrounding working myocardium into a specialized fast-conducting , influenced by hemodynamic forces and epicardial-derived signals. In murine embryos, His-Purkinje specification initiates around embryonic day (E) 8.5, with discrete fiber expression detectable by E10.5 via reporter genes like LacZ; functional activation patterns, including apical breakthroughs, emerge by E10.5-12.5 prior to interventricular septation completion. Development proceeds in phases: an initial scaffold forms from first heart field-derived trabeculae (left-sided) and second heart field contributions (right-sided), followed by postnatal recruitment of additional cardiomyocytes to expand the network. In chick embryos, Purkinje fibers differentiate subendocardially along nascent coronary branches from stage HH25 onward, suggesting vascular induction via endothelin signaling converts myocytes to conduction phenotype. Human data remain limited, but analogous processes occur, with ventricular conduction maturing in late fetal stages alongside gap junction expression (e.g., Cx40, Cx43) for electrical coupling; cardiac contractions begin around day 23 post-conception, though network specifics align with murine timelines scaled to gestation. Key regulators include transcription factors Nkx2-5, essential for early specification and late recruitment; Tbx5 and Irx3 for patterning; and ETV1 for fiber identity. Neuregulin-1/ signaling from promotes trabecular origins, while from vascular drives Purkinje differentiation and . Disruptions, such as Nkx2-5 mutations, impair network formation and yield arrhythmias, underscoring causal roles in . The network achieves insulation via adjacent fibroblasts and fibrous tissue, ensuring insulated propagation distal to the His bundle.

Molecular and Genetic Regulation

Transcription Factors and Gene Networks

The core transcriptional regulation of heart development relies on interconnected gene regulatory networks orchestrated by key transcription factors that bind cooperatively to enhancers, driving cardiac specification, differentiation, and morphogenesis. Central to these networks are NKX2-5, GATA4, and TBX5, which exhibit extensive interdependent genomic occupancy, with their binding sites overlapping at thousands of cardiac enhancers to coordinately activate genes essential for cardiogenesis, such as those involved in sarcomere assembly and chamber maturation. This heterotypic cooperation ensures robust gene expression, as disruption of one factor redistributes the others, highlighting a causal mechanism where mutual reinforcement amplifies transcriptional output beyond individual contributions. NKX2-5, among the earliest markers of the cardiac lineage, is indispensable for myocardial differentiation and restricts cardiac progenitor proliferation through BMP signaling modulation, with knockout mice exhibiting arrested heart tube formation and embryonic lethality around E9.5. It synergizes with GATA4 to promote cardiomyocyte maturation and with HAND2 for ventricular identity, while human heterozygous mutations occur in up to 4% of congenital heart disease (CHD) cases, often manifesting as atrial septal defects (ASD) or tetralogy of Fallot due to impaired conduction system development. GATA4 initiates cardiac gene expression during mesoderm fusion and endocardial cushion formation, physically interacting with NKX2-5 and TBX5 to regulate targets like Nppa (); its haploinsufficiency in mice leads to thin ventricular myocardium and perinatal lethality, while human variants associate with ventricular septal defects (VSD) and atrioventricular septal defects (AVSD), particularly when compounded with TBX5 mutations. TBX5 drives atrial and left ventricular expansion, patterning the and conduction system via activation of genes like Myh6; cause Holt-Oram , featuring , VSD, and conduction blocks, with mouse models showing dose-dependent defects in outflow tract alignment. It antagonizes repressors like TBX2 in the atrioventricular canal to balance and . Upstream regulators like Mesp1 act as master switches for multipotent cardiovascular progenitors, activating downstream networks including Isl1 for second heart field contributions to the right ventricle and outflow tract.00288-9) Additional factors such as HAND2 specify right ventricular fate, MEF2C promotes chamber , and Tbx20 enhances synergy among the core triad for sarcomeric , forming hierarchical GRNs that integrate with signaling pathways (e.g., , FGF) for temporal control. These networks' evolutionary conservation is evident in models, where perturbations reveal motifs enriched for Nkx2-5 and Tbx5 during looping. Disruptions, often from heterozygous loss-of-function, underscore causal links to CHD, with empirical lineage tracing confirming restricted progenitor contributions.

Signaling Pathways and Crosstalk

(BMP) signaling, particularly through and BMP4, promotes cardiogenic specification and myocardial during early heart tube formation in vertebrates. BMP activity gradients from the posterior induce expression of cardiac transcription factors like Nkx2.5 and Gata4, with studies in mice showing that null embryos exhibit cardia bifida due to failed heart tube fusion. (FGF) signaling, via FGF8 and , drives proliferation of cardiac progenitors in the second heart field (SHF) and supports outflow tract elongation, as evidenced by Fgf8 hypomorphic mutants displaying tetralogy of Fallot-like defects. Canonical Wnt/β-catenin signaling inhibits early cardiac but activates later in SHF progenitors to promote proliferation and deployment, with temporal shifts from repressive to inductive roles confirmed in chick and mouse models where β-catenin stabilization disrupts chamber morphogenesis. Notch signaling regulates endocardial-mesenchymal transition (EMT) in atrioventricular canal (AVC) and outflow tract (OFT) cushions, essential for and septal formation, through ligands like Dll4 and Jag1 activating 1 in endocardial cells to induce expression and EMT. Inactivation of 1 in mice leads to hypocellular cushions and , underscoring its necessity. Transforming growth factor-β (TGF-β) and activin/Nodal pathways cooperate with to drive EMT in cushions, with TGF-β2 promoting matrix production in mesenchymal cells post-EMT. Crosstalk between these pathways integrates spatial and temporal control of cardiogenesis. and Wnt signaling exhibit antagonistic interactions in cardiac progenitors, where activates non-overlapping gene sets from Wnt/β-catenin, as shown in explant assays where combined inhibition disrupts progenitor specification without redundancy. synergizes with to enhance from embryonic stem cells, with crosstalk via Smad amplifying induction. In valve development, intersects with and TGF-β to coordinate , where upregulates TGF-β receptors in , and signaling modulates via Hey1 repressors to prevent excessive cushion invasion. Disruption of this - axis in and models results in thickened valves, highlighting balanced crosstalk for proper remodeling. Additionally, and pathways interact in trabeculation, with promoting myocardial proliferation that feeds -mediated patterning, as conditional knockouts reveal interdependent roles in compact versus trabecular myocardium formation. These interactions ensure robust patterning, with dysregulated crosstalk implicated in congenital defects like from FGF-Wnt imbalances.

Left-Right Asymmetry Mechanisms

Left-right (L-R) asymmetry in heart development ensures the rightward looping of the initially linear heart tube, proper alignment of atria and ventricles, and correct positioning of great vessels relative to systemic circulation. This process begins during , prior to heart tube formation, and integrates , signaling cascades, and tissue-intrinsic to establish situs-specific . Disruptions in L-R patterning contribute to heterotaxy syndromes, where heart looping may be randomized or reversed, often co-occurring with defects in outflow tract septation and valve formation. In and embryos, L-R asymmetry initiates at the embryonic (or Hensen's node in chicks), a transient structure where monociliated cells generate directional motility. These motile cilia, tilted posteriorly and rotating clockwise when viewed from above, produce a leftward extracellular fluid flow—termed nodal flow—at speeds of approximately 10–20 μm/s, persisting for 12–24 hours around embryonic day 7.5–8.5 in mice. This hydrodynamic cue breaks bilateral symmetry by asymmetrically distributing extracellular vesicles or inducing transient left-sided calcium fluxes in perinodal crown cells, which express polycystin-2 channels. Immotile sensory cilia in the same region may further transduce mechanical signals into intracellular responses, though the precise sensor remains debated. Mutations in ciliary genes like Kif3b abolish nodal flow, resulting in 50% randomization of heart looping direction. Nodal flow triggers asymmetric gene expression in the lateral plate mesoderm (LPM), with the TGF-β family member Nodal upregulated transiently in left LPM cells via autoinduction and inhibition of right-sided expression by antagonists like Lefty-1 and Cerberus. Nodal signaling, mediated by activin-like kinase receptors and Smad2/3 transcription factors, directly induces the bicoid-related homeobox gene Pitx2 (isoform Pitx2c) exclusively in left LPM by embryonic day 8.0 in mice. Pitx2 expression extends into the left cardiogenic mesoderm and persists through heart tube elongation, where it modulates proliferation and migration of second heart field (SHF) progenitors. In Pitx2 knockout mice, SHF cells fail to contribute asymmetrically to the left ventricle and outflow tract, leading to right isomerism and impaired rightward looping, though initial tube bending occurs via Nodal-independent actomyosin contractility in myocardium.30798-X) Beyond signaling, tissue-intrinsic mechanisms reinforce during looping. Left myocardial cells exhibit higher actomyosin tension and oriented cell divisions, driven by planar pathways and RhoA/ROCK signaling, which elongate and bend the tube rightward. Pitx2 represses right-sided genes like Tbx5 in left domains, ensuring differential chamber maturation, while SHF recruitment adds cells preferentially to the rightward bend. In , where nodal flow is absent, Kupffer's vesicle cilia still impose asymmetry via similar Nodal-Pitx2 cascades, highlighting evolutionary conservation. Human ciliopathies, such as due to DNAH5 mutations, disrupt nodal equivalents and cause 50–100% heterotaxy incidence with cardiac defects like transposition of great arteries.

Pathological Disruptions and Anomalies

Etiology of Congenital Heart Defects

Congenital heart defects (CHDs) represent the most common class of major birth defects, affecting approximately 1% of live births annually , or about 40,000 infants per year. Globally, the among children under five years old exceeded 4.18 million cases in 2021, reflecting a gradual increase over prior decades due to improved detection and survival rates. The of CHDs is predominantly multifactorial, with genetic factors implicated in roughly 40% of cases, environmental influences in about 5%, and the majority remaining idiopathic or involving complex gene-environment interactions during critical cardiogenic windows from embryonic weeks 3 to 8. Genetic contributions to CHDs include chromosomal abnormalities, which occur in 16-30% of affected individuals depending on the cohort studied. is the most frequent, present in up to 16.8% of chromosomal cases in some series, with 40-50% of individuals with trisomy 21 developing CHDs, particularly atrioventricular septal defects. Other aneuploidies, such as (Edwards syndrome), (Patau syndrome), and (Turner syndrome), are associated with high rates of conotruncal and left-sided obstructive lesions, though overall prevalence is lower due to reduced viability. Submicroscopic copy number variants and single-gene mutations further account for syndromic CHDs; for instance, mutations in NKX2-5, GATA4, or TBX5 disrupt transcription factor networks essential for cardiac septation, contributing to 1-4% of isolated septal defects or syndromes like Holt-Oram. Syndromic associations, including 22q11.2 deletion () with interrupted and with , highlight recurrent genetic lesions in 10-20% of extracardiac anomaly cases. Environmental risk factors, while less dominant, exert causal influence through teratogenic exposures during . Maternal pregestational or elevates CHD risk with odds ratios of 3-5, particularly for conotruncal defects like transposition of the great arteries and left obstructions, via hyperglycemia-induced and disrupted cardiogenic signaling. Maternal during the periconceptional period to first increases odds of septal defects (ventricular and atrial) by 10-20%, attributable to and carbon monoxide-mediated impairing endothelial function and vascular remodeling. (BMI ≥30 kg/m²) and (>35 years) confer 1.5-2-fold risks, potentially through inflammatory cytokines or epigenetic modifications affecting fetal cardiac progenitors. Other exposures, including certain antiepileptics (e.g., ) and infections like , demonstrate stronger causal links in historical cohorts, though modern and have diminished their incidence. Gene-environment interactions amplify susceptibility; for example, polymorphisms in metabolism genes (e.g., MTHFR) interact with periconceptional nutrient deficiencies to heighten conotruncal defect risk, underscoring the protective role of preconceptional folic acid supplementation in reducing select CHDs by up to 10-20% in randomized trials. Socioeconomic and pollutant exposures, such as air particulates, correlate with higher live-born CHD incidence in deprived areas, likely via additive epigenetic or inflammatory pathways, though requires further longitudinal validation. Despite advances in genomic sequencing identifying variants in up to 10% of sporadic cases, over half of CHDs elude singular explanations, emphasizing the need for integrated models of polygenic risk and stochastic developmental perturbations.

Genetic Mutations and Environmental Factors

Genetic mutations contribute to approximately 20-30% of congenital heart defects (CHD), with single-gene defects identified in 3-5% of cases, often involving transcription factors essential for cardiogenesis. Mutations in NKX2-5, a homeobox gene critical for cardiac progenitor specification and conduction system development, are linked to atrial septal defects (ASD), tetralogy of Fallot, and atrioventricular conduction delays, disrupting ventricular myocyte proliferation and chamber maturation. Similarly, heterozygous mutations in GATA4 impair endocardial cushion formation, leading to atrioventricular septal defects and pulmonary stenosis, while TBX5 variants underlie Holt-Oram syndrome, featuring ASD and limb anomalies due to defective septation and patterning. Chromosomal anomalies, such as 22q11.2 deletion in DiGeorge syndrome, account for conotruncal defects like interrupted aortic arch, affecting neural crest migration during outflow tract septation. Environmental factors act primarily during the embryonic window of heart development (3-8 weeks ), often through teratogenic disruption of signaling pathways like Wnt or . Maternal pregestational elevates CHD risk 3- to 5-fold, particularly for transposition of the great arteries and ventricular septal defects, via hyperglycemia-induced and altered cardiogenic in the embryo. similarly heightens critical CHD incidence, with odds ratios up to 1.5-2.0 for outflow tract anomalies, independent of . Other teratogens include infection, which causes pulmonary stenosis and through inflammatory interference with endothelial function, and maternal untreated by diet, yielding 10-15% CHD rates via hyperphenylalaninemia disrupting cells. Gene-environment interactions amplify risk, as seen in folate pathway variants (MTHFR polymorphisms) where insufficient periconceptional folic acid supplementation correlates with a 1.5-2-fold increase in conotruncal defects, though supplementation mitigates this in high-risk groups. Prenatal exposure to air pollutants like PM2.5 elevates CHD odds by 10-20%, potentially via epigenetic modifications to cardiac genes, while maternal smoking doubles left obstructive lesions through nicotine-mediated vascular disruption. These factors underscore multifactorial , with empirical studies emphasizing causal verification over associative claims from observational data prone to .

Recent Advances and Experimental Models

Stem Cell Organoids and Synthetic Embryology

-derived cardiac organoids are three-dimensional, self-organizing structures generated from pluripotent s, such as human induced pluripotent s (hiPSCs) or embryonic s (hESCs), that mimic key aspects of early heart development, including cardiogenic specification, chamber-like , and electromechanical function. These models typically involve directed protocols to induce cardiac cells, followed by aggregation into embryoid bodies or scaffold-free cultures that promote tissue-like architecture with aligned cardiomyocytes exhibiting organization and calcium transients. By 2023, advancements enabled the generation of patterned primitive heart organoids from hiPSCs, recapitulating anterior-posterior patterning and primitive atrium-like structures through geometric confinement and signaling modulation with BMP4 and FGF2. Recent progress has focused on enhancing physiological relevance, such as integrating vascularization to address limitations in nutrient and in avascular organoids. In June 2025, researchers at Stanford Medicine reported the first vascularized human heart organoids derived from hiPSCs, co-cultured with endothelial progenitors to form perfusable vessel networks that support long-term culture beyond 30 days and improve cardiomyocyte maturation via and . These vascularized models have demonstrated applications in modeling ischemic responses and drug screening, with organoids exhibiting synchronized beating rates of 60-80 and responsiveness to agents like isoproterenol. Multicellular cardiac organoids incorporating fibroblasts, endothelial cells, and epicardial layers have further advanced disease modeling, replicating fibrotic remodeling in patient-derived lines with increased collagen deposition and stiffness. Synthetic embryology extends these approaches by engineering embryo-like aggregates, such as , from stem cells to simulate pre-implantation and stages without oocytes or sperm, providing insights into heart . , generated via geometric patterning of hiPSCs with WNT and agonists, have been shown to form cardiac crescents resembling embryonic day 18-20 structures, expressing NKX2-5 and TBX5 in anterior domains while specifying hepatic progenitors posteriorly, thus capturing bilateral cardiogenic . By November 2024, optimized protocols demonstrated competence for both cardiac and lineages, with 20-30% of aggregates developing contractile cardiac tissue responsive to electrical pacing, enabling dissection of mesodermal fate decisions via single-cell sequencing. These models have elucidated causal mechanisms in cardiogenesis, such as the role of NODAL signaling in formation preceding cardiac , validated by perturbations reducing MESP1+ progenitors by over 50%. In systems, synthetic embryos from extended pluripotent cells achieved beating hearts by embryonic day 8.5 equivalents in 2022, forming yolk-sac vasculature and neural tubes alongside cardiac tubes, though human equivalents remain limited to pre-gastrulation stages due to ethical and technical constraints. Applications include for etiologies, with organoids from trisomy 21 hiPSCs showing delayed chamber septation mirroring phenotypes. Despite maturity gaps—organoids rarely exceed fetal-like states with immature sarcomeres and absent Purkinje networks—these platforms offer scalable, ethically favorable alternatives to animal models for in developmental timing and signaling .

Advanced Imaging and Lineage Tracing

Advanced imaging techniques, such as two-photon microscopy and light-sheet microscopy, have enabled real-time visualization of dynamic cellular processes during embryonic heart tube formation, revealing coordinated movements of cardiac progenitors in embryos. These methods allow whole-embryo tracking at cellular resolution, identifying three distinct phases of heart tube assembly characterized by progenitor migration and fusion events between embryonic day (E) 7.5 and E8.5. Light-sheet microscopy facilitates rapid 3D imaging of embryonic hearts within one minute, supporting high-throughput analysis of at least 30 samples per session with the Lightsheet Z.1 system. Genetic lineage tracing, primarily via Cre-loxP systems, has elucidated the of cardiac progenitors, demonstrating that first heart field (FHF) derivatives predominate in left ventricular cardiomyocytes (>90%) under Wnt-based protocols in human pluripotent models combined with single-cell sequencing (scRNA-seq). Intersectional and multicolor reporter strategies, including analysis with double markers and rainbow systems, provide clonal resolution to trace multipotent progenitors like cKit+ cells of origin contributing to cardiomyocytes and other lineages. These tools have clarified second heart field (SHF) contributions to right ventricular and outflow tract regions, challenging earlier models of strict FHF-SHF segregation. Integration of lineage tracing with live protocols, such as two-photon excitation for early cardiac progenitors in embryos, permits observation of differentiation trajectories from E8.5 onward, highlighting migration patterns and environmental influences on cell fate. Noninvasive biomicroscopy at 40 MHz images developing hearts from E8.5 to E13.5, quantifying morphological changes without disrupting embryogenesis. Recent advancements, including adaptive optical gating for prolonged time-lapse , capture functional cardiac contractions and reveal developmental shifts in electromechanical properties. Such combined approaches underscore the multipotency of progenitors like Tbx5+ cells, which persist post-injury in adult hearts and contribute to regeneration models.

References

  1. [1]
    Development of the human heart - PMC - PubMed Central - NIH
    Cardiac development is initiated at gastrulation at the end of the second week of human development (Carnegie Stage [CS] 7) (Figure 1). During gastrulation the ...
  2. [2]
    Embryology, Heart - StatPearls - NCBI Bookshelf - NIH
    The cardiovascular system's embryological development begins with cardiac progenitor cells' migration in the epiblast, just lateral to the primitive streak.
  3. [3]
    A pictorial account of the human embryonic heart between 3.5 and 8 ...
    Mar 11, 2022 · We describe the conspicuous morphological features of the developing human heart as they appear in consecutive stages of development. The ...
  4. [4]
    The molecular mechanisms of cardiac development and related ...
    Dec 23, 2024 · This review focuses primarily on prenatal development. Cardiac development initiates when cardiac mesoderm progenitors migrate away from the ...
  5. [5]
    Human Cardiac Development in the First Trimester | Circulation
    Jul 28, 2009 · Using MRI and EFIC imaging, we conducted a systematic analysis of human cardiovascular development in the first trimester.Mri And Efic · Cardiac Loop · Atrial Septation (ega 6
  6. [6]
    Embryonic Heart Progenitors and Cardiogenesis - PMC - NIH
    The cardiogenic mesoderm, which harbors the so-called first and second heart fields (FHF and SHF), forms the major proportion of the ventricular, atrial, and ...
  7. [7]
    Early Signals in Cardiac Development | Circulation Research
    Moreover, coculture experiments suggest that the heart-inducing activity originates from the anterior endoderm and not from the cardiogenic mesoderm itself.
  8. [8]
    BMP4 triggers regulatory circuits specifying the cardiac mesoderm ...
    We show that Bmp4 induces cardiac mesoderm formation in murine embryonic stem cells in vitro. Bmp4 first activates Wnt3 and upregulates Nodal.
  9. [9]
    Heart development: molecular insights into cardiac specification and ...
    Jun 1, 2003 · Requirement for BMP and FGF signaling during cardiogenic induction in non-precardiac mesoderm is specific, transient, and cooperative. Dev ...
  10. [10]
    Inhibition of Wnt activity induces heart formation from posterior ... - NIH
    Dkk-1, a Wnt inhibitor of a different protein family, similarly induces heart-specific gene expression in posterior lateral plate mesoderm.Crescent Is A Wnt-8c... · Anterior Endoderm Induces... · Wnt Inhibition Promotes...
  11. [11]
    Mesp1 patterns mesoderm into cardiac, hematopoietic, or skeletal ...
    Further gene expression characterization of Mesp1-induced cells showed that Mesp1 induction from day 3-8 led to the upregulation of paraxial and presomitic ...
  12. [12]
    Physiology of Cardiac Development: From Genetics to Signaling to ...
    BMP signaling is mediated, at least in part, by induction of GATA4, MEF2c, SRF, and NKX2.5 [55, 56]. Specifically, BMP2 and BMP4, secreted by underlying ...
  13. [13]
    Id genes are essential for early heart formation - PMC
    Id1 promotes cardiogenic mesoderm differentiation by inhibiting Tcf3 and Foxa2. Id proteins do not directly bind DNA but regulate transcription by antagonizing ...
  14. [14]
    Synthetic embryology of the human heart - Frontiers
    Jan 27, 2025 · The formation of the human heart is driven by precisely timed and regulated events initiated at gastrulation at the end of the second week of ...
  15. [15]
    Sox17 is essential for the specification of cardiac mesoderm ... - PNAS
    Here, we show that the Wnt/β-catenin pathway also is essential for cardiac myogenesis to occur in ES cells, acting at a gastrulation-like stage, mediating ...
  16. [16]
    Development Timeline of Human Heart Embryology
    Cephalocaudal and lateral folding brings the bilateral endocardial tubes into the ventral midline of the embryo. 21-22, Heart tube fusion. 22, Heart begins to ...
  17. [17]
    Myocardial Lineage Development | Circulation Research
    Dec 10, 2010 · The early myocardial tube comprises primarily cells destined to ... endocardial tube. The trough then gradually closes in a caudal-to ...
  18. [18]
    Functional Morphology of the Cardiac Jelly in the Tubular Heart of ...
    The early embryonic heart is a multi-layered tube consisting of (1) an outer myocardial tube; (2) an inner endocardial tube; and (3) an extracellular matrix ...
  19. [19]
    Formation of the primitive myo- and endocardial tubes in the chicken ...
    At the 8 to 9 somite stage, fusion of the lateral heart primordia proceeds to include the outflow tract and atrial primordia; the pacemaker site migrates into ...
  20. [20]
    Endocardium is necessary for cardiomyocyte movement during ...
    Jun 15, 2007 · ... primitive heart tube. This simple cylinder immediately becomes responsible for driving embryonic circulation; therefore,the precise ...
  21. [21]
    Altered VEGF Signaling Leads to Defects in Heart Tube Elongation ...
    Oct 9, 2018 · ... myocardial tube fusion can be noted in the SU5416-treated embryo. ... Heart looping in the absence of normal endocardial tube morphology ...
  22. [22]
    Bending of the Looping Heart: Differential Growth Revisited - PMC
    In the early embryo, the primitive heart tube (HT) undergoes the morphogenetic process of c-looping as it bends and twists into a c-shaped tube.
  23. [23]
    Bending and twisting the embryonic heart: a computational model ...
    The morphogenetic process of cardiac looping transforms the straight heart tube into a curved tube that resembles the shape of the future four-chambered ...
  24. [24]
    On the Biomechanics of Cardiac S-Looping in the Chick - NIH
    Cardiac looping is an important embryonic developmental stage where the primitive heart tube (HT) twists into a configuration that more closely resembles ...
  25. [25]
    Pitx2 regulates cardiac left–right asymmetry by patterning second ...
    The Pitx2 homeobox gene functions at the final stages of this cascade to direct asymmetric morphogenesis of selected organs including the heart. We previously ...
  26. [26]
    Left-right asymmetry in heart development and disease
    Nov 22, 2018 · Summary: This Review compares mechanisms underlying asymmetric heart morphogenesis and the dynamics thereof in fish, chick and mouse, ...
  27. [27]
    Pitx2 regulates cardiac left-right asymmetry by patterning ... - PubMed
    The Pitx2 homeobox gene functions at the final stages of this cascade to direct asymmetric morphogenesis of selected organs including the heart. We previously ...
  28. [28]
    The Role of Left-Right Asymmetry in Congenital Heart Defects
    Aug 30, 2025 · Unlike the transient expression of NODAL in the node and the left LPM [107, 108], expression of PITX2 is established by NODAL in the left LPM ...
  29. [29]
    The Functional Significance of Cardiac Looping - PubMed Central
    Looping morphogenesis leads to the transformation of the initially straight embryonic heart tube into a looped tube whose geometric configuration is described ...
  30. [30]
    Extracellular Matrix and Heart Development - PMC
    This cardiac jelly is critically important for the proper formation of the endocardial cushions at the atrioventricular (AV) junction. The cells in the tubular ...
  31. [31]
    Compositional and structural heterogenicity of the cardiac jelly of the ...
    During the early stages of heart development the extracellular matrix, 'cardiac jelly' (CJ) (Davis, 1924), constitutes the major component of this organ.<|separator|>
  32. [32]
    Regulation of cardiac cushion development by hyaluronan - PMC
    Immediately following looping of the heart tube, the cardiac jelly expands, forming the endocardial cushions. A subpopulation of the endocardial cells ...
  33. [33]
    Mechanical regulation of cardiac development - Frontiers
    During S-looping, regional wall thickenings of cardiac jelly form endocardial cushions (ECs) in the AV canal and OFT regions. At this stage, ECs function as ...
  34. [34]
    Partitioning the heart: mechanisms of cardiac septation and valve ...
    In addition, the AVC endocardial cushions develop into atrioventricular (mitral and tricuspid) valves, whereas the OFT endocardial cushions give rise to ...
  35. [35]
    Cardiac Embryology and Molecular Mechanisms of Congenital ...
    In this paper we aim to review current knowledge pertaining to genetic alterations and their cellular effects that are involved in the formation of congenital ...
  36. [36]
    Regulation of atrial and ventricular cardiomyocyte identity
    The first recognized molecular step toward cardiogenesis is Mesp1 expression, which indicates cardiovascular specification [23]. Such a multimodal process ...Review Article · 4. Acm Identity And... · 5. Vcm Identity And...
  37. [37]
    Embryology and Developmental Defects of the Interatrial Septum | AJR
    Starting the fifth week of gestation, the septum primum begins to develop, growing toward the endocardial cushions.
  38. [38]
    Development of the Cardiovascular System - TeachMeAnatomy
    Jun 10, 2024 · The development of the heart begins with the formation of the primitive heart tube following the folding of the embryo during the end of the third week.Primitive Heart Tube · Aortic Arches · Septation Of The Heart
  39. [39]
    Embryology, Atrioventricular Septum - StatPearls - NCBI Bookshelf
    The atrial septum is then formed by a fusion of the septum primum and secundum. A flap covering the foramen ovale is formed from the septum primum. Shortly ...
  40. [40]
    [PDF] 4 Combined genetic investigations of Atrioventricular Septal Defects ...
    The final mesenchymal set of cells required for AV canal septation in the dorsal mesenchymal protrusion (DMP) comes from the second heart field (SHF) which ...
  41. [41]
    Development of the myocardium of the atrioventricular canal and the ...
    Septation of the atrioventricular junction is brought about by downgrowth of the primary atrial septum, fusion of the endocardial cushions, and forward ...
  42. [42]
    Atrioventricular Septal Defect - StatPearls - NCBI Bookshelf - NIH
    Atrioventricular septal defect (AVSD) is a congenital cardiac anomaly characterized by incomplete formation of the atrial and ventricular septa.
  43. [43]
    Neural Crest Cells Contribute to Normal Aorticopulmonary Septation
    Bilateral removal of this neural crest population prior to migration causes malformation of the aorticopulmonary septum resulting in common arterial outflow ...
  44. [44]
    How to Make a Heart Valve: From Embryonic Development to ...
    Cardiac valves develop from endocardial cushions that become populated by valve precursor mesenchyme formed by an epithelial–mesenchymal transition (EMT). The ...
  45. [45]
    Heart Valve Development: Regulatory networks in ... - NIH
    Here, we review the cellular and molecular mechanisms underlying the early development of valve progenitors and establishment of normal valve structure and ...
  46. [46]
    The Development of the Atrioventricular Junction in the Human Heart
    Our findings show that the nonmuscular part of the developing leaflets of the atrioventricular valves derives from the atrioventricular cushions and that the ...
  47. [47]
    The development of the semilunar valves in the human heart
    The development of human semilunar valves was studied in a range of specimens including embryos before the appearance of cellular semilunar valves at stage 17.
  48. [48]
    Development of the cardiac pacemaker - PMC - PubMed Central
    Oct 21, 2016 · The sinoatrial node (SAN) is the dominant pacemaker of the heart. Abnormalities in SAN formation and function can cause sinus arrhythmia.
  49. [49]
    Detailed Cardiac - Sinus Node - UNSW Embryology
    Feb 19, 2017 · The sinoatrial node (SAN) is the primary pacemaker of the heart that initially develops from part of pro-pacemaking embryonic venous pole.
  50. [50]
    Development of the Pacemaker Tissues of the Heart
    Feb 5, 2010 · This review discusses current insights into the cellular origin of the nodal pacemaker tissues, their formation and function in relation to ...
  51. [51]
    Development of the Cardiac Conduction Tissue in Human Embryos ...
    HNK-1 is present in the sinoatrial node (SAN), located in front of the superior caval vein. Three tracts of HNK-1–positive myocardium converge in this area. One ...
  52. [52]
    Formation of the Sinus Node Head and Differentiation of Sinus Node ...
    Dec 18, 2008 · Our data establish a functional order for Tbx18 and Tbx3 in SAN formation, in which Tbx18 controls the formation of the SAN head from mesenchymal precursors.
  53. [53]
    Molecular Pathway for the Localized Formation of the Sinoatrial Node
    Pitx2c-deficient fetuses form sinoatrial nodes with indistinguishable molecular signatures at both the right and left sinuatrial junction, indicating that Pitx ...
  54. [54]
    New Insights into the Development and Morphogenesis of the ...
    Aug 7, 2021 · During embryogenesis, the VCS, consisting of the His bundle, bundle branches, and the distal Purkinje network, originates from two independent ...
  55. [55]
    Embryology of the Conduction System for the Electrophysiologist
    Aug 15, 2010 · Distal to the His bundle, the Purkinje fibers also develop through the differentiation of cardiomyocytes yet are intimately linked to the ...
  56. [56]
    Hemodynamics Is a Key Epigenetic Factor in Development of the ...
    The His-Purkinje system (HPS) is a specialized network of conduction tissues that ensures coordinated and reliable activation of the ventricular myocardium.
  57. [57]
    Visualization and functional characterization of the developing ... - NIH
    Moreover, optical mapping studies of embryonic hearts demonstrate that a murine His-Purkinje system is functioning well before septation has completed. Thus, ...
  58. [58]
    The development of cardiac rhythm - PMC - NIH
    PURKINJE FIBRE DIFFERENTIATION. In the maturing chick embryonic heart Purkinje fibres develop in the subendocardium along coronary artery branches, suggesting a ...
  59. [59]
  60. [60]
    Signaling and Transcriptional Networks in Heart Development and ...
    Heterozygous mutations of several transcription factor genes that are important in early heart development cause human congenital heart disease (CHD) (Bruneau ...
  61. [61]
    Investigating the transcriptional control of cardiovascular development
    Transcriptional regulation of thousands of genes instructs complex morphogenetic and molecular events for heart development.
  62. [62]
    Heart morphogenesis gene regulatory networks revealed by ...
    Summary: Transcriptome profiling during zebrafish heart looping reveals novel and evolutionarily conserved gene regulatory networks and cohorts of genes.Results · Motif Enrichment In Specific... · Nkx2-5 And Tbx5 Rna-Seq...
  63. [63]
    Signaling Pathways Governing Cardiomyocyte Differentiation - PMC
    Jun 18, 2024 · We have reviewed the major signaling pathways involved in cardiomyocyte differentiation, including the BMP, Notch, sonic hedgehog, Hippo, and Wnt signaling ...
  64. [64]
    Signaling Pathways and Gene Regulatory Networks in ...
    Retinoid signaling, for instance, may regulate the atrial versus ventricular differentiation of hPSCs although more studies are warranted. Human H7 ESCs ...
  65. [65]
    Wnt signaling in cardiac development and heart diseases - PubMed
    May 6, 2024 · Both canonical and non-canonical Wnt signaling pathways play a critical role during embryonic heart development in a region- and stage-specific manner.<|separator|>
  66. [66]
    Heart Valve Development | Circulation Research
    Aug 28, 2009 · Notch signaling also is required for expression of TGFβ2 and multiple TGFβ receptors in AVC and OFT endocardial cushion endothelial cells, ...
  67. [67]
    NOTCH Signaling in Aortic Valve Development and Calcific ... - NIH
    The NOTCH1 mutations are the first identified human genetic variants that cause congenital bicuspid aortic valve (BAV) and calcific aortic valve disease (CAVD).Missing: FGF | Show results with:FGF
  68. [68]
    Wnt/β-catenin and Bmp signals control distinct sets of ... - PNAS
    We show that Wnt and Bmp4 signaling activate specific and nonoverlapping cardiac-specific genes in the cardiac progenitors.
  69. [69]
    Mechanisms of heart valve development and disease
    Jul 3, 2020 · Notch signaling also has been implicated in endocardial cushion EndoMT, acting upstream of TGFβ signaling and intersecting with BMP signaling.The initiation of valve... · Endocardial cushion... · The development of cardiac...
  70. [70]
    Co-ordinating Notch, BMP, and TGF-β signaling during heart valve ...
    We discuss BMP, TGF-β, and Notch signaling pathways during mouse cardiac cushion formation and how they together produce a coordinated EMT response in the ...Missing: FGF | Show results with:FGF
  71. [71]
    Left Right Patterning, Evolution and Cardiac Development - PMC
    The embryo has evolved a finely tuned system to tell left from right and this axis is established in the mesoderm before formation of the linear heart tube.
  72. [72]
    Randomization of left-right asymmetry due to loss of nodal cilia ...
    Dec 11, 1998 · Randomization of left-right asymmetry due to loss of nodal cilia generating leftward flow of extraembryonic fluid in mice lacking KIF3B motor ...Missing: cardiac | Show results with:cardiac<|control11|><|separator|>
  73. [73]
    Left–right asymmetry: cilia stir up new surprises in the node - PMC
    May 7, 2013 · The 'two-cilia model' posits that the motile cilia generate a leftward-directed fluid flow that is somehow sensed by the immotile sensory cilia on the ...Missing: cardiac | Show results with:cardiac
  74. [74]
    Left–Right Determination: Involvement of Molecular Motor KIF3, Cilia ...
    The discovery of nodal flow generated by the rotation of cilia, which plays a fundamental role in L/R asymmetry, was based on studies of molecular motors. Cells ...
  75. [75]
    Expression and Function of Pitx2 in Chick Heart Looping - NCBI - NIH
    It was demonstrated in mice that the asymmetric expression of Pitx2 is directly induced by Nodal signaling through the action of the transcription factor FAST ...
  76. [76]
    A Nodal-independent and tissue-intrinsic mechanism controls heart ...
    Nov 11, 2013 · When actomyosin activity is blocked, even in the presence of asymmetric Nodal signalling, directional heart looping is prevented (shown in Fig.
  77. [77]
    Randomization of Left-Right Asymmetry and Congenital Heart Defects
    Oct 22, 2019 · We highlight that mutations in DNAH5 are not only associated with classical randomization of left-right body asymmetry but also with severe laterality defects ...
  78. [78]
    Data and Statistics | Congenital Heart Defects (CHDs) - CDC
    Oct 21, 2024 · Heart defects affect nearly 1% of―or about 40,000―births per year in the United States. The prevalence of some heart defects, especially mild ...
  79. [79]
    Global, regional, and national epidemiology of congenital heart ...
    May 16, 2025 · In 2021, the global prevalence of CHD in children under five years was over 4.18 million, reflecting a 3.4% increase since 1990.
  80. [80]
    Genetic detection of congenital heart disease - ScienceDirect.com
    Genetic variation contributes to an estimated 40% of CHD cases, environmental factors contribute to an estimated 5%, and the remaining 55% of CHD cases are of ...
  81. [81]
    Chromosomal Abnormalities in Patients with Congenital Heart ... - NIH
    Chromosomal abnormalities were observed in 50 subjects (16.8%) and the full trisomy of chromosome 21 was the most frequent of them (n = 40). Of the remaining ...
  82. [82]
    Chromosomal Abnormalities in Congenital Heart Disease Causing ...
    Dec 1, 2024 · Our review of the literature revealed that numerical chromosomal anomalies, such as DS, ES, PS, and TS, can be associated with CHD defects and ...
  83. [83]
    Genetics of congenital heart disease: a narrative review of recent ...
    The etiology of CHD is multifactorial as both genetic and environmental factors have been implicated in its etiology (20). Specific genetic causes can be ...Introduction · Recent advances in the... · Clinical implications of recent... · Footnote
  84. [84]
    Heart-Related Genetic Syndromes in Children - OHSU
    Types of genetic syndromes that affect the heart · Loeys-Dietz syndrome · Marfan syndrome · Noonan syndrome · Trisomy 21 (Down syndrome) · Turner syndrome · 22q11.2 ...
  85. [85]
    Maternal Diabetes and Overweight and Congenital Heart Defects in ...
    Jan 5, 2024 · This cohort study investigates the association of maternal diabetes and body mass index with congenital heart defects in offspring.
  86. [86]
    Impact of Maternal Diabetes on the Incidence of Critical Congenital ...
    Sep 22, 2025 · Factors associated with CCHD, including maternal diabetes, illness, phenylketonuria, and maternal exposure to toxins, drugs, or viral infections ...
  87. [87]
    Maternal Smoking and Congenital Heart Defects | Pediatrics
    Apr 1, 2008 · Women who smoked anytime during the month before pregnancy to the end of the first trimester were more likely to have infants with septal heart defects.
  88. [88]
    Systematic Review and Meta-analysis of Prenatal Risk Factors for ...
    There exists robust evidence for increased risk of CHD in the presence of obesity, maternal diabetes, maternal smoking and increased maternal age.
  89. [89]
    Environmental Risk Factors and Congenital Heart Disease - Frontiers
    This umbrella review shows that many environmental factors have substantial evidence in relation to the risk of developing CHD.Abstract · Introduction · Methods · Discussion
  90. [90]
    Genetic and environmental risk factors in congenital heart disease ...
    There are many environmental risk factors for CHD, including pesticides and therapeutic agents (5), and some of these are known to target particular genes.
  91. [91]
    Environmental and Socioeconomic Factors Influence the Live‐Born ...
    Apr 19, 2020 · Increased social deprivation and exposure to environmental pollutants are associated with the incidence of live‐born CHD in California.
  92. [92]
    Unraveling the genomic basis of congenital heart disease - JCI
    Jan 19, 2021 · The genetic, epigenetic, and environmental etiologic basis of congenital heart disease (CHD) for most heart anomalies remains unexplained.
  93. [93]
    Review Cardiovascular disease associated with genetic defects
    Congenital heart defects may have a genetic component, with a genetic cause identified in 20 % to 30 % of patients with CHD, including single gene defects in 3 ...
  94. [94]
    Genetics of Congenital Heart Defects: The NKX2-5 Gene, a Key Player
    Jan 23, 2016 · The mutant NKX2-5 factor can regulate a number of off-targets downstream to facilitate CHD development. This review summarizes the genetic ...
  95. [95]
    Genetic Basis for Congenital Heart Disease: Revisited: A Scientific ...
    Sep 27, 2018 · This review provides an updated summary of the state of our knowledge of the genetic contributions to the pathogenesis of congenital heart disease.
  96. [96]
    Genetics of congenital heart disease: a narrative review of recent ...
    In this review, we provide an updated summary of the established genetic contributors to CHD and discuss recent advances in our understanding of the genetic ...
  97. [97]
    Maternal Obesity and Diabetes Mellitus as Risk Factors for ...
    Apr 20, 2020 · In large, population‐based studies, maternal diabetes mellitus appears to be a strong risk factor for any and all subtypes of CHD.
  98. [98]
    Environmental Risk Factors for Congenital Heart Disease - PMC - NIH
    Such factors can be either an excess of a toxic substance, or the lack of an essential nutrient. In both cases, the factor can act directly on the embryo itself ...
  99. [99]
    Environmental Risk Factors for Congenital Heart Disease - PubMed
    Mar 2, 2020 · In this review, we describe the most important environmental CHD risk factors, and what is known about how they cause CHD.
  100. [100]
    Prenatal exposure to air pollutants and the risk of congenital heart ...
    Jul 23, 2024 · Prenatal exposure to air pollution and heavy metals has been suggested as a potential contributing factor to the development of CHD. Several ...
  101. [101]
    Advances in cardiac organoid research - PubMed Central - NIH
    Jan 18, 2025 · This review comprehensively surveys the various fabrication techniques used to create cardiac organoids and their diverse applications in modeling a range of ...
  102. [102]
    Cardiac organoids: a new tool for disease modeling and drug ...
    May 19, 2025 · Cardiac organoid is a miniature and simplified three-dimensional (3D) cellular model system grown from progenitor cells or stem cells.
  103. [103]
    A patterned human primitive heart organoid model generated by ...
    Dec 12, 2023 · Recently, we and others have described methodologies to create human heart organoids from pluripotent stem cells. These methods enable the study ...
  104. [104]
    Advance in creating organoids could aid research, lead to treatment
    Jun 5, 2025 · Stanford Medicine researchers developed a way to create the first heart and liver organoids that generate their own blood vessels.Share Article · Recipe Testing · Other Organs
  105. [105]
    Advancements in Stem Cell Research: Creating Vascularized ...
    Jul 22, 2025 · Wu, MD, PhD, conducted a new study designed to co-create blood vessels within heart and liver organoids using human pluripotent stem cells, ...
  106. [106]
    Progress in multicellular human cardiac organoids for clinical ... - NIH
    Here, we highlight the recent remarkable progress on multicellular cardiac organoids and review the current status for their practical applications.Introduction · Contractility And... · Cardiac Organoid To Become...
  107. [107]
    Synthetic embryology of the human heart - PMC - PubMed Central
    Jan 28, 2025 · Synthetic embryology refers to in vitro embryo models that via stem cell gastruloid technology mimic discrete developmental stages and thus by ...
  108. [108]
    Gastruloids are competent to specify both cardiac and skeletal ...
    Nov 23, 2024 · In this work, we unveil the potential of gastruloids to undergo specification into both cardiac and skeletal muscle lineages.Missing: blastoids | Show results with:blastoids
  109. [109]
    Capturing Cardiogenesis in Gastruloids: Cell Stem Cell
    These axially patterned embryonic organoids (gastruloids) mimic embryonic development and support the generation of cardiovascular progenitors, including first ...Missing: blastoids | Show results with:blastoids
  110. [110]
    'Synthetic' embryo with brain and beating heart grown from stem ...
    Aug 25, 2022 · Researchers have created model embryos from mouse stem cells that form a brain, a beating heart, and the foundations of all the other organs ...
  111. [111]
    Human cardiac organoids for disease modeling and drug discovery
    Sep 5, 2025 · Recent advancements in cardiac organoid modeling have enabled further understanding of cardiogenesis, cardiovascular disease, and drug-induced ...Review · Highlights · Abstract
  112. [112]
    Live imaging of heart tube development in mouse reveals ... - eLife
    Dec 5, 2017 · Here, we established a whole-embryo live-imaging method based on two-photon microscopy that allows whole-tissue tracking at cellular resolution.Missing: techniques | Show results with:techniques
  113. [113]
    A low-cost, high-throughput pipeline for 3D imaging of embryonic ...
    This system enables rapid 3D imaging of a mouse embryonic heart within 1 min using the Zeiss Lightsheet Z.1 microscopy and supports imaging of at least 30 ...
  114. [114]
    Combined lineage tracing and scRNA-seq reveals unexpected first ...
    Jun 7, 2023 · This study presents elegant lineage tracing results demonstrating that first heart field (FHF) generates a dominance (>90%) of left ventricular cardiomyocytes ...
  115. [115]
    Cre-loxP-mediated genetic lineage tracing: Unraveling cell fate and ...
    Feb 26, 2023 · Widespread lineage-tracing systems have significantly advanced research in cardiac development, homeostasis, and tissue regeneration.
  116. [116]
    cKit+ cardiac progenitors of neural crest origin - PMC - NIH
    A high-resolution genetic lineage-tracing study in mice reveals that cKit identifies multipotent progenitors of cardiac neural crest (CNC) origin.
  117. [117]
    Combined lineage tracing and scRNA-seq reveals unexpected first ...
    Jun 7, 2023 · During mammalian development, the left and right ventricles arise from early populations of cardiac progenitors known as the first and second ...
  118. [118]
    Live Imaging of Early Cardiac Progenitors in the Mouse Embryo
    Jul 12, 2022 · To facilitate a broader use of live imaging in mouse developmental research, this paper presents a detailed protocol for two-photon live ...Missing: techniques | Show results with:techniques
  119. [119]
    Noninvasive, In Utero Imaging of Mouse Embryonic Heart ...
    For this study, 40-MHz UBM was used to image the developing hearts of live mouse embryos in utero from gestational age 8.5 to 13.5 days (E8. 5 to E13. 5).
  120. [120]
    Adaptive prospective optical gating enables day-long 3D time-lapse ...
    Nov 15, 2019 · Rapid three-dimensional imaging and analysis of the beating embryonic heart reveals functional changes during development. Dev. Dyn. 235 ...
  121. [121]
    Return of the Tbx5; lineage-tracing reveals ventricular ... - Nature
    Mar 3, 2023 · We identify a Tbx5-expressing ventricular cardiomyocyte-like precursor population, in the injured adult mammalian heart.