Advanced Technologies In Cardiovascular Bioengineering Jianyi Zhang
1. Advanced Technologies In Cardiovascular
Bioengineering Jianyi Zhang download
https://ebookbell.com/product/advanced-technologies-in-
cardiovascular-bioengineering-jianyi-zhang-38310660
Explore and download more ebooks at ebookbell.com
2. Here are some recommended products that we believe you will be
interested in. You can click the link to download.
Advanced Technologies In Wastewater Treatment Food Processing Industry
Angelo Basile
https://ebookbell.com/product/advanced-technologies-in-wastewater-
treatment-food-processing-industry-angelo-basile-49169422
Advanced Technologies In Ad Hoc And Sensor Networks Proceedings Of The
7th China Conference On Wireless Sensor Networks 1st Edition Xue Wang
https://ebookbell.com/product/advanced-technologies-in-ad-hoc-and-
sensor-networks-proceedings-of-the-7th-china-conference-on-wireless-
sensor-networks-1st-edition-xue-wang-4664606
Advanced Technologies In Biodiesel New Advances In Designed And
Optimized Catalysts Aminul Islam
https://ebookbell.com/product/advanced-technologies-in-biodiesel-new-
advances-in-designed-and-optimized-catalysts-aminul-islam-5067610
Advanced Technologies In Biohydrometallurgy For Recovery And Recycling
Of Metals Stefano Ubaldini
https://ebookbell.com/product/advanced-technologies-in-
biohydrometallurgy-for-recovery-and-recycling-of-metals-stefano-
ubaldini-54691972
3. Advanced Technologies In Biodiesel Introduction To Principles And
Emerging Trends Chan
https://ebookbell.com/product/advanced-technologies-in-biodiesel-
introduction-to-principles-and-emerging-trends-chan-5846698
Advanced Technologies In Practical Applications For National Security
Bereska Damian
https://ebookbell.com/product/advanced-technologies-in-practical-
applications-for-national-security-bereska-damian-6753438
Advanced Technologies In Modern Robotic Applications 1st Edition
Chenguang Yang
https://ebookbell.com/product/advanced-technologies-in-modern-robotic-
applications-1st-edition-chenguang-yang-10304002
Advanced Technologies In Robotics And Intelligent Systems Proceedings
Of Itr 2019 1st Ed 2020 Sergey Yu Misyurin
https://ebookbell.com/product/advanced-technologies-in-robotics-and-
intelligent-systems-proceedings-of-itr-2019-1st-ed-2020-sergey-yu-
misyurin-10801202
Advanced Technologies For Sustainable Systems Selected Contributions
From The International Conference On Sustainable Vital Technologies In
Engineering And Informatics Bue Ace1 2016 79 November 2016 Cairo Egypt
1st Edition Yehia Baheieldin
https://ebookbell.com/product/advanced-technologies-for-sustainable-
systems-selected-contributions-from-the-international-conference-on-
sustainable-vital-technologies-in-engineering-and-informatics-bue-
ace1-2016-79-november-2016-cairo-egypt-1st-edition-yehia-
baheieldin-5675906
9. v
Preface
In recent decades, the convergence of discoveries in biological sciences and engi-
neering have resulted in the development of new industries that offer the promise of
revolutionary changes in society, as part of the Convergence Revolution (the Fourth
Industrial Revolution). These advancements have offered the potential new man-
agement options for some of humanity’s most intractable and deadly diseases.
Within the cardiovascular sciences, many of the most provocative discoveries have
emerged from studies of pluripotent stem cells, whose roles in the medical sciences
and physiological and injury response are becoming increasingly acknowledged. In
2016, the National Institutes of Health (NIH) established the Progenitor Cell
Translational Consortium (PCTC) to support research into the use of stem and pro-
genitor cells for both biology and therapeutic applications. This book was inspired
by the thought-provoking ideas and observations presented at the 2019 PCTC
Cardiovascular Bioengineering (CVBE) Symposium and was written by leading
scientists and physicians whose work in the CVBE field spans decades and was
conducted on four different continents.
Cardiomyocytes in the hearts of humans and other mammals are largely incapa-
ble of self-replicating; thus, although advancements in the clinical management of
cardiovascular conditions have led to substantial improvements in patient longevity
and quality of life, the scarring caused by cardiac disease or injury is essentially
permanent. Myocardial integrity can be fully restored via whole-heart transplanta-
tion surgery, but the supply of donated hearts is far smaller than the number of
patients who require treatment, so alternative strategies for replacing the myocardial
scar with functional contractile tissue are urgently needed. The lack of cell-cycle
activity in adult mammalian cardiomyocytes also severely restricts their availability
for investigational work, so early studies of myocardial cell therapy were frequently
conducted with stem cells which, though obtained from a variety of sources (e.g.,
the bone marrow, adipose tissue), were expected to differentiate into cardiomyo-
cytes after transplantation. However, the benefits observed in subsequent clinical
trials were only marginal and likely evolved from the cells’ paracrine activity, rather
than through the production of new cardiomyocytes.
10. vi
The scarcity of cardiomyocytes for therapeutic investigations was alleviated by
the isolation of human embryonic stem cells (ESCs) and, especially, by the develop-
ment of techniques for reprogramming somatic cells into induced-pluripotent stem
cells (iPSCs). Both cell types can proliferate indefinitely and are capable of differ-
entiating into diverse cellular lineage; however, direct stem cell transplantation can
lead to tumor formation; so, ESCs and iPSCs must be differentiated into more spe-
cialized cell types before administration to patients, and only in recent years the
differentiation protocols achieved the adequate efficiency to meet such demands. In
general, the most effective protocols are modeled after the mechanisms that regulate
cell specification during embryogenesis, when the four major lineages of cardiac
cells evolve from progenitor cells of the first and second heart fields, the proepicar-
dial organ, and the cardiac neural crest. These protocols may become even more
efficient as researchers continue to refine and develop novel methods for determin-
ing the identity, ancestry, and progeny of progenitor cells during development and
as the heart recovers from injury.
Only a small fraction of transplanted cells are engrafted within the native tissue
and survives for more than a few days after administration, which is perhaps not
surprising, since the cytotoxic conditions responsible for the loss of endogenous
cells are likely to endure longer than the initial injury. One of the chief requirements
of a more salubrious environment for transplanted cells is adequate perfusion. Both
the size and thickness of engineered tissues are typically limited by the access of
nutrients and signaling molecules to the cells within the tissue. Thus, the success of
cell-based regenerative therapies for treatment of cardiac disease, as well as periph-
eral artery disease, critical limb ischemia, and other predominantly vascular condi-
tions, will depend on understanding the mechanisms by which the vascular cell
differentiation and proliferation can be manipulated to promote vessel growth.
Tissues constructed from human ESC- or iPSC-derived cells can also provide
researchers with an entirely human-specific platform for studying the pathogenesis
of disease and for testing new pharmaceutical products. Notably, iPSC-derived cell
and tissue models are powerful tools for personalized therapies, because the iPSCs
can be reprogrammed from the patient’s own somatic cells and, consequently, reca-
pitulate all of the genetic factors that regulate disease pathology and progression, as
well as the patient’s response to treatment. Autologous iPSC-derived cells are also
expected to be minimally immunogenic when re-administered to the same patient
for treatment of chronic conditions such as heart failure; however, the reprogram-
ming and differentiation procedures take several weeks, so cell-based treatments for
emergency situations, such as acute myocardial infarction, will require the use of
allogeneic cells, which have rarely been studied. Furthermore, one of the primary
concerns associated with cardiac cell therapy is the potential for arrhythmogenic
complications caused by inadequate electromechanical coupling between the
endogenous and transplanted cells. Thus, researchers continue to develop increas-
ingly sophisticated tools for assessing the integration and electrophysiological func-
tion of engrafted cells and tissues, such as epicardial electrode arrays, genetically
encoded fluorescent reporters, and catheter-based electroanatomic mapping.
Preface
11. vii
Although the regenerative capacity of adult mammalian hearts is extremely lim-
ited, the hearts of at least some neonatal mammals (e.g., mice and pigs) can fully
repair the damage caused by myocardial injury, provided that the injury occurs
within the first few days after birth. Existing evidence suggests that this recovery is
driven primarily by the proliferation of pre-existing cardiomyocytes, rather than the
activity of stem or progenitor cells, which suggests that the cardiomyocytes of adult
hearts may retain some latent proliferative capacity that could be therapeutically
re-activated to improve cardiac performance in patients with heart disease. The
mechanisms responsible for inducing proliferation in cardiomyocytes are just
beginning to be explored. These works will be facilitated by advancements in
single-
cell genomics, which can characterize the gene expression profiles of thou-
sands of individual cells; however, the resulting datasets are typically so enormous
that they require the use of modern data science techniques, such as dimensionality
reduction and clustering analysis, to identify the genes and pathways that are dif-
ferentially activated in proliferating and non-proliferating cardiomyocytes.
Machine-learning algorithms can even be applied to the text mined from the Medline
database and other unstructured sources to identify relationships among specific
genes, diseases, and disease symptoms, including those that may explain why out-
comes of COVID-19 treatment are worse for patients with cardiovascular
comorbidities.
In summary, many of the greatest advancements in science, and in civilization as
a whole, have occurred when previously disparate lines of inquiry come together in
unanticipated ways. The fields of personal and public health will soon reap the ben-
efits of the unprecedented degree of synergy that has recently developed among the
life and physical sciences, computing, and engineering. The authors of this book
hope to foster these advancements by sharing their knowledge and expertise with
the broader community of scientists, engineers, and clinicians.
Birmingham, AL, USA Jianyi Zhang
Atlanta, GA, USA Vahid Serpooshan
Preface
12. ix
Contents
Part I
Cardiac Development and Morphogenesis
From Simple Cylinder to Four-Chambered Organ:
A Brief Overview of Cardiac Morphogenesis������������������������������������������������ 3
Carissa Lee, Sharon L. Paige, Francisco X. Galdos, Nicholas Wei,
and Sean M. Wu
Lineage Tracing Models to Study Cardiomyocyte
Generation During Cardiac Development and Injury �������������������������������� 15
Kamal Kolluri, Bin Zhou, and Reza Ardehali
Mechanisms that Govern Endothelial Lineage Development
and Vasculogenesis������������������������������������������������������������������������������������������ 31
Daniel J. Garry and Javier E. Sierra-Pagan
Part II
Cellular Approaches to Cardiac Repair and Regeneration
Remuscularization of Ventricular Infarcts
Using the Existing Cardiac Cells�������������������������������������������������������������������� 51
Yang Zhou and Jianyi Zhang
Allogeneic Immunity Following Transplantation
of Pluripotent Stem Cell-
Derived Cardiomyocytes�������������������������������������� 79
Yuji Shiba
Vascular Regeneration with Induced Pluripotent Stem Cell-Derived
Endothelial Cells and Reprogrammed Endothelial Cells���������������������������� 87
Sangho Lee and Young-sup Yoon
The Guinea Pig Model in Cardiac Regeneration Research;
Current Tissue Engineering Approaches and Future Directions���������������� 103
Tim Stüdemann and Florian Weinberger
13. x
Part III
Genetic Approaches to Study Cardiac
Differentiation and Repair
Analysing Genetic Programs of Cell Differentiation
to Study Cardiac Cell Diversification������������������������������������������������������������ 125
Zhixuan Wu, Sophie Shen, Yuliangzi Sun, Tessa Werner,
Stephen T. Bradford, and Nathan J. Palpant
Recombinant Adeno-Associated Virus for Cardiac Gene Therapy������������ 169
Cindy Kok, Dhanya Ranvindran, and Eddy Kizana
Part IV
Bioengineering Approaches to Cardiovascular
Tissue Modeling and Repair
Microfabricated Systems for Cardiovascular Tissue Modeling������������������ 193
Ericka Jayne Knee-Walden, Karl Wagner, Qinghua Wu,
Naimeh Rafatian, and Milica Radisic
Bioengineering of Pediatric Cardiovascular Constructs:
In Vitro Modeling of Congenital Heart Disease�������������������������������������������� 233
Holly Bauser-Heaton, Carmen J. Gil, and Vahid Serpooshan
Biomaterial Interface in Cardiac Cell and Tissue Engineering������������������ 249
Chenyan Wang and Zhen Ma
Stem Cell-Based 3D Bioprinting for Cardiovascular
Tissue Regeneration���������������������������������������������������������������������������������������� 281
Clara Liu Chung Ming, Eitan Ben-Sefer, and Carmine Gentile
Creating and Validating New Tools to Evaluate
the Electrical Integration and Function of hPSC-Derived
Cardiac Grafts In Vivo������������������������������������������������������������������������������������ 313
Wahiba Dhahri, Fanny Wulkan, and Michael A. Laflamme
Part V Clinical Perspectives
Understanding the Molecular Interface of Cardiovascular
Diseases and COVID-
19: A Data Science Approach������������������������������������ 335
Dibakar Sigdel, Dylan Steinecke, Ding Wang, David Liem,
Maya Gupta, Alex Zhang, Wei Wang, and Peipei Ping
Clinical Application of iPSC-Derived Cardiomyocytes
in Patients with Advanced Heart Failure������������������������������������������������������ 361
Jun Fujita, Shugo Tohyama, Hideaki Kanazawa, Yoshikazu Kishino,
Marina Okada, Sho Tanosaki, Shota Someya, and Keiichi Fukuda
Cell Therapy with Human ESC-Derived Cardiac Cells:
Clinical Perspectives���������������������������������������������������������������������������������������� 375
Philippe Menasché
Index������������������������������������������������������������������������������������������������������������������ 399
Contents
16. 4
development due to its similarity to human models and popularity in recent and
ongoing embryology research.
1.1
Early Gastrulation and Formation of the Cardiac Crescent
(E5.0 – E7.5)
Prior to gastrulation at approximately embryonic day E5.0, the mouse embryo
resembles an elongating cylinder consisting of a single proximal-distal axis. The
TGF-beta signaling protein nodal growth differentiation factor (NODAL), expressed
along a concentration gradient with its antagonists (Cer1 and Lefty1), in both
embryonic and extraembryonic tissues induces patterning along a second anterior-
posterior axis by E5.5 as shown in Fig. 1 [2].
Gastrulation is an essential process for embryogenesis, beginning at approxi-
mately E6.0 in mice [3]. During this period, a pluripotent group of embryonic cells
called the epiblast ingresses through a strip-like structure known as the primitive
streak (PS) to generate the three germ layers of the early embryo: endoderm, meso-
derm, and ectoderm. While cells from these germ layers collectively give rise to the
body and all its organs, the heart is specifically established by a group of myocardial
Fig. 1 At E5.5, NODAL expression originates from the Node, a structure located at the posterior
side of the mouse embryo. NODAL antagonists Lefty1 and Cer1 are expressed anteriorly, allowing
for formation of the primitive streak on the posterior side. Upon induction of the primitive streak
around E6.5–7.5, NODAL, Bmp, and canonical Wnt signaling gradients direct commitment of
migrating epiblast cells to various endoderm and mesodermal lineages, including cardiac meso-
derm that gives rise to the heart. (Adapted from [2])
C. Lee et al.
17. 5
progenitor cells which derive from the mesoderm. Notably, the heart is the first
functioning organ of the embryo, as it pumps blood carrying oxygen and nutrients
necessary for embryonic development [4].
By E6.5, these precardiac mesodermal progenitors on the posterior end of the
embryonic “cylinder” will migrate anteriorly and laterally to a region named the
anterior lateral plate mesoderm (ALPM). Along the way, these cells acquire cardiac
fates due to the patterned expression of bone morphogenic protein (BMP), wingless-
related integration site (Wnt), fibroblast growth factor (FGF), and other signaling
molecules that guide their organization at the ALPM into the two main cardiac
progenitor cell populations: the first and second heart fields. These two cell popula-
tions contribute to distinct structures of the developing heart. Fate-mapping studies
have shown that the FHF lineage primarily establishes the myocardium of the left
ventricle (LV), while the SHF lineage gives rise to the right ventricle (RV) and out-
flow tract (OFT) [5]. Initially, FHF precursors differentiate rapidly, becoming beat-
ing cardiomyocytes (CMs) which form the early cardiac crescent [6]. As SHF
precursors settle medially to the FHF, they together form the completed cardiac
crescent, which is typically visible by E7.5 [2]. The temporal delay between the
formation of FHF and SHF progenitors from the precardiac mesoderm, FHF pre-
ceding SHF, is the primary driver of organization into each heart field.
Recent findings have shown that coordinated flow of calcium ions between CMs
triggers the first heartbeat [7]. This explains evidence of primitive pacemaker activ-
ity originating near the inflow tract and sinus venosus of the linear heart tube at this
stage of development [8]. Furthermore, this portion of the heart tube includes the
primordium of the sinus node, which later becomes the chief pacemaker of the
mature cardiac conduction system [9].
1.2
The Linear Heart Tube (E8.0)
As development proceeds, the next few processes—heart tube formation, heart tube
elongation, and early chamber establishment —all overlap in time across the embry-
onic heart. Additionally, these morphogenic stages from E8.0–11.0 contribute sig-
nificantly to growth, facilitating a 100-fold increase in CM number and cardiac
volume [10].
Following formation of the cardiac crescent, a process characterized by rapid
differentiation of FHF progenitors into CMs, the embryonic heart enters a period of
more extensive morphogenesis. Splanchnic mesoderm slides over endoderm, tem-
porarily pausing CM differentiation as the heart tube (HT) is assembled. When dif-
ferentiation resumes upon completion of the primitive HT, newly formed CMs
instead contribute to SHF-derived regions of the HT and initiate closure of the dor-
sal side. The influx of SHF cells also strengthen the heartbeat such that by E8.25,
the embryonic “cylinder” is transformed into a beating linear HT [6].
The linear HT is made up of three distinct layers, with the inner endocardial layer
and outer myocardial layer separated by a thick band of extracellular matrix (ECM)
From Simple Cylinder to Four-Chambered Organ: A Brief Overview of Cardiac…
18. 6
referred to as the cardiac jelly [8]. The ECM plays a role in the maturation of cardiac
cells into highly vascularized, densely compacted myocardium.
Immunohistochemistry techniques have designated four essential ECM proteins—
collagen types I and IV (COLI, COLIV), elastin (ELN), and fibronectin (FN)—that
are found within the LV of the mouse heart [11]. The first beating CMs are found in
the outer heart tube, while the cells of the inner heart tube retain an endothelial cell
identity [2].
Subsequent migration of SHF progenitors to the arterial and venous poles facili-
tates gradual elongation of the heart tube as these cells undergo proliferation. The
proepicardial organ (PEO), a transitory mesenchymal structure responsible for gen-
erating the embryonic epicardial cell lineage, also appears near the venous pole by
E8.5 [12]. Importantly, this cluster of coelomic cells is highly conserved among
vertebrates [13].
1.3
Cardiac Looping (E8.5)
As elongation slows, the linear heart tube undergoes a characteristic process of
rightwards looping in which the posterior regions begin to move anteriorly as shown
in Fig. 2. This establishes the structural basis of the heart’s four distinct chambers:
right atrium (RA), right ventricle (RV), left atrium (LA), and left ventricle (LV) [3].
Inversion of the “S” loop during this phase is a common malformation observed
in patients with heterotaxy syndrome (HS), a disorder characterized by develop-
mental abnormalities in the left-right axis [14]. Early defects in the looping process
underscore its importance to proper heart formation as patients with HS may pres-
ent with dextrocardia, a condition in which the apex of the heart points towards the
right instead of the left.
Cardiac valve formation also initiates around E8.5 with the formation of the
dorsal and ventral endocardial cushions following heart looping [15]. These “swol-
len” protrusions of the ECM are lined with endocardial cells, a portion of which will
migrate into the cushion ECM and adopt mesenchymal identities through a process
known as the endothelial-to-mesenchymal transition (EndMT). This thickening of
the endocardial cushions provides the foundation for later remodeling into valve
leaflets.
Finally, epicardium development proceeds simultaneously as vesicles from the
PEO either directly adhere to the surface of the beating heart or gradually drift
towards the myocardium following their release into the pericardial cavity [13].
Upon making contact, the attached cells will collapse and proliferate, generating a
primitive layer of epicardium that covers the heart. These epicardially derived cells
(EPDCs), a subset of which will undergo an epithelial-to-mesenchymal transition
(EMT) and migrate into the myocardium, have the potential to differentiate into
coronary smooth muscle cells and interstitial fibroblasts while reports of their dif-
ferentiation into coronary endothelium and cardiomyocytes require further investi-
gation [16].
C. Lee et al.
19. 7
1.4
The Four-Chambered Heart (E9.5)
The looped heart tube consists of four main structural elements: the atrium, atrio-
ventricular canal (AVC), ventricle, and outflow tract [5]. Partitioning the left from
right and the atrial from ventricular regions of the heart constitutes the most com-
plex stage of heart morphogenesis, beginning around E9.5 and lasting approxi-
mately until E14.5 as depicted in Fig. 3. Proper chamber formation is crucial to the
Fig. 2 Diagram illustrating the process of cardiac looping beginning at E8.5, whereby the linear
heart tube is first transformed into an S-shaped curve before organization into primitive chambers.
(a) The linear heart tube. (b) Looping. (c) The primitive 4-chambered heart. (Key: T truncus arte-
riosus, BC bulbus cordis, SV sinus venosus, PV primitive ventricle, PA primitive atrium, pRV prim-
itive right ventricle, pLV primitive left ventricle, pRA primitive right atrium, pLA primitive left
atrium, OFT outflow tract). (Adapted from [17])
From Simple Cylinder to Four-Chambered Organ: A Brief Overview of Cardiac…
20. 8
developmental pathway, as failure to establish structures capable of sustaining sys-
temic circulation results in defects such as atrial septal defects (ASDs), ventricular
septal defects (VSDs), and atrioventricular septal defects (AVSDs) that can progress
to end-stage heart failure.
At E9.5, active proliferation and migration of the SHF leads to the formation and
elongation of the outflow tract (OFT), a transient structure which, in its most primi-
tive state, connects the developing right ventricle to the aortic sac [18].
Subsequent infiltration of cNCCs into the looped heart initiates septation of the
elongated OFT into the aorta (Ao) and pulmonary trunk (PT) [19]. From their start-
ing point in the cardiac neural crest, these cells migrate through the aortic arches
and cluster near the distal OFT, forming truncal cushions. Interactions between
these truncal cushions and the proximally-located conal cushions of the OFT cre-
ates a spiral septum that divides the OFT into the Ao and PT, allowing for separate
systemic and pulmonary circulations [5]. Congenital malformations of the OFT
may result in conotruncal defects, which include conditions such as Tetralogy of
Fallot (TOF) and Transposition of the Great Arteries (TGA).
Fig. 3 Illustrations depicting ventral surface cuts of embryonic mouse hearts between E9.5 and
E17.5. (Key: AVC atrioventricular canal). (Adapted from [10])
C. Lee et al.
21. 9
By E10.5, “well-defined chambers” are visible in the heart despite persistence of
the primitive tubular structure [3]. Histological samples indicate the presence of a
functioning sinoatrial node, which is responsible for initiating heart beats [8]. The
epicardium is fully formed, creating a protective envelope around the heart. At this
point, the atrial and ventricular chambers septate, a process that begins with the
expansion of the mesenchymal cushions, leading to the formation of the right and
left atria and ventricles.
The development of the two major atrioventricular (AV) cushions, the inferior
and superior cushions, in the centralAVC is facilitated by EndMT [20]. Endocardium
derived cells (ENDCs) populate the cushions, displacing the existing ECM within
the AV canal. As such, lineage tracing studies have shown that the majority of mes-
enchymal cells infiltrating the cushions are derived from endocardium [21].
As shown in Fig. 4, atrial septation occurs from E10.5–E13.5, beginning when
the major AV cushions fuse at the AVC along with two mesenchymal structures: the
vestibular spine and mesenchymal cap. Muscularization of the mesenchymal tissue
results in two muscular tissue structures, the septum primum and septum secundum,
which together septate the atrial chamber into right and left [22]. At E11.5, within
the ventricular chamber, an outgrowth called the interventricular muscular septum
fuses with the AV cushions to create distinct right and left ventricles [5]. The
“minor” left and right lateral AV cushions, which form after the inferior and supe-
rior AV cushions, are also formed from ENDCs. These minor cushions become the
Fig. 4 Depiction of the atrial septation process, occurring approximately from E10.5–E13.5. (a)
Formation of the first of two muscular septa, the septum primum, begins at the roof of the primitive
atrial chamber. (b) As the septum primum elongates, the foramen primum and foramen secundum
allow for continued communication between the right and left sides of the atrium. (c) The septum
secundum grows to the right of the septum primum, forming an oval-shaped hole called the fora-
men ovale. (d) Both septa begin to fuse. (e) The foramen ovale remains open, allowing for blood
flow from the right to left atrium. (Key: RA right atrium, LA left atrium). (Adapted from [25])
From Simple Cylinder to Four-Chambered Organ: A Brief Overview of Cardiac…
22. 10
septal leaflets of the mitral and tricuspid valves, which are necessary to prevent
retrograde flow of blood from the ventricles to the atria [23]. Failure of the tricuspid
valve tissue to delaminate from the ventricular myocardium at this stage results in
right ventricular myopathy and a apically-displaced tricuspid valve, which are char-
acteristic of a rare congenital defect called Ebstein’s anomaly [24].
Myocardial trabeculation, which also involves the endocardium, is another
essential developmental step that begins at this timepoint, extending through the end
of the embryonic stage. Within the heart wall, the myocardial layer projects into the
cardiac jelly, the ECM layer between the endocardium and myocardium, as endo-
cardial cells invaginate, the resulting finger-like structures are called trabeculae.
This process aids in the gradual dissipation of the cardiac jelly as the trabeculae
mature and eventually collapse to join with the compact myocardium, completing
the inner wall of the heart.
1.5
The Mature Embryonic Heart (E15.0)
The conclusion of chamber and OFT septation around E15.0 prepares the heart for
postnatal separation of the pulmonary and systemic circulatory pathways of the
blood [3]. In the fetal heart, oxygenated blood flows from the placenta to the umbili-
cal vein, entering the ductus venosus and passing through the inferior vena cava
(IVC) before entering the RA. It then travels across the foramen ovale to the LA,
down to the LV, and out the Ao to the brain and upper body. Deoxygenated blood
from the superior vena cava (SVC) drains to the RA, down to the RV, through the
pulmonary artery, and across the ductus arteriosus to the rest of the developing
embryo. The fetal circulation pathway ensures that the most oxygenated blood in
the fetus goes to the brain, with limited blood entering the lungs as oxygenation of
this area occurs postnatally. In the final fetal morphogenic phase, the heart tissue
undergoes “fine tuning” modifications that improve cardiac conduction, coronary
circulation, and control of blood flow.
Cardiac Conduction System The cellular origins of the cardiac conduction system
have yet to be detailed in full. Currently, it is known that signaling from arterial
endothelial cells induces the differentiation of Purkinje conduction cells from myo-
cardium [26]. Fast-conducting chamber myocardium makes up the contractile fibers
of the bundle of His, while slow-conducting myocardium from the inflow tract and
AV canal creates the SA and AV nodes [4]. The sinus venosus region’s important
role as a primitive pacemaker provides evidence of SAN progenitors, but the mech-
anisms behind the formation of the AV node remain poorly understood.
Coronary Vessels The appearance of coronary endothelial cells has been recorded
as early as E12.5, with endocardium giving rise to coronary arterial endothelium
and the sinus venosus generating coronary venous endothelium [27]. SV-derived
“sprouts” of venous cells cover the fetal heart before proliferating to form the
C. Lee et al.
23. 11
immature coronary plexus [28]. Recent single-cell RNA sequencing experiments
found that a subset of these venous endothelial cells dedifferentiate and undergo
pre-
arterial specification via transcriptional changes that take place prior to the
establishment of blood flow [29]. The connecting of the plexus to the Ao initiates
blood flow and is crucial to arterial morphogenesis. Epicardial cells from the PEO
also play a role in coronary vessel formation, assembling the smooth muscle wall of
the coronary vasculature through EMT [30].
Valves The mitral and tricuspid valves are derived from the AV endocardial cush-
ions. As the superior and inferior AV cushions fuse and divide the AVC, they give
rise to the anterior mitral and septal tricuspid leaflets. Further remodeling of these
primitive leaflets results in formation of the mature mitral and tricuspid valves that
ensure unidirectional atrial to ventricular blood flow.
Atrioventricular EPDCs (AV-EPDCs) give rise to the AV sulcus, a transient mes-
enchymal structure that separates the atrial and ventricular myocardial walls. A por-
tion of the AV-EPDCs within the sulcus infiltrate the AV myocardial junction where
they begin to form the annulus fibrosus, a divider made up of fibrous tissue respon-
sible for physically and conductively isolating the working atrial myocardium from
its ventricular counterpart. Yet another group of AV-EPDCs will continue on from
the annulus fibrosus to merge with the parietal AV valve leaflets and eventually
become valve interstitial cells [23].
Trabeculation By E19.0, trabeculation concludes with the complete degradation
of the cardiac jelly layer and resultant compaction of the ventricular wall [8]. This
compaction is associated with greater strength of contraction, allowing the blood to
penetrate deeper layers of the myocardium before the coronary vasculature fully
develops in the post-embryonic stage. With the completion of trabeculation, the
prenatal mouse heart is ready for postnatal modification following gestation, which
typically occurs 20 days post-fertilization.
2 Challenges and Opportunities
While much of early heart development has been documented through fate-mapping
and histological examination of mice, chick, and zebrafish embryos, a number of
challenges still remain in documenting human heart morphogenesis, including lack
of data, inconclusive literature, and poor imaging capabilities.
Due to a combination of technical, legal, and ethical complications, human fetal
heart cells are exceedingly difficult to obtain for data collection. Use of human
induced-pluripotent stem cells (hiPSCs), while generally considered more ethically
sound, suffer from difficulties with chamber and cell type identification. Ongoing
research focuses on single-cell RNA sequencing or lineage tracing-based solutions
that enable determination of distinct genetic markers within the embryonic heart.
From Simple Cylinder to Four-Chambered Organ: A Brief Overview of Cardiac…
24. 12
Recent studies have used sequencing data to identify candidate genes that may con-
tribute to CHDs such as HS [14].
An emerging topic of research is the transcriptomic correlation between murine
and human embryonic heart cells. A recent single-cell RNA sequencing (scRNA-
seq) study of cardiac cells from 18 human embryos showed several differences
between developing mouse and human cardiac cells in terms of certain gene expres-
sion levels, targets, and corresponding developmental timepoints, suggesting that
correlating mouse transcriptomics to human may not be practical. However, pre-
liminary research into using mouse scRNA-seq data in human cardiac cell classifi-
cation models have yielded promising results. Ongoing research is looking into
whether machine learning algorithms can be trained on mouse scRNA-seq data and
fine-tuned on preliminary human data to identify human embryonic heart cells, as
well as heart cells derived from induced pluripotent stem cells. The impact of this
research would be two-fold. First, a mouse-to-human classification model can lever-
age the vast amount of mouse transcriptomic data that exists (as well as future epig-
enomic data) to create a highly useful tool providing insights into the mechanisms
that control the signaling pathways of human cardiac development and regenera-
tion. Second, a successful classification model could be used to identify cardiac
cells derived from human induced pluripotent stem cells (hiPSCs), enabling scien-
tists to use these findings in order to better study cardiac cells in vitro.
Another area of interest is improving existing functional and molecular defini-
tions of certain cell types and processes. For instance, EndMT remains poorly
understood in comparison to EMT [31] as cell culture conditions severely impact
the process. Furthermore, endocardial precursors originate from a variety of regions,
making it difficult to establish precise molecular criteria. Standardizing markers for
both the presence of and definitive stages of ongoing EndMTs may improve under-
standing of endothelial dysfunctions that cause both CHDs and adult cardiovascular
diseases.
The “fine tuning” steps that occur during the septation phase, most notably the
fusion of the OFT and AVC, are complex and difficult to record. Additionally, while
heart formation is thus far understood to unfold continuously, viewing morphoge-
netic events in real time may reveal new insight into the kinetics of development.
Although limited by embryo survivability, whole-embryo live-imaging methods,
such as the two-photon microscopy approach used by Ivanovitch and colleagues [6],
may harbor potential to capture these nuances at cellular resolution.
In summary, cardiac morphogenesis involves an extensive process of looping,
septation, and remodeling that transforms the early heart fields into a matured, four-
chambered organ capable of systemic and pulmonary circulation. As the intricacies
of the embryonic heart render it susceptible to disruption, the understanding of
human fetal heart development will remain a popular topic that continues to gener-
ate novel research directions in hopes of finding solutions to prevalent the develop-
ment of CHDs.
C. Lee et al.
25. 13
References
1. Virani, S., Alonso, A., Benjamin, E., Bittencourt, M., Callaway, C., Carson, A., et al.: Heart
disease and stroke statistics—2020 update: a report from the American Heart Association.
Circulation (New York, NY). 141(9), e139–ee51 (2020). https://doi.org/10.1161/
CIR.0000000000000757
2. Galdos, F.X., Wu, S.M.: Development of cardiac muscle. Elsevier Inc (2015)
3. Zaffran, S., Meilhac, S., Buckingham, M.: Building the mammalian heart from two sources of
myocardial cells. Nat. Rev. Genet. 6(11), 826–837 (2005). https://doi.org/10.1038/nrg1710
4. Hill, M.A., Aug. Cardiac Embryology (2020)
5. Lin, C.-J., Lin, C.-Y., Chen, C.-H., Zhou, B., Chang, C.-P.: Partitioning the heart: mecha-
nisms of cardiac septation and valve development. Development. 139(18), 3277–3299 (2012).
https://doi.org/10.1242/dev.063495
6. Ivanovitch, K., Temiño, S., Torres, M.: Live imaging of heart tube development in mouse
reveals alternating phases of cardiac differentiation and morphogenesis. Elife. 6 (2017).
https://doi.org/10.7554/elife.30668
7. Tyser, R.C.V., Miranda, A.M.A., Chen, C.-M., Davidson, S.M., Srinivas, S., Riley, P.R.:
Calcium handling precedes cardiac differentiation to initiate the first heartbeat. Elife. 5 (2016).
https://doi.org/10.7554/elife.17113
8. Goodyer, W.R., Wu, S.M.: Fates aligned: origins and mechanisms of ventricular conduction
system and ventricular wall development. Pediatr. Cardiol. 39(6), 1090–1098 (2018). https://
doi.org/10.1007/s00246-018-1869-9
9. Christoffels, V., Moorman, A.: Development of the cardiac conduction system: why are some
regions of the heart more arrhythmogenic than others? Circ. Arrhythm. Electrophysiol. 2(2),
195–207 (2009). https://doi.org/10.1161/CIRCEP.108.829341
10. de Boer, B.A., van den Berg, G., de Boer, P.A.J., Moorman, A.F.M., Ruijter, J.M.: Growth of
the developing mouse heart: an interactive qualitative and quantitative 3D atlas. Dev. Biol.
368(2), 203–213 (2012). https://doi.org/10.1016/j.ydbio.2012.05.001
11. Hanson, K.P., Jung, J.P., Tran, Q.A., Hsu, S.-P.P., Iida, R., Ajeti, V., et al.: Spatial and tem-
poral analysis of extracellular matrix proteins in the developing murine heart: a blueprint for
regeneration. Tissue Eng. Part A. 19(9–10), 1132–1143 (2013). https://doi.org/10.1089/ten.
tea.2012.0316
12. Vincent, S.D., Buckingham, M.E.: How to make a heart: the origin and regulation of car-
diac progenitor cells. Curr. Top. Dev. Biol. 90, 1–41 (2010). https://doi.org/10.1016/
S0070-2153(10)90001-X
13. Simões, F.C., Riley, P.R.: The ontogeny, activation and function of the epicardium during heart
development and regeneration. Development (Cambridge). 145(7), dev155994 (2018). https://
doi.org/10.1242/dev.155994
14. Liang, S., Shi, X., Yu, C., Shao, X., Zhou, H., Li, X., et al.: Identification of novel candi-
date genes in heterotaxy syndrome patients with congenital heart diseases by whole exome
sequencing. Biochim. Biophys. Acta Mol. basis Dis. 2020(12), 165906 (1866). https://doi.
org/10.1016/j.bbadis.2020.165906
15. Kim, D.H., Xing, T.,Yang, Z., Dudek, R., Lu, Q., Chen,Y.-H.: Epithelial mesenchymal transi-
tion in embryonic development, tissue repair and cancer: a comprehensive overview. J. Clin.
Med. 7(1), 1 (2017). https://doi.org/10.3390/jcm7010001
16. Zhou, B., Pu, W.T.: More than a cover: epicardium as a novel source of cardiac progenitor
cells. Regen. Med. 3(5), 633–635 (2008). https://doi.org/10.2217/17460751.3.5.633
17. Young, K.A., Wise, J.A., DeSaix, P., Kruse, D.H., Poe, B., Johnson, E., et al.: Anatomy and
physiology, 1st edn, p. 1335. OpenStax (2013)
18. Anderson, R.H., Mori, S., Spicer, D.E., Brown, N.A., Mohun, T.J.: Development and morphol-
ogy of the ventricular outflow tracts. World J. Pedia. Congenit. Heart Surg. 7(5), 561–577
(2016). https://doi.org/10.1177/2150135116651114
From Simple Cylinder to Four-Chambered Organ: A Brief Overview of Cardiac…
26. 14
19. Mirzoyev, S., McLeod, C.J., Asirvatham, S.J.: Embryology of the conduction system for the
electrophysiologist. Ind. Pacing Electrophysiol J. 10(8), 329–338 (2010)
20. Markwald, R.R., Fitzharris, T.P., Manasek, F.J.: Structural development of endocardial cush-
ions. Am. J. Anat. 148(1), 85–119 (1977). https://doi.org/10.1002/aja.1001480108
21. Snarr, B.S., Kern, C.B., Wessels, A.: Origin and fate of cardiac mesenchyme. Dev. Dyn.
237(10), 2804–2819 (2008). https://doi.org/10.1002/dvdy.21725
22. Krishnan, A., Samtani, R., Dhanantwari, P., Lee, E., Yamada, S., Shiota, K., et al.: A detailed
comparison of mouse and human cardiac development. Pediatr. Res. 76(6), 500–507 (2014).
https://doi.org/10.1038/pr.2014.128
23. Lockhart, M.M., van den Hoff, M., Wessels, A., Nakanishi, T., Markwald, R.R., Baldwin,
H.S., et al.: The role of the epicardium in the formation of the cardiac valves in the mouse,
pp. 161–170. Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function
and Cellular Interaction to Morphology. Springer Open (2016)
24. Possner, M., Gensini, F.J., Mauchley, D.C., Krieger, E.V., Steinberg, Z.L.: Ebstein’s anomaly
of the tricuspid valve: an overview of pathology and management. Curr. Cardiol. Rep. 22(12),
157 (2020). https://doi.org/10.1007/s11886-020-01412-z
25. Obgynkey.com; Cardiac Development, Cardac Septation. Chapter 414.3.
26. Hatcher, C.J., Basson, C.T.: Specification of the cardiac conduction system by transcription fac-
tors. Circ. Res. 105(7), 620–630 (2009). https://doi.org/10.1161/CIRCRESAHA.109.204123
27. Wu, B., Zhang, Z., Lui, W., Chen, X., Wang, Y., Chamberlain, A.A., et al.: Endocardial cells
form the coronary arteries by angiogenesis through myocardial-endocardial VEGF signaling.
Cell (Cambridge). 151(5), 1083–1096 (2012). https://doi.org/10.1016/j.cell.2012.10.023
28. Red-Horse, K., Ueno, H., Weissman, I.L., Krasnow, M.A.: Coronary arteries form by develop-
mental reprogramming of venous cells. Nature (London). 464(7288), 549–553 (2010). https://
doi.org/10.1038/nature08873
29. Su, T., Stanley, G., Sinha, R., D’Amato, G., Das, S., Rhee, S., et al.: Single-cell analysis of
early progenitor cells that build coronary arteries. Nature (London). 559(7714), 356–362
(2018). https://doi.org/10.1038/s41586-018-0288-7
30. Diman, N., Brooks, G., Kruithof, B., Elemento, O., Seidman, J.G., Seidman, C., et al.: Tbx5
is required for avian and mammalian epicardial formation and coronary Vasculogenesis. Circ.
Res. 115(10), 834–844 (2014). https://doi.org/10.1161/CIRCRESAHA.115.304379
31. Kovacic, J.C., Dimmeler, S., Harvey, R.P., Finkel, T., Aikawa, E., Krenning, G., et al.:
Endothelial to mesenchymal transition in cardiovascular disease: JACC state-of-the-art review.
J. Am. Coll. Cardiol. 73(2), 190–209 (2019). https://doi.org/10.1016/j.jacc.2018.09.089
C. Lee et al.
28. 16
any cell expressing those genes as well as all subsequent progeny. By using markers
expressed by cardiomyocytes or cardiac progenitors, researchers can gain insight
into how these cells progress over the course of development and growth, or in
response to injury by analyzing their proliferative capacity as well as the expression
profile of the generated clones. In this chapter we will review two powerful lineage
tracing tools (Mosaic Analysis with Double Markers [MADM] and Rainbow) that
have been successfully used for clonal analysis of cardiomyocytes during normal
development and after injury. Both models allow for precise fate tracking and lin-
eage tracing, making them applicable to many different types of studies.
2
Mosaic Analysis with Double Markers (MADM)
The extent of cardiomyocyte proliferation during development and in the postnatal
heart remains an area of controversy. Recent studies suggest that there is a low turn-
over of cardiomyocytes that declines with age in mice and humans [2–6]. It has
come to consensus in the field that newly generated cardiomyocytes originate from
pre-existing cardiomyocytes through proliferation rather than differentiation from
stem cells [5]. Studies of cardiomyocyte proliferation have been limited by reliance
on indirect assays of markers for cell proliferation and on surrogates for cell divi-
sion [7, 8]. These studies are particularly challenging to interpret, due to the con-
founding issues of cardiomyocyte polyploidy, multi-nucleation, and DNA repair
after injury in adult hearts [9, 10]. Therefore, it is important to develop a system that
differentiates between karyokinesis and cytokinesis in adult cardiomyocytes.
Mosaic Analysis with Double Markers (MADM) uses Cre-Recombinase technol-
ogy that induces genetic recombination upon cell division, which indelibly labels
clones of proliferating cells with a single fluorescent marker. Analysis of clonal
expansion using MADM has numerous applications in studying cellular prolifera-
tion in development, stem cell biology, and regenerative medicine.
2.1 Methodology
The MADM strategy uses two reciprocally chimeric genes that are knocked into the
same location on homologous chromosomes, with each containing the N-terminus
of one reporter and the C-terminus of the other reporter, interrupted by a lox-P site.
After DNA replication, genetic recombination induced by Cre-LoxP creates func-
tional reporter genes (green fluorescent protein [GFP] or red fluorescent protein
[RFP]). Upon G2-X-segregation during cell division, the divided cells express
either GFP or RFP, and this feature allows MADM to be used for genetically record-
ing cytokinesis events upon G2-X segregation. Considering that inter-chromosomal
Cre-LoxP recombination after S phase is a rare event, labeling a fraction of cells of
interest allows for clonal analysis of marked cells and their progeny. Since G2-X
K. Kolluri et al.
29. 17
Fig. 1 Mosaic Analysis with Double Markers (MADM). (a) Schematic detailing how MADM
works. Upon DNA replication and MADM recombination, the cell can result in three different
situations. Upon G2-X segregation, two single-labeled daughter cells will arise, with one being
GFP+
, the other RFP+
. G2-Z segregation results in one double-labeled (GFP+
RFP+
) cell and one
unlabeled (GFP−
RFP−
) cell. G0 recombination with no division results in one double-labeled cell
(GFP+
RFP+
). (b) Two distinctly, single-labeled, sibling cardiomyocytes exhibiting intimate end-on
contact (Scale bar, 10 μm) [13]. (c) Myocardial infarction (MI) was induced by Left Anterior
Descending Artery (LAD) ligation. The number of cardiomyocytes in Sham versus MI is similar,
suggesting that MI does not result in any appreciable increase of cardiomyocyte proliferation in the
left ventricle (LV). White arrowheads point to single-labeled cells (Scale bars, 120 μm) [13]. (d)
The utility of MADM in post-mitotic and mitotic tissues. Hprt-Cre was used to generate MADM
labeled cells in post-mitotic tissues (liver, kidney, heart, spleen). Actin-Cre was used to label cells
in the epidermis while keratin5-Cre was used to label keratinocytes in the epidermis (Scale bars:
(Aa)-(Da) and (Ab)–(Db), 2 mm; (Ac)–(Dc), 50 μm; (Ea) and (Fa), 100 μm; (Eb) and (Fb), 10 μm)
[12]. (e) Two single-labeled daughter cardiomyocytes generated by a MADM recombination/divi-
sion event in the Myh6/MADM model (Scale bar, 15 μm) [13]. (f) Section from a P12 Myh6/
MADM heart revealing the presence of sparse single-labeled and double-labeled cells (Scale bar,
100 μm). (i) GFP+
single-labeled cardiomyocyte with its sibling RFP+
single-labeled cell, and
double-labeled cells. (ii) GFP+
single-labeled cell with a double-labeled cell. The GFP+
cell con-
tains visible sarcomeric elements, demonstrating the fidelity of this model in marking cardiomyo-
cytes. (iii) RFP+
single-labeled cardiomyocyte (Scale bars (i-iii), 10 μm) [13]
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
30. 18
mitosis is required to generate single-labeled cells, the MADM system unambigu-
ously labels divided cells [11, 12]. Figure 1a details a schematic for how
MADM works.
In addition, MADM allows for asymmetric labeling of daughter cells so that a
relationship between precursor-progeny lineages can be established [14]. This is
particularly important in studies involving stem cell differentiation, organ develop-
ment and tissue regeneration in response to injury. Since MADM couples mitosis
with labeling, MADM can be used to identify the progenitor cell or single cells that
can be tracked for clonal expansion. The two differentially labeled MADM daugh-
ter cells can be retrospectively analyzed to investigate clonal analysis and patterns
of cell division. MADM has been successfully used to track stem cell division [12,
15, 16]. In the case of an asymmetric stem cell division where a daughter stem cell
and a differentiated cell are generated, each daughter cell is differentially labeled. If
the differentiated cell continues to proliferate, then a clone of single-labeled cells
can be identified. This is particularly useful for in vivo stem cell tracking, since
MADM events are rare, hence labeling a small number of cells and their progeny.
This allows for retrospective tracing of cellular expansion through easily identifi-
able sparse clones. In addition, multilineage cell potential can be studied by track-
ing whether certain clones generate multiple different cell types [14].
The labeling efficiency of MADM is partly dependent on the Cre line used.
Obviously, ubiquitous Cre lines result in more doublelabeled cells, which is due to
the increase in post-mitotic cells, which undergo G0 recombination [12]. Tissue-
specific inducible Cre lines are routinely used for two main reasons: 1) to control
the extent of recombination events, and 2) to control the timing of recombination
events according to the study design. When experimental planning requires clonal
analysis at specific time points (i.e. at precise stages of development or after injury),
temporal control of MADM labeling can be achieved using transiently expressed
Cre lines [12]. While the amount of tamoxifen induces a desired recombination
event, the timing of tamoxifen administration allows for temporal control of recom-
bination and fluorescent labeling of cells.
2.2
Labeling Rare Populations and Lineage Tracing
The first study to use MADM for clonal analysis in the heart was reported by Ali
et al. [13]. First, they used an HprtCre+/−
/MADM-11GT/TG
model, which can label
any cell, and observed distinct clusters of RFP+
and GFP+
cells in the heart. In addi-
tion, Tasic et al. generated another transgenic mouse model where they inserted the
reconstituted GFP gene into one Hipp11 locus and the reconstituted RFP gene into
the other Hipp11 locus of each cell [17]. In this model, MADM-11GG/TT
, they
observed only double-labeled cells and found no evidence of single or unlabeled
cells. This further validated the use of MADM in the heart because there was no
observed silencing of the MADM transgene, thereby ensuring no false negative
results. After confirming the validity of MADM in the heart, Ali and colleagues
K. Kolluri et al.
31. 19
created an inducible Myh6CreERT2;MADM-11GT/TG
mouse model in which upon
tamoxifen induction, Myh6 dividing cells may undergo homologous recombina-
tion, resulting in two distinctly labeled daughter cells (Fig. 1b). Newborn pups were
given tamoxifen and their hearts were analyzed at P12. They observed that approxi-
mately 11% of labeled cells were single-labeled, which were progeny of Myh6-
expressing cardiomyocytes. These cells all expressed alpha-actinin and contained
sarcomeric elements, demonstrating that dividing cardiomyocytes gave rise to new
cardiomyocytes. In addition, they observed equivalent frequencies of GFP+
and
RFP+
labeled cells, demonstrating that cardiomyocytes divide to generate further
cardiomyocytes in a symmetric fashion. In many cases, the cardiomyocyte clones
were noncontiguous, separating from each other after division.
They next induced MADM recombination at E13.5 and analyzed the labeling of
Myh6 expressing cells during development. They found that a majority of the cells
were single-labeled, confirming that a majority of these cells are mitotically active.
In contrast, they observed a significant number of double-labeled cardiomyocytes
after birth, likely arising from G0 inter-chromosomal recombination, indicating that
after birth, cardiomyocytes are not mitotically active. While their work showed that
cardiomyocytes were the source of proliferating cells during development, Ali et al.
demonstrated that only a very small portion of cardiomyocytes divide in the adult
heart. Their lineage tracing experiments confirmed that pre-existing cardiomyo-
cytes generate cardiomyocytes in adults at a low rate after birth.
Next, they used a β-ActinCreER/MADM-11GT/TG
model, which permits MADM
recombination in any cell-type. The use of the β-ActinCreER/MADM model
allowed the investigators to determine if there is a stem/progenitor cell source for
cardiomyocytes, and their findings clearly and unambiguously argued against the
existence of a multipotent progenitor cell in the adult heart akin to canonical stem
cells in other tissues. To investigate the proliferative behavior of cardiomyocytes
after injury, MI was induced by ligation of the left anterior descending artery (LAD)
at 8 weeks of age, with tamoxifen administration for 2 weeks. They analyzed the
hearts 4 weeks after MI and found similar frequencies of single-labeled cells in both
sham and MI, suggesting that injury in mice does not necessarily induce cardiomyo-
cyte proliferation above the basal level (Fig. 1c). An alternative explanation could
be the inefficient inter-chromosomal recombination in the setting of induced Cre
recombinase in the adult cardiomyocytes. Further iteration of MADM for more
efficient recombination would improve our understanding of adult cardiomyocyte
proliferation in homeostasis and after injuries. The study by Ali et al. demonstrated
for the first time how cardiomyocytes can be labeled through MADM and how their
fate can be tracked both in response to injury and through several stages of
development.
MADM has also been used to study cardiomyocyte division stimulated by cell-
cycle gene induction. Mohamed and colleagues used a combination of four cell
cycle regulators (CDK1:CCNB and CDK4:CCND complexes) to induce cardio-
myocyte proliferation and growth in vitro and in vivo [18]. Particularly, they used
MADM lineage tracing and demonstrated that adult cardiomyocyte division could
be induced in vivo at an efficiency of at least 15%.
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
32. 20
2.3
Utility of MADM in Other Organ Systems
Zong et al. first reported the development of the MADM system [12]. In their
groundbreaking report, they demonstrated that Cre-dependent inter-chromosomal
recombination can be induced efficiently in vivo in mitotic and post-mitotic cells
(Fig. 1d). They used this system for conditional gene knockout, lineage analysis,
and neural connection tracing. To illustrate the utility of MADM, they studied the
fate of granule cell progenitors in the cerebellar cortex. Using the MADM system,
they identified 26 distinct subclusters of granule cells in the cerebellar cortex. They
showed that these granule cell clusters exhibit limited dispersion, that there was a
low frequency of generating these clusters, and that there was a small chance that
clusters were generated from two separate clonal lineages. They reasoned that each
cluster was likely the progeny of a single-labeled clone. Thus, Zong et al. success-
fully used the MADM system for the first time to demonstrate that granule cell
progenitors are fated to give rise to adult granule cells which distinctly localize and
project axons to specific sublayers of the cerebellar cortex.
MADM has been used extensively for developmental studies in the field of neu-
roscience. Mihalas and Hevner used MADM to study the differentiation of early
intermediate progenitors (IP) and their role in the developing cerebral cortex [16].
IP cells are derived from radial glial progenitor cells and give rise to pyramidal
projection neurons in the cerebral cortex. Their data suggested three main models
for IP cell differentiation. Their analysis revealed that IP cells can have asymmetric
fates and generate multilayered clones, or undergo rapid or delayed terminal dif-
ferentiation to produce either upper or lower-layer cortical neurons, respectively. In
all of the suggested processes, the authors observed asymmetric cell death.
MADM has since been used in other systems, particularly cancer. Liu et al. used
a MADM-based model for glioma to lineage trace neural stem cells (NSC) and
distinguish between cell-of-mutation and cell-of-origin [15]. The lineage tracing
feature of MADM allowed this group to track clones throughout the process of
tumorigenesis. The mutant cells were labeled GFP+
, whereas the wild-type cells,
which served as internal controls, were labeled RFP+
. Importantly, the MADM sys-
tem was utilized to trace the cells at pre-malignant stages. They induced p53 and
NF1 mutations in NSCs and observed that oligodendrocyte progenitor cells exhib-
ited higher proliferative capacity, thus pointing to these as the cell-of-origin in their
glioma model. By using MADM to differentiate between mutant and wild type
cells, Liu et al. were able to conduct a detailed analysis of the precise physiological
changes that occur during tumor formation and identify a cell-of-origin in their
glioma model.
The MADM system has also been used in a variety of genetic imprinting studies.
A particular advantage is that MADM can be used to study uniparental disomy
(UPD) by indelibly and unambiguously labeling either unimaternal or unipaternal
disomic cells. Hippenmeyer et al. studied the effects of genomic imprinting in chro-
mosome 7 and 12 and explored chromosome and cell-type specific imprinting [19].
In addition, Laukoter et al. found that UPD in the neocortex results in highly
K. Kolluri et al.
33. 21
cell-type specific genome-wide changes [20]. They also used MADM to reveal dif-
ferences in paternal dominant and maternal dominant UPD within cortical astrocytes.
2.4 Limitations/Future Directions
The major limitation of traditional systems for conditional gene knockout has been
the difficulty to achieve strict coupling of knockout and labeling. Since there is a
single chromosomal exchange event in MADM, generation of homozygous cells
and labeling is coupled, hence leaving little chance for ambiguity. However, there
are several limitations with MADM. First, the efficiency of interchromosomal
recombination is much less than intrachromosomal recombination used in tradi-
tional knockout systems. Although this could be a desirable feature for analyzing
single-cell autonomous gene function, it may become a problem where high fre-
quency of gene knockout is desired. Also, since MADM is based on the availability
of a pair of MADM knock-ins between the gene of interest and the centromere,
there is a need to generate knock-in cassettes for other chromosomes, an effort that
has been realized in recent publications [12, 15, 19, 20].
For lineage tracing and clonal analysis studies, a potential limitation of the
MADM system is performing event quantification. A binucleated daughter cell
resulting from G2-Z segregation without cytokinesis would be double-labeled, an
observation that is frequently encountered when analyzing heart tissue during
development (Fig. 1e). However, if cytokinesis occurs, G2-Z segregation could lead
to one double-labeled daughter cell while its sibling would be unlabeled (Fig. 1f).
This ultimately could cause underestimation of the number of proliferated cells.
Another limitation is the fact that MADM cannot be used as effectively in post-
mitotic cells as it depends on mitosis in order to label cells [12]. Terminally differ-
ential cells in G0 can also undergo recombination without cytokinesis, leading to
the presence of a double-labeled cell.
3 Rainbow Reporter
The rainbow reporter system is a novel stochastic four-color Cre-dependent reporter
system that has been used for clonal analysis studies. The advantage of this system
lies in its ability to randomly assign different fluorescent labels to cells of interest,
allowing for retrospective tracing of their progeny with easily distinguishable clones
in vivo. When used in combination with an inducible tissue-specific Cre mouse line,
recombination events can be controlled in a spatiotemporal manner. Random recom-
bination events in proliferating cells will result in clones of cells that retain the same
fluorescent label as the parental cell. When rare recombination events occur (i.e. as
a result of limited amount of tamoxifen administration), sparse clones are direct
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
34. 22
evidence of cell proliferation, whereas the size of clones is suggestive of their pro-
liferative capacity through a specific time window.
3.1 Methodology
The Rainbow system relies on Cre-dependent recombination to induce indelible
labeling of one of three random fluorescent markers. Rainbow mice carry a cassette
of four fluorescent genes (GFP, mCerulean, mOrange, and mCherry) inserted in the
Rosa26 (R26) locus. Without Cre-mediated recombination, all tissues express the
default GFP reporter label. When the Rainbow mouse is crossed with a tissue-
specific inducible Cre line, tamoxifen administration leads to random excision of a
pair of the mutated LoxP sites, resulting in permanent and exclusive expression of
one of the three fluorescent proteins. Rainbow can be used to retrospectively trace
cell lineages by identifying and counting same-colored clones that arise from a
common progenitor. When crossed with a mouse line that expresses Cre/CreER
under the promoter of a certain marker gene, this model can be used to label and
track the fate of a population of cells of interest. The permanent labeling feature also
allows for analysis over a long period of time in processes such as cellular dynamics
and symmetry of cell divisions in a single cell lineage [21]. Figure 2a illustrates a
schematic of how the Rainbow reporter works.
We previously reported the utility of the Rainbow system to retrospectively iden-
tify the source of new cardiomyocytes during fetal and neonatal development, as
well as in adult hearts after injury [23]. Through 3D clonal analysis of cardiovascu-
lar progenitors and cardiomyocytes, we demonstrated that cardiac progenitors are
the main source of cardiomyocytes during murine cardiac development. The lineage
tracing experiments revealed that immature cardiomyocytes maintain their prolif-
erative potential throughout embryonic development, however, there is a decline in
their proliferation as they progress to more mature cardiomyocytes. In this study,
several inducible mouse models were used in combination with the Rainbow system
which allowed for distinguishable reporter expression in the heart as opposed to a
mosaic pattern generated by a non-inducible Cre model. Clones of cells were per-
manently labeled with one of the three fluorescent proteins and further staining for
α-sarcomeric actinin confirmed their cardiomyocyte identity. In order to determine
the size of the generated clonal clusters, the cell counter tool on ImageJ software
was used to quantify the number of cells per clone (Fig. 2b). Additionally, for a
more detailed three-dimensional clone volume analysis and anatomical localization,
a modified CLARITY technique was used in which the heart was transformed into
an optically translucent but structurally preserved organ. The cleared hearts were
subsequently imaged by confocal and light-sheet fluorescence microscopy. This
advanced imaging modality facilitated an accurate measurement of clone volumes
at different time points during heart development.
K. Kolluri et al.
35. 23
Fig. 2 Rainbow Reporter: (a) Schematic of how Rainbow works. When a Rosa26 Rainbow mouse
is crossed with a tissue-specific CreER mouse, expression of the tissue-specific marker results in
Cre expression, which allows for the excision of a random pair of loxP sites and results in expres-
sion of one of three fluorescent proteins (mCerulean, mOrange, or mCherry). Cells that do not
undergo recombination express GFP. In the absence of cell proliferation, the tissue gives a mosaic
appearance. When cell proliferation occurs, a clear abundance of same-colored cells is visible. (b)
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
36. 24
3.2
Labeling of Rare Cell Populations and Lineage Tracing
Sereti et al. were the first to use Rainbow mice to study the regenerative capacity of
the heart [22]. In order to first demonstrate the utility of Rainbow in the heart, they
utilized 3 different transgenic Cre mouse models, under the control of either the
cardiovascular progenitor genes Mesp1 and Nkx2.5 or the more mature cardiomyo-
cyte marker αMHC. Analysis of hearts at embryonic day 14.5 (E14.5), postnatal
day 1 (P1) and P21 revealed that the hearts marked by progenitor markers formed
clonal clusters while those marked by αMHC showed a mosaic pattern of singletons
(Fig. 2c). The use of constitutively active Cre lines, although very informative, nor-
mally produces high levels of recombination, and one cannot exclude the possibility
that the observed single-color cell clusters are the results of random recombination
events. On the other hand, a tamoxifen-inducible Cre line permits spatiotemporal
control of recombination events. By administering a limiting amount of tamoxifen,
one can achieve a handful of labeled cells in an organ and follow their fate
retrospectively.
The authors next crossed Rainbow mice with mice harboring an inducible Cre
under the control of a βactin, Nkx2.5, or αMHC promoter. This approach allowed
for distinction between non-cardiomyocyte-derived clonal expansion (βactinCreER
;
R26VT2/GK
), cardiac progenitor-derived clonal expansion (Nkx2.5CreER
; R26VT2/GK
), or
mature cardiomyocyte (αMHCCreER
; R26VT2/GK
) clonal expansion. When a limiting
amount of tamoxifen was administered at E9.5 or E12.5 to βactinCreER
; R26VT2/GK
mice, postnatal heart analysis revealed clear clones of cardiomyocytes, fibroblasts,
endothelial and vascular smooth muscle cells (Fig. 2d). Clonal analysis of
Nkx2.5CreER
; R26VT2/GK
mice labeled at E9.5 or E12.5, and analyzed postnatally, also
revealed a similar pattern of clonal expansion with comparable clone size and vol-
ume to those observed in βactinCreER
; R26VT2/GK
mice. These findings supported the
Fig. 2 (continued) Marking a cardiomyocyte clone for counting purposes. Cells are pseudo-col-
ored red and WGA staining is used to mark cell boundaries (Scale bar, 100 μm) [22]. (c)
Longitudinal sections from E14.5, P1 and P21 transgenic mice under the control of the progenitor
markers Mesp1 (i-iii) and Nkx2.5 (iv-vi) or the adult CM marker αMHC (vii-ix). Fluorescent
microscope images from hearts under the control of progenitor markers (ii, v) revealed the pres-
ence of clear clonal clusters. Images from hearts under the control of αMHC (viii) revealed a
mosaic pattern of singletons, with no definite clonal clusters (Scale bar (iii, vi, ix), 500 μm; all
others 50 μm) [22]. (d) The utility of the βactinCreER
; R26VT2/GK
model in marking other cell types
in the heart. (i) shows a close up confocal image of a clone (blue) containing vascular smooth
muscle cells in a P7 heart stained for smooth muscle Myosin Heavy Chain (smMHC). (ii) shows a
confocal image of cardiomyocyte clones in a P15 Nkx2.5CreER
; R26VT2/GK
heart stained
for α-sarcomeric actinin (Scale bar (i), 50 μm; (ii), 100 μm) [22]. (e) Limited tamoxifen adminis-
tration allows for rare recombination events. Section of a βactinCreER
; R26VT2/GK
adult heart, in
which recombination was induced at E9.5, shows the presence of sparse single-colored clones
labeled with mOrange and mCherry (Scale bar, 500 μm) [22]. (f) Representative confocal images
of sections from βactinCreER
; R26VT2/GK
(i), Nkx2.5CreER
; R26VT2/GK
(ii) and αMHCCreER
; R26VT2/GK
(iii) neonatal mice that received LAD ligation at P0. Clonal analysis performed 21 days after MI
reveals the presence of sparse single-labeled clones, suggesting that neonatal mice undergo cardio-
myocyte regeneration in response to injury [22]
K. Kolluri et al.
37. 25
proliferative capacity of progenitor cells to generate cardiomyocytes during early
fetal development (Fig. 2e).
In order to explore the proliferative capacity of cardiomyocytes during fetal
development, recombination was induced in cardiomyocytes at different embryonic
time points using αMHCCreER
; R26VT2/GK
mice. When tamoxifen was administered at
E12.5, postnatal analysis revealed mostly singleton cardiomyocytes with few small
size clones. However, interrogation of αMHCCreER
; R26VT2/GK
mice labeled at E9.5
revealed similar size cardiomyocyte clones compared to βactinCreER
; R26VT2/GK
or
Nkx2.5CreER
; R26VT2/GK
mice. These data suggest that αMHC-expressing cardiomyo-
cytes at E9.5 retain the ability to proliferate and that this capacity is significantly
diminished by E12.5.
They went on to perform single cell transcriptional analysis of αMHC-expressing
cardiomyocytes at E9.5, E12.5 and P1. Their investigation demonstrated the exis-
tence of a heterogeneous population of cardiomyocytes within the early stages of
cardiac development and their transition into a mature, less proliferative, and
homogenous population by the early postnatal period. Overall, the use of the
Rainbow model revealed that clonal dominance of differentiating progenitors medi-
ates cardiac development, while a distinct subpopulation of cardiomyocytes may
have the potential for limited proliferation during late fetal and early postnatal life.
Such precise analyses at a single cell resolution would be challenging and prone to
inaccurate interpretations if traditional lineage tracing experiments were utilized. It
would be important to incorporate new technology in future, eg. DNA barcoding,
for high-throughput analysis of a large number of individual cardiomyocytes and
their progenies in developing hearts.
Sereti et al. also used Rainbow to study clonal expansion in neonatal and adult
cardiomyocytes in response to cardiac injury. Newborn (P0) αMHCCreER
; R26VT2/GK
,
βactinCreER
; R26VT2/GK
and Nkx2.5CreER
; R26VT2/GK
mice received tamoxifen followed
by LAD ligation or Sham operation at P1. At 21 days after MI, hearts were analyzed
and frequent clones of cardiomyocytes were observed in the infarct and border zone
areas of αMHCCreER
; R26VT2/GK
mice (Fig. 2f). Similar observations were made with
βactinCreER
; R26VT2/GK
and Nkx2.5CreER
; R26VT2/GK
mice after injury. This suggested
that regeneration of the heart after injury in neonates was largely due to cardiomyo-
cyte proliferation, which was confirmed by recent fate mapping studies of cardio-
myocytes and non-cardiomyocytes in neonates [24]. In adults, LAD ligation was
performed at 8 weeks of age and analysis at 21 days post-MI revealed the presence
of sparse single-labeled clones in the infarct and border zone areas. These observa-
tions suggested that injury induces cardiomyocyte proliferation in the neonatal heart
but not in the adult heart.
Wang et al. used the Rainbow system to study the clonal expansion of smooth
muscle cells (SMC) in atherosclerosis [25]. Myh11CreERT2
; R26VT2/GK
; ApoE−/−
mice
were fed a high-fat Western diet to induce atherogenesis. The authors observed that
during early atherogenesis, there was a distinct subpopulation of SMCs that de-
differentiated and upregulated Sca1 (a stem cell marker). Sca1 staining was most
intense within the core of the dominant clone and Sca1+
cells appeared to colocalize
near the necrotic core of the atherosclerotic plaque. Single cell RNA sequencing
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
38. 26
analysis of this Sca1+
population revealed that these cells down-regulated SMC
marker genes and upregulated genes relating to inflammation and the complement
cascade (complement C3 was one of the most significantly up-regulated factors).
These findings translated to human specimens as well. Histological analysis of post-
mortem carotid artery specimens revealed localization of C3 expression to the
necrotic core of human atherosclerotic plaque, similar to mice. Furthermore, this
Sca1+
SMC signature was found in humans and was associated with coronary artery
disease, with an enriched expression of inflammatory genes (including C3).
Production of C3 by the Sca1+
SMC population may play a role in triggering further
SMC proliferation and vascular inflammation, which could explain the rapid expan-
sion of SMCs during early atherogenesis. Overall, Wang et. al’s study reveals the
potential of the Rainbow system to identify rapidly proliferating and differentiating
populations of cells to gain insight into the pathophysiology of complex diseases
such as atherosclerosis.
3.3
Utility of Rainbow in Other Organ Systems
Rinkevich et al. used an inducible lineage tracing mouse model (βactinCreER
; R26VT2/
GK
) to perform lineage tracing and clonal analysis of individual cells of mouse hind
limb tissue during regeneration of the digit tip, cutaneous wound healing, and nor-
mal maintenance [26]. They removed nerve supply and observed clonal expansion,
revealing that cellular regeneration remains largely intact in the absence of nerve
supply. In a study of the kidney, Rinkevich et al. used the Rainbow mouse model for
clonal analysis and lineage tracing of cells that contribute to the development, main-
tenance and regeneration of the kidney [27]. In all three processes, they found that
cells generating distinct parts of the nephron (i.e. Proximal, Distal tubules or col-
lecting duct) were fate-restricted and stayed within their lineage. Furthermore, they
used an Axin2CreER
; R26VT2/GK3
mouse line to track Wnt pathway responsive cells
(WRC). They showed that WRCs increased their proliferative capacity and that
their clones were restricted to either a proximal tubule or collecting duct fate.
Recent studies using dual recombinases and Confetti reporter (also random
labeling by one of the three fluorescences) demonstrated that bronchioalveolar stem
cells residing in the bronchioalveolar duct junction could clonally expand to form
bronchial epithelial cells and/or alveolar type I and II cells during lung repair and
regeneration [28, 29]. This demonstrates the utility of clonal analysis by rainbow/
confetti reporters for resolving the uni- or bi-differentiation potential of stem cells
in tissue regeneration.
Interestingly, Rainbow has also been used in cancer models. For example, Corey
et al. lineage traced endothelial cells within the tumor microenvironment and con-
cluded that clonal expansion within the microvasculature is crucial to an invasive
melanoma phenotype [30]. Particularly, their studies showed that there is a dimin-
ishing of founder clones to produce subclones, with tumor blood vessels upregulat-
ing genes associated with angiogenesis and a downregulation of lymphocyte
K. Kolluri et al.
39. 27
adhesion molecules. Thus, the Rainbow mouse model allowed lineage tracing of
single cells and their progeny to conclude that clonal evolution within melanoma
can induce changes within the microvasculature to confer cancer cells an advantage.
3.4 Limitations/Future Directions
The ability of Rainbow to precisely track the fate of certain cells across multiple
organ systems makes it a strong candidate for clonal analysis and lineage tracing
within the heart. This model allows researchers to differentiate between proliferat-
ing and quiescent cardiomyocytes, a phenomenon that remains controversial in the
field of cardiac development and regeneration. However, it is not without limita-
tions. A major consideration when using the Rainbow mouse model is the efficiency
of recombination events that partially depends on the Cre line used. While in many
mouse lines, Rainbow faithfully induces equal expression of fluorescent proteins in
labeled cells, some Cre lines have been reported to show uneven expression of the
markers. Additionally, the model’s dependence on tamoxifen could leave the pos-
sibility that a smaller dose does not induce enough recombination to mark all pos-
sible clones, thus underestimating the number of clones [22]. Furthermore, since in
clonal expansion studies, only a small number of cells are labeled and their fate is
monitored retrospectively, it is possible that rare populations of cells cannot be
labeled by this strategy. The Rainbow system can be used for clonal analysis studies
and the presence of clusters of single-colored cells confidently supports the exis-
tence of proliferating cells. However, the absence of an observation does not con-
firm its lack of existence.
References
1. Kretzschmar, K., Watt, F.M.: Lineage tracing. Cell. 148(1-2), 33–45 (2012). https://doi.
org/10.1016/j.cell.2012.01.002
2. Bergmann, O., Bhardwaj, R.D., Bernard, S., Zdunek, S., Barnabé-Heider, F., Walsh, S., et al.:
Evidence for cardiomyocyte renewal in humans. Science. 324(5923), 98–102 (2009). https://
doi.org/10.1126/science.1164680
3. Bergmann, O., Zdunek, S., Felker, A., Salehpour, M., Alkass, K., Bernard, S., et al.: Dynamics
of cell generation and turnover in the human heart. Cell. 161(7), 1566–1575 (2015). https://
doi.org/10.1016/j.cell.2015.05.026
4. Cai, C.L., Molkentin, J.D.: The elusive progenitor cell in cardiac regeneration: slip Slidin’
away. Circ. Res. 120(2), 400–406 (2017). https://doi.org/10.1161/CIRCRESAHA.116.309710
5. Eschenhagen, T., Bolli, R., Braun, T., Field, L.J., Fleischmann, B.K., Frisén, J., et al.:
Cardiomyocyte regeneration: a consensus statement. Circulation. 136(7), 680–686 (2017).
https://doi.org/10.1161/CIRCULATIONAHA.117.029343
6. Vagnozzi, R.J., Molkentin, J.D., Houser, S.R.: New myocyte formation in the adult heart:
endogenous sources and therapeutic implications. Circ. Res. 123(2), 159–176 (2018). https://
doi.org/10.1161/CIRCRESAHA.118.311208
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
40. 28
7. Wei, K., Serpooshan, V., Hurtado, C., Diez-Cuñado, M., Zhao, M., Maruyama, S., et al.:
Epicardial FSTL1 reconstitution regenerates the adult mammalian heart. Nature. 525(7570),
479–485 (2015). https://doi.org/10.1038/nature15372
8. Yu, W., Huang, X., Tian, X., Zhang, H., He, L., Wang,Y., et al.: GATA4 regulates Fgf16 to pro-
mote heart repair after injury. Development. 143(6), 936–949 (2016). https://doi.org/10.1242/
dev.130971
9. Laflamme, M.A., Murry, C.E.: Heart regeneration. Nature. 473(7347), 326–335 (2011). https://
doi.org/10.1038/nature10147
10. Soonpaa, M.H., Kim, K.K., Pajak, L., Franklin, M., Field, L.J.: Cardiomyocyte DNA synthesis
and binucleation during murine development. Am. J. Phys. 271(5 Pt 2), H2183–H2189 (1996).
https://doi.org/10.1152/ajpheart.1996.271.5.H2183
11. Soonpaa, M.H., Field, L.J.: Survey of studies examining mammalian cardiomyocyte DNA
synthesis. Circ. Res. 83(1), 15–26 (1998). https://doi.org/10.1161/01.res.83.1.15
12. Zong, H., Espinosa, J.S., Su, H.H., Muzumdar, M.D., Luo, L.: Mosaic analysis with double
markers in mice. Cell. 121(3), 479–492 (2005). https://doi.org/10.1016/j.cell.2005.02.012
13. Ali, S.R., Hippenmeyer, S., Saadat, L.V., Luo, L., Weissman, I.L., Ardehali, R.: Existing car-
diomyocytes generate cardiomyocytes at a low rate after birth in mice. Proc. Natl. Acad. Sci.
U. S. A. 111(24), 8850–8855 (2014). https://doi.org/10.1073/pnas.1408233111
14. Weissman, I.L.: Stem cells: units of development, units of regeneration, and units in evolution.
Cell. 100(1), 157–168 (2000). https://doi.org/10.1016/s0092-8674(00)81692-x
15. Liu, C., Sage, J.C., Miller, M.R., Verhaak, R.G., Hippenmeyer, S., Vogel, H., et al.: Mosaic
analysis with double markers reveals tumor cell of origin in glioma. Cell. 146(2), 209–221
(2011). https://doi.org/10.1016/j.cell.2011.06.014
16. Mihalas, A.B., Hevner, R.F.: Clonal analysis reveals laminar fate multipotency and daugh-
ter cell apoptosis of mouse cortical intermediate progenitors. Development. 145(17) (2018).
https://doi.org/10.1242/dev.164335
17. Tasic, B., Miyamichi, K., Hippenmeyer, S., Dani, V.S., Zeng, H., Joo, W., et al.: Extensions
of MADM (mosaic analysis with double markers) in mice. PLoS One. 7(3), e33332 (2012).
https://doi.org/10.1371/journal.pone.0033332
18. Mohamed, T.M.A., Ang, Y.S., Radzinsky, E., Zhou, P., Huang, Y., Elfenbein, A., et al.:
Regulation of cell cycle to stimulate adult cardiomyocyte proliferation and cardiac regenera-
tion. Cell. 173(1), 104–116 (2018). https://doi.org/10.1016/j.cell.2018.02.014
19. Hippenmeyer, S., Johnson, R.L., Luo, L.: Mosaic analysis with double markers reveals
cell-type-specific paternal growth dominance. Cell Rep. 3(3), 960–967 (2013). https://doi.
org/10.1016/j.celrep.2013.02.002
20. Laukoter, S., Pauler, F.M., Beattie, R., Amberg, N., Hansen, A.H., Streicher, C., et al.: Cell-
type specificity of genomic imprinting in cerebral cortex. Neuron. 107(6), 1160–1179 (2020).
https://doi.org/10.1016/j.neuron.2020.06.031
21. He, L., Nguyen, N.B.,Ardehali, R., Zhou, B.: Heart regeneration by endogenous stem cells and
cardiomyocyte proliferation: controversy, fallacy, and Progress. Circulation. 142(3), 275–291
(2020). https://doi.org/10.1161/CIRCULATIONAHA.119.045566
22. Sereti, K.I., Nguyen, N.B., Kamran, P., Zhao, P., Ranjbarvaziri, S., Park, S., et al.: Analysis of
cardiomyocyte clonal expansion during mouse heart development and injury. Nat. Commun.
9(1), 754 (2018). https://doi.org/10.1038/s41467-018-02891-z
23. Nguyen, N., Fernandez, E., Ding, Y., Hsiai, T., Ardehali, R.: In vivo clonal analysis of car-
diomyocytes. In: Poss, K., Kühn, B. (eds.) Cardiac Regeneration: Methods and Protocols.
Methods in Molecular Biology, vol. 2158, pp. 243–256. Springer (2021)
24. Li, Y., Lv, Z., He, L., Huang, X., Zhang, S., Zhao, H., et al.: Genetic tracing identifies early
segregation of the cardiomyocyte and nonmyocyte lineages. Circ. Res. 125(3), 343–355
(2019). https://doi.org/10.1161/CIRCRESAHA.119.315280
25. Wang, Y., Nanda, V., Direnzo, D., Ye, J., Xiao, S., Kojima, Y., et al.: Clonally expanding
smooth muscle cells promote atherosclerosis by escaping efferocytosis and activating the
K. Kolluri et al.
41. 29
complement cascade. Proc. Natl. Acad. Sci. U. S. A. 117(27), 15818–15826 (2020). https://
doi.org/10.1073/pnas.2006348117
26. Rinkevich, Y., Montoro, D.T., Muhonen, E., Walmsley, G.G., Lo, D., Hasegawa, M., et al.:
Clonal analysis reveals nerve-dependent and independent roles on mammalian hind limb tis-
sue maintenance and regeneration. Proc. Natl. Acad. Sci. U. S. A. 111(27), 9846–9851 (2014).
https://doi.org/10.1073/pnas.1410097111
27. Rinkevich, Y., Montoro, D.T., Contreras-Trujillo, H., Harari-Steinberg, O., Newman, A.M.,
Tsai, J.M., et al.: In vivo clonal analysis reveals lineage-restricted progenitor characteristics in
mammalian kidney development, maintenance, and regeneration. Cell Rep. 7(4), 1270–1283
(2014). https://doi.org/10.1016/j.celrep.2014.04.018
28. Liu, Q., Liu, K., Cui, G., Huang, X.,Yao, S., Guo, W., et al.: Lung regeneration by multipotent
stem cells residing at the bronchioalveolar-duct junction. Nat. Genet. 51(4), 728–738 (2019).
https://doi.org/10.1038/s41588-019-0346-6
29. Liu, J., Cao, R., Xu, M., Wang, X., Zhang, H., Hu, H., et al.: Hydroxychloroquine, a less
toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell
Discov. 6, 16 (2020). https://doi.org/10.1038/s41421-020-0156-0
30. Corey, D.M., Rinkevich, Y., Weissman, I.L.: Dynamic patterns of clonal evolution in tumor
vasculature underlie alterations in lymphocyte-endothelial recognition to Foster tumor
immune escape. Cancer Res. 76(6), 1348–1353 (2016). https://doi.org/10.1158/0008-5472.
CAN-15-1150
Lineage Tracing Models to Study Cardiomyocyte Generation During Cardiac…
43. 32
HSC Hematopoietic stem cell
Mlc2v Myosin light chain 2v
Myf5 Myogenic factor 5
MyHC Myosin heavy chain
Myod Myoblast determination protein 1
Nkx2-5 Homeobox Protein Nkx2–5
OE Overexpression
Pdx1 Pancreatic and duodenal homeobox 1
qPCR Quantitative polymerase chain reaction
scRNA-seq Single cell RNA sequencing
Tie2 Endothelial cell specific receptor tyrosine kinase 2
Ve-Cad Vascular endothelial cadherin
1
Cardiovascular Diseases Are Common and Have
Considerable Morbidity and Mortality
Peripheral artery disease affects more than 10 M Americans resulting in more than
150,000 limb amputations each year in the U.S. In addition, more than 300,000
patients have coronary artery bypass grafting (surgical revascularization) [1]. These
diseases collectively are amplified by the rising incidence of diabetes, obesity and
cardiovascular disease. These complications result in considerable morbidity and
mortality [1, 2]. Current medical therapies for vascular diseases include limb ampu-
tation and vascular bypass grafting. However, these therapeutic interventions have
significant limitations. These diseases are chronic, debilitating, lethal and they war-
rant new and novel therapies. The definition of the molecular mechanisms that gov-
ern the endothelial lineage and vascular development will provide a platform to
modulate these pathways and promote vasculogenesis as a therapeutic initiative.
The overall goal for this chapter is to highlight the key regulators that govern endo-
thelial and vascular development.
2
Master Regulators Govern Fate Decisions
and Lineage Development
Loss of function and gain of function genetic studies have defined essential factors
that govern cell fate and lineage development (Fig. 1a). These factors also known as
master regulators occupy or sit at the top of a regulatory hierarchy [3, 4]. Perhaps a
prototypic example are members of the MYOD family of transcription factors. The
MYOD family consists of bHLH transcription factors that have distinct and over-
lapping functional roles for the regulation of the myogenic lineage [5]. These mas-
ter regulators also have the capacity to convert another differentiated cell type
(usually a fibroblast) to a specific lineage (i.e. skeletal muscle) using a
D. J. Garry and J. Sierra-Pagan
44. 33
promoter-
reporter construct that demonstrates expression with lineage specific dif-
ferentiation (Fig. 1) [6–9]. These assays were initially referred to as conversion
assays. Using conversion assays in combination with gene disruption and transgenic
technologies, hundreds of master regulators have been described [3]. In addition to
the MYOD family, PDX1 (pancreas), OCT4/SOX2/NANOG (pluripotency), SCL/
TAL1 (blood), HIF1 (hypoxia), and others have been identified (Fig. 1b) [6–8, 10–
12]. ETV2 is a recently identified master regulator that has been shown to be essen-
tial for the specification and development of the endothelial lineage (Fig. 1b)
[13, 14].
Fig. 1 Master regulators specify lineage-specific development. (a) Adaptation of Waddington’s
landscape that outlines the role of master regulators to govern fate decisions during embryogene-
sis. (b) Schematic outlining examples of master regulators for specific lineages. PDX1 is a master
regulator for pancreatogenesis and ETV2 is master regulator for hematoendothelial lineages
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
45. 34
3
Developmental Milestones for Endothelial Development
The initial developmental stage for vasculogenesis occurs as specified progeni-
tors (i.e. angioblasts) that migrate from the primitive streak in the developing mouse
embryo to the yolk sac [15]. Later, these angioblasts migrate from the extraembry-
onic yolk sac to the embryo proper to form cord like structures, lumens (a process
known as tubulogenesis) and ultimately form a vascular plexus (Fig. 2) [15]. This
process continues and is associated with the onset of cardiac contractility in the
E8.25 heart tube, the appearance of primitive blood cells associated with the primi-
tive circulation (E8.5) and the establishment of a complete circulation with the
propagation of blood throughout the E10-E10.25 mouse embryo [16]. This circula-
tion is impacted by the growth of the embryo and the transition from diffusion to the
circulation of blood in response to hypoxic signals (HIF1 and HIF2) and signaling
pathways (VEGF1-FLK1/FLT1/FLT2, SHH-GLI1/2/3, SRC-CDH5, ANG1/2-
TIE2, etc.) [16, 17]. This latter process is termed angiogenesis, which is character-
ized by the formation of new vessels that originate or sprout from pre-established or
Fig. 2 Overview of hematoendothelial development. (a) Schematic highlighting the role of angio-
blasts being recruited to form a vascular plexus, tubulogenesis and sprouts to generate vascular
networks and remodeling. (b) Schematic highlighting the role of hemogenic endothelium during
primitive (yolk sac) and definitive (AGM) hematopoiesis
D. J. Garry and J. Sierra-Pagan
46. 35
pre-existing vessels [17]. The vasculature continues to architecturally evolve and
mature with the determination of venous and arterial endothelial fates. This matura-
tion phase is marked by the expression of integrins, the balance of apoptosis and cell
proliferation and the impact of the extracellular matrix [15]. Overall, the endothelial
and vascular lineages are coordinated and responsive to microenvironmental cues
and signals.
The endothelial-endocardial relationship is established early during embryogen-
esis. The endothelial lineage is characterized by a single cell thickness of develop-
ing vascular networks that form a tight syncytium separating the luminal space with
the underlying vascular wall. This endothelial lining is contiguous with the endocar-
dium, which lines the four-chambered heart. The ontogeny of the endocardium is
distinct from the endothelial lineage and is reflected in the expression of a lineage
specific molecular program [18].
4
The Common Origin of Endothelial
and Hematopoietic Lineages
The endothelial and hematopoietic lineages are both derivatives of the mesodermal
germ layer [19]. As both lineages develop in close proximity (i.e., blood islands of
the yolk sac and the blood containing vessels) and express overlapping molecular
programs, studies support a common origin for the lineages [19]. The extraembry-
onic yolk sac is the source for primitive hematopoiesis (Fig. 2b) [20]. Indeed, stud-
ies have demonstrated that multilineage hematopoietic stem cells are derivatives of
hemogenic endothelium. Hemogenic endothelium, despite its endothelial gene
expression profile, loses its endothelial potential early in development and only
gives rise to blood [21]. Hemogenic endothelium are flat shaped cells that undergo
endothelial-to-hematopoietic transition (EHT and marked by CD44 expression) and
acquire a spherical shape characteristic of blood cells [20, 22]. These hemogenic
endothelial cells are found within the allantois, the yolk sac, the endocardium and
the aorta-gonad-mesonephros (AGM) of the developing embryo [20, 23]. The AGM
is closely associated with the ventral wall of the dorsal aorta (~E9.5-E11.5) and has
been shown to produce hematopoietic stem cells that are capable of engrafting and
reconstituting the irradiated bone marrow. Therefore, the AGM is recognized for its
critical role in definitive hematopoiesis (Fig. 2b) [23]. Single cell RNA-seq of the
AGM in the developing mouse have identified multiple cell types and defined the
expression of GATA2, RUNX1, LYL1, ERG, FLI1, LMO2 and TAL1 transcription
factors, which result in the loss of endothelial gene expression and acquisition of
hematopoietic gene expression [22].
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
47. 36
5
ETV2 Is Necessary and Sufficient for Endothelial
Lineage Development
ETV2 (Ets Variant Transcription Factor 2) was initially sequenced from the testis by
Steve McKnight’s laboratory [24]. Later, ETV2 was co-discovered as an essential
factor for hematoendothelial (HE) lineage development by the Choi laboratory and
the Garry laboratory [25, 26]. These independent efforts resulted in studies by the
Choi laboratory, which defined a BMP/NOTCH/WNT-ETV2 axis during mouse
embryogenesis and demonstrated that Etv2 null embryos were lethal and lacked
hematopoietic and vascular lineages [25]. Similarly, the Garry laboratory used a
Nkx2-5-reporter transgenic strategy and identified Etv2 as a putative downstream
target. Further, they demonstrated that Etv2 null embryos were nonviable at E9-E9.5
and had an absence of blood and endothelial lineages and defined that ETV2 was a
direct upstream regulator of the Tie2 gene [26]. Studies further demonstrated the
essential role for ETV2 in zebrafish, xenopus, pig and human [13, 14, 27–35]. These
latter results support the evolutionary conserved role for ETV2 as an essential factor
for hematoendothelial development. Furthermore, forced overexpression (OE) of
ETV2 using mouse embryonic stem cells/embryoid bodies (ES/EB) differentiation
assays demonstrated that it was sufficient for HE development (Fig. 3) [25, 35–37].
Collectively, these studies provided a foundation for ETV2 as a master regulator for
the HE lineages.
6
ETV2 Expression during Mouse Embryogenesis
and the Postnatal Period
Using in situ hybridization, PCR, immunohistochemistry and the 3.9Kb Etv2-EYFP
reporter transgenic expression, ETV2 expression was restricted to the angioblasts at
E6.5, hematoendothelial cells and endocardium until ~E10-E10.5 after which it was
rapidly extinguished with the exception of a small subpopulation of cells associated
with the dorsal aorta (Fig. 4) [25, 26, 36–41]. Similarly, using the ES/EB differen-
tiation assay, ETV2 was robustly expressed on Days 3–4 following differentiation
after which it was extinguished (Fig. 3) [25, 42]. These expression studies support
the notion that ETV2 functions as a “rheostat” to tightly regulate HE lineage devel-
opment similar to other master regulators for the specification of other lineages.
While the mechanisms that regulate extinguished expression of ETV2 are incom-
pletely defined, the feedback mechanisms involving FLT1 contribute, in part, to the
negative regulation of Etv2 gene expression [42]. Future studies will need to focus
on the definition of these mechanisms to enhance our understanding of ETV2
expression.
ETV2 is expressed postnatally in the testis and in the HSC population (Lin-
Sca1+
cKit+
cells) in adult mouse bone marrow [44, 45]. ETV2 is induced and upreg-
ulated following tissue and vascular injury without sustained long-term or persistent
D. J. Garry and J. Sierra-Pagan
48. 37
expression [46]. Furthermore, recent studies support the notion that ETV2 is
expressed in tumorigenic tissues and may be associated with the angiogenic
response observed with various solid tumors (as outlined below) [47–49]. These
expression patterns and its role as a master regulator suggest that ETV2 may be an
important target to promote or repress angiogenesis depending on the physiological
context.
7
ETV2 Is Dynamically and Transcriptionally Regulated by
Upstream Factors
Previous studies by our laboratory demonstrated that the 3.9 kb upstream fragment
of the Etv2 gene harbored all the modules and motifs necessary for the spatial and
temporal expression pattern of endogenous ETV2 (Fig. 5) [26, 31, 36, 41, 42, 50,
51]. These studies established that the EYFP reporter expression pattern recapitu-
lated endogenous ETV2 activity demonstrating the onset of expression and
Fig. 3 Embryoid body (EBs) differentiation assays recapitulate developmental mechanisms. (a)
Schematic highlighting ES cells differentiating to EBs and forming mesodermal derivatives
including: hematoendothelial, cardiac and skeletal muscle lineages. (b) FACS profile of dissoci-
ated EBs stained for FLK1 and PDGFR-a demonstrate the HE (FLK1+
/ PDGFR-a−
), cardiac
(FLK1+
/PDGFR-a+
) and skeletal muscle (FLK1−
/PDGFR-a+
) lineages. (c) Schematic of the
expression profile of Etv2 during mesodermal EB differentiation showing its transient pattern of
expression
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
49. 38
extinguished activity (Fig. 4) [41]. Bioinformatics analysis revealed evolutionary
conservation of two modules (CRI and CRII) within the 3.9 kb fragment and the
deletion of this fragment phenocopied the global deletion of Etv2 (Fig. 5b) [42].
Furthermore, the characterization of the Etv2 cis-regulatory modules in vitro
revealed the importance of the CRI and CRII modules using luciferase assays [26,
42, 52]. Collectively, these studies confirmed that the upstream regulation of this
gene utilized the binding motifs contained within the 3.9 kb fragment. Using gene
disruption models, transcriptional assays, EMSAs and mutagenesis, transcriptional
regulators of Etv2 gene expression included: ETV2, GATA2, VEFGF/FLK1-
Calcineurin-
NFAT, CREB1, MESP1, NKX2-5 and other signaling pathways (BMP,
Notch and Wnt signaling pathways) (Fig. 5c) [25, 26, 39, 42, 52, 53]. While all
Fig. 4 ETV2 fate mapping identifies ETV2 expressing lineages and its descendants in the heart.
Using the 3.9 kb Etv2-Cre transgenic mouse model, the ETV2 contribution to embryogenesis was
mapped during cardiogenesis in the developing mouse. E12.5 heart co-stained with NKX2–5 (red),
DAPI (blue), and ETV2 cells/descendants (green) [43] demonstrate that every endothelial and
endocardial cell is labeled with GFP including the developing valves (arrowhead) and aorta (open
arrowhead) (a, atria and v, ventricle)
D. J. Garry and J. Sierra-Pagan
50. 39
Fig. 5 The regulatory mechanisms that govern the Etv2 gene. (a) Schematic highlighting the ATG
start site and seven exons associated with the Etv2 gene. (b) Schematic demonstrating the CRI and
CRII modules that regulate Etv2 gene expression. Deletion of the CRI and CRII modules pheno-
copies the Etv2 global gene KO with embryonic lethality and absence of blood and vasculature. (c)
Upstream cis transcriptional regulators of the Etv2 gene and their binding motifs are outlined. (d)
Schematic highlighting the ETV2 protein with its transcriptional activation domain (TAD) and
DNA binding or Ets domain
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
51. 40
these factors were important regulators, it appeared that MESP1-CREB1 may initi-
ate Etv2 gene expression in the mesodermal lineage. Gene activation may also be
context dependent, as in zebrafish, the overexpression of Nkx2-5 functions to
repress Etv2 gene activity whereas in the mouse it appears to function as a direct
upstream regulator of Etv2 in the endocardium [54].
8
Definition of Transcriptional Targets for ETV2 Mediate
Distinct Developmental Events
Master regulators have a number of functions that are mediated by their respective
downstream targets. ETV2 binds a canonical GGAA/T Ets motif and transactivates
the hematoendothelial molecular program including: Scl/Tal1, Lmo2, Tie2/Tek,
VE-Cadherin/Cdh5, Pecam1/CD31, Gata1, Gata2, Flk1, Elk3, Fli1, Sox7, Cepd,
and others (Fig. 6) [14, 26, 34, 37, 42, 43, 53, 55–61]. These targets were identified
and confirmed using CHiP-seq, transcriptional assays, EMSA, Standard ChIP and
other molecular techniques [14, 55, 62]. More recent studies have demonstrated that
ETV2 promotes cell proliferation by regulating Yes1 gene expression, which inter-
acts with the Hippo signaling pathway (Fig. 6) [63]. ETV2 also transcriptionally
regulates Rhoj gene expression to modulate endothelial progenitor cell migration
during embryogenesis (Fig. 6) [64]. Finally, ETV2 regulates microRNAs (i.e.
miR130a), which govern fate determination by promoting endothelial differentia-
tion but not hematopoietic differentiation (Fig. 6) [59, 60]. Context specific gene
regulation is observed in the testis where ETV2 is a direct upstream regulator of
Sox9 gene expression. In a positive feed-back loop, SOX9 binds to the Etv2 pro-
moter and serves to transactivate its regulator thereby maintaining the sertoli cell
phenotype [65]. ETV2-Chip-seq datasets are publicly available and are continuing
to be mined to further define and explore additional targets and functional roles for
this master regulator.
9
Protein-Protein Interacting Factors for ETV2 Are
Important Coregulators
The Etv2 gene harbors seven exons and encodes a protein that has carboxy terminal
domain, a DNA binding domain (amino acids 316–336 which overlaps with the
ETS domain that spans from 231 to 315 aa) and an amino terminal domain (amino
acids 1–157) (Fig. 5d) [44]. Using an array of biochemical and molecular tech-
niques (i.e. mass spectrometry, yeast two hybrid screening assay, FRET, etc.) inter-
acting factors have been identified for ETV2. Early studies identified FOXC2 as an
interacting factor for ETV2 and suggested that adjacent Ets-Fox binding motifs
(Ets-Fox enhancer motif) in more than 20% of endothelial specific genes were
D. J. Garry and J. Sierra-Pagan
52. 41
potent coactivators of gene expression [66]. Furthermore, GATA2 has been shown
using multiple assays to interact with ETV2 and coactivate hematoendothelial gene
expression [53]. Other factors (OVol11311) have also been shown to cooperate with
ETV2 [67]. For example, forced overexpression of ETV2 and GATA2 in hiPSCs
promoted a hematopoietic fate [68]. These interacting factors are important context
dependent cofactors that function to amplify and modify the functional role
of ETV2.
10
ETV2 Is a Master Regulator for Hematoendothelial
Lineages Using Conversion Assays
Previous studies have demonstrated that forced overexpression of ETV2 promotes a
hematoendothelial cell fate (Fig. 7) [9, 36, 37]. Forced overexpression of ETV2
alone or in combination with other factors (i.e. GATA2, SCL, etc.) during murine
EB differentiation promotes a hematoendothelial fate (FLK1+
cells) (Fig. 7) [14, 36,
37, 55, 68]. Furthermore, the delivery of ETV2 converted cell populations into
Fig. 6 ETV2 functions as an upstream regulator of gene expression. Schematic which demon-
strates direct downstream targets of ETV2 and their functional role during vasculogenesis and
angiogenesis
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
53. 42
ischemic hindlimbs demonstrated that these converted cells were endothelial cells
as they participated in the repair of ischemic hindlimb mouse models [9].
Additionally, the delivery of lentivirus overexpressing ETV2 promoted repair in
response to ischemic injury and reduced the fibroproliferative response in mouse
hearts [69]. Collectively, these preliminary studies provide an important platform
for therapeutic initiatives focused on the overexpression of ETV2.
11
ETV2 Overexpression in Tumor Angiogenesis
Previous studies have demonstrated that master regulators not only promote fate
determination during embryogenesis but also are overexpressed in the context of
cancer [70]. Solid tumors require an enhanced vascular supply for growth, cell pro-
liferation and metastasis. Therefore, if ETV2 is at the top of the transcriptional
hierarchy for vasculogenesis/angiogenesis, then ETV2 should be expressed at dis-
tinct time-periods during tumorigenesis. Expression analysis demonstrated coex-
pression of ETV2 and the histone dymethylase, Junonji domain containing 2A in
Fig. 7 The Etv2 network regulates hematoendothelial lineage development. Upstream and down-
stream regulators and ETV2 effectors specify HE lineage development. ETV2 also represses non-
hematoendothelial lineages
D. J. Garry and J. Sierra-Pagan
54. 43
neuroendocrine prostate tumors [49]. In a separate study, the knockdown of Etv2
using siRNA nanoparticles resulted in the inhibition of tumor angiogenesis and the
inhibition of tumor growth [48]. Furthermore, using a zebrafish xenotransplantation
model, ETV2 and FLI1b were shown to have redundant roles in promoting tumor
angiogenesis [71–73]. Collectively, these initial studies support the conclusion that
strategies focused on the inhibition of ETV2 may be effective therapies that target
tumor growth and angiogenesis.
12
ETV2 Functions to Repress Nonhematoendothelial
Fate Decisions
Master regulators function to reprogram the fate of differentiated cells [3, 4]. They
also have the capacity to repress other lineages and direct progenitor cell popula-
tions down a specified pathway as outlined in the Waddington’s landscape (Fig. 1)
[4, 74]. In the Etv2 global knockout mouse model, mesodermal progenitors that
typically are destined for the hematoendothelial lineage are redirected to the cardio-
myocyte lineage using genetic fate mapping techniques (Fig. 7) [26, 41, 75].
Similarly, the Etv2 null zebrafish endothelial progenitors are redirected to the skel-
etal muscle lineage (Fig. 7) [29, 30]. These studies using a gene disruption strategy
emphasize that ETV2 represses nonhematopoietic lineage formation during early
stages of embryogenesis.
13 Summary
The endothelial and vascular lineages require a complex network of gene expres-
sion that governs fate determination. ETV2 is an important master regulator for the
endothelial and vascular lineages. Definition of the ETV2 mediated molecular net-
works provide a platform for future therapeutic interventions.
Acknowledgements The authors acknowledge Cynthia Faraday for her assistance with the prep-
aration of the figures.
References
1. Roger, V.L., Go, A.S., Lloyd-Jones, D.M., Benjamin, E.J., Berry, J.D., Borden, W.B., et al.:
Executive summary: heart disease and stroke statistics–2012 update: a report from the
American Heart Association. Circulation. 125(1), 188–197 (2012). https://doi.org/10.1161/
CIR.0b013e3182456d46
Mechanisms that Govern Endothelial Lineage Development and Vasculogenesis
60. This ebook is for the use of anyone anywhere in the United States
and most other parts of the world at no cost and with almost no
restrictions whatsoever. You may copy it, give it away or re-use it
under the terms of the Project Gutenberg License included with this
ebook or online at www.gutenberg.org. If you are not located in the
United States, you will have to check the laws of the country where
you are located before using this eBook.
Title: An Unsentimental Journey through Cornwall
Author: Dinah Maria Mulock Craik
Illustrator: Charles Napier Hemy
Release date: January 1, 2014 [eBook #44557]
Most recently updated: October 23, 2024
Language: English
Credits: Produced by Chris Curnow and the Online Distributed
Proofreading Team at http://www.pgdp.net (This file was
produced from images generously made available by The
Internet Archive)
*** START OF THE PROJECT GUTENBERG EBOOK AN
UNSENTIMENTAL JOURNEY THROUGH CORNWALL ***
63. WITH ILLUSTRATIONS
BY
C. NAPIER HEMY
London
MACMILLAN AND CO.
1884
The Right of Translation and Reproduction is Reserved
LONDON:
R. Clay, Sons, and Taylor,
BREAD STREET HILL, E.C.
64. CONTENTS
PAGE
Day the First 1
Day the Second 9
Day the Third 25
Day the Fourth 45
Day the Fifth 53
Day the Sixth 59
Day the Seventh 67
Day the Eighth 75
Day the Ninth 86
Day the Tenth 101
Day the Eleventh 110
Day the Twelfth 118
Day the Thirteenth 127
Days Fourteenth, Fifteenth, and Sixteenth 133
65. LIST OF ILLUSTRATIONS
PAGE
ST. MICHAEL'S MOUNT Frontispiece
FALMOUTH, FROM FLUSHING 1
ST. MAWE'S CASTLE, FALMOUTH BAY 5
VIEW OF FLUSHING FROM THE GREEN BANK HOTEL,
FALMOUTH
7
A FISHERMAN'S CELLAR NEAR THE LIZARD 11
THE CORNISH COAST: FROM YNYS HEAD TO BEAST
POINT
15
THE LIZARD LIGHTS BY NIGHT 23
CORNISH FISH 24
POLTESCO 29
CADGWITH COVE 32
THE DEVIL'S FRYING PAN, NEAR CADGWITH 34
MULLION COVE, CORNWALL 38
A CRABBER'S HOLE, GERRAN'S BAY 41
STEAM SEINE BOATS GOING OUT 46
HAULING IN THE BOATS—EVENING 50
HAULING IN THE LINES 55
THE LIZARD LIGHTS BY DAY 60
THE FISHERMAN'S DAUGHTER—A CORNISH STUDY 63
KYNANCE COVE, CORNWALL 68
THE STEEPLE ROCK, KYNANCE COVE 71
THE LION ROCKS—A SEA IN WHICH NOTHING CAN
LIVE
76
HAULING IN THE BOATS 79
ENYS DODNAN AND PARDENICK POINTS 83
JOHN CURGENVEN FISHING 87
66. THE ARMED KNIGHT AND THE LONG SHIP'S
LIGHTHOUSE
94
CORNISH FISHERMAN 100
THE SEINE BOAT—A PERILOUS MOMENT 103
ST. IVES 108
THE LAND'S END AND THE LOGAN ROCK 114
SENNEN COVE, WAITING FOR THE BOATS 119
ON THE ROAD TO ST. NIGHTON'S KEEVE 124
TINTAGEL 128
CRESWICK'S MILL IN THE ROCKY VALLEY 135
BOSCASTLE 139
THE OLD POST-OFFICE, TREVENA 145
AN UNSENTIMENTAL JOURNEY
THROUGH CORNWALL
68. DAY THE FIRST
I believe in holidays. Not in a frantic rushing about from place to
place, glancing at everything and observing nothing; flying from
town to town, from hotel to hotel, eager to do and to see a
country, in order that when they get home they may say they have
done it, and seen it. Only to say;—as for any real vision of eye,
heart, and brain, they might as well go through the world blindfold.
It is not the things we see, but the mind we see them with, which
makes the real interest of travelling. Eyes and No Eyes,—an old-
fashioned story about two little children taking a walk; one seeing
everything, and enjoying everything, and the other seeing nothing,
and thinking the expedition the dullest imaginable. This simple tale,
which the present generation has probably never read, contains the
essence of all rational travelling.
So when, as the old hen, (which I am sometimes called, from my
habit of going about with a brood of chickens, my own or other
people's) I planned a brief tour with two of them, one just entered
upon her teens, the other in her twenties, I premised that it must be
a tour after my own heart.
In the first place, my children, you must obey orders implicitly. I
shall collect opinions, and do my best to please everybody; but in
travelling one only must decide, the others coincide. It will save
them a world of trouble, and their 'conductor' also; who, if
competent to be trusted at all, should be trusted absolutely.
Secondly, take as little luggage as possible. No sensible people travel
with their point-lace and diamonds. Two 'changes of raiment,' good,
useful dresses, prudent boots, shawls, and waterproofs—these I
shall insist upon, and nothing more. Nothing for show, as I shall take
you to no place where you can show off. We will avoid all huge
hotels, all fashionable towns; we will study life in its simplicity, and
69. make ourselves happy in our own humble, feminine way. Not
'roughing it' in any needless or reckless fashion—the 'old hen' is too
old for that; yet doing everything with reasonable economy. Above
all, rushing into no foolhardy exploits, and taking every precaution to
keep well and strong, so as to enjoy the journey from beginning to
end, and hinder no one else from enjoying it. There are four things
which travellers ought never to lose: their luggage, their temper,
their health, and their spirits. I will make you as happy as I possibly
can, but you must also make me happy by following my rules:
especially the one golden rule, Obey orders.
So preached the old hen, with a vague fear that her chickens
might turn out to be ducklings, which would be a little awkward in
the region whither she proposed to take them. For if there is one
place more risky than another for adventurous young people with a
talent for perpetuating themselves down prejudices, as Mrs.
Malaprop would say, it is that grandest, wildest, most dangerous
coast, the coast of Cornwall.
I had always wished to investigate Cornwall. This desire had existed
ever since, at five years old, I made acquaintance with Jack the
Giantkiller, and afterwards, at fifteen or so, fell in love with my life's
one hero, King Arthur.
Between these two illustrious Cornishmen,—equally mythical,
practical folk would say—there exists more similarity than at first
appears. The aim of both was to uphold right and to redress wrong.
Patience, self-denial; tenderness to the weak and helpless, dauntless
courage against the wicked and the strong: these, the essential
elements of true manliness, characterise both the humble Jack and
the kingly Arthur. And the qualities seem to have descended to more
modern times. The well-known ballad:—
And shall they scorn Tre, Pol, and Pen?
And shall Trelawny die?
There's twenty thousand Cornishmen
Will know the reason why,
70. has a ring of the same tone, indicating the love of justice, the spirit
of fidelity and bravery, as well as of that common sense which is at
the root of all useful valour.
I wanted to see if the same spirit lingered yet, as I had heard it did
among Cornish folk, which, it was said, were a race by themselves,
honest, simple, shrewd, and kind. Also, I wished to see the Cornish
land, and especially the Land's End, which I had many a time beheld
in fancy, for it was a favourite landscape-dream of my rather
imaginative childhood, recurring again and again, till I could almost
have painted it from memory. And as year after year every chance of
seeing it in its reality seemed to melt away, the desire grew into an
actual craving.
After waiting patiently for nearly half a century, I said to myself, I
will conquer Fate; I will go and see the Land's End.
And it was there that, after making a circuit round the coast, I
proposed finally to take my chickens.
We concocted a plan, definite yet movable, as all travelling plans
should be, clear in its dates, its outline, and intentions, but subject
to modifications, according to the exigency of the times and
circumstances. And with that prudent persistency, without which all
travelling is a mere muddle, all discomfort, disappointment, and
distaste—for on whatever terms you may be with your travelling
companions when you start, you are quite sure either to love them
or hate them when you get home—we succeeded in carrying it out.
The 1st of September, 1881, and one of the loveliest of September
days, was the day we started from Exeter, where we had agreed to
meet and stay the night. There, the previous afternoon, we had
whiled away an hour in the dim cathedral, and watched, not without
anxiety, the flood of evening sunshine which poured through the
great west window, lighting the tombs, old and new, from the
Crusader, cross-legged and broken-nosed, to the white marble bas-
relief which tells the story of a not less noble Knight of the Cross,
Bishop Patteson. Then we wandered round the quaint old town, in
71. such a lovely twilight, such a starry night! But—will it be a fine day
to-morrow? We could but live in hope: and hope did not deceive us.
To start on a journey in sunshine feels like beginning life well. Clouds
may come—are sure to come: I think no one past earliest youth
goes forth into a strange region without a feeling akin to Saint Paul's
not knowing what things may befall me there. But it is always best
for each to keep to himself all the shadows, and give his companions
the brightness, especially if they be young companions.
And very bright were the eyes that watched the swift-moving
landscape on either side of the railway: the estuary of Exe; Dawlish,
with its various colouring of rock and cliff, and its pretty little sea-
side houses, where family groups stood photographing themselves
on our vision, as the train rushed unceremoniously between the
beach and their parlour windows; then Plymouth and Saltash, where
the magnificent bridge reminded us of the one over the Tay, which
we had once crossed, not long before that Sunday night when,
sitting in a quiet sick-room in Edinburgh, we heard the howl outside
of the fearful blast which destroyed such a wonderful work of
engineering art, and whirled so many human beings into eternity.
But this Saltash bridge, spanning placidly a smiling country, how
pretty and safe it looked! There was a general turning to carriage-
windows, and then a courteous drawing back, that we, the
strangers, should see it, which broke the ice with our fellow-
travellers. To whom we soon began to talk, as is our conscientious
custom when we see no tangible objection thereto, and gained,
now, as many a time before, much pleasant as well as useful
information. Every one evinced an eager politeness to show us the
country, and an innocent anxiety that we should admire it; which we
could honestly do.
I shall long remember, as a dream of sunshiny beauty and peace,
this journey between Plymouth and Falmouth, passing Liskeard,
Lostwithiel, St. Austell, c. The green-wooded valleys, the rounded
hills, on one of which we were shown the remains of the old castle
of Ristormel, noted among the three castles of Cornwall; all this,
72. familiar to so many, was to us absolutely new, and we enjoyed it and
the kindly interest that was taken in pointing it out to us, as happy-
minded simple folk do always enjoy the sight of a new country.
ST. MAWE'S CASTLE, FALMOUTH BAY.
Our pleasure seemed to amuse an old gentleman who sat in the
corner. He at last addressed us, with an unctuous west-country
accent which suited well his comfortable stoutness. He might have
fed all his life upon Dorset butter and Devonshire cream, to one of
73. which counties he certainly belonged. Not, I think, to the one we
were now passing through, and admiring so heartily.
So you're going to travel in Cornwall. Well, take care, they're sharp
folk, the Cornish folk. They'll take you in if they can. (Then, he must
be a Devon man. It is so easy to sit in judgment upon next-door
neighbours.) I don't mean to say they'll actually cheat you, but
they'll take you in, and they'll be careful that you don't take them in
—no, not to the extent of a brass farthing.
We explained, smiling, that we had not the slightest intention of
taking anybody in, that we liked justice, and blamed no man,
Cornishman or otherwise, for trying to do the best he could for
himself, so that it was not to the injury of other people.
Well, well, perhaps you're right. But they are sharp, for all that,
especially in the towns.
We replied that we meant to escape towns, whenever possible, and
encamp in some quiet places, quite out of the world.
Our friend opened his eyes, evidently thinking this a most singular
taste.
Well, if you really want a quiet place, I can tell you of one, almost
as quiet as your grave. I ought to know, for I lived there sixteen
years. (At any rate, it seemed to have agreed with him.) Gerrans is
its name—a fishing village. You get there from Falmouth by boat.
The fare is —(I regret to say my memory is not so accurate as his
in the matter of pennies), and mind you don't pay one farthing
more. Then you have to drive across country; the distance is—and
the fare per mile— (Alas! again I have totally forgotten.) They'll be
sure to ask you double the money, but never you mind! refuse to
pay it, and they'll give in. You must always hold your own against
extortion in Cornwall.
I thanked him, with a slightly troubled mind. But I have always
noticed that in travelling with such measure as ye mete it shall be
meted to you again, and that those who come to a country
expecting to be cheated generally are cheated. Having still a
74. lingering belief in human nature, and especially in Cornish nature, I
determined to set down the old gentleman's well-meant advice for
what it was worth, no more, and cease to perplex myself about it.
For which resolve I have since been exceedingly thankful.
He gave us, however, much supplementary advice which was rather
useful, and parted from us in the friendliest fashion, with that air of
bland complaisance natural to those who assume the character of
adviser in general.
Mind you go to Gerrans. They'll not take you in more than they do
everywhere else, and you'll find it a healthy place, and a quiet place
—as quiet, I say, as your grave. It will make you feel exactly as if
you were dead and buried.
That not being the prominent object of our tour in Cornwall, we
thanked him again, but as soon as he left the carriage determined
among ourselves to take no further steps about visiting Gerrans.
VIEW OF FLUSHING FROM THE GREEN BANK HOTEL,
FALMOUTH.
75. However, in spite of the urgency of another fellow-traveller—it is
always good to hear everybody's advice, and follow your own—we
carried our love of quietness so far that we eschewed the
magnificent new Falmouth Hotel, with its table d'hôte, lawn tennis
ground, sea baths and promenade, for the old-fashioned Green
Bank, which though it had no green banks, boasted, we had been
told, a pleasant little sea view and bay view, and was a resting-place
full of comfort and homely peace.
Which we found true, and would have liked to stay longer in its
pleasant shelter, which almost conquered our horror of hotels; but
we had now fairly weighed anchor and must sail on.
You ought to go at once to the Lizard, said the friend who met us,
and did everything for us at Falmouth—and the remembrance of
whom, and of all that happened in our brief stay, will make the very
name of the place sound sweet in our ears for ever. The Lizard is
the real point for sightseers, almost better than the Land's End. Let
us see if we can hear of lodgings.
She made inquiries, and within half an hour we did hear of some
most satisfactory ones. The very thing! We will telegraph at once—
answer paid, said this good genius of practicality, as sitting in her
carriage she herself wrote the telegram and despatched it.
Telegrams to the Lizard! We were not then at the Ultima Thule of
civilisation.
Still, she said, you had better provide yourself with some food,
such as groceries and hams. You can't always get what you want at
the Lizard.
So, having the very dimmest idea what the Lizard was—whether a
town, a village, or a bare rock—when we had secured the desired
lodgings (quite ideal lodgings, remarked our guardian angel), I
proceeded to lay in a store of provisions, doing it as carefully as if
fitting out a ship for the North Pole—and afterwards found out it was
a work of supererogation entirely.
76. The next thing to secure was an ideal carriage, horse, and man,
which our good genius also succeeded in providing. And now, our
minds being at rest, we were able to write home a fixed address for
a week, and assure our expectant and anxious friends that all was
going well with us.
Then, after a twilight wander round the quaint old town—so like a
foreign town—and other keen enjoyments, which, as belonging to
the sanctity of private life I here perforce omit, we laid us down to
sleep, and slept in peace, having really achieved much; considering
it was only the first day of our journey.
77. DAY THE SECOND
Is there anything more delightful than to start on a smiling morning
in a comfortable carriage, with all one's impedimenta (happily not
much!) safely stowed away under one's eyes, with a good horse,
over which one's feelings of humanity need not be always agonising,
and a man to drive, whom one can trust to have as much sense as
the brute, especially in the matter of refreshment. Our letters that
morning had brought us a comico-tragic story of a family we knew,
who, migrating with a lot of children and luggage, and requiring to
catch a train thirteen miles off, had engaged a driver who refreshed
himself so successfully at every public-house on the way, that he
took five hours to accomplish the journey, and finally had to be left
at the road-side, and the luggage transferred to another vehicle,
which of course lost the train. We congratulated ourselves that no
such disaster was likely to happen to us.
Yes; I've been a teetotaller all my life, said our driver, a bright-
looking, intelligent young fellow, whom, as he became rather a
prominent adjunct to our life and decidedly to our comfort, I shall
individualise by calling him Charles. I had good need to avoid
drinking. My father drank through a small property. No fear of me,
ma'am.
So at once between him and us, or him and we, according to the
Cornish habit of transposing pronouns, was established a feeling of
fraternity, which, during the six days that we had to do with him,
deepened into real regard. Never failing when wanted, never
presuming when not wanted, straightforward, independent, yet full
of that respectful kindliness which servants can always show and
masters should always appreciate, giving us a chivalrous care,
which, being unprotected females, was to us extremely valuable, I
here record that much of the pleasure of our tour was owing to this
78. honest Cornishman, who served us, his horse, and his master—he
was one of the employés of a livery-stable keeper—with equal
fidelity.
Certainly, numerous as were the parties he had driven—(I go to the
Lizard about three times a week, he said)—Charles could seldom
have driven a merrier trio than that which leisurely mounted the
upland road from Falmouth, leading to the village of Constantine.
Just turn and look behind you, ladies (we had begged to be shown
everything and told everything); isn't that a pretty view?
It certainly was. From the high ground we could see Falmouth with
its sheltered bay and glittering sea beyond. Landward were the
villages of Mabe and Constantine, with their great quarries of
granite, and in the distance lay wide sweeps of undulating land,
barren and treeless, but still beautiful—not with the rich pastoral
beauty of our own Kent, yet having a charm of its own. And the air,
so fresh and pure, yet soft and balmy, it felt to tender lungs like the
difference between milk and cream. To breathe became a pleasure
instead of a pain. I could quite understand how the semi-tropical
plants that we had seen in a lovely garden below, grew and
flourished, how the hydrangeas became huge bushes, and the
eucalyptus an actual forest tree.
But this was in the sheltered valley, and we had gained the hill-top,
emerging out of one of those deep-cut lanes peculiar to Devon and
Cornwall, and so pretty in themselves, a perfect garden of wild
flowers and ferns, except that they completely shut out the view.
This did not much afflict the practical minds of my two juniors. Half
an hour before they had set up a shout—
Stop the carriage! Do stop the carriage! Just look there! Did you
ever see such big blackberries? and what a quantity! Let us get out;
we'll gather them for to-morrow's pudding.
Undoubtedly a dinner earned is the sweetest of all dinners. I
remember once thinking that our cowslip tea (I should not like to
drink it now) was better than our grandmother's best Bohea or
79. something out of her lovely old tea-caddy. So the carriage, lightened
of all but myself, crawled leisurely up and waited on the hill-top for
the busy blackberry-gatherers.
While our horse stood cropping an extempore meal, I and his driver
began to talk about him and other cognate topics, including the
permanent one of the great advantage to both body and soul in
being freed all one's life long from the necessity of getting
something to drink stronger than water.
Yes, he said, I find I can do as much upon tea or coffee as other
men upon beer. I'm just as strong and as active, and can stand
weather quite as well. It's a pretty hard life, winter and summer,
driving all day, coming in soaked, sometimes in the middle of the
night, having to turn in for an hour or two, and then turn out again.
And you must look after your horse, of course, before you think of
yourself. Still, I stand it well, and that without a drop of beer from
years end to years end.
I congratulated and sympathised; in return for which Charles
entered heart and soul into the blackberry question, pointed out
where the biggest blackberries hung, and looked indeed—he was
still such a young fellow!—as if he would have liked to go blackberry-
hunting himself.
I put, smiling, the careless question, Have you any little folks of
your own? Are you married?
How cautious one should be over an idle word! All of a sudden the
cheerful face clouded, the mouth began to quiver, with difficulty I
saw he kept back the tears. It was a version in every-day life of
Longfellow's most pathetic little poem, The Two Locks of Hair.
My wife broke her heart after the baby, I think. It died. She went
off in consumption. It's fifteen months now—(he had evidently
counted them)—fifteen months since I have been alone. I didn't like
to give up my home and my bits of things; still, when a man has to
come in wet and tired to an empty house——
80. A FISHERMAN'S CELLAR NEAR THE LIZARD.
He turned
suddenly
away and
busied
himself over
his horse, for
just that
minute the
two girls
came
running
back,
laughing
heartily, and
showing
their baskets
full of the
very biggest
blackberries
you ever
saw! I took
them back
into the
carriage; the
driver
mounted his
box, and
drove on for
some miles in total silence. As, when I had whispered that little
episode to my two companions, so did we.
There are two ways of going from Falmouth to the Lizard—the
regular route through the town of Helstone, and another, a trifle
longer, through the woods of Trelowarren, the seat of the old
Cornish family of Vyvyan.
81. Welcome to our website – the perfect destination for book lovers and
knowledge seekers. We believe that every book holds a new world,
offering opportunities for learning, discovery, and personal growth.
That’s why we are dedicated to bringing you a diverse collection of
books, ranging from classic literature and specialized publications to
self-development guides and children's books.
More than just a book-buying platform, we strive to be a bridge
connecting you with timeless cultural and intellectual values. With an
elegant, user-friendly interface and a smart search system, you can
quickly find the books that best suit your interests. Additionally,
our special promotions and home delivery services help you save time
and fully enjoy the joy of reading.
Join us on a journey of knowledge exploration, passion nurturing, and
personal growth every day!
ebookbell.com