Next Article in Journal
Regulation of Cardiac Conduction and Arrhythmias by Ankyrin/Spectrin-Based Macromolecular Complexes
Next Article in Special Issue
Role of the Epicardium in the Development of the Atrioventricular Valves and Its Relevance to the Pathogenesis of Myxomatous Valve Disease
Previous Article in Journal
A Heterozygous Mutation in Cardiac Troponin T Promotes Ca2+ Dysregulation and Adult Cardiomyopathy in Zebrafish
Previous Article in Special Issue
Transcriptome Analysis Reveals Differential Gene Expression between the Closing Ductus Arteriosus and the Patent Ductus Arteriosus in Humans
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Cell Tracing and Fate Mapping Experiments in Cardiac Outflow Tract Development, New Opportunities through Emerging Technologies

1
Department Anatomy & Embryology, Leiden University Medical Center, 2300RC Leiden, The Netherlands
2
Department Cellular and Chemical Biology, Leiden University Medical Center, 2300RC Leiden, The Netherlands
3
Department of Cardiology, Leiden University Medical Center, 2300RC Leiden, The Netherlands
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Cardiovasc. Dev. Dis. 2021, 8(5), 47; https://0-doi-org.brum.beds.ac.uk/10.3390/jcdd8050047
Submission received: 29 March 2021 / Revised: 18 April 2021 / Accepted: 22 April 2021 / Published: 26 April 2021

Abstract

:
Whilst knowledge regarding the pathophysiology of congenital heart disease (CHDs) has advanced greatly in recent years, the underlying developmental processes affecting the cardiac outflow tract (OFT) such as bicuspid aortic valve, tetralogy of Fallot and transposition of the great arteries remain poorly understood. Common among CHDs affecting the OFT, is a large variation in disease phenotypes. Even though the different cell lineages contributing to OFT development have been studied for many decades, it remains challenging to relate cell lineage dynamics to the morphologic variation observed in OFT pathologies. We postulate that the variation observed in cellular contribution in these congenital heart diseases might be related to underlying cell lineage dynamics of which little is known. We believe this gap in knowledge is mainly the result of technical limitations in experimental methods used for cell lineage analysis. The aim of this review is to provide an overview of historical fate mapping and cell tracing techniques used to study OFT development and introduce emerging technologies which provide new opportunities that will aid our understanding of the cellular dynamics underlying OFT pathology.

1. General Introduction

The development of the cardiac outflow tract (OFT), a region of the heart connecting the ventricles to the great arteries of the heart, has been studied for more than a century [1]. Cellular lineage tracing experiments laid the foundation for our knowledge regarding congenital heart disease (CHD). Whilst major progress has been made and CHDs as a whole are understood increasingly better, the degree of understanding varies greatly among the individual congenital defects. CHDs affecting the OFT like bicuspid aortic valve, tetralogy of Fallot, transposition of the great arteries, double outlet right ventricle and pulmonary atresia are well known disorders, yet it remains challenging to explain such morphological emergences with our current models of OFT development. Common among many of these congenital diseases is a broad phenotypical variation arising from a singular genetic deficiency. Murine knockout models used to study such aforementioned CHDs, such as Notch1 [2], Gata5 [3] and Nos3 [4] mutant mice, have been known for their incomplete penetration, yet any agreement addressing such phenotypical variation remains elusive. Whilst lineage tracing studies have contributed significantly to our understanding of a particular cell type heritage, it still falls short to explain, how a cell’s lineage primes cellular behavior to allow for proper cell adaptation (such as cell maturation) upon environmental cues. Epigenetic modification of chromatin have been described to affect gene expression and could act to consolidate directional developmental gradients [5,6]. As such a cell’s identity may arise not only from a genetic component but from a combination of expressed genes and cell lineage epigenetic signatures [7].
A single genetic mutation could give rise to multiple phenotypical outcomes if the imprinted epigenetics are dissimilar among cell lineages [8]. Currently we are limited in our understanding about the role of cell identity in CHD. However given that a cell’s identity is highly plastic [9], understanding the role of the identity of a cell might explain the broad phenotypical variation observed in these congenital heart diseases and could aid with disease risk stratification. Lineage tracing experiments built the foundation of our current understanding and new developments in methods could provide more detailed definitions of cell types and cell lineage dynamics to better explain how aberrant cell behavior contributes to the multivariate phenotypes seen in CHD.

1.1. Basic Cardiac Development and Anatomy of the OFT

The heart is formed from the anterior splanchnic mesoderm of the embryonic plate. During gastrulation, mesodermal cells arise from the primitive streak, and migrate cranially and laterally to the notochord to form the cardiogenic plates expressing Nkx2.5 [10] and GATA4 [11]. The first sign of cardiomyocyte differentiation is seen in this region at Hamburger and Hamilton (HH) stage 8–9 in chick and between embryonic day (E) 6.5–7.5 in mice, when cardiac troponin-I (cTnI) and sarcomeric myosin (MF20) are first detected. Fusion of the bilateral plates of splanchnic mesoderm establishes the primary heart tube (PHT), that shows peristaltic contraction at 3 weeks of development in a human, E8.0–8.5 in mouse and HH stage 10–11 in chick embryos [12,13,14,15]. During further development, cells from the splanchnic mesoderm differentiate into cardiomyocytes and are added to the PHT. The splanchnic mesoderm that gives rise to the cardiomyocytes of the cardiac crescent and PHT is called the first heart field (FHF). The FHF will contribute to part of the atria, atrioventricular canal (AVC), inlet of the right ventricle and left ventricle [12]. In the next phase of heart formation, cells are added to specific structures at the venous (atrial septum and smooth myocardial wall of the atrium) and arterial poles (outflow tract and the right ventricle). These cells are considered to be derived from a distinct cardiogenic field, the second heart field (SHF) [12]. How the distinction between FHF and SHF is related to the ultimate phenotype of the left and right ventricular myocardium is an intriguing question but is not the focus of this review. The addition of cells at the arterial and venous poles initiates the process of cardiac looping, the transformation of the linear heart tube into an S-shaped heart tube with primitive ventricular and atrial chambers [16]. These primitive chambers are connected by the AVC. In the AVC and the OFT, the myocardium secretes cardiac jelly, a hydrophilic substance rich in hyaluronic acid, proteoglycans and extracellular matrix. This results in the formation of localised swellings called the endocardial cushions acting as primitive valves, aiding in maintaining unidirectional flow in the heart tube [17,18]. The endocardial cushions of the OFT mature into aortic and pulmonary valves through contribution of cell lineages of neural crest [19,20], endothelial [21,22] and second heart field cells [23,24,25], but are also crucial to the development of the interventricular septum [18,26]. The separation of the aorta and the pulmonary trunk is the result of OFT septation. During OFT septation, extracardiac neural crest cells migrate into the endocardial cushions forming a central mass of condensed mesenchyme, resulting in an aortopulmonary septum followed by invagination of cardiomyocytes. The rotation and elongation of the aortic and pulmonary orifices is the result of the pulmonary push driven by cells of the second heart field lineage [27]. OFT septation is completed when the aortic orifice connects to the left ventricular outflow tract whilst the pulmonary orifice remains connected above the right ventricle [28]. In addition to cardiac growth, several processes, including the outgrowth and remodelling of the different chambers, coronary artery development and formation of the cardiac conduction system, will establish the mature electro-mechanically functional 4-chambered heart with separated pulmonary and systemic circulations (reviewed in [29,30]).

1.2. OFT Development

The OFT can first be observed between E8.0–E8.5 in mice and week 3 of development in humans during formation of the PHT where it connects the primitive ventricle with the aortic sac. During cardiac looping the OFT elongates and remodels forming the OFT cushions. Aortopulmonary septation occurs at the level of: (1) the great arteries, (2) the valvular level and (3) of the outflow tract [31].
The aortopulmonary septum is formed by contributions of neural crest cells and second heart field cells [32,33,34]. The initially unseparated vascular part of the outflow tract is called the aortic sac. For adequate separation of the different levels of the developing outflow tract, development of the aortopulmonary septum and adequate fusion of the outflow cushions are mandatory [35]. Neural crest cells will be positioned at the level of the aortic sac, as well as in the condensed mesenchyme of the septal outflow tract cushion at and below the orifice level [36]. As a result of this process, separation of the aortic sac will extend from the arterial orifice level (i.e., the level of the developing arterial valve) to the myocardial outflow tract.
The aortopulmonary septum will gradually separate the common aortopulmonary trunk into the aorta and pulmonary trunk. At the valvular level, this process will also separate the OFT cushions at the aortic and pulmonary orifices giving rise to the semilunar valves. Later the coronary arteries will grow into the aortic sinuses of Valsalva and the myocardial and smooth muscle components of the OFT will mature.
Historically, the developing OFT has been studied using classical light microscopy, histological staining and electron microscopy [14,37,38]. A comprehensive basis providing insight in OFT development was founded using these techniques. Unfortunately, as cells undergo intrinsic and phenotypical changes during development, analysing the developmental origin of the OFT was not possible using these early methods. To examine cell lineage development, several techniques such as genetic tracing and vital dye labelling of cells have been developed. Extensive lineage tracing experiments examining OFT morphogenesis have revealed an important role for multiple cell types and signaling pathways in OFT development.

2. Cell Tracing Techniques Applied to OFT Development

In the next section, the different techniques used to study OFT development will be described briefly. The main focus of this section will be to review fate mapping experiments that contributed to our current understanding of OFT development.

2.1. Vital Dye and Viral Labelling Experiments

With vital dye it is possible to directly label clusters of cells. This method is widely used in developmental biology in several species and has provided essential information on the developmental origin of the OFT.

2.1.1. Basics of Vital Dye and Viral Labelling Experiments during Heart Development

Labelling of cells is performed with a glass capillary mounted to micromanipulators, and a pneumatic micro-injector to inject small amounts of dye or virus in the region of interest.
Vital dye labelling is performed with lipophilic dyes, such as diI-C18-(3) [39], which can label distinct cell compartments (Figure 1A) [40]. Commercial dyes are available in different colours, which enables simultaneous labelling of different clusters of cells and studying the direction of migration and potential intermingling of different cell clusters.
Viral labelling experiments are aimed at induction of reporter gene expression in specific regions of the embryo. This technique utilises the ability of retroviruses to infect the cell and to incorporate its genome in the genome of the infected cell [41,42,43]. Retroviral vectors driving the expression of a reporter gene (such as LacZ) enable cell tracing of the infected cells (Figure 1B). More targeted viral tracing can be performed by placing expression of the reporter gene under the transcriptional control of a cell type specific promoter, which enables tracing of cells that express the gene of interest (Figure 1C) [44].

2.1.2. Advantages of Vital Dye Labelling Experiments

With physical labelling of a progenitor pool, it is possible to select that specific group of cells based on location and to time the exact moment of labelling. An important advantage of this technique is the opportunity to select a cluster of cells based without prior knowledge of the cell type. Performing physical labelling experiments is relatively cost efficient and easy, especially in chick, fish and amphibian embryos. Furthermore, labelling can be combined with other microsurgical procedures.

2.1.3. Limitations of Vital Dye and Viral Labelling

To perform microinjection of minute quantities of either vital dye or viral constructs, the embryo needs to be physically accessible to the researcher. For this reason most labelling experiments examining heart development using micro-injections have been performed in oviparous vertebrates, such as zebrafish [45], xenopus [46] and chick embryos [47,48]. For use in mice, it is first necessary to extract the embryo from the uterus, followed by re-incubation in culture medium, for example in rolling bottles [49].
Physical labelling experiments require a progenitor population of interest to study. Knowledge about the location of a possible progenitor pool is required to correctly target these cells. Moreover, when targeting a progenitor population, the lineage trace is limited to cells solely derived from that original cluster. This complicates examining structures derived from multiple progenitor clusters because non-labelled cell contributions could be interpreted as inefficient labelling.
Furthermore, physically labelling cells is relatively invasive, and manipulation of the embryo may induce developmental defects. It is therefore necessary to carefully select embryos that show no macroscopic abnormalities and to include control embryos. Toxicity of the dyes itself appears to be negligible [50,51].
With viral labelling, there is random integration of the viral genome in the host cell’s DNA, which could result in damage or abnormal functioning of the infected cell [52]. Leakage of dye or infection of cells in the vicinity of the designated progenitor pool could result in incorrect interpretation regarding the fate of the progenitor cell population. It is therefore essential to evaluate the location of dye directly after administration in great detail [40,53]. Histological evaluation of embryos directly after labelling is crucial, but this also terminates lineage tracing in those embryos. Live-imaging of vital dyes [51,54,55] could help to minimise this limitation. Furthermore, delay in reporter gene expression with viral labelling omits direct evaluation of the location of labelling. In addition, efficiency of reporter gene expression in the desired cells has to be tested prior to ascertain activation of expression in the specific cell type within the animal model (i.e., species differences in expression levels in specific cell types). Rapid cell division in early embryos results in dilution of the vital dyes, which complicates long-term follow-up of the labelled cells [56]. The extent of dilution is dependent on the rate of proliferation and the amount of dye administered, which therefore necessitates titration of the dye with every new experiment. With retroviral labelling, integration of the reporter gene in the host cell’s DNA results in stable and high expression of the reporter gene in the daughter cells [57], which overcomes the problem of dilution. Therefore, a disadvantage of vital dye labelling is dilution, whilst the possibility of direct evaluation after labelling is an important advantage. The opposite is true for viral labelling, where direct evaluation is not possible, but dilution of dye does not occur after stable integration of the viral vector. Therefore, a possible solution to overcome these limitations is to combine vital dye labelling with viral labelling. Finally, to perform viral labelling experiments, advanced biosafety laboratories are crucial to safely perform experiments with potentially hazardous viral constructs.

2.1.4. Vital Dye and Viral Labelling Experiments Aimed at Understanding OFT Development

Studies performing dye injected lineage tracing have aided in understanding heart development by carefully fate mapping groups of cardiac precursor cells [58,59]. Prior to the concept of vital dye lineage tracing descriptive embryological studies in the early 20th century examining human embryos postulated the concept of the primitive cardiac cavities [38]. These primitive cardiac cavities where demarcated by local folds. Using these folds as fixed borders, the outgrowth of cardiac structures such as the development of the atria and ventricles could be described. The introduction of vital dye injected lineage tracing pioneered by de la Cruz refuted the concept of primitive cardiac cavities and showed that structures such as the primordium of the trabeculated portion of the right ventricle could be determined at the straight heart tube stage prior to the formation of primitive cardiac cavities [47].

2.2. Lineage Tracing Using the Quail-Chick Chimeric System

The use of chimeric models is an alternative lineage tracing approach as compared to the previous mentioned vital dye injections. The quail-chick chimeric system proved a vital tool for understanding cell lineage development.

2.2.1. Basics of the Quail-Chick Chimeric System

The quail chick chimera system was devised in 1969 [60,61,62]. In the quail-chick chimera system a subset of cells derived from quail, i.e., cells of the neural fold, are transplanted in stage-matched chick embryos (or vice versa) (Figure 2). As a result, the quail derived cells develop into functional tissues resulting in viable chimeric quail-chick offspring. The quail-chick marker system relies on the differences in heterochromatic DNA in the nuclei of chick and quail. With the use of a nuclear Feulgen staining or a donor specific antibody [63] distinction could be made between host and donor cells, allowing researchers to study embryonic development at later stages as compared to vital dye labelling. In addition to the quail-chic chimeric system, xenopus hybrids were also used to study early stages of embryonic development [64].

2.2.2. Advantages of the Quail-Chick Chimeric System

The quail-chick chimera system allows for cell lineage tracing using the genetic footprint of the host cells. As a result, this method of lineage tracing results in a permanent change through which chimeric models overcome the limitation of vital dye dilution mentioned previously. Therefore, chimeric systems allow researchers to examine a specific cell population for longer time periods as compared to injecting cells with vital dyes. Moreover, whilst vital dyes may suffer from dye leakage during injection, chimeric models have no such issues. Another advantage of chimeric models is that it enabled researchers to perform in-vitro cell manipulation prior to transplantation.

2.2.3. Disadvantages of the Quail-Chick Chimeric System

Similar to vital dyes, chimeric models require physical access to an embryo favouring oviparous vertebrates over mammalian embryos. Nevertheless, lineage tracing through cell transplantation has often been applied in mice studies examining multilineage differentiation through bone marrow transplantation in irradiated mice but also to study early mammalian development through injection of cells in the developing morula [65]. Moreover, with the recent developments in stem cell biology the study of heart development using mice-rat chimeric models could lead to new insights [66]. In contrast to vital dye injections, in chimeric models a graft versus host rejection may occur in postnatal studies. Another challenge when making interspecies grafts is the need to perform microsurgery on two stage-matched embryos increasing the experimental complexity. Moreover, chimeric models are generally best used when cells are transplanted superficially, such is the case with cells of the spinal cord or epicardium. Transplantation of difficult to reach mesodermal cells is likely to complicate experimental reproducibility.

2.2.4. The Quail-Chick Chimeric System and Insight into OFT Development

Chimeric models like the chick-quail system were paramount for understanding the role of NCCs in cardiac development. Le Lievre and LeDouarin showed that NCCs formed the tunica media of the great arteries [67]. Kirby and Steward used quail-chick chimeras to observe migration of NCCs into the heart, contributing to the aortopulmonary septum and showed how ablation of the NCC population resulted in persistent truncus arteriosus [34]. Moreover, transplantation studies using chimeric models made the initial observation of cellular pluripotency because quail NCCs could expand into subsets of totipotent and pluripotent progenitors [68]. The quail-chick chimera system was also used to determine that neural crest cells were contributing to the formation of semilunar valves [69] as well as to elastogenesis in the developing cardiovascular system [70]. A role for NCCs in coronary artery formation was demonstrated by Arima et al. which found NCCs contributing to heart development and smooth muscle heterogeneity within a coronary artery using the quail-chick chimeric model [71].

2.3. Genetic Lineage Tracing

A widely used tool in developmental biology is based on site-specific recombination within the genome. Most mouse models developed for genetic lineage tracing utilise the P1 Bacteriophage derived enzyme Cre-recombinase, which recognises a 34-bp sequence called the loxP site, and induces recombination between pairs of these sites [72]. The Cre-loxP system, combined with the ability to knock-in sequences at specific sites in the genome has been used to study the origin of the developing OFT.

2.3.1. Basics of Genetic Lineage Tracing

To use the Cre-loxP system for genetic tracing, several steps are necessary (Figure 3). The basis of the Cre-loxP system is crossing a Cre transgenic mouse strain with a mouse in which a reporter gene is flanked by LoxP sequences (“floxed”). First, the sequence coding for Cre-recombinase is knocked into the locus of the gene of interest, for example Isl1. The next step is crossing the Isl1Cre mouse line with a reporter line (Figure 3A). The genetic tracing experiments in this review were mostly performed in reporter lines that utilise the Rosa26 locus (R26R), but different loci (e.g., Gata4 [73]) have also been used in other studies. Important characteristics of a potential reporter locus are ubiquitous expression of the gene and viability and fertility of the mouse strain after homozygous inactivation of the reporter locus by insertion of a reporter gene, such as LacZ or a single or multiple fluorescent proteins. To control expression of the reporter gene, a floxed sequence preventing transcriptional read-through (also referred to as a “stop-cassette”) is placed before the reporter gene sequence (Figure 3A).
Crossing the floxed reporter line with the cell type specific Cre line results in offspring carrying both constructs (Figure 3B). Cells expressing the gene of interest will produce Cre-recombinase, which will result in permanent deletion of the stop-cassette and expression of the reporter gene (Figure 3C). Since expression is under control of the ubiquitous Rosa26 promoter, these cells and their progeny will retain reporter gene expression, thereby enabling tracing of these cells over a long period of time. The above-described method is referred to as “constitutive” genetic tracing in this review (Figure 3D).
A variation of genetic tracing, referred to as “inducible” genetic tracing, enables temporal control of recombination (Figure 3D). This system is based on fusion of a mutated ligand-binding domain (LBD) of the human estrogen receptor (ER) and Cre-recombinase. The resulting protein is only sensitive to the anti-estrogen Tamoxifen, not to the endogenously present estradiol [74]. The fusion gene is inserted in the locus of the gene of interest, which results in production of the fusion protein upon expression of the gene of interest. The Cre fusion protein is located in the cytoplasm and is unable to enter the nucleus, due to the presence of the mutated LBD. Administration of Tamoxifen results in binding to the LBD, which enables translocation of Cre-recombinase to the nucleus, where it induces gene specific recombination [75]. Thus, recombination can only occur after administration of Tamoxifen, resulting in temporal control of lineage labelling (Figure 3D).
In 2007, multicolour lineage tracing using the Brainbow labelling cassettes was introduced to the genetic lineage tracing toolset [76]. The Brainbow cassettes (Brainbow-1,2 and 3.2) are genetic labelling constructs exploiting the Cre-loxP system to allow for stochastic recombination events to obtain single cell resolution lineage labelling. This method is based on combining multiple fluorescent reporter gene sequences in the Brainbow cassettes flanked by a combination of cis- and trans-loxP sites. Upon Cre expression, driven by the gene of interest, the fluorescent reporter genes randomly change configuration within the cassette affecting expression of the cell marker. Because multiple copies of fluorescent reporters are embedded within the Brainbow cassettes (i.e., CFP, YFP, RFP) it allows for multiple fluorescent signatures related to the combination of reporters being stochastically expressed after recombination (i.e., Expression of a CFP-CFP-CFP combination would allow for a “Blue” signal, whilst a RFP-RFP-CFP combination would result in a “Magenta” signal). Combining multicolour lineage tracing with tamoxifen induction can result in a multicolour lineage tracing model with temporal control.

2.3.2. Advantages of Genetic Tracing

The most important advantage of genetic tracing is the ability to visualise the complete heritage of a progenitor population driven by a single marker gene of interest. Combination of these results with previous known expression patterns (e.g., based on immunohistochemical staining patterns) allows for the reconstruction a genetic fate map during development of the OFT.
Furthermore, genetic lineage tracing is non-invasive, as long as insertion of the sequence for Cre does not impair normal development of the embryo. This is an important advantage as compared to performing prospective tracing with microinjection of vital dyes or viral constructs (see Section 2.1.3).
Inducible genetic tracing has an important advantage over constitutive genetic tracing, as it allows for temporal control of Cre expression. Constitutive genetic tracing is solely dependent on the natural timing of Cre expression related to the driving promotor. Expression of the gene of interest at any given (and frequently unknown) time point will result in reporter gene expression (if the level of Cre expression is sufficient to induce reporter gene expression, see below). With inducible tracing, reporter gene expression will only occur in the presence of Tamoxifen [74].

2.3.3. Disadvantages of Genetic Tracing

Reporter gene expression in the tissue or structure of interest can result from several scenarios. In the ideal situation, it can show that at time point 0, a certain structure with known expression of the gene of interest is a progenitor for the structure with reporter gene expression at a later time point. However, a possible pitfall can arise from interference resulting from aspecific lineage markers. In scenarios when a gene of interest gives rise to multiple progenitor populations of different origin, interpretation of developing structures would be troubled, which could lead to an inaccurate conclusion regarding the lineage developmental contribution. This difficulty in interpretation of genetic tracing experiments was demonstrated by constitutive genetic tracing of Tbx18. Tbx18 is commonly used as a marker for the sinus venosus and epicardium. Constitutive genetic tracing was performed with a Tbx18Cre in the R26RLacZ background and reporter gene expression was found in myocardial cells, which led to the conclusion that these cardiomyocytes derived from epicardial cells [77]. However, later experiments showed active expression of Tbx18 in cardiomyocytes, which was put forward as an explanation for the observed reporter gene expression [78]. Using constitutive genetic tracing, reporter gene expression in cells shows that either these cells have expressed or still actively express the gene of interest.
Even though timing is far more accurately determined using inducible genetic tracing, exact timing of recombination and reporter gene expression remains difficult. It was shown that after intraperitoneal injection of Tamoxifen at E8.5 in pregnant dams, the first signs of LacZ activity in the embryos were seen 6 h after injection, with an increase in expression at 12, 24 and 48 h [79]. Analysis of reporter gene expression after Tamoxifen injection therefore is dependent on the duration of Tamoxifen exposure, which complicates exact timing. Furthermore, the dosage of Tamoxifen has also been shown to influence reporter gene expression, with higher doses resulting in reporter gene expression in more cells [79]. Recent findings also revealed that the basal activity of Tamoxifen induced Cre (CreERT2) can be sufficient to induce genetic recombination even in the absence of tamoxifen induction [80]. Depending on the reporter line this may result in aspecific cell labelling [80]. This shows that analysis of reporter gene expression and interpretation of these results can be a challenge and might require further substantiation to guarantee efficacy.
The complete absence of reporter gene expression is also difficult to interpret as this can be caused by absence of expression of the gene of interest, or absence of recombination due to sub-threshold levels of Cre expression, not sufficient to induce reporter gene expression.
Recent studies might raise questions regarding the accuracy of using Cre driven genes as a reliable cell lineage marker. In genetic tracing experiments, interpreting specific gene expression can be problematic, but a far more complex problem arises in case of lack of reporter gene expression. A large number of genetic tracing experiments utilise the R26R locus, which is also the most commonly used reporter locus described in the current review. Genetic tracing experiments comparing the R26R locus with a Gata4 based reporter system showed differences in reporter gene expression between the two lines [73]. Constitutive genetic tracing of Isl1 and Nkx2-5 (both genes are discussed in more detail below) showed more extensive reporter gene expression in the Gata4 reporter line, indicating that the Gata4 locus was more sensitive to recombination then the R26R locus [73]. The difference in reporter gene expression shows that different reporter strains have different thresholds for reporter gene expression, resulting in different conclusions based on the reporter used. An example of the importance of this is shown by reassessment of the fate of Isl1+ progenitors [73]. This gene is commonly used as marker for the SHF [81,82]. However, the expanded pattern of reporter gene expression in the Gata4 reporter line showed that nearly all cells (including the LV, which is commonly described as being derived from the FHF) of the heart derive from progenitor cells that did express Isl1, albeit perhaps at a low level for a brief period. This suggest that Isl1 is either not specific enough or not reliable as a SHF marker [73]. In this case, mRNA [83] and protein [40] expression of Isl1 in the cardiac crescent, classically considered to be a FHF structure, further support these observations.
Therefore, when performing constitutive or inducible genetic tracing experiments, one should be cautious with interpretation of the results (Figure 4 illustrates this in more detail). In constitutive genetic tracing, reporter gene expression demonstrates that labelled cells have expressed or still actively express the promotor driven Cre. It does not provide information on the initial source/location of the progenitor cells. With inducible genetic tracing, temporal control of Cre expression is possible. Expression of the reporter gene shows that Cre is present in the labelled cell between the time of Tamoxifen administration and the moment of analysis. Finally, interpretation of negative results in genetic tracing is challenging and results need to be verified by additional tracing techniques and gene/protein expression experiments at sequential stages of development.

2.3.4. Using Genetic Lineage Tracing to Study OFT Development

Genetic lineage tracing has been applied extensively to identify cell lineages contributing to OFT formation. The early observations of neural crest derived cells contributing to the developing heart were further substantiated using the neural crest specific WntCre transgenic mouse (Figure 4A) [20]. This model, when crossed with a Cre-specific reporter line, allowed for lineage tracing of NCCs revealing their role in semilunar valve development as well as the cardiac conduction system [85]. The appearance of the OFT cushions and a role for endothelial cells was observed in early studies using electron microscopy [86]. However, the development of the Tie2Cre transgenic model was essential for the study of endothelial cell lineages in vivo (Figure 4B) [22]. Other genetic markers have been extensively used to define cell lineages such as the Isl1+ SHF [81,82] and the Mef2cCre SHF lineage model (Figure 4C) [87]. Table 1 shows an overview of genetic lineage tracing models that are often used in OFT studies, the morphological structures these lineages contribute to, and the possible conflicts arising from the genetic lineage interpretations.
Recent developments in genetic lineage tracing have introduced a dual genetic tracing system to address the dynamic origins of cardiac valve mesenchyme [88]. Dual genetic tracing allows for the study of multiple cell lineages simultaneously by using multiple site-specific recombinases such as Nigri-nox to complement the Cre-loxP system [89]. Dual genetic lineage tracing could aid in addressing the specificity concerns of traditional Cre-loxP lineage tracing systems.
Table 1. An overview of common genetic lineage tracing models used in OFT studies.
Table 1. An overview of common genetic lineage tracing models used in OFT studies.
Transgenic Mouse LinesCommonly Used as Cell Lineage Marker for:Observed Tissue ExpressionPossible Lineage Conflicts
Hcn4Cre-ert2First heart field [90] Cardiac conduction system [90]
Myocardium [90]
Second heart field
Hoxa1CreCardiac precursors [91]Aortopulmonary septum [91]
Cardiac conduction system [91]
Coronary arteries [91]
Endothelial lining [91]
Myocardium [91]
Semilunar valves [91]
Endothelial
Neural Crest
Second heart field
Isl1CreSecond heart field [81]Atrioventricular valves [73]
Cardiac conduction System [90]
Cushion mesenchyme [73]
Endocardium [73]
Myocardium [92]
Proepicardium [93]
Semilunar Valves [73]
Endothelial
Epicardial
First heart field
Neural Crest
Krox20CreNeural Crest [94]Endocardium [94]
Semilunar valves [94]
Endothelial
Mef2cCreSecond heart field [87] Ascending aorta [95]
Coronary arteries [96]
Cushion mesenchyme [95]
Endocardium [95]
Myocardium [87,95]
Semilunar valves [95]
Endothelial
Epicardial
Nkx2-5CreFirst and Second heart field [97]Ascending aorta [98]
Coronary artery [98]
Endocardium [73]
Epicardium [93]
Myocardium [97],
Semilunar valves [98]
Endothelial
Epicardial
Pax3CreNeural Crest [99]Aortopulmonary septum [100]
Ascending aorta [99]
Cushion mesenchyme [100]
Semilunar valves [100]
Tbx18CreProepicardium/epicardium [77]Cardiac conduction system [90]
Epicardium [77]
Myocardium [77]
First heart field
Second heart field
Tbx2CreProepicardium/epicardium [101]Cardiac conduction system [102]
Coronary arteries [101]
Epicardium [101]
Myocardium [101,103]
First heart field
Second heart field
Tie2CreEndothelium [22] Atrioventricular valves [22,104]
Coronary arteries [105]
Cushion mesenchyme [22,104]
Endocardium [22,104]
Hematopoietic cells [106]
Semilunar valves [104]
Hematopoietic
Tnnt2CreMyocardium [107]Ascending aorta [25]
Myocardium [25,107]
Semilunar valves [25]
Wnt11CreERCardiac precursors [108]Endocardium [108]
Epicardium [108]
Myocardium [108]
Semilunar valves [108]
First heart field
Endothelial
Epicardial
Second heart field
Wnt1CreNeural Crest [20]Aortopulmonary septum [20]
Ascending aorta [20]
Cardiac conduction system [109]
Coronary arteries [71]
Cushion mesenchyme [20]
Epicardium [110]
Myocardium [110,111]
Semilunar valves [20]
Epicardial
Second heart field
WT1CreProepicardium/epicardium [112]Coronary arteries [112]
Epicardium [93]
Myocardium [93]
Endothelial lineage
First heart field
Second heart field

2.4. Retrospective Clonal Analysis

The aforementioned techniques can all be described as prospective fate mapping techniques. Knowledge on gene and protein expression profile, timing of expression and/or location of progenitors is essential to perform subsequent prospective labelling of this progenitor pool [52]. The following section focuses on retrospective clonal analysis.

2.4.1. Basics of Retrospective Clonal Analysis

Retrospective clonal analysis is based on infrequent and spontaneous recombination of the nLaacZ reporter [113]. This reporter gene encodes the LacZ gene, with an inactivating duplication inserted in the reading frame, thereby inhibiting transcription of functional β-galactosidase. A spontaneous and rare recombination event can result in removal of the duplication and subsequent production of β-galactosidase (Figure 5A) [113]. Reporter gene expression can be followed in progeny of cells in which spontaneous recombination of the nLaacZ construct occurred [113]. Targeting of this construct to an allele which is highly expressed in cardiac muscle, (e.g., α-cardiac-actin) enables retrospective clonal analysis of cardiac cells, irrespective of gene expression [114,115,116].
Clonal analysis is based on statistical evaluation of the chance that reporter gene expression in different cells is based on separate spontaneous recombination events, or that these different cells are progeny of one recombination event (and thus share a common progenitor during development). A statistical method commonly used is the fluctuation test of Luria and Delbrück [117]. This famous experiment showed that mutations in bacteria occur spontaneously (not induced by selection) and at a constant rate, which was used to formulate a probability distribution [117]. This distribution is used to calculate the probability of one or more recombination events, and will therefore determine whether cells showing reporter gene expression are most likely clonally related or are derived from different recombination events (Figure 5B) [113,114,115,118]. Early recombination (i.e., during early stages of development) will give large clusters of cells with reporter gene expression (Figure 5C), while late spontaneous recombination will give smaller clusters of LacZ positive cells (Figure 5D).
Recent studies performing retrospective clonal analysis have been adopting next-generation sequencing techniques to study lineage tracing. Recent advances in single cell sequencing technologies allow transcriptome profiling of thousands of single cells. This allowed for the collection of large datasets detailing cellular expression profiles with unprecedented resolution. Alongside advances in sequencing technologies, computational methods designed to examine lineage trajectory reconstruction based on single-cell transcriptomics data have also evolved [119,120]. Single-cell transcriptomics allows for the investigation of the transcriptional state of thousands of individual single cells. As a result, cell-type diversity in heterogeneous samples can be reliably captured. As cells transition between different states during embryologic development, their expression profiles give insight into their lineage fate and cellular identity.
If sufficient amounts of cells in these transition states are captured, differentiation trajectories through which tissues are derived can be reconstructed through hierarchical clustering of cells based on the gradient in expressional similarities (Figure 5E).

2.4.2. Advantages of Retrospective Clonal Analysis

To perform prospective lineage tracing, some basic knowledge on the progenitor population that will be traced is required. This information is not necessary when performing retrospective clonal analysis. Studying whether certain structures or clusters of cells are clonally related is done independent of gene expression or any other preconceived idea of a possible progenitor pool. Retrospective clonal analysis can therefore establish clonal relationships that are less apparent at first sight, such as the clonal relationship between head musculature and cardiomyocytes from the outflow tract and right ventricle [121,122].
Furthermore, as described previously, prospective genetic tracing is hampered by the difficulty to draw conclusions from the absence of reporter gene expression. With the retrospective clonal approach, analysis is based on LacZ positive cells. LacZ expression in different structures or tissues is mapped and statistical analysis is performed to calculate whether these different tissues or structures are clonally related [113]. Moreover, as with genetic tracing, retrospective clonal analysis is non-invasive.
When performing retrospective lineage tracing using single-cell sequencing, an advantage is the ability to perform lineage tracing using naturally occurring somatic mutations or copy number variations (CNVs). CNVs can be used as genetic markers to link clones of cells to a common progenitor. This approach allows for lineage tracing without manual labelling or genetic modification. Even though CNVs are relatively uncommon in healthy tissue, they are highly abundant in cancerous tissue and have been used to study tumour evolution [123,124]. Alternative to CNVs is lineage tracing using single-nucleotide variations (SNVs) and genomic insertion/deletions (indels). Both SNVs and Indels are often present in non-coding regions of the genome and have been used for the reconstruction of phylogenetic trees of tumours from bulk DNA [125]. This technique, however, is not limited to tumour lineage analysis as lineage tracing using somatic SNVs in mitochondrial DNA can be performed with any eukaryotic cell [126]. Moreover, combining the genetic variation in non-coding regions of DNA with the RNA expression profiles when performing lineage tracing has recently been successfully demonstrated [127].

2.4.3. Limitations of Retrospective Clonal Analysis

The retrospective approach has several drawbacks. Since data is analysed retrospectively, it is not possible to locate the common progenitor, either in time or space. Therefore, it is not possible to perform further experiments with these progenitors (e.g., characterisation or ablation). Retrospective clonal analysis has to be performed in conjunction with other fate mapping techniques to build the complete lineage tree of a structure or organ. In this sense, the most important advantage of this technique is also its most important drawback. Starting with a certain progenitor pool based on timing, location and/or gene/protein expression profile excludes contributions from other (sometimes unexpected) progenitors but does directly pinpoint a possibly important progenitor pool for the structure/tissue of interest. Furthermore, spontaneous recombination of the nlaacZ gene is rare and it is therefore necessary to obtain a large library of embryos with LacZ positive clones and as a result, large quantities of embryos or adult mice will have to be analysed. A challenge when performing single cell sequencing for lineage tracing lies in the sparse distribution of SNVs and CNVs within the genome. To achieve confident detection of SNVs, large quantities of reads will have to be generated to achieve adequate sequencing depth. A large portion of the generated data is unsuitable for lineage tracing as these would contain identical sequences. However, enrichment of regions of DNA with greater odds of developing nucleotide variations such as microsatellites could apprehend these challenges.

2.4.4. Retrospective Clonal Analysis Aimed at the Development of the OFT

Many of the cardiac lineages contributing to OFT development have been found using prospective lineage tracing techniques (Table 1). However, a recent study examining cell populations at the aortic root and pulmonary trunk determined a novel SHF derived cell population called arterial mesothelial cells (AMC) populating a local niche at the base of the great arteries [128]. Whilst AMCs have first been observed as a distinctive cell type using chick-quail chimera’s [129], the study of Lioux et al. used a clonal analysis strategy based on the ubiquitous, low-frequency random recombination of two independent reporters to distinguish this population from the frequency of colour matching between labelled cells in different clusters or different cell types within single clusters [130].This approach allowed for an unbiased characterisation of the fate of SHF precursors at the base of the great arteries and detailed the contribution of SHF to the coronary lymphatic vasculature [128]. Moreover, another study examining the cardiac outflow tract using single cell RNA sequencing (scRNA-seq) found convergent development of the vascular smooth muscle cell lineage suggesting a method of myocardial-to-VSMC differentiation or mesenchymal-to-VSMC differentiation [131].

3. New Technologies for Lineage Tracing in OFT Development

Lineage tracing has contributed greatly to our understanding of OFT development. Nevertheless, there is still much to be learned regarding multifactorial CHDs. To advance our understanding of phenotypical variation, research efforts should be aimed at understanding the relation between cell lineage and cellular identity. The methods previously described in this review are inadequate to address such issues, as this would require detailed description of the variation among single cells within a lineage population. The introduction of novel analysis methods which combine lineage tracing and single cell analysis could provide new opportunities to examine cell lineage plasticity and phenotypical variation in CHD. These methods will be discussed in the following section.

3.1. Spatially Resolved Transcriptomics

Current lineage tracing techniques, such as genetic lineage tracing, rely primarily on spatial information to reconstruct OFT development over multiple embryonic stages. The tracking of cell lineage movements can give insight into the environment in which these cells function. However, evaluating the internal cellular state of these cells within such environments can be challenging. Single cell RNA sequencing (scRNA-seq) is a technique which can be used to examine a cell’s innerworkings because it allows for quantification of all the RNA molecules within a single cell. This cellular transcriptome can be used to examine gene expression patterns and to make cell type specific expression profiles. By collecting expression profiles of multiple cells of the same cell type, it is possible to examine the cellular variation in gene expression within a specific cell population. This allows researchers to evaluate the variation in cell states within a particular cell lineage, and reconstruct subclusters structural to a cell lineage population. Whilst scRNA-seq can be used to obtain genome-wide expression data of cells allowing for unprecedented cell type identification, it does not provide any spatial resolution to determine where these cells were located within the embryo. As a result, experiments using scRNA-seq to identify new cell types and cell states still require the use of visualisation techniques such as in-situ hybridisation using the genetic markers found during sequencing. However, in-situ hybridisation can only provide spatial information regarding a few genes at any given moment. Tomo-seq is a technique which combines genome-wide expression data with spatial information [132,133]. This is achieved by performing RNA-seq on individual cryosections of an embryo or tissue allowing the generation of genome-wide spatial expression maps in 3D. By sequencing sequential tissue sections, spatial information regarding the head to tail axis can be maintained. As a result, gene expression data can be projected with respect to the head to tail axis. Performing this approach using multiple axis (dorsal-ventral, right-left) allows for the reconstruction of gene expression patterns in 3D in the developing embryo [133]. Although Tomo-seq allows for 3D expression pattern analysis, one limitation is that it is unable to provide single cell resolution in 3D. An alternative method called transcriptome in vivo analysis (TIVA) does allow for single cell sequencing whilst maintaining single cell spatial resolution. TIVA is a noninvasive tool enabling capture of mRNA from single spatially defined cells in living and intact tissue for transcriptome analysis [134]. This is achieved by addition of a cell permeable TIVA tag to the tissue sample which upon photo activation is able to capture cellular mRNA. This technique is an excellent advance over more traditional laser capture techniques or pipette cell isolation adapted for scRNA-seq due to the non-invasive design. The limited throughput, however, might restrict explorative research to specific cases in which the number of cells of interest might be relatively low. A practical example in which TIVA might prove very valuable for OFT studies, is to examine the properties of valvular interstitial cells (VICs) contributing to the aortic and pulmonary valves. VICs may derive from multiple cell lineages such as SHF, NCC or endothelial lineages and VIC dysfunction has been suggested to underly valvular diseases such as calcification [135]. TIVA could enable researchers to examine how cell lineage background influence VIC formation and how cell lineage deficiencies could relate to valvular disease by assigning a spatial criterium to scRNA-seq.

3.2. Lineage Tracing through DNA Barcoding

DNA-barcoding technologies are another potential tool for lineage tracing. Lineage tracing using DNA barcoding is a technique that combines aspects of both prospective and retrospective lineage tracing. Similar to genetic lineage tracing using fluorescent reporters, DNA-barcoding technologies introduce genetic modifications into progenitor cells to study cell lineage development. However, whilst lineage tracing using fluorescent reporters is limited to only a number of reporter genes, DNA barcodes allow for more complex labelling methods. Techniques such as scGESTALT [136], ScarTrace [137] and LINNAEUS [138] have demonstrated how the CRISPR-Cas9 system could be used to generate genetic barcodes to perform whole-organism lineage tracing in zebrafish embryos. The CRISPR-Cas9 system allows genomic editing on a specific DNA locus determined by the supplied guide RNA. In the absence of a template for homologous repair, Cas9 may introduce short DNA insertions or deletions at the targeted site [139]. These genetic aberrations, which vary in size and position, can then be used as a genetic barcoded label to evaluate a cell’s offspring. If the guide RNA was designed specific to a region within a constitutive expressed gene, then scRNA-seq can capture the barcode within the transcriptome of a cell. Since the process of barcoding is randomly determined, a cell’s lineage can only be evaluated after single cell sequencing. This can be achieved through computational reconstruction of full lineage trees from the single cell level. Even though these methods still rely on microinjection of Cas9 and guide RNAs in an early stage zebrafish embryos, DNA-barcoding has also been achieved using retroviral integration [140] or polylox recombination [141] in mice. For an in dept comparison of barcoding techniques, the reader is referred to a recent review of Wagner and Klein [142]. Lineage tracing using barcodes could be an excellent method to examine cell lineage commitment because it allows to retrospectively evaluate cell lineage branching events. The differences in lineage trajectories among FHF and SHF lineages would be an interesting topic to be evaluated using such methods. Moreover, if barcoding events are temporally restricted and multiple stages of embryonic development are analysed, then it could enable reconstruction of all cell lineage trajectories contributing to heart development simultaneously. Genetic mutations or epigenetic influences affecting cellular identity could also be examined as these would influence the relative distributions among (sub)populations of cell lineages. As such, these methods could aim to elucidate the relationship between phenotypical variation and cell lineage variation.

3.3. Multi-Omics Lineage Tracing

Whilst barcoding techniques for lineage tracing require aspects of a prospective approach to lineage tracing, recent technological developments using a multi-omics sequencing approach for lineage tracing could allow for complete retrospective lineage tracing. RETrace is an example of a recent lineage tracing technique which captures both DNA microsatellite loci and methylation-informative cytosines from a single cell in order to simultaneously characterise both lineage and cell type [143]. The utilisation of microsatellite regions as markers for retrospective lineage tracing was successfully demonstrated at single cell level [144]. However, enabling DNA methylation profiling of the same cell allows for better cell characterisation than either method alone. Even though still in early development, a multi-omics approach to lineage tracing would ideally combine the cell lineage identification capability using natural occurring CNV and SNVs captured using whole genome sequencing whilst allowing for accurate cell type identification through gene expression analysis using scRNA-seq and also include epigenetic profiling techniques such as methylome analysis [145] or include histone signatures [146] to investigate cellular identity and cell lineage dynamics. Multi-omics lineage tracing would enable researchers to investigate the relations between cellular variation, epigenetic modification and disease susceptibility related to CHD. This knowledge could potentially translate to novel methods of treatment and therapy aimed at influencing the epigenome of patients.

3.4. Reference Maps for Lineage Tracing

The large amounts of data derived from lineage tracing using transcriptomics are likely to extent the depth at which sub-lineages and cell states will be defined. To facilitate accurate definitions of such sub lineages and intermediate cell states, large quantities of single cell data could be combined. A project called the Human Cell Atlas (HCA) aims to characterise all human cell types using scRNA-seq and single cell transposase accessible chromatin sequencing (scATAC-seq), and combine it with data regarding cell lineage, location and cell states [147]. The scope extents to unify single cell sequencing data towards common reference models similar to the efforts of the Human Genome Project [148]. Having the infrastructure to navigate cell lineage development will greatly contribute to the understanding of human health and treatment of disease. (For more information regarding the HCA project, the reader is referred to the HCA white paper [149]). Initiatives such as these could allow for the comparison of many cell lineage populations among the various types of CHD to determine key processes critical to human cardiovascular development.

4. Future Perspectives, Combining Old Strengths with New Technologies

With the advances in lineage tracing, new insights regarding cell fate came forth and the role of prospective lineage tracing paved the way for modern developmental biology. In this review we describe the use of lineage tracing techniques and their limitations with respect to OFT development. The recent technological methods described in the final section of this review have the potential to allow more fateful reconstruction of lineage dynamics than previous methods.
The development of next-generation sequencing technology has enabled new and exciting research opportunities for lineage tracing experiments. From our perspective we expect that results obtained from retrospective lineage tracing using naturally occurring mutations and single cell transcriptomics will challenge our current definitions of cell lineages and advance our understanding of cellular dynamics and lineage plasticity. Assuming technological improvements will continue to be developed in the coming years, we can expect an integration of (epi)genetics signatures with single cell sequencing to aid with tracing cellular heritage. Continued development and a multi-omics approach will allow future research to expand the scope of cellular identity and resolve the mechanisms explaining congenital variation in more detail and with better accuracy than current available methods. As a result, a multi-omics approach will help to elucidate the mechanisms for normal OFT development and aid in understanding the pathophysiological pathways leading to the multivariate phenotypes common to OFT diseases. As sequencing techniques become more and more accessible, multi-omics sequencing approaches combined with single cell resolution will become the new frontier for future lineage tracing studies.

Author Contributions

J.C.P. and T.P.K. drafted the manuscript and designed the figures. M.C.D., M.R.M.J. and M.J.T.H.G. were involved in planning, supervision and critical review of the manuscript. All authors were involved in the assessment of relevant literature and substantially contributed to the conception and design of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Funding and salary support were provided by The Dutch Heart foundation (Project code: 31190BAV).

Acknowledgments

We are very grateful for all the support provided by Adriana C. Gittenberger-de Groot. Her work fueled our passions for cardiac development and congenital heart disease. The rigorous scientific discussions carry warm memories and helped pave the road to academic excellence. We thank her for all her support throughout the years and the care she took for all her colleagues and collaborators.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMCarterial mesothelial cells
SHFsecond heart field
AVCatrioventricular canal
SNVsingle nucleotide variation
CHDcongenital heart disease
TIVAtranscriptome in vivo analysis
CNVcopy number variations
VICvalvular interstitial cell
Eembryonic day
ERestrogen receptor
FHFfirst heart field
GFPgreen fluorescent protein
HHHamburger and Hamilton
HCAHuman Cell Atlas
LBDligand-binding domain
mT/mGmembrane-bound Tomato/membrane-bound GFP
NCCsneural crest cells
OFToutflow tract
PHTprimary heart tube
R26RRosa26 reporter
scRNA-seqsingle cell RNA sequencing

References

  1. Thompson, P. Description of a Human Embryo of Twenty-Three Paired Somites. J. Anat. Physiol. 1907, 41, 159–171. [Google Scholar] [PubMed]
  2. Garg, V.; Muth, A.N.; Ransom, J.F.; Schluterman, M.K.; Barnes, R.; King, I.N.; Grossfeld, P.D.; Srivastava, D. Mutations in NOTCH1 cause aortic valve disease. Nature 2005, 437, 270–274. [Google Scholar] [CrossRef]
  3. Laforest, B.; Andelfinger, G.; Nemer, M. Loss of Gata5 in mice leads to bicuspid aortic valve. J. Clin. Investig. 2011, 121, 2876–2887. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Peterson, J.C.; Chughtai, M.; Wisse, L.J.; Gittenberger-de Groot, A.C.; Feng, Q.; Goumans, M.-J.T.H.; VanMunsteren, J.C.; Jongbloed, M.R.M.; DeRuiter, M.C. Bicuspid aortic valve formation: Nos3 mutation leads to abnormal lineage patterning of neural crest cells and the second heart field. Dis. Model. Mech. 2018, 11, 655–658. [Google Scholar] [CrossRef] [Green Version]
  5. Klemm, S.L.; Shipony, Z.; Greenleaf, W.J. Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 2019, 20, 207–220. [Google Scholar] [CrossRef] [PubMed]
  6. Perino, M.; Veenstra, G.J.C. Chromatin Control of Developmental Dynamics and Plasticity. Dev. Cell 2016, 38, 610–620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Fisher, A.G. Cellular identity and lineage choice. Nat. Rev. Immunol. 2002, 2, 977–982. [Google Scholar] [CrossRef]
  8. Peaston, A.E.; Whitelaw, E. Epigenetics and phenotypic variation in mammals. Mamm. Genome 2006, 17, 365–374. [Google Scholar] [CrossRef] [Green Version]
  9. Merrell, A.J.; Stanger, B.Z. Adult cell plasticity in vivo: De-differentiation and transdifferentiation are back in style. Nat. Rev. Mol. Cell Biol. 2016, 17, 413–425. [Google Scholar] [CrossRef]
  10. Olson, E.N.; Srivastava, D. Molecular pathways controlling heart development. Science 1996, 272, 671–676. [Google Scholar] [CrossRef]
  11. Laverriere, A.C.; MacNeill, C.; Mueller, C.; Poelmann, R.E.; Burch, J.B.E.; Evans, T. GATA-4/5/6, a subfamily of three transcription factors transcribed in developing heart and gut. J. Biol. Chem. 1994, 269, 23177–23184. [Google Scholar] [CrossRef]
  12. Buckingham, M.; Meilhac, S.; Zaffran, S. Building the mammalian heart from two sources of myocardial cells. Nat. Rev. Genet. 2005, 6, 826–835. [Google Scholar] [CrossRef]
  13. Abu-Issa, R.; Kirby, M.L. Heart field: From mesoderm to heart tube. Annu. Rev. Cell Dev. Biol. 2007, 23, 45–68. [Google Scholar] [CrossRef] [PubMed]
  14. DeRuiter, M.C.; Poelmann, R.E.; VanderPlas-de Vries, I.; Mentink, M.M.T.; Gittenberger-de Groot, A.C. The development of the myocardium and endocardium in mouse embryos—Fusion of two heart tubes? Anat. Embryol. 1992, 185, 461–473. [Google Scholar] [CrossRef] [PubMed]
  15. Tyser, R.C.V.; Srinivas, S. The first heartbeat—Origin of cardiac contractile activity. Cold Spring Harb. Perspect. Biol. 2020, 12, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Moorman, A.F.M.; Christoffels, V.M. Cardiac chamber formation: Development, genes, and evolution. Physiol. Rev. 2003. [Google Scholar] [CrossRef]
  17. Combs, M.D.; Yutzey, K.E. Heart valve development: Regulatory networks in development and disease. Circ. Res. 2009, 105, 408–421. [Google Scholar] [CrossRef] [Green Version]
  18. Kirby, M.L. Cardiac Development; Oxford University Press: Oxford, UK, 2007. [Google Scholar]
  19. Waldo, K.; Miyagawa-Tomita, S.; Kumiski, D.; Kirby, M.L. Cardiac neural crest cells provide new insight into septation of the cardiac outflow tract: Aortic sac to ventricular septal closure. Dev. Biol. 1998, 196, 129–144. [Google Scholar] [CrossRef]
  20. Jiang, X.; Rowitch, D.H.; Soriano, P.; McMahon, A.P.; Sucov, H.M. Fate of the mammalian cardiac neural crest. Development 2000, 127, 1607–1616. [Google Scholar] [CrossRef]
  21. Eisenberg, L.M.; Markwald, R.R. Molecular regulation of atrioventricular valvuloseptal morphogenesis. Circ. Res. 1995, 77, 1–6. [Google Scholar] [CrossRef]
  22. Kisanuki, Y.Y.; Hammer, R.E.; Miyazaki, J.; Williams, S.C.; Richardson, J.A.; Yanagisawa, M. Tie2-Cre transgenic mice: A new model for endothelial cell-lineage analysis in vivo. Dev. Biol. 2001, 230, 230–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Zaffran, S.; Kelly, R.G. New developments in the second heart field. Differentiation 2012, 84, 17–24. [Google Scholar] [CrossRef]
  24. Eley, L.; Alqahtani, A.M.S.; Macgrogan, D.; Richardson, R.V.; Murphy, L.; Salguero-jimenez, A.; Sintes, M.; San, R.; Tiurma, S.; Mccutcheon, L.; et al. A novel source of arterial valve cells linked to bicuspid aortic valve without raphe in mice. eLife 2018, 7, 1–27. [Google Scholar] [CrossRef]
  25. Mifflin, J.J.; Dupuis, L.E.; Alcala, N.E.; Russell, L.G.; Kern, C.B. Intercalated cushion cells within the cardiac outflow tract are derived from the myocardial troponin T type 2 (Tnnt2) Cre lineage. Dev. Dyn. 2018, 247, 1005–1017. [Google Scholar] [CrossRef] [PubMed]
  26. Miquerol, L.; Kelly, R.G. Organogenesis of the vertebrate heart. Wiley Interdiscip. Rev. Dev. Biol. 2013. [Google Scholar] [CrossRef]
  27. Scherptong, R.W.; Jongbloed, M.R.; Wisse, L.J.; Vicente-Steijn, R.; Bartelings, M.M.; Poelmann, R.E.; Schalij, M.J.; Gittenberger-De Groot, A.C. Morphogenesis of outflow tract rotation during cardiac development: The pulmonary push concept. Dev. Dyn 2012, 241, 1413–1422. [Google Scholar] [CrossRef]
  28. Gittenberger-De Groot, A.C.; Bartelings, M.M.; Deruiter, M.C.; Poelmann, R.E. Basics of cardiac development for the understanding of congenital heart malformations. Pediatric Res. 2005, 57, 169–176. [Google Scholar] [CrossRef] [Green Version]
  29. Gittenberger-de Groot, A.C.; Bartelings, M.M.; Poelmann, R.E.; Haak, M.C.; Jongbloed, M.R.M. Embryology of the heart and its impact on understanding fetal and neonatal heart disease. Semin. Fetal Neonatal Med. 2013, 18, 237–244. [Google Scholar] [CrossRef]
  30. Gittenberger-de Groot, A.C.; Calkoen, E.E.; Poelmann, R.E.; Bartelings, M.M.; Jongbloed, M.R.M. Morphogenesis and molecular considerations on congenital cardiac septal defects. Ann. Med. 2014, 46, 640–652. [Google Scholar] [CrossRef] [PubMed]
  31. Bartelings, M.M.; Gittenberger-de Groot, A.C. Morphogenetic considerations on congenital malformations of the outflow tract. Part 1: Common arterial trunk and tetralogy of Fallot. Int. J. Cardiol. 1991, 32, 213–230. [Google Scholar] [CrossRef]
  32. Poelmann, R.E.; Gittenberger-de Groot, A.C.; Biermans, M.W.M.; Dolfing, A.I.; Jagessar, A.; van Hattum, S.; Hoogenboom, A.; Wisse, L.J.; Vicente-Steijn, R.; de Bakker, M.A.G.; et al. Outflow tract septation and the aortic arch system in reptiles: Lessons for understanding the mammalian heart. Evodevo 2017, 8, 9. [Google Scholar] [CrossRef] [Green Version]
  33. Poelmann, R.E.; Mikawa, T.; Gittenberger-De Groot, A.C. Neural crest cells in outflow tract septation of the embryonic chicken heart: Differentiation and apoptosis. Dev. Dyn. 1998, 212, 373–384. [Google Scholar] [CrossRef]
  34. Kirby, M.L.; Gale, T.F.; Stewart, D.E. Neural crest cells contribute to normal aorticopulmonary septation. Science 1983, 220, 1059–1061. [Google Scholar] [CrossRef]
  35. Anderson, R.; Mohun, T.; Spicer, D.; Bamforth, S.; Brown, N.; Chaudhry, B.; Henderson, D. Myths and Realities Relating to Development of the Arterial Valves. J. Cardiovasc. Dev. Dis. 2014, 1, 177–200. [Google Scholar] [CrossRef] [Green Version]
  36. Gittenberger-de Groot, A.C.; Bartelings, M.M.; Bogers, A.J.J.C.; Boot, M.J.; Poelmann, R.E. The embryology of the common arterial trunk. Prog. Pediatric Cardiol. 2002, 15, 1–8. [Google Scholar] [CrossRef]
  37. DeHaan, R.L. Migration patterns of the precardiac mesoderm in the early chick embryo. Exp. Cell Res. 1963, 29, 544–560. [Google Scholar] [CrossRef]
  38. Davis, C. Development of the human heart from its first appearance to the stage found in embryos of twenty paired somites. Carnegie Inst. Contr. Embryol. 1927, 19, 245–284. [Google Scholar]
  39. Axelrod, D. Carbocyanine dye orientation in red cell membrane studied by microscopic fluorescence polarization. Biophys. J. 1979, 26, 557–573. [Google Scholar] [CrossRef] [Green Version]
  40. Kelder, T.P.; Vicente-Steijn, R.; Harryvan, T.J.T.; Kosmidis, G.; Gittenberger-de Groot, A.C.; Poelmann, R.R.E.; Schalij, M.M.J.; DeRuiter, M.M.C.; Jongbloed, M.R.M. The sinus venosus myocardium contributes to the atrioventricular canal: Potential role during atrioventricular node development? J. Cell. Mol. Med. 2015, 19, 1375–1389. [Google Scholar] [CrossRef] [PubMed]
  41. Sanes, J.R. Analysing cell lineage with a recombinant retrovirus. Trends Neurosci. 1989, 12, 21–28. [Google Scholar] [CrossRef]
  42. Sanes, J.R.; Rubenstein, J.L.; Nicolas, J.F. Use of a recombinant retrovirus to study post-implantation cell lineage in mouse embryos. EMBO J. 1986, 5, 3133–3142. [Google Scholar] [CrossRef]
  43. Price, J.; Turner, D.; Cepko, C. Lineage analysis in the vertebrate nervous system by retrovirus-mediated gene transfer. Proc. Natl. Acad. Sci. USA 1987, 84, 156–160. [Google Scholar] [CrossRef] [Green Version]
  44. Pacak, C.A.; Byrne, B.J. AAV vectors for cardiac gene transfer: Experimental tools and clinical opportunities. Mol. Ther. 2011, 19, 1582–1590. [Google Scholar] [CrossRef] [Green Version]
  45. Stainier, D.Y.R.; Lee, R.K.; Fishman, M.C. Cardiovascular development in the zebrafish: I. Myocardial fate map and heart tube formation. Development 1993, 119, 31–40. [Google Scholar] [CrossRef]
  46. Krotoski, D.M.; Fraser, S.E.; Bronner-Fraser, M. Mapping of neural crest pathways in Xenopus laevis using inter- and intra-specific cell markers. Dev. Biol. 1988, 127, 119–132. [Google Scholar] [CrossRef]
  47. De la Cruz, M.V.; Sánchez Gómez, C.; Arteaga, M.M.; Argüello, C. Experimental study of the development of the truncus and the conus in the chick embryo. J. Anat. 1977, 123, 661–686. [Google Scholar]
  48. Kelder, T.P.; Vicente-Steijn, R.; Poelmann, R.E.; Mummery, C.L.; DeRuiter, M.C.; Jongbloed, M.R.M. The avian embryo to study development of the cardiac conduction system. Differentiation 2016, 91, 90–103. [Google Scholar] [CrossRef]
  49. Domínguez, J.N.; Meilhac, S.M.; Bland, Y.S.; Buckingham, M.E.; Brown, N.A. Asymmetric fate of the posterior part of the second heart field results in unexpected left/right contributions to both poles of the heart. Circ. Res. 2012, 111, 1323–1335. [Google Scholar] [CrossRef] [Green Version]
  50. Honig, M.G.; Hume, R.I. Fluorescent carbocyanine dyes allow living neurons of identified origin to be studied in long-term cultures. J. Cell Biol. 1986, 103, 171–187. [Google Scholar] [CrossRef] [PubMed]
  51. Progatzky, F.; Dallman, M.J.; Lo Celso, C. From seeing to believing: Labelling strategies for in vivo cell-tracking experiments. Interface Focus 2013, 3, 20130001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Buckingham, M.E.; Meilhac, S.M. Tracing cells for tracking cell lineage and clonal behavior. Dev. Cell 2011, 21, 394–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Bressan, M.; Liu, G.; Mikawa, T. Early mesodermal cues assign avian cardiac pacemaker fate potential in a tertiary heart field. Science 2013, 340, 744–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Lo Celso, C.; Fleming, H.E.; Wu, J.W.; Zhao, C.X.; Miake-Lye, S.; Fujisaki, J.; Côté, D.; Rowe, D.W.; Lin, C.P.; Scadden, D.T. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature 2009, 457, 92–96. [Google Scholar] [CrossRef] [Green Version]
  55. Sipkins, D.A.; Wei, X.; Wu, J.W.; Runnels, J.M.; Côté, D.; Means, T.K.; Luster, A.D.; Scadden, D.T.; Lin, C.P. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 2005, 435, 969–973. [Google Scholar] [CrossRef]
  56. Honig, M.G.; Hume, R.I. Dil and diO: Versatile fluorescent dyes for neuronal labelling and pathway tracing. Trends Neurosci. 1989, 12, 333–335, 340–341. [Google Scholar] [CrossRef] [Green Version]
  57. Mikawa, T.; Hyer, J.; Itoh, N.; Wei, Y. Retroviral vectors to study cardiovascular development. Trends Cardiovasc. Med. 1996, 6, 79–86. [Google Scholar] [CrossRef]
  58. Barbosky, L.; Lawrence, D.K.; Karunamuni, G.; Wikenheiser, J.C.; Doughman, Y.-Q.; Visconti, R.P.; Burch, J.B.E.; Watanabe, M. Apoptosis in the developing mouse heart. Dev. Dyn. 2006, 235, 2592–2602. [Google Scholar] [CrossRef]
  59. Schaefer, K.S.; Doughman, Y.Q.; Fisher, S.A.; Watanabe, M. Dynamic patterns of apoptosis in the developing chicken heart. Dev. Dyn. 2004, 229, 489–499. [Google Scholar] [CrossRef]
  60. Le Douarin, N. Particularites du noyau interphasique chez la caille Japonaise (Coturnix coturnix japonica). Bull. Biol. Fr. Belg. 1969, 103, 435–452. [Google Scholar]
  61. Le Douarin, N. A biological cell labeling technique and its use in experimental embryology. Dev. Biol. 1973, 30, 217–222. [Google Scholar] [CrossRef]
  62. Le Douarin, N.M. The avian embryo as a model to study the development of the neural crest: A long and still ongoing story. Mech. Dev. 2004, 121, 1089–1102. [Google Scholar] [CrossRef]
  63. Selleck, M.A.; Bronner-Fraser, M. Origins of the avian neural crest: The role of neural plate-epidermal interactions. Development 1995, 121, 525–538. [Google Scholar] [CrossRef]
  64. Koga, M.; Kageura, H.; Yamana, K. Use of Hybrids between Xenopus laevis and Xenopus borealis in Chimera Formation: Dorsalization of Ventral Cells: (cell lineage/chimera/hybrid/Xenopus/dorsalization). Dev. Growth Differ. 1986, 28, 177–183. [Google Scholar] [CrossRef]
  65. Tam, P.P.L.; Rossant, J. Mouse embryonic chimeras: Tools for studying mammalian development. Development 2003, 130, 6155–6163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sereti, K.I.; Nguyen, N.B.; Kamran, P.; Zhao, P.; Ranjbarvaziri, S.; Park, S.; Sabri, S.; Engel, J.L.; Sung, K.; Kulkarni, R.P.; et al. Analysis of cardiomyocyte clonal expansion during mouse heart development and injury. Nat. Commun. 2018, 9, 1–13. [Google Scholar] [CrossRef] [PubMed]
  67. Le Lievre, C.S.; Le Douarin, N.M. Mesenchymal derivatives of the neural crest: Analysis of chimaeric quail and chick embryos. J. Embryol. Exp. Morphol. 1975, 34, 125–154. [Google Scholar] [PubMed]
  68. Baroffio, A.; Dupin, E.; Le Douarin, N.M. Common precursors for neural and mesectodermal derivatives in the cephalic neural crest. Development 1991, 112, 301–305. [Google Scholar] [CrossRef] [PubMed]
  69. Takamura, K.; Okishima, T.; Ohdo, S.; Hayakawa, K. Association of cephalic neural crest cells with cardiovascular development, particularly that of the semilunar valves. Anat. Embryol. 1990, 182, 263–272. [Google Scholar] [CrossRef] [PubMed]
  70. Rosenquist, T.H.; McCoy, J.R.; Waldo, K.L.; Kirby, M.L. Origin and propagation of elastogenesis in the developing cardiovascular system. Anat. Rec. 1988, 221, 860–871. [Google Scholar] [CrossRef] [PubMed]
  71. Arima, Y.; Miyagawa-Tomita, S.; Maeda, K.; Asai, R.; Seya, D.; Minoux, M.; Rijli, F.M.; Nishiyama, K.; Kim, K.S.; Uchijima, Y.; et al. Preotic neural crest cells contribute to coronary artery smooth muscle involving endothelin signalling. Nat. Commun. 2012, 3, 1–11. [Google Scholar] [CrossRef] [Green Version]
  72. Sauer, B. Inducible gene targeting in mice using the Cre/lox system. Methods 1998, 14, 381–392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ma, Q.; Zhou, B.; Pu, W.T. Reassessment of Isl1 and Nkx2-5 cardiac fate maps using a Gata4-based reporter of Cre activity. Dev. Biol. 2008, 323, 98–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Feil, R.; Brocard, J.; Mascrez, B.; LeMeur, M.; Metzger, D.; Chambon, P. Ligand-activated site-specific recombination in mice. Proc. Natl. Acad. Sci. USA 1996, 93, 10887–10890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Indra, A.K.; Warot, X.; Brocard, J.; Bornert, J.M.; Xiao, J.H.; Chambon, P.; Metzger, D. Temporally-controlled site-specific mutagenesis in the basal layer of the epidermis: Comparison of the recombinase activity of the tamoxifen-inducible Cre-ER(T) and Cre-ER(T2) recombinases. Nucleic Acids Res. 1999, 27, 4324–4327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Livet, J.; Weissman, T.A.; Kang, H.; Draft, R.W.; Lu, J.; Bennis, R.A.; Sanes, J.R.; Lichtman, J.W. Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system. Nature 2007, 450, 56–62. [Google Scholar] [CrossRef]
  77. Cai, C.-L.; Martin, J.C.; Sun, Y.; Cui, L.; Wang, L.; Ouyang, K.; Yang, L.; Bu, L.; Liang, X.; Zhang, X.; et al. A myocardial lineage derives from Tbx18 epicardial cells. Nature 2008, 454, 104–108. [Google Scholar] [CrossRef] [Green Version]
  78. Christoffels, V.M.; Grieskamp, T.; Norden, J.; Mommersteeg, M.T.M.; Rudat, C.; Kispert, A. Tbx18 and the fate of epicardial progenitors. Nature 2009, 458, E8–E9. [Google Scholar] [CrossRef]
  79. Hayashi, S.; McMahon, A.P. Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: A tool for temporally regulated gene activation/inactivation in the mouse. Dev. Biol. 2002, 244, 305–318. [Google Scholar] [CrossRef] [Green Version]
  80. Álvarez-Aznar, A.; Martínez-Corral, I.; Daubel, N.; Betsholtz, C.; Mäkinen, T.; Gaengel, K. Tamoxifen-independent recombination of reporter genes limits lineage tracing and mosaic analysis using CreERT2 lines. Transgenic Res. 2020, 29, 53–68. [Google Scholar] [CrossRef] [Green Version]
  81. Cai, C.-L.; Liang, X.; Shi, Y.; Chu, P.-H.; Pfaff, S.L.; Chen, J.; Evans, S. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev. Cell 2003, 5, 877–889. [Google Scholar] [CrossRef] [Green Version]
  82. Sun, Y.; Liang, X.; Najafi, N.; Cass, M.; Lin, L.; Cai, C.-L.; Chen, J.; Evans, S.M. Islet 1 is expressed in distinct cardiovascular lineages, including pacemaker and coronary vascular cells. Dev. Biol. 2007, 304, 286–296. [Google Scholar] [CrossRef] [Green Version]
  83. Yuan, S.; Schoenwolf, G.C. Islet-1 marks the early heart rudiments and is asymmetrically expressed during early rotation of the foregut in the chick embryo. Anat. Rec. 2000, 260, 204–207. [Google Scholar] [CrossRef]
  84. Baardman, M.E.; Zwier, M.V.; Wisse, L.J.; Gittenberger-De Groot, A.C.; Kerstjens-Frederikse, W.S.; Hofstra, R.M.W.; Jurdzinski, A.; Hierck, B.P.; Jongbloed, M.R.M.; Berger, R.M.F.; et al. Common arterial trunk and ventricular non-compaction in Lrp2 knockout mice indicate a crucial role of LRP2 in cardiac development. DMM Dis. Model. Mech. 2016, 9, 413–425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Nakamura, T.; Colbert, M.C.; Robbins, J. Neural crest cells retain multipotential characteristics in the developing valves and label the cardiac conduction system. Circ. Res. 2006, 98, 1547–1554. [Google Scholar] [CrossRef] [Green Version]
  86. Bolender, D.L.; Markwald, R.R. Epithelial-mesenchymal transformation in chick atrioventricular cushion morphogenesis. Scan. Electron. Microsc. 1979, 3, 313–321. [Google Scholar]
  87. Verzi, M.P.; McCulley, D.J.; De Val, S.; Dodou, E.; Black, B.L. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev. Biol. 2005, 287, 134–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Liu, K.; Yu, W.; Tang, M.; Tang, J.; Liu, X.; Liu, Q.; Li, Y.; He, L.; Zhang, L.; Evans, S.M.; et al. A dual genetic tracing system identifies diverse and dynamic origins of cardiac valve mesenchyme. Development 2018, 145. [Google Scholar] [CrossRef] [Green Version]
  89. Liu, K.; Jin, H.; Zhou, B. Genetic lineage tracing with multiple DNA recombinases: A user’s guide for conducting more precise cell fate mapping studies. J. Biol. Chem. 2020, 295, 6413–6424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Liang, X.; Wang, G.; Lin, L.; Lowe, J.; Zhang, Q.; Bu, L.; Chen, Y.; Chen, J.; Sun, Y.; Evans, S.M. HCN4 dynamically marks the first heart field and conduction system precursors. Circ. Res. 2013, 113, 399–407. [Google Scholar] [CrossRef]
  91. Makki, N.; Capecchi, M.R. Hoxa1 lineage tracing indicates a direct role for Hoxa1 in the development of the inner ear, the heart, and the third rhombomere. Dev. Biol. 2010, 341, 499–509. [Google Scholar] [CrossRef] [Green Version]
  92. Yang, L.; Cai, C.L.; Lin, L.; Qyang, Y.; Chung, C.; Monteiro, R.M.; Mummery, C.L.; Fishman, G.I.; Cogen, A.; Evans, S. Isl1 Cre reveals a common Bmp pathway in heart and limb development. Development 2006, 133, 1575–1585. [Google Scholar] [CrossRef] [Green Version]
  93. Zhou, B.; Ma, Q.; Rajagopal, S.; Wu, S.M.; Domian, I.; Rivera-Feliciano, J.; Jiang, D.; von Gise, A.; Ikeda, S.; Chien, K.R.; et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 2008, 454, 109–113. [Google Scholar] [CrossRef] [Green Version]
  94. Odelin, G.; Faure, E.; Coulpier, F.; Di Bonito, M.; Bajolle, F.; Studer, M.; Avierinos, J.-F.; Charnay, P.; Topilko, P.; Zaffran, S. Krox20 defines a subpopulation of cardiac neural crest cells contributing to arterial valves and bicuspid aortic valve. Development 2017, 145, dev151944. [Google Scholar] [CrossRef] [Green Version]
  95. Crucean, A.; Alqahtani, A.; Barron, D.J.; Brawn, W.J.; Richardson, R.V.; O’Sullivan, J.; Anderson, R.H.; Henderson, D.J.; Chaudhry, B. Re-evaluation of hypoplastic left heart syndrome from a developmental and morphological perspective. Orphanet J. Rare Dis. 2017, 12, 1–10. [Google Scholar] [CrossRef] [Green Version]
  96. Tian, X.; Hu, T.; Zhang, H.; He, L.; Huang, X.; Liu, Q.; Yu, W.; He, L.; Yang, Z.; Zhang, Z.; et al. Subepicardial endothelial cells invade the embryonic ventricle wall to form coronary arteries. Cell Res. 2013, 23, 1075–1090. [Google Scholar] [CrossRef] [Green Version]
  97. Moses, K.A.; DeMayo, F.; Braun, R.M.; Reecy, J.L.; Schwartz, R.J. Embryonic expression of an Nkx2-5/Cre gene using ROSA26 reporter mice. Genesis 2001, 31, 176–180. [Google Scholar] [CrossRef] [PubMed]
  98. Harmon, A.W.; Nakano, A. Nkx2-5 lineage tracing visualizes the distribution of second heart field-derived aortic smooth muscle. Genesis 2013, 51, 862–869. [Google Scholar] [CrossRef] [Green Version]
  99. Epstein, J.A.; Li, J.; Lang, D.; Chen, F.; Brown, C.B.; Jin, F.; Lu, M.M.; Thomas, M.; Liu, E.; Wessels, A.; et al. Migration of cardiac neural crest cells in Splotch embryos. Development 2000, 127, 1869–1878. [Google Scholar] [CrossRef]
  100. Epstein, J.; Buck, C.A. Transcriptional regulation of cardiac development: Implications for congenital heart disease and DiGeorge syndrome. Pediatric Res. 2000, 48, 717–724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Greulich, F.; Rudat, C.; Farin, H.F.; Christoffels, V.M.; Kispert, A. Lack of Genetic Interaction between Tbx18 and Tbx2/Tbx20 in Mouse Epicardial Development. PLoS ONE 2016, 11, e0156787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Aanhaanen, W.T.J.; Brons, J.F.; Domínguez, J.N.; Rana, M.S.; Norden, J.; Airik, R.; Wakker, V.; De Gier-De Vries, C.; Brown, N.A.; Kispert, A.; et al. The Tbx2+ primary myocardium of the atrioventricular canal forms the atrioventricular node and the base of the left ventricle. Circ. Res. 2009, 104, 1267–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Singh, R.; Hoogaars, W.M.; Barnett, P.; Grieskamp, T.; Rana, M.S.; Buermans, H.; Farin, H.F.; Petry, M.; Heallen, T.; Martin, J.F.; et al. Tbx2 and Tbx3 induce atrioventricular myocardial development and endocardial cushion formation. Cell. Mol. Life Sci. 2012, 69, 1377–1389. [Google Scholar] [CrossRef] [Green Version]
  104. De Lange, F.J.; Moorman, A.F.M.; Anderson, R.H.; Männer, J.; Soufan, A.T.; De Gier-De Vries, C.; Schneider, M.D.; Webb, S.; van den Hoff, M.J.B.; Christoffels, V.M.; et al. Lineage and morphogenetic analysis of the cardiac valves. Circ. Res. 2004, 95, 645–654. [Google Scholar] [CrossRef]
  105. Payne, S.; De Val, S.; Neal, A. Endothelial-specific cre mouse models is your cre CREdibile? Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2550–2561. [Google Scholar] [CrossRef] [Green Version]
  106. Tang, Y.; Harrington, A.; Yang, X.; Friesel, R.E.; Liaw, L. The contribution of the Tie2+ lineage to primitive and definitive hematopoietic cells. Genesis 2010, 48, 563–567. [Google Scholar] [CrossRef] [Green Version]
  107. Jiao, K.; Kulessa, H.; Tompkins, K.; Zhou, Y.; Batts, L.; Baldwin, H.S.; Hogan, B.L.M. An essential role of Bmp4 in the atrioventricular septation of the mouse heart. Genes Dev. 2003, 17, 2362–2367. [Google Scholar] [CrossRef] [Green Version]
  108. Sinha, T.; Lin, L.; Li, D.; Davis, J.; Evans, S.; Wynshaw-Boris, A.; Wang, J. Mapping the dynamic expression of Wnt11 and the lineage contribution of Wnt11-expressing cells during early mouse development. Dev. Biol. 2015, 398, 177–192. [Google Scholar] [CrossRef] [Green Version]
  109. Poelmann, R.E.; Jongbloed, M.R.M.; Molin, D.G.M.; Fekkes, M.L.; Wang, Z.; Fishman, G.I.; Doetschman, T.; Azhar, M.; Gittenberger-De Groot, A.C. The neural crest is contiguous with the cardiac conduction system in the mouse embryo: A role in induction? Anat. Embryol. 2004, 208, 389–393. [Google Scholar] [CrossRef] [PubMed]
  110. Stottmann, R.W.; Choi, M.; Mishina, Y.; Meyers, E.N.; Klingensmith, J. BMP receptor IA is required in mammalian neural crest cells for development of the cardiac outflow tract and ventricular myocardium. Development 2004, 131, 2205–2218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Hatzistergos, K.E.; Durante, M.A.; Valasaki, K.; Wanschel, A.C.B.A.; William Harbour, J.; Hare, J.M. A novel cardiomyogenic role for Isl1+ neural crest cells in the inflow tract. Sci. Adv. 2020, 6, eaba9950. [Google Scholar] [CrossRef]
  112. Zhou, B.; Pu, W.T. Genetic Cre-loxP assessment of epicardial cell fate using Wt1-Driven cre alleles. Circ. Res. 2012, 111, e276. [Google Scholar] [CrossRef] [Green Version]
  113. Bonnerot, C.; Nicolas, J.F. Clonal analysis in the intact mouse embryo by intragenic homologous recombination. Comptes Rendus Acad. Sci. III 1993, 316, 1207–1217. [Google Scholar]
  114. Meilhac, S.M.; Kelly, R.G.; Rocancourt, D.; Eloy-Trinquet, S.; Nicolas, J.-F.; Buckingham, M.E. A retrospective clonal analysis of the myocardium reveals two phases of clonal growth in the developing mouse heart. Development 2003, 130, 3877–3889. [Google Scholar] [CrossRef] [Green Version]
  115. Miquerol, L.; Moreno-Rascon, N.; Beyer, S.; Dupays, L.; Meilhac, S.M.; Buckingham, M.E.; Franco, D.; Kelly, R.G. Biphasic development of the mammalian ventricular conduction system. Circ. Res. 2010, 107, 153–161. [Google Scholar] [CrossRef]
  116. Miquerol, L.; Bellon, A.; Moreno, N.; Beyer, S.; Meilhac, S.M.; Buckingham, M.; Franco, D.; Kelly, R.G. Resolving cell lineage contributions to the ventricular conduction system with a Cx40-GFP allele: A dual contribution of the first and second heart fields. Dev. Dyn. 2013, 242, 665–677. [Google Scholar] [CrossRef]
  117. Luria, S.E.; Delbrück, M. Mutations of Bacteria from Virus Sensitivity to Virus Resistance. Genetics 1943, 28, 491–511. [Google Scholar] [CrossRef] [PubMed]
  118. Lescroart, F.; Mohun, T.; Meilhac, S.M.; Bennett, M.; Buckingham, M. Lineage tree for the venous pole of the heart: Clonal analysis clarifies controversial genealogy based on genetic tracing. Circ. Res. 2012, 111, 1313–1322. [Google Scholar] [CrossRef] [Green Version]
  119. Cannoodt, R.; Saelens, W.; Saeys, Y. Computational methods for trajectory inference from single-cell transcriptomics. Eur. J. Immunol. 2016, 46, 2496–2506. [Google Scholar] [CrossRef] [PubMed]
  120. Todorov, H.; Cannoodt, R.; Saelens, W.; Saeys, Y. Network Inference from Single-Cell Transcriptomic Data. In Methods in Molecular Biology; Humana Press: New York, NY, USA, 2019; Volume 1883, pp. 235–249. [Google Scholar]
  121. Lescroart, F.; Kelly, R.G.; Le Garrec, J.-F.; Nicolas, J.-F.; Meilhac, S.M.; Buckingham, M. Clonal analysis reveals common lineage relationships between head muscles and second heart field derivatives in the mouse embryo. Development 2010, 137, 3269–3279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Bredman, J.J.; Weijs, W.A.; Korfage, H.A.; Brugman, P.; Moorman, A.F. Myosin heavy chain expression in rabbit masseter muscle during postnatal development. J. Anat. 1992, 180 Pt 2, 263–274. [Google Scholar]
  123. Navin, N.; Kendall, J.; Troge, J.; Andrews, P.; Rodgers, L.; McIndoo, J.; Cook, K.; Stepansky, A.; Levy, D.; Esposito, D.; et al. Tumour evolution inferred by single-cell sequencing. Nature 2011, 472, 90–94. [Google Scholar] [CrossRef] [Green Version]
  124. Wang, Y.; Waters, J.; Leung, M.L.; Unruh, A.; Roh, W.; Shi, X.; Chen, K.; Scheet, P.; Vattathil, S.; Liang, H.; et al. Clonal evolution in breast cancer revealed by single nucleus genome sequencing. Nature 2014, 512, 155–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, J.; Nuno, K.; Litzenburger, U.M.; Qi, Y.; Corces, M.R.; Majeti, R.; Chang, H.Y. Single-cell lineage tracing by endogenous mutations enriched in transposase accessible mitochondrial DNA. eLife 2019, 8, e45105. [Google Scholar] [CrossRef] [PubMed]
  127. Ding, J.; Lin, C.; Bar-Joseph, Z. Cell lineage inference from SNP and scRNA-Seq data. Nucleic Acids Res. 2019, 47, e56. [Google Scholar] [CrossRef] [Green Version]
  128. Lioux, G.; Liu, X.; Temiño, S.; Oxendine, M.; Ayala, E.; Ortega, S.; Kelly, R.G.; Oliver, G.; Torres, M. A Second Heart Field-Derived Vasculogenic Niche Contributes to Cardiac Lymphatics. Dev. Cell 2020, 52, 350–363. [Google Scholar] [CrossRef] [PubMed]
  129. Gittenberger-De Groot, A.C.; Vrancken Peeters, M.P.F.M.; Bergwerff, M.; Mentink, M.M.T.; Poelmann, R.E. Epicardial outgrowth inhibition leads to compensatory mesothelial outflow tract collar and abnormal cardiac septation and coronary formation. Circ. Res. 2000, 87, 969–971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Arques, C.G.; Doohan, R.; Sharpe, J.; Torres, M. Cell tracing reveals a dorsoventral lineage restriction plane in the mouse limb bud mesenchyme. Development 2007, 134, 3713–3722. [Google Scholar] [CrossRef] [Green Version]
  131. Liu, X.; Chen, W.; Li, W.; Li, Y.; Priest, J.R.; Zhou, B.; Wang, J.; Zhou, Z. Single-Cell RNA-Seq of the Developing Cardiac Outflow Tract Reveals Convergent Development of the Vascular Smooth Muscle Cells. Cell Rep. 2019, 28, 1346–1361. [Google Scholar] [CrossRef] [Green Version]
  132. Kruse, F.; Junker, J.P.; van Oudenaarden, A.; Bakkers, J. Tomo-seq: A method to obtain genome-wide expression data with spatial resolution. Methods Cell Biol. 2016, 135, 299–307. [Google Scholar] [CrossRef]
  133. Junker, J.P.; Noel, E.S.; Guryev, V.; Peterson, K.A.; Shah, G.; Huisken, J.; McMahon, A.P.; Berezikov, E.; Bakkers, J.; Van Oudenaarden, A.; et al. Genome-wide RNA Tomography in the Zebrafish Embryo. Cell 2014, 159, 662–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lovatt, D.; Ruble, B.K.; Lee, J.; Dueck, H.; Kim, T.K.; Fisher, S.; Francis, C.; Spaethling, J.M.; Wolf, J.A.; Grady, M.S.; et al. Transcriptome in vivo analysis (TIVA) of spatially defined single cells in live tissue. Nat. Methods 2014, 11, 190–196. [Google Scholar] [CrossRef] [Green Version]
  135. Rutkovskiy, A.; Malashicheva, A.; Sullivan, G.; Bogdanova, M.; Kostareva, A.; Stensløkken, K.O.; Fiane, A.; Vaage, J. Valve interstitial cells: The key to understanding the pathophysiology of heart valve calcification. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef] [PubMed]
  136. Raj, B.; Wagner, D.E.; McKenna, A.; Pandey, S.; Klein, A.M.; Shendure, J.; Gagnon, J.A.; Schier, A.F. Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain. Nat. Biotechnol. 2018, 36, 442–450. [Google Scholar] [CrossRef] [PubMed]
  137. Alemany, A.; Florescu, M.; Baron, C.S.; Peterson-Maduro, J.; Van Oudenaarden, A. Whole-organism clone tracing using single-cell sequencing. Nature 2018, 556, 108–112. [Google Scholar] [CrossRef]
  138. Spanjaard, B.; Hu, B.; Mitic, N.; Olivares-Chauvet, P.; Janjuha, S.; Ninov, N.; Junker, J.P. Simultaneous lineage tracing and cell-type identification using CrIsPr-Cas9-induced genetic scars. Nat. Biotechnol. 2018, 36, 469–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Junker, J.P.; Spanjaard, B.; Peterson-Maduro, J.; Alemany, A.; Hu, B.; Florescu, M.; van Oudenaarden, A. Massively parallel clonal analysis using CRISPR/Cas9 induced genetic scars. bioRxiv 2016, 56499. [Google Scholar] [CrossRef] [Green Version]
  140. Weinreb, C.; Rodriguez-Fraticelli, A.; Camargo, F.D.; Klein, A.M. Lineage tracing on transcriptional landscapes links state to fate during differentiation. Science 2020, 367, eaaw3381. [Google Scholar] [CrossRef]
  141. Pei, W.; Feyerabend, T.B.; Rössler, J.; Wang, X.; Postrach, D.; Busch, K.; Rode, I.; Klapproth, K.; Dietlein, N.; Quedenau, C.; et al. Polylox barcoding reveals haematopoietic stem cell fates realized in vivo. Nature 2017, 548, 456–460. [Google Scholar] [CrossRef] [Green Version]
  142. Wagner, D.E.; Klein, A.M. Lineage tracing meets single-cell omics: Opportunities and challenges. Nat. Rev. Genet. 2020, 21, 410–427. [Google Scholar] [CrossRef]
  143. Wei, C.J.Y.; Zhang, K. Retrace: Simultaneous retrospective lineage tracing and methylation profiling of single cells. Genome Res. 2020, 30, 602–610. [Google Scholar] [CrossRef] [Green Version]
  144. Biezuner, T.; Spiro, A.; Raz, O.; Amir, S.; Milo, L.; Adar, R.; Chapal-Ilani, N.; Berman, V.; Fried, Y.; Ainbinder, E.; et al. A generic, cost-effective, and scalable cell lineage analysis platform. Genome Res. 2016, 26, 1588–1599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Liu, Y.; Siejka-Zielińska, P.; Velikova, G.; Bi, Y.; Yuan, F.; Tomkova, M.; Bai, C.; Chen, L.; Schuster-Böckler, B.; Song, C.X. Bisulfite-free direct detection of 5-methylcytosine and 5-hydroxymethylcytosine at base resolution. Nat. Biotechnol. 2019, 37, 424–429. [Google Scholar] [CrossRef] [PubMed]
  146. Ku, W.L.; Nakamura, K.; Gao, W.; Cui, K.; Hu, G.; Tang, Q.; Ni, B.; Zhao, K. Single-cell chromatin immunocleavage sequencing (scChIC-seq) to profile histone modification. Nat. Methods 2019, 16, 323–325. [Google Scholar] [CrossRef] [PubMed]
  147. Regev, A.; Teichmann, S.A.; Lander, E.S.; Amit, I.; Benoist, C.; Birney, E.; Bodenmiller, B.; Campbell, P.; Carninci, P.; Clatworthy, M.; et al. Science forum: The human cell atlas. eLife 2017, 6, e27041. [Google Scholar] [CrossRef] [PubMed]
  148. Pullen, L.C. Human Cell Atlas Poised to Transform Our Understanding of Organs. Am. J. Transplant. 2018. [Google Scholar] [CrossRef] [Green Version]
  149. Regev, A.; Teichmann, S.; Rozenblatt-Rosen, O.; Stubbington, M.; Ardlie, K.; Amit, I.; Arlotta, P.; Bader, G.; Benoist, C.; Biton, M.; et al. The Human Cell Atlas White Paper. arXiv 2018, arXiv:1810.05192. [Google Scholar]
Figure 1. Vital dye and viral labeling techniques. (A) Lipophilic dyes are used to visualise and track distinct cell compartments over time. (B) Alternatively, retroviral vectors can allow for expression of a reporter gene (such as LacZ) to enable cell tracing of infected cells. (C) Placing the reporter gene under the transcriptional control of a cell specific promoter allows for increased accuracy in targeting cells through gene specific viral labelling.
Figure 1. Vital dye and viral labeling techniques. (A) Lipophilic dyes are used to visualise and track distinct cell compartments over time. (B) Alternatively, retroviral vectors can allow for expression of a reporter gene (such as LacZ) to enable cell tracing of infected cells. (C) Placing the reporter gene under the transcriptional control of a cell specific promoter allows for increased accuracy in targeting cells through gene specific viral labelling.
Jcdd 08 00047 g001
Figure 2. The quail-chick chimeric system. A schematic representation of heterospecific transplantation of the neural crest tissue common to chimeric lineage tracing. The dorsal part of the neural tube of the quail, containing the neural crest cells, is excised and interchanged into a chick host of a comparable embryonic stage at the same locus. 1: somite 1, 2: somite 2, OV: otic vesicle. Image obtained with permission from TP Kelder & R Vincente-Steijn et al. [48].
Figure 2. The quail-chick chimeric system. A schematic representation of heterospecific transplantation of the neural crest tissue common to chimeric lineage tracing. The dorsal part of the neural tube of the quail, containing the neural crest cells, is excised and interchanged into a chick host of a comparable embryonic stage at the same locus. 1: somite 1, 2: somite 2, OV: otic vesicle. Image obtained with permission from TP Kelder & R Vincente-Steijn et al. [48].
Jcdd 08 00047 g002
Figure 3. Genetic lineage tracing. (A) Genetic lineage tracing can be achieved using the Cre-LoxP system. Upon expression, the Cre recombinase specifically targets LoxP DNA motives and allows for excision of DNA elements floxed by LoxP sites. The reporter line contains a stop-cassette inhibiting expression of the reporter gene in the absence of Cre. (B) Crossing the reporter line with the Cre line results in offspring carrying both constructs. (C) Cells expressing the gene of interest will express the Cre-recombinase. Cre will then permanently remove the stop-cassette and thus allow expression of the reporter gene, resulting in cell lineage labelling. (D) Modified Cre-recombinases can be used to allow for more temporal control in genetic labelling. Cre-ER variants can prevent Cre-recombinase activity by restricting mobility to the cells cytoplasm in absence of Tamoxifen. Upon stimulation of Tamoxifen-Cre activity is restored resulting in an inducible system for genetic lineage tracing.
Figure 3. Genetic lineage tracing. (A) Genetic lineage tracing can be achieved using the Cre-LoxP system. Upon expression, the Cre recombinase specifically targets LoxP DNA motives and allows for excision of DNA elements floxed by LoxP sites. The reporter line contains a stop-cassette inhibiting expression of the reporter gene in the absence of Cre. (B) Crossing the reporter line with the Cre line results in offspring carrying both constructs. (C) Cells expressing the gene of interest will express the Cre-recombinase. Cre will then permanently remove the stop-cassette and thus allow expression of the reporter gene, resulting in cell lineage labelling. (D) Modified Cre-recombinases can be used to allow for more temporal control in genetic labelling. Cre-ER variants can prevent Cre-recombinase activity by restricting mobility to the cells cytoplasm in absence of Tamoxifen. Upon stimulation of Tamoxifen-Cre activity is restored resulting in an inducible system for genetic lineage tracing.
Jcdd 08 00047 g003
Figure 4. Transversal sections of immunofluorescent outflow tracts show challenges with genetic lineage tracing. (A) A Wnt1Cre; R26RmTmG embryo showing neural crest cell derived cells (green). Interestingly, Wnt1 derived cells with NKX2.5 stained nuclei can also be observed at the so-called flow divider [84], in front of the primitive foregut which is the central part of the second heart filed contribution (white arrowhead) as well as in the atrial myocardium (white arrow). (B) a Tie2Cre; R26RmTmG embryo showing endothelial derived cells (green). Endothelial cells undergoing epithelial to mesenchymal transition give rise to cells within the septal and parietal cushions. Cells derived from myeloid lineages also express Tie2 resulting in lineage positive blood cells (yellow arrowhead). (C) A Mef2cCre; R26RmTmG embryo showing second heart field derived cells (green). Cre positive endothelial cells can also be found lining the heart (red arrowhead). Mef2c derived cells also contribute directly to the septal cushion (red arrow). Ra: Right atrium, Ao: Aorta, PT: Pulmonary trunk, Pc: Parietal cushion, Sc: Septal cushion. Scalebar: 50 µm.
Figure 4. Transversal sections of immunofluorescent outflow tracts show challenges with genetic lineage tracing. (A) A Wnt1Cre; R26RmTmG embryo showing neural crest cell derived cells (green). Interestingly, Wnt1 derived cells with NKX2.5 stained nuclei can also be observed at the so-called flow divider [84], in front of the primitive foregut which is the central part of the second heart filed contribution (white arrowhead) as well as in the atrial myocardium (white arrow). (B) a Tie2Cre; R26RmTmG embryo showing endothelial derived cells (green). Endothelial cells undergoing epithelial to mesenchymal transition give rise to cells within the septal and parietal cushions. Cells derived from myeloid lineages also express Tie2 resulting in lineage positive blood cells (yellow arrowhead). (C) A Mef2cCre; R26RmTmG embryo showing second heart field derived cells (green). Cre positive endothelial cells can also be found lining the heart (red arrowhead). Mef2c derived cells also contribute directly to the septal cushion (red arrow). Ra: Right atrium, Ao: Aorta, PT: Pulmonary trunk, Pc: Parietal cushion, Sc: Septal cushion. Scalebar: 50 µm.
Jcdd 08 00047 g004
Figure 5. Retrospective lineage tracing. (A,B) Traditional retrospective lineage tracing exploits spontaneous and rare recombination events. The nlaacZ sequence was designed to be inactive by default but increase the odds of recombination events within the region. Upon spontaneous recombination, β-galactosidase is expressed resulting in a lineage label. (C,D) Early recombination (i.e., during early stages of development) will give large clusters of cells with reporter gene expression, whilst late spontaneous recombination results in smaller clusters of LacZ positive cells. (E) Modern retrospective lineage tracing derives lineage hierarchy from single cell transcriptomics data. Principle component analysis (PC) aids in clustering groups of cells based on similarity in gene expression profiles. Follow up algorithms can then produce a phylogenetic lineage tree derived from expressional gradients among cell clusters to reconstruct cell lineage development.
Figure 5. Retrospective lineage tracing. (A,B) Traditional retrospective lineage tracing exploits spontaneous and rare recombination events. The nlaacZ sequence was designed to be inactive by default but increase the odds of recombination events within the region. Upon spontaneous recombination, β-galactosidase is expressed resulting in a lineage label. (C,D) Early recombination (i.e., during early stages of development) will give large clusters of cells with reporter gene expression, whilst late spontaneous recombination results in smaller clusters of LacZ positive cells. (E) Modern retrospective lineage tracing derives lineage hierarchy from single cell transcriptomics data. Principle component analysis (PC) aids in clustering groups of cells based on similarity in gene expression profiles. Follow up algorithms can then produce a phylogenetic lineage tree derived from expressional gradients among cell clusters to reconstruct cell lineage development.
Jcdd 08 00047 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Peterson, J.C.; Kelder, T.P.; Goumans, M.J.T.H.; Jongbloed, M.R.M.; DeRuiter, M.C. The Role of Cell Tracing and Fate Mapping Experiments in Cardiac Outflow Tract Development, New Opportunities through Emerging Technologies. J. Cardiovasc. Dev. Dis. 2021, 8, 47. https://0-doi-org.brum.beds.ac.uk/10.3390/jcdd8050047

AMA Style

Peterson JC, Kelder TP, Goumans MJTH, Jongbloed MRM, DeRuiter MC. The Role of Cell Tracing and Fate Mapping Experiments in Cardiac Outflow Tract Development, New Opportunities through Emerging Technologies. Journal of Cardiovascular Development and Disease. 2021; 8(5):47. https://0-doi-org.brum.beds.ac.uk/10.3390/jcdd8050047

Chicago/Turabian Style

Peterson, Joshua C., Tim P. Kelder, Marie José T. H. Goumans, Monique R. M. Jongbloed, and Marco C. DeRuiter. 2021. "The Role of Cell Tracing and Fate Mapping Experiments in Cardiac Outflow Tract Development, New Opportunities through Emerging Technologies" Journal of Cardiovascular Development and Disease 8, no. 5: 47. https://0-doi-org.brum.beds.ac.uk/10.3390/jcdd8050047

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop