ChapterPDF AvailableLiterature Review

Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac Development, Degeneration and Regeneration

Authors:

Abstract and Figures

Cardiovascular diseases are the most common cause of human death in the developing world. Extensive evidence indicates that various toxic environmental factors and unhealthy lifestyle choices contribute to the risk, incidence and severity of cardiovascular diseases. Alterations in the genetic level of myocardium affects normal heart development and initiates pathological processes leading to various types of cardiac diseases. Homeobox genes are a large and highly specialized family of closely related genes that direct the formation of body structure, including cardiac development. Homeobox genes encode homeodomain proteins that function as transcription factors with characteristic structures that allow them to bind to DNA, regulate gene expression and subsequently control the proper physiological function of cells, tissues and organs. Mutations in homeobox genes are rare and usually lethal with evident alterations in cardiac function at or soon after the birth. Our understanding of homeobox gene family expression and function has expanded significantly during the recent years. However, the involvement of homeobox genes in the development of human and animal cardiac tissue requires further investigation. The phenotype of human congenital heart defects unveils only some aspects of human heart development. Therefore, mouse models are often used to gain a better understanding of human heart function, pathology and regeneration. In this review, we have focused on the role of homeobox genes in the development and pathology of human heart as potential tools for the future development of targeted regenerative strategies for various heart malfunctions.
Content may be subject to copyright.
Adv Exp Med Biol Cell Biology and Translational Medicine
https://doi.org/10.1007/5584_2019_349
#Springer Nature Switzerland AG 2019
Homeobox Genes and Homeodomain
Proteins: New Insights into Cardiac
Development, Degeneration
and Regeneration
Rokas Miksiunas, Ali Mobasheri, and Daiva Bironaite
Abstract
Cardiovascular diseases are the most common
cause of human death in the developing world.
Extensive evidence indicates that various toxic
environmental factors and unhealthy lifestyle
choices contribute to the risk, incidence and
severity of cardiovascular diseases. Alterations
in the genetic level of myocardium affects
normal heart development and initiates patho-
logical processes leading to various types of
cardiac diseases. Homeobox genes are a large
and highly specialized family of closely related
genes that direct the formation of body struc-
ture, including cardiac development. Homeo-
box genes encode homeodomain proteins that
function as transcription factors with character-
istic structures that allow them to bind to DNA,
regulate gene expression and subsequently con-
trol the proper physiological function of cells,
tissues and organs. Mutations in homeobox
genes are rare and usually lethal with evident
alterations in cardiac function at or soon after
the birth. Our understanding of homeobox gene
family expression and function has expanded
signicantly during the recent years. However,
the involvement of homeobox genes in the
development of human and animal cardiac tis-
sue requires further investigation. The pheno-
type of human congenital heart defects unveils
only some aspects of human heart develop-
ment. Therefore, mouse models are often used
to gain a better understanding of human heart
function, pathology and regeneration. In this
review, we have focused on the role of homeo-
box genes in the development and pathology of
human heart as potential tools for the future
development of targeted regenerative strategies
for various heart malfunctions.
Keywords
Cardiac development · Cardiac regeneration ·
Heart disease · Homeobox genes
Abbreviations
AMHC1 atrial myosin heavy chain-1
ANTP Antennapedia
BMP bone morphogenetic protein
Cdh2 cadherin 2
CDK cyclin-dependent kinases
Cited2 Cbp/P300 interacting
transactivator with Glu/Asp Rich
Carboxy-Terminal Domain 2
CNS central nerve system
R. Miksiunas, A. Mobasheri, and D. Bironaite (*)
Department of Regenerative Medicine, State Research
Institute Centre for Innovative Medicine, Vilnius,
Lithuania
e-mail: daibironai@gmail.com;
daiva.bironaite@imcentras.lt
ESC embryonic stem cells
FGF broblast growth factor
FHF rst heart eld
Flk1 fetal liver kinase 1
GJA5 gap junction protein alpha 5
GSC goosecoid
H3K27me3 histone H3 methylation on the
amino (N) terminal tail
Hcn4 hyperpolarization-activated cyclic
nucleotide-gated channel 4 gene
HOXL homeobox transcription factor
Hox-like
Irx Iroquois family of homeobox
genes
ISL1 LIM-homeodomain transcription
factor islet 1/insulin gene enhancer
protein ISL-1
JMJD3 JmjC domain-containing protein 3
MEF2C myocyte-specic enhancer factor
2C
MESP1 mesoderm posterior BHLH tran-
scription factor 1
MSCs mesenchymal stem cells
Myocd myocardin
NKL NK-like
Nkx2-5 homeobox protein NK-2 homolog
E
Nodal nodal growth differentiation factor
Nppa natriuretic peptide A
OFT outow tract
PCBP2 poly(rC)-binding protein 2
Pitx2 paired like homeodomain 2
Pitx2c paired-like homeodomain tran-
scription factor 2
PROS prospero
RA retinoic acid
SAN sinoatrial node
SHF second heart eld
Shox2 short stature homeobox 2
SMAD main signal transducers for
receptors of the transforming
growth factor beta (TGF-β)
superfamily;
TALE three-amino-acid loop extension
Tbx5 T-box transcription factor 5
TF transcription factors
TGF-βtransforming growth factor beta;
VCS ventricular conduction system
ZEB2 zinc nger E-box binding homeo-
box 2
ZF zinc nger
Ziro zebrash iroquois homeobox
genes
ZO-3 tight junction protein 3
1 Introduction
Homeobox genes are a large family of genes that
direct the formation of body structures along the
head-tail axis in multicellular animal species
(Innis 1997; Shashikant et al. 1991). It is also
known that homeobox genes (Hox genes), as an
ancient class of transcription factors, are impor-
tant for the body patterning during embryo devel-
opment (Innis 1997; Shashikant et al. 1991).
Many of the homeobox genes play very important
part in the spatiotemporal development of human
heart (Lage et al. 2010). Likewise, some of these
genes shape the human heart and control its mul-
tistep developmental process from simple cres-
cent cells to a fully functional organ. For
example, homeobox genes like homeobox protein
NK-2 homolog E (Nkx2-5), LIM-homeodomain
transcription factor islet 1 (Isl1), paired like
homeodomain 2 (Pitx2) are widely known to be
important for the proper development of human
heart (Akazawa and Komuro 2005; Luo et al.
2014; Franco et al. 2017). However, there are
many more homeobox genes that play substantial
roles in cardiac function but thus far, there are less
known and/or less investigated.
Specic inherited gene mutations cause con-
genital heart defects such as atrial or ventricle
septal defects, abnormalities of outow tract and
etc. (Bao et al. 1999). Similarly, various patho-
logical lifestyle factors like smoking, low physi-
cal activity, toxic and noxious agents and other
environmental factors might also negatively
affect cardiovascular function and promote heart
failure (OToole et al. 2008; Nayor and Vasan
R. Miksiunas et al.
2015). Since it is impossible to exactly pinpoint
how certain gene mutations inuence develop-
ment of human heart at the earliest stages, differ-
ent mouse models have been created to better
understand regulation of human heart develop-
ment and its relation to various diseases
(Camacho et al. 2016). Many of the genes studies
in mouse models have similar vital roles in the
development and function of human heart
(Xu and Baldini 2007). Therefore, investigation
of human disease and cues from mouse heart
development models have revealed an important
role of homeobox genes, including those that
encode transcription factors.
Aside from already known homeobox genes,
there are more homeobox genes that are essential
for the formation of human and/or mouse myo-
cardium. Some of these homeobox genes code
transcription factors (TF), whereas other form a
tight network regulating heart development and
fate of heart progenitors. Several review articles
have explored individual families of homeobox
gene and their roles in embryo development.
However, knowledge concerning the involve-
ment of homeobox genes and homeodomain TF
in the development of human heart referring
mouse models are still lacking. Therefore, in this
review we describe the role of more than
20 homeobox genes that are mainly involved in
heart development and around 15 homeobox
genes that are known to play minor or less
investigated, but nonetheless important roles in
cardiac development. Data summarized in this
review will help to broaden the possible future
applications of homeobox genes and their coded
TF in targeted therapeutic strategies for cardiac
regeneration and therapy.
2 Development of the Human
Heart
Starting from the day rst of fertilization, the
zygote undergoes multiple cell divisions leading
to the formation of third germ layer, known as the
mesoderm (Moorman et al. 2003). Later
mesodermal cells migrate towards anterior part
of embryo to form a distinct crescent-shaped epi-
thelium, named the cardiac crescent
(Buckingham et al. 2005). Cells situated in the
distinct anterior-lateral territory within the cardiac
crescent contribute to the formation of rst heart
eld (FHF), distinguished by the expression of
hyperpolarization-activated cyclic nucleotide-
gated channel 4 gene (Hcn4) (Liang et al. 2013).
Cardiac progenitor cells also develop into second
heart eld (SHF), which is located medially to the
cardiac crescent and extend posteriorly (Cai et al.
2003). Formation of SHF is marked by the
expression of LIM-homeodomain transcription
factor islet 1 (ISL1) (Cai et al. 2003). Sometimes
progenitors of FHF and SHF are called cardio-
genic or cardiac mesoderm (Dupays et al. 2015;
Liu et al. 2014; Kitajima et al. 2000). These
distinct heart elds fuse to form heart tube,
which eventually develops into functional heart
(Fig. 1) (Moorman et al. 2003; Nemer 2008).
During this time, the primitive cardiac conduction
system, including sinoatrial node (SAN), ventric-
ular conduction system and other, starts to form
(van Weerd and Christoffels 2016). The FHF
cells develop into the left ventricle, as well as
into the atrioventricular canal and part of the
atria, whereas SHF cells develop into the right
ventricle and outow tract, with contribution to
the formation of atria and inow vessels
(Buckingham et al. 2005). Once the heart elds
are formed, they fuse into heart tube and undergo
process called heart looping. During this phase
the whole heart tube twists in the rightward direc-
tion eventually forming clearly visible, but still
primitive, heart chambers (Santini et al. 2016).
Later on, the heart undergoes septation to fully
separated left and right sides of the heart.
There are many factors regulating human and
mouse heart development, however only some of
them may be considered to be core regulators of
cardiogenesis. One of the most important TF is
GATA4 which orchestrates expression of multi-
ple transcriptions including other major
determinants of cardiomyogenesis like Nkx2-5,
T-box transcription factor 5 (Tbx5), heart- and
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
neural crest derivatives-expressed protein
1 (HAND1/2) and others (Bruneau et al. 2001a;
Belaguli et al. 2000; Sepulveda et al. 1998).
GATA4 integrates bone morphogenetic proteins
(BMP) and SMAD, the main signal transducers
for receptors of the transforming growth factor
beta (TGF-β) superfamily, to ensure cardiac cell
survival and stable lineage during cardiac devel-
opment (Benchabane and Wrana 2003). Of
course, there are other factors that promote devel-
opment of various structures within the heart. For
example, it is know that Tbx5 controls atrial gene
expression, whereas myocyte-specic enhancer
factor 2C (MEF2C) promotes development of
ventricle and vasculogenesis (Bruneau et al.
2001a; Lin et al. 1997). Altogether, the develop-
ment of human heart is a carefully controlled
multistep process involving many genes, intracel-
lular and extracellular signalling factors leading
to proper cardiac function. The miss-controlled
heart development process leads to various
inherited or acquired cardiac disorders. This
review focuses mostly on the homeodomain
proteins, as one of the most important group of
transcription factors regulating heart develop-
ment, function and impairment.
3 Homeobox Genes
Homeodomain proteins are one of the most
important group of proteins/transcription factors
regulating plan of body structure and
organogenesis in eukaryotes including heart
development and disorders. DNA binding
proteins have been extensively studied, but even
today there are no established rules for predicting
the specicity of DNA sequence based upon the
amino acid sequence of the proteins.
Homeodomain proteins are characterized by spe-
cic 60 amino acid long helix-turn-helix DNA
binding homeodomain motif (Seifert et al.
2015). The homeodomain is a very highly
conserved structure and consists of three helical
regions folded into a tight globular structure that
binds a 50-TAAT-30core motif. The high degree
of conservation of homeodomain proteins is an
ideal model to study specic protein-DNA
interactions. The DNA sequence that encodes
the homeodomain is called the homeoboxand
homeobox-containing genes are known as hox
genes.
Most of the transcription factors belonging to
this group are not only structurally but also evolu-
tionary conserved and play crucial roles in embry-
onic patterning and differentiation (Pearson et al.
2005). The main role of homeodomain proteins
in vivo is to control the genetic determination of
development and implementation of the genetic
body plan. There are 102 homeobox gene families
that represent 235 active human homeodomain
proteins, but only some homeodomain classes
have close association with cardiac development
and/or diseases (Bürglin and Affolter 2016). This
review covers description of around 20 homeobox
genes that up today are known to have a major
Fig. 1 Schematic representation of heart development in human and mouse. FGF rst heart eld, SHF second heart
eld, RV right ventricle, LV left ventricle, RA right atrium, LA left atrium. (Scheme adapted from (Nemer, 2008))
R. Miksiunas et al.
impact in the regulation of heart development and
functioning (Fig. 2).
4 Homeobox Genes in Mouse
Heart Development
and Human Disease
Humans have more than 67 genes that are impor-
tant for cardiac hypertrophy and over 92 genes
that control cardiovascular system, therefore it is
reasonable to assume that some of the genes
might be responsible for cardiac development
and disease (van der Harst et al. 2016; Smith
and Newton-Cheh 2015). Congenital heart dis-
ease (CHD) have structural heart anomaly,
including atrial or ventricle septal defects,
overriding aorta, right atrium isomerism and
other structural changes in new born heart.
Patients do usually display multiple symptoms,
like, rapid breathing, bluish skin, poor weight
gain and feeling tired in general (Sun et al.
2015). The main cause of these abnormalities is
reduced blood oxygen levels in whole organism,
which is a result of improper heart septation lead-
ing to the mixture of oxygenated and deoxygen-
ated blood in systemic blood circulation.
Additionally, CHD could be caused by genetical
or environmental factors like infections during
pregnancy such as Rubella, drugs and maternal
illness (Sun et al. 2015). Since homeobox genes
are important for the heart development, some
mutation of the homeobox genes including
MEIS2,Nkx2-5 and others can potentially cause
CHD (Zakariyah et al. 2017; Johansson et al.
2014). Of course, homeobox gene mutations is
not a sole cause of heart defect present at human
birth. For example, mutations in GATA4 and Tbx5
can also affect integrity of heart tissue (McCulley
and Black 2012). In addition, impaired gene func-
tioning might be also related to the other heart
diseases like cardiomyopathy, hypertrophy,
Fig. 2 Hierarchy of homeobox genes involved in heart
development. Pink boxes indicate homeobox gene classes,
yellow box indicates TALE gene superclass. Red boxes
indicate genes with major involvement in heart develop-
ment and diseases. Blue boxes indicate genes having less
important role in heart development and diseases
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
defects of the heart rhythm and other
abnormalities (Kathiresan and Srivastava 2012).
However, the impact of homeobox genes in heart
disorders is still not clear and needs further inves-
tigation to clarify their role not only in the cardiac
development but also in the physiological and
pathophysiological conditions.
5 ANTP Class of Homeobox
Genes
The Antennapedia (ANTP)-class of homeobox
genes are involved in the determination of pattern
formation along the anterior-posterior axis of the
animal embryo. NK-like (NKL) and homeobox
transcription factor Hox-like (HOXL) are an
ancient subclasses of homeobox genes that
belong to an ANTP homeobox gene class (Hol-
land et al. 2007). It is likely that HOXL gene
clusters originate from NKL, since NKL genes
are widespread throughout the genome as tight
clustered Hox genes (Bürglin and Affolter 2016).
Both subclasses of the genes (NKL and HOXL)
are evolutionarily conserved and play
predetermined roles in heart patterning and
disease.
6 Hox Gene Families of HOXL
Subclass
Hox gene families belong to the HOXL subclass of
ANTP class of homeobox. Hox genes code tran-
scription factors which are important for whole
body patterning and development (Pearson et al.
2005). It total, there are 39 human Hox genes
grouped in HOXA, HOXB, HOXC and HOXD
gene clusters. Hox genes are highly conserved,
because they play a vital role in anterior-posterior
formation of body axis (Pearson et al. 2005). The
precise function of these genes is achieved by their
specic temporal and spatial expression over the
life course. During early mouse cardiac develop-
ment, the retinoic acid (RA) might be responsible
for the anterior-posterior patterning in SHF
(Bertrand et al. 2011). Hoxb1, Hoxa1, and Hoxa3
act as downstream targets of RA and participate in
forming outow tract (OFT) and normal SHF
development (Bertrand et al. 2011). Hoxb1/
or Hoxa1/,Hoxb1+/mouse embryos
develop shortened OFT and display abnormal pro-
liferation and premature differentiation of cardiac
progenitors (Bertrand et al. 2011). This is probably
related to the altered broblast growth factor
(FGF) and BMP signaling pathways in developing
mouse embryo (Roux et al. 2015). Clinical studies
have revealed that Hoxa1 mutations might cause
congenital human heart defects and other
abnormalities like, mental retardation, deafness,
horizontal gaze restriction and etc. (Bosley et al.
2008). Several other studies have shown that Hox
genes might be also related to the human heart
diseases, however more research is needed to
unveil exact functions of these genes in
human heart development (Gong et al. 2005;
Haas et al. 2013).
7 Nk4 Gene Family of NKL
Subclass
There are multiple NKL genes in mouse and
humans regulating various developmental pro-
cesses, however only some of them contribute to
the development of the heart (Larroux et al.
2007). The Nkx2-5 and Nkx2-6 genes are the
members of the NK4 homeobox gene family of
NKL subclass and are closely related to the Dro-
sophila tinman gene (Bürglin and Affolter 2016;
Harvey 1996). To our knowledge, only Nkx2-6
and Nkx2-5 relate to the mouse and human heart
development and disease, whereas Nkx2-3,Nkx2-
7,Nkx2-8 and Nkx2-10 might be important for the
heart development of zebrash, frog or chicken
(Newman and Krieg 1998; Wang et al. 2014;Tu
et al. 2009; Allen et al. 2006; Brand et al. 1997).
During the early stages of embryogenesis,
Nkx2-5 is expressed in myocardium and pharyn-
geal endoderm, whereas Nkx2-6 can be found in
sinus venosus, pharyngeal endoderm and myo-
cardium of the outow tract (Lints et al. 1993).
R. Miksiunas et al.
Moreover, during normal heart development,
Nkx2-5 expression is essential for the looping of
vertebrate embryonic heart, heart septation and
formation of cardiac conduction system, whereas
most of the Nkx2-5 mutations are related to
human congenital heart disease and conduction
defects (Tanaka et al. 1999). Inactivation of Nkx2-
5arrested heart formation at the looping stage
revealing its critical role in cardiac development
(Lyons et al. 1995). However, targeted disruption
of Nkx-2.6 did not cause any abnormalities in the
heart suggesting a possible compensatory func-
tion of Nkx-2.5 (Tanaka et al. 2000).
It is important to note that Nkx2-5 mutations
lead to an altered spatiotemporal development of
human heart, improper heart septation and forma-
tion of cardiac conduction system (Dupays et al.
2015; McCulley and Black 2012; McElhinney
et al. 2003). Analysis of human Nkx2-5 mutants
and gene truncations showed that most of the
mutations affected Nkx2-5 binding to DNA or
its localization but not protein-protein
interactions (McCulley and Black 2012;
Reamon-Buettner et al. 2004). Several different
studies of mice Nkx2-5 knockout and human
embryonic stem cells (ESC) revealed that Nkx2-
5mutations might alter gene expression of spe-
cic transcription factors like SP1, SRY, JUND,
STAT6, MYCN,PRDM16,HEY2 and others
(Anderson et al. 2018; Li et al. 2015). Also
some studies support an idea that, Nkx2-5 mutant
proteins might alter space and time specic
human cardiac development by dysregulating
BMP, Notch and Wnt signalling pathways
(Anderson et al. 2018; Wang et al. 2011; Luxán
et al. 2016; Cambier et al. 2014). There is a
possibility that Nkx2-5 modulates these pathways
by interacting with multiple transcription factors
in time-dependent mode. For example, in mouse
heart Nkx2-5 interacts with Hand2 transcriptions
factor to activate Irx4, which is necessary for the
ventricular identity (Yamagishi et al. 2001). Con-
versely, Nkx2-5 expression is also timely
regulated since Nkx2-5 overexpression leads to
an improper SAN formation in early mouse
development (Roux et al. 2015). Mammalian
heart development is also regulated by the com-
bination of cardiac transcription factors having
specic DNA motifs in their centrally located
DNA binding domains. It was also shown that
Nkx2-5, GATA4 and Tbx5 can physically inter-
act and synergistically regulate targeted genes
(Hiroi et al. 2001; Pradhan et al. 2016). Since
these genes are the master regulators of heart
development, functional mutations in these
genes are linked to various types of congenital
heart diseases (Benson 2002; Hatcher et al. 2003).
Taken together, Nkx2-5 and other transcription
factors like Isl1, GATA4, Tbx5, Hand2, MEF2C,
Irx4 form a core of transcription factors essential
for the heart development and congenital heart
disease (Fig. 3) (McCulley and Black 2012).
8 HHEX Gene Family of NKL
Subclass
Proline rich homeodomain protein or homeobox
protein (PRH/HHEX) expressed by
hematopoietic system is a transcription factor
belonging to the family of NKL subclass gene
(Bedford et al. 1993). As the name implies, it is
important for the development of hematopoietic
cell, but not less is essential for the development
of other systems, including heart (Bedford et al.
1993). Mouse double HHex mutants have multi-
ple developmental issues, including defective
vasculogenesis, hypoplasia of the right ventricle,
aberrant development of the compact myocar-
dium and other complications related to forebrain,
thyroid and liver developmental disorders (Hallaq
et al. 1998). Additional studies have revealed that
HHex plays distinct role in mouse cardiac meso-
derm specication and development. HHex
expression is controlled by Sox17 transcription
factor, which is known to be essential for the
formation of mouse cardiac mesoderm (Liu
et al. 2014). Several studies of human population
have shown that common variants of HHex gene
(rs7923837 and rs1111875) may also be
associated with diabetes (Karns et al. 2013;
Kelliny et al. 2009; Pechlivanis et al. 2010).
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
9 PRD and PRD-Like Homeobox
Class
The PRD class is the second largest of the homeo-
box gene classes in animal genomes and, like the
ANTP class, these genes have been found only in
animals. The PRD class derives its name from the
Paired (Prd) gene of Drosophila. Multiple gene
families belong to the PRD homeobox class,
including Shox2,Hopx,GSC,Pitx2 and others,
which are important for the heart development
and disease.
10 Shox Gene Family of PRD Class
Short stature homeobox 2 (Shox2) is a homeobox
gene belonging to the PRD class of homeobox.
Shox2 is an essential for the development of limb
and cardiac conduction systems, including forma-
tion of sinoatrial node (SAN) in mice and humans
(Gu et al. 2008; Blaschke et al. 2007; Liu et al.
2011). Studies of Shox2 function during the
mouse development revealed several cues how
this homeodomain transcription factor in particu-
lar controls formation of SAN (Blaschke et al.
2007). Shox2 mice null mutants displayed severe
cardiac conduction defects, such as low heart
rhythm rate and drastically reduced cell prolifera-
tion (Espinoza-Lewis et al. 2009). This phenotype
is probably related to the downregulation of
HCN4,Tbx3 and the upregulation of natriuretic
peptide A (Nppa), gap junction protein alpha
5(GJA5) and Nkx2-5 gene expressions
(Espinoza-Lewis et al. 2009). It is also known
that HCN channels play a vital role in autonomic
control of heart rate, so it is no surprise why
Shox2 null mutants do not develop SAN (Alig
et al. 2009). During the normal development of
mouse cardiac expression of Shox2 is also tightly
controlled by several transcription factors. For
example, transcription factor Tbx5 activate
Shox2 expression, however transcription factors
like Pitx2c and NKX2-5 potentially silence
Shox2 expression (Espinoza-Lewis et al. 2011;
Puskaric et al. 2010). Paired-like homeodomain
transcription factor 2 (Pitx2c) also can potentially
inhibit left-sided pacemaker specication by
suppressing Shox2 expression in left atrium,
therefore SAN develops only in the region of
right atrium (Wang et al. 2010). All these results
indicate that Shox2 is essential for the maintaining
pacemaker cell program during the heart develop-
ment. On the other hand, in adulthood Nkx2-5
Fig. 3 Core transcription factors important for the heart development and congenital heart disease. (Scheme adapted
from (McCulley and Black, 2012))
R. Miksiunas et al.
antagonizes Shox2 and promotes cardiomyocyte
formation (Fig. 4) (Liang et al. 2017).
Taken together, these studies indicate that
Shox2 might be a good candidate to develop
biological pacemaker. Results from mouse ESCs
and canine mesenchymal stem cells (MSCs) have
shown that cells overexpressing Shox2 induce
expression of SAN markers such as HCN4,
Cx45 and Tbx3 (Ionta et al. 2015; Feng et al.
2016). In addition, mouse embryonic bodies
overexpressing Shox2 showed better contractile
phenotype compared to the control group of
embryonic bodies (Ionta et al. 2015). Moreover,
human patients with an early-onset atrial brilla-
tion had signicantly downregulated expression
of Shox2 gene (Hoffmann et al. 2016). These
results are promising for patients suffering from
heart rhythm defects, however, more studies are
needed to test functions of biological pacemaker
in order to treat human arrhythmias.
11 HOPX Gene Family of PRD
Class
HOPX is another PRD-class homeobox gene
family important for the multiple organ
development and tissue homeostasis in adults
(Chen et al. 2015; Schneider et al. 2015; Mariotto
et al. 2016). Since it lacks a DNA binding
domain, HOPX can only modulate gene expres-
sion by forming complexes with other regulatory
proteins (Kook et al. 2006). In general HOPX acts
as a cell proliferation inhibitor in humans cancer
cells, however its function in mouse cardiac cell
differentiation is not entirely clear (Chen et al.
2015; Waraya et al. 2012; Yap et al. 2016). Stud-
ies of mouse development have shown that
HOPX plays a critical function in early formation
of cardiomyocyte progenitors. HOPX integrates
BMP and WNT signalling in developing mouse
heart by interacting with SMAD proteins and
inhibiting WNT signalling pathway leading to
the formation and differentiation of
cardiomyocyte progenitors (Jain et al. 2015).
On the other hand, there are some cues that
HOPX might act as a negative regulator of car-
diac differentiation in mice. HOPX interacts with
HDAC2, thus reducing GATA4 transcriptional
activity by deacetylation (Trivedi et al. 2010).
These ndings are consistent with previous
reports that overexpression of HDAC2 inhibits
the development of cardiomyocytes by down-
regulating the expression of GATA4 and Nkx2-5
Fig. 4 Signaling networks governing pacemaker and atrial differentiation of cardiomyocyte in developing mouse heart.
(Scheme adapted from (Liang et al. 2017))
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
genes (Kawamura et al. 2005; Karamboulas et al.
2006). All these results highlight the complex
nature of HOPX and its partners in heart develop-
ment. HOPX undeniably plays a critical role in
early heart development, because some mouse
mutants cannot develop a functional myocardium
and display cardiac conduction defects (Chen
et al. 2002; Ismat et al. 2005). HOPX also might
be related to the human heart failure, since HOPX
is downregulated in patients having cardiac
hypertrophy (Güleç et al. 2014; Trivedi et al.
2011).
12 Goosecoid Gene Family of PRD
Class
Goosecoid (GSC) is another protein that belongs
to the bicoid related paired (PRD) homeobox
class of genes. Goosecoid is often associated
with limb, skeletal and craniofacial development,
although, it might be important for cardiac meso-
derm formation, since its expression is controlled
by Mesp1 (Zhu et al. 1998). Mesoderm posterior
BHLH transcription factor 1 (MESP1) preferen-
tially binds to two variations of E-box sequences
and activates critical mesoderm modulators,
including Gata4, mix paired-like homeobox
(Mixl1) and GSC homeobox (Soibam et al.
2015). In addition, mesoderm formation can be
induced with l-proline and trans-4-hydroxy-l-pro-
line resulting in increased expression of Mixl1
and GSC (Date et al. 2013). GSC also is impor-
tant for the cell migration in early embryonic
development, therefore the overexpression of
goosecoid enhances oncogenic cell growth and
metastasis (Kang et al. 2014).
13 Pitx Gene Family of PRD-Like
Class
Paired like homeodomain 2 (Pitx2) is a PRD-like
homeobox class gene which is important for the
establishment of the left-right axis and for the
asymmetrical development of the mouse and
probably human heart, lung, and spleen, twisting
of the gut and stomach, as well as the develop-
ment of the eyes (Campione et al. 1999; Shiratori
et al. 2006; Evans and Gage, 2005). There are
several alternative Pitx2 transcripts, however only
Pitx2c isoform plays determined role in the asym-
metric development of mouse heart (Liu et al.
2002). Higher vertebrates, at an early heart devel-
opment stage and after the heart tube formation,
undergo embryonic heart looping, which is the
rst visual evidence of embryo asymmetry
(Harvey 2002). Transcription factors like nodal
growth differentiation factor (Nodal) and Cbp/P300
interacting transactivator with Glu/Asp Rich
Carboxy-Terminal Domain 2 (Cited2) activate
Pitx2 transcription leading to the rightward twist
of the heart tube and forming prospective embry-
onic atrial and ventricular chambers. Deletion of
Pitx2c in mouse caused drastic alteration of
looping process leading to various heart defects
including the isomerism of right atrium and ven-
tricle (Lin et al. 1999; Yu et al. 2001).
Humans with Pitx2c mutations develop vari-
ous heart abnormalities, including an improper
formation of ventricle and atrial chambers septa,
atrial brillation and others. It is quite likely that
septation defects are caused by the
downregulation of transcription factors down-
stream of Pitx2, since certain Pitx2 mutants
displayed reduced cardiac transcriptional activity
in human patients (Wang et al. 2013; Wei et al.
2014). Surprisingly, the overexpression of Pitx2c
in mouse R1-embryonic stem cells results in ele-
vated gene expression of essential cardiac tran-
scription factors like GATA4, MEF2C, Nkx2-5
and others (Lozano-Velasco et al. 2011). Conse-
quently, Pitx2c might be a good candidate for
heart regeneration, since it positively regulates
multiple transcription factors important for car-
diac development. Recently it was shown that
mouse embryonic stem cells overexpressing
Pitx2c could restore mouse heart function after a
myocardium infarct through the multiple
mechanisms including efcient terminal
R. Miksiunas et al.
differentiation, regulation of action potentials of
cardiomyocytes and positive paracrine effects
(Guddati et al. 2009). However, more studies
need to be done to determine the utility of
Pitx2c in human heart regeneration strategies.
14 TALE Homeobox Superclass
Three-amino-acid loop extension (TALE) is
another superclass of homeobox genes, which
codes for highly conserved transcription
regulators essential for various developmental
programs. These genes encode proteins with
atypical homeodomain structure, dened by hav-
ing three additional amino acids in
homeodomain. TALE homeobox gene superclass
includes the main zinc nger (ZF), PBC and Meis
homeobox 1 (Meis) classes. Out of 20 human
homeodomains only Meis1, Meis2 and Iroquois
homeobox proteins 1-6 (Irx1-6) have their clearly
dened function in heart development and
disease.
15 Meis Genes Family of Meis
Class
Meis1 encodes the TALE superclass homeobox
transcription factor implicated in cardiac,
hematopoietic and neural development (Mariotto
et al. 2013; Azcoitia et al. 2005; Hisa et al. 2004).
Meis1 decient mice have malformed cardiac
outow tracts with overriding aorta and ventricu-
lar septal defect (Stankunas et al. 2008).
Downregulation of Meis1 leads to cardiac hyper-
trophy in humans and mice. Meis1 binds poly
(rC)-binding protein 2 (PCBP2) gene promoter
and activates its expression in order to suppress
human or mouse heart hypertrophy (Zhang et al.
2016). In turn, PCBP2 represses angiotensin II,
which enhances hypertrophic human or mouse
cardiac growth (Zhang et al. 2015). There are
around 79 cardiac specic genes that have
Meis1 and NKX2-5 binding sites in developing
mouse heart, some of them are associated with
cell signaling and cardiac progenitor differentia-
tion, like Tbx20, myocardin, cadherin 2 (Cdh2),
Wnt11, and Wnt2 (Dupays et al. 2015). Adult
mouse cardiomyocytes with mutant MEIS1
exhibit increased proliferation and progression
of the cell cycle (Mariotto et al. 2013). This
function is emphasized in adult mouse hearts
since Meis1 activates inhibitors of cyclin-
dependent kinases (CDK) like p15, p16 and p21
(Mariotto et al. 2013). In humans
non-synonymous Meis1 gene variants might be
associated with congenital heart defects, whereas
patients carrying 2p14 microdeletions show
symptoms of deafness and cardiomyopathy
(Mathieu et al. 2017; Arrington et al. 2012). It is
likely that Meis1 is required for the control of
spatiotemporal cell proliferation in early develop-
ing heart to prevent hypertrophy, however more
studies need to be done to fully understand the
role of Meis1 in cardiac development and disease.
Meis2 encodes TALE homeobox superclass
transcription factor essential for the development
of mouse cranial and cardiac neural crest (Machon
et al. 2015). Recent ndings indicate that Meis2
might be an important factor for the proliferation of
fetal human cardiomyocyte cells (Wu et al. 2015).
Reduction of Meis2 gene expression by miR-134
results in slowed progression of human
cardiomyocyte progenitor cell cycle (Wu et al.
2015). A clinical and genetic study also revealed
that small Meis2 deletion can negatively affect
several developmental processes: human patients
with small Meis2 non-frame shift deletion
(c.998_1000del:p.Arg333del) had serious cleft
palate and cardiac septal defects (Louw et al.
2015). It is known that Meis2 interacts with
DNA and forms multimeric complexes with Hox
and Pbx proteins (Louw et al. 2015). Single dele-
tion of arginine residue affects the ability of Meis2
to bind DNA leading to serious developmental
problems of human heart (Louw et al. 2015). Clin-
ical studies have shown that patients having only
one functioning Meis2 gene copy survive, how-
ever they have similar phenotype such as clefting
and ventricular septal defects leading to delayed
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
motor development and learning disability
(Johansson et al. 2014).
16 Irx Gene Family of IRX Class
Iroquois homeobox genes and their coded
homeodomain proteins are another class of tran-
scription factors belonging to TALE superclass of
homeobox genes. Iroquois-class homeodomain TF
(Irx) dening feature is atypical homeodomain
structure and specicIroquois(IRO)
homeodomain family sequence motif, which is
important for the recognition of DNA sequence
(Gómez-Skarmeta and Modolell 2002; Cavodeassi
et al. 2001). Humans and mice have six Irx
proteins, which are important for the development
of lung, nervous system, eye, pancreas, female
gonad, early limb and, of course, heart patterning
(Cavodeassi et al. 2001; Cheng et al. 2005;
Schwab et al. 2006; van Tuyl et al. 2006; Ragvin
et al. 2010; Jorgensen and Gao 2005;McDonald
et al. 2010). Irx1 and Irx2 are expressed in inter-
ventricular septum from E14.5 onward, however,
mouse Irx2 mutants are viable and display no
notable phenotype defects in the developing heart
(Christoffels et al. 2000; Lebel et al. 2003). Irx1
gene variants might be related to the congenital
heart disease in humans (Guo et al. 2017).
Irx3 gene in mice seems to be very important
for the ventricular conduction system (VCS)
(Christoffels et al. 2000). Various studies suggest
that Irx3 is required to maintain rapid electric
conduction through the VCS for proper ventricu-
lar activation, via antithetical regulation of Cx40
and Cx43 expression (Zhang et al. 2011;
Kasahara et al. 2003). Clinical studies have
revealed that defects of Irx3 gene can cause lethal
cardiac arrhythmias in human patients (Koizumi
et al. 2016). Irx3 function appears to be evolu-
tionary conserved, since expression of Ziro3a, a
Irx3 homologue in zebrash, is detected in devel-
oping sh heart (Zhang et al. 2011).
Irx4 is associated with the formation of ven-
tricular myocardium in mouse and humans
(Christoffels et al. 2000; Cheng et al. 2011).
Data from mouse and chicken indicate that Irx4
suppresses atrial gene expression by down
regulating atrial myosin heavy chain-1
(AMHC1) (Bao et al. 1999; Bruneau et al.
2001b). Several Irx4 mutations have been
identied that might be associated with human
congenital heart disease, particularly ventricular
septal defect (Cheng et al. 2011).
Irx5 is expressed in adult mouse heart and
maintains proper action potentials, particularly
regulates T-wave seen in ECG (Costantini et al.
2005). Mice lacking Irx5 develop properly with-
out any structural abnormalities in the heart
(Costantini et al. 2005). This indicates that Irx5
is not required for cardiac development or that
other Irx genes can compensate for the loss
of Irx5.
Irx6 is detectable in mouse developing heart,
however its expression is relatively weak com-
pared to other Irx genes (Christoffels et al. 2000).
17 Zeb Gene Family of ZF Class
ZEB2 or zinc nger E-box binding homeobox 2 is
a gene coding transcription factor belonging to
class of ZF homeobox gene and homeodomain
class of ZN proteins (Bürglin and Affolter, 2016).
It has multiple functional domains (E-box, Zinc
nger, homeobox), so naturally it can control
gene expression with a variety of transcription
factors (Gheldof et al. 2012). The complex nature
of Zeb2 shows that it drives multiple processes
including the development of heart and neural
systems, however, it usually acts as a transcrip-
tion repressor rather than activator (Hegarty et al.
2015). Systematic study of mouse and human
ESC transcriptome differentiation proles
revealed that Zeb2 might play important role in
cardiac specialization. Human ESC with silenced
Zeb2 gene proliferate more slowly and fail to
differentiate into mature cardiomyocytes com-
pared to the wild cells (Busser et al. 2015). In
addition, cardiomyocytes with silenced Zeb2 do
not show any contractile properties, although car-
diac differentiation program is activated. More
R. Miksiunas et al.
detailed analysis has revealed that silencing of
Zeb2 gene negatively affects human striated mus-
cle contraction program, including genes related
to calmodulin pathway, HCN and potassium
channels (Busser et al. 2015). Targeted regulation
of Zeb2 gene expression improves
cardiomyogenic processes and heart regeneration.
Zeb2 mutation is also often associated with
Mowat-Wilson syndrome (Garavelli and
Mainardi 2007). Major signs of this disorder fre-
quently include distinctive facial features, intel-
lectual disability, delayed development, an
intestinal disorder called Hirschsprung disease,
Congenital Heart Disease and other types of
birth defects (Garavelli and Mainardi 2007). All
mentioned disorders are related to the improper
heart development caused by the Zeb2 defective
heart cells. In addition, Zeb2 repress epithelial
genes (claudins, tight junction protein 3 (ZO-3),
connexins, E-cadherin, plakophilin
2, desmoplakin, and crumbs3) in order to induce
epithelial to mesenchymal transition (EMT),
which is crucial for the developmental processes
such as gastrulation, neural crest formation, heart
morphogenesis, formation of the musculoskeletal
system, and craniofacial structures (Vandewalle
et al. 2009; Garavelli et al. 2017).
18 LIM Homeobox Class
LIM homeobox class genes encode two Lim
domains and one homeodomain. Lim domain is
a5060 amino acid length zinc nger motif,
which is primarily involved in protein-protein
interactions, so naturally LIM transcription
factors can interact with multiple proteins in
cell, thus regulating its phenotype.
19 Isl Gene Family of LIM
Homeobox Class
Isl1 is a LIM homeobox class member that
encodes a homeodomain transcription factor
important for cell differentiation, fate determina-
tion and generation of cell diversity in multiple
mouse and human tissues including central nerve
system (CNS), pancreas and heart (Zhuang et al.
2013). During early cardiac development, Isl1,
Nkx2-5 and fetal liver kinase 1 (Flk1) support
the formation of SHF, which gives rise to the
right ventricle, outow tract and part of the atria
(Dyer and Kirby, 2009). Isl1 promotes expansion,
migration and proliferation of SHF progenitor
cells during the development of the mouse heart
(Witzel et al. 2012). Additionally, Isl1+ mouse
heart cells have potential to differentiate into
multiple cell types within the heart, including
cardiomyocytes, smooth muscle, pacemaker and
endothelial cells (Laugwitz et al. 2007).
There are multiple mechanisms explaining
how Isl1 can promote expression of target
genes, which suggests the expression of Isl1 is
tightly controlled during the mouse heart devel-
opment. For example, Nkx2-5 homeodomain
transcription factor downregulates Isl1 expression
in order to promote ventricular development in
mouse heart (Witzel et al. 2012; Prall et al. 2007).
The newest studies indicate, that Isl1 may repress
development of mouse heart ventricle in order to
promote the development of SAN (Dorn et al.
2015). Mouse embryos overexpressing Isl1
develop SAN-like cells instead of ventricle myo-
cardium (Dorn et al. 2015). It is likely that the
expression of Isl1 activates Nkx2-5 expression in
SHF progenitor cells, however, in later staged of
heart development Nkx2-5 shuts down ISL1
expression to promote ventricular development
(Dorn et al. 2015). Isl1 orchestrates the expres-
sion of hundreds of potential genes implicated in
cardiac differentiation, mainly through epigenetic
mechanisms (Wang et al. 2016). Isl1 in mouse
ESCs acts together with JmjC domain-containing
protein 3 (JMJD3) histone demethylase to pro-
mote the demethylation or tri-methylation of core
histone H3 on the amino (N) terminal tail
(H3K27me3) at the enhancers place of key
downstream target genes, such as myocardin
(Myocd), MEF2C and others (Wang et al. 2016).
In addition, Isl1 may reduce histone methylation
near GATA4 and Nkx2-5 genes after the expres-
sion of Isl1 lentiviral gene, and can also recruit
p300 histone acetyltransferase to the promoter of
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
MEF2C gene in order to promote Mef2c expres-
sion in developing mouse embryo (Yu et al.
2013). Other data suggest that lentiviral-induced
overexpression of Isl1 gene promotes not only
MEF2C gene acetylation, but also GATA4 and
Nkx2-5 in C3H10T1/2 mouse cell line (Xu et al.
2016). The tight control of Ils1 gene expression is
required, since it acts as a positive
cardiomyogenic gene regulator reducing methyl-
ation and increasing acetylation levels of genes
and histones by direct and indirect methods. Most
of the published data concerning Isl1 function
have come from the studies of mouse develop-
ment, however there are some studies that link
Isl1 gene expression with the susceptibility to
human congenital heart disease (Luo et al. 2014;
Stevens et al. 2010). Development of mechanisms
that could control expression of Isl1 might be
important target in further regulation of heart
regeneration.
20 PROS Homeobox Class
Homeobox prospero (PROS) genes code atypical
C terminal prospero domain and belongs to a
distinctive class of Prospero homeodomain
proteins (Yousef and Matthews, 2005). The
PROS domain is a DNA binding domain of
approximately 100 amino acids. In addition,
PROS homeobox genes code additional three
amino acids in their HD domain (Yousef and
Matthews 2005).
21 PROX Gene Family of PROS
Homeobox Class
Prospero homeobox 1 or Prox1 is a gene coding a
transcription factor that plays important role in
the development of mouse heart, CNS, eye, liver
and lymphatic system (Elsir et al. 2012). Firstly, it
was discovered in Drosophila as an important
player in the development of central nervous sys-
tem in insects. However, later Prox1 homologues
were found in vertebrates and mammals (Elsir
et al. 2012). In mouse heart development of
Prox1 is important for the sarcomere formation
and muscle contraction (Risebro et al. 2009).
Mouse Prox1 conditional mutants show increased
number of fast twitch bers compared to slow
twitch bers. Prox1 mutant mice develop fatal
dilated cardiomyopathy and die around 714th
week (Petchey et al. 2014). It was shown that in
mice Prox1 acts as a transcriptional repressor of
genes like Tnnt3,Tnni2 and Myl1 that are essen-
tial for the formation of fast twitch bbers
(Petchey et al. 2014). Prox1 might also be impor-
tant for the maintenance of cardiac conduction
system in adult mice. It was also shown that
uncontrolled Nkx2-5 expression led to cardiac
conduction defects, surprisingly suggesting that
Prox1 might act as a direct upstream modier of
Nkx2-5 gene expression (Risebro et al. 2012). In
humans dysregulation of Prox1 gene expression
might also lead to congenital heart disease, like,
hypoplastic left heart (Gill et al. 2009). Thus, the
close connection of Prox1 with Nkx2-5 and other
heart development and diseases regulating genes
makes it an attractive target in cardiac regenera-
tion eld.
22 Role of Homeobox Genes
in Cardiomyogenesis
The summarized and reviewed data of estimated
involvement of homeobox genes in the heart
development, diseases and/or regeneration pro-
cesses suggest that some homeobox genes play
more important role than the other. Data
summarized in Table 1show the homeobox
genes that have been most commonly
investigated with important roles in
cardiomyogenesis.
It is quite evident that dozens of homeobox
genes are required for early cardiomyogenesis,
heart septation, formation of pacemaker cell,
cardiomyocyte and etc. Some of the homeobox
genes are directly related to the development of
CHD, atrial brillations and other cardiac
pathologies. However, there are much more
R. Miksiunas et al.
homeobox genes related to the heart develop-
ment, that so far have been less investigated or
in one or another model system showed less direct
involvement in cardiomyogenic processes
(Table 2). Data summarized in Table 2also high-
light the fact that many more studies are needed to
understand regulation of homeobox genes and
their role in cardiomyogenic processes.
Table 1 Homeobox genes with major involvement in heart development and diseases
Gene Development Disease Reference
Hox Hoxa1, Hoxb2 and Hoxb2 is
important for anterior-posterior
patterning in SHF. Integrating FGF
and BMP signalling.
Hoxa1 mutations might
cause CHD.
Pearson et al. (2005), Bertrand et al.
(2011), Bosley et al. (2008), Gong
et al. (2005) and Haas et al. (2013)
HOXB13, and HOXC5
mutations might be related
to heart disease.
Nkx25Heart looping, heart septation and
cardiac conduction system formation.
Integrates BMP, notch and WNT
signaling during development.
Multiple gene variants and
truncations are related to
CHD.
McCulley and Black (2012), Tanaka
et al. (1999), McElhinney et al.
(2003), Anderson et al. (2018),
Wang et al. (2011), Luxán et al.
(2016) and Cambier et al. (2014)
Hhex Cardiac mesoderm specication. HHex gene variants might
be associated with diabetes.
Liu et al. (2014), Karns et al. (2013),
Kelliny et al. (2009) and Pechlivanis
et al. (2010)
Shox2 Cardiac conduction system
development.
Downregulation during
early-onset atrial brillation.
Blaschke et al. (2007) and Hoffmann
et al. (2016)
Hopx Cardiomyocyte progenitor formation
in mouse early heart. Negative
regulator of GATA4 expression.
Downregulated in patients
having cardiac hypertrophy.
Jain et al. (2015), Trivedi et al.
(2010) and Trivedi et al. (2011)
GSC Cardiac mesoderm specication. Zhu et al. (1998)
Pitx2c Establishment of the left-right axis in
heart development. Heart looping and
chamber septation.
Mutations cause improper
ventricle and atrial
chambers septa formation,
atrial brillation.
Liu et al. (2002), Wang et al. (2013)
and Wei et al. (2014)
Meis1
and
Meis2
Meis1 and Meis2 control of cell cycle
progression during heart
development.
Meis1 and Meis2 gene
variants might be associated
with CHD.
Mariotto et al. (2013), Arrington
et al. (2012), Wu et al. (2015) and
Louw et al. (2015)
Irx1-6 Irx3 very important for ventricular
conduction system.
Irx1 gene variants might be
associated with CHD.
Christoffels et al. (2000), Guo et al.
(2017), Koizumi et al. (2016) and
Cheng et al. (2011)
Irx4 is associated with the formation
of ventricular myocardium in mouse
and humans.
Irx3 gene defects can cause
lethal cardiac arrhythmias
in human patients.
ZEB2 Controls striated muscle
development and contraction.
Gene variants cause Mowat-
Wilson syndrome. Patients
display CHD and other
defects.
Busser et al. (2015) and Garavelli
and Mainardi (2007)
Islet1 Cell expansion, migration and
proliferation. Marks formation of
SHF. Repress ventricular fate in order
to promote sinoatrial node
development. Positive gene
regulator, which reduces gene and
histone methylation levels and
increase acetylation.
Gene variants might be
related to CHD.
Witzel et al. (2012), Dorn et al.
(2015), Wang et al. (2016) and
Stevens et al. (2010)
Prox1 Important for sarcomere formation
and muscle contraction.
Dysregulation of Prox1
might lead to CHD.
Elsir et al. (2012) and Petchey et al.
(2014)
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
23 Concluding Remarks
Heart development is a complex process requir-
ing strict spatiotemporal development to form a
healthy organ providing properly functioning
organism. Multiple transcription factors, signal-
ling pathways, morphogens and other stimuli
govern the heart development process. However,
it is possible to state that multiple homeobox
genes and their coded transcription factors come
into the heart developmental stages when their
function is needed (Tables 1and 2). None of
these homeodomain transcription factors can be
separated from each other, since their ability to
bind DNA affects patterns of multiple gene thus
resulting in changed transcriptome level of multi-
ple cells.
Homeodomain proteins are not the only tran-
scription factors important for cardiac develop-
ment. The transcription factors of other gene
families also signicantly contribute heart devel-
opment. For example, GATA, Tbx, HAND,
Mef2c and other accompany homeodomain
factors like Nkx2-5, Isl1, etc. (Hiroi et al. 2001;
Gao et al. 2011; Maves et al. 2009; Skerjanc et al.
1998). Most of these homeodomain transcription
factors are conserved and display coexistence and
codependence in heart development of human as
well as simple invertebrates like fruit yor
ascidians (Jensen et al. 2013a; Olson 2006).
Only birds and mammals display fully separated
heart, however reptilians still have no septum
between right and left ventricles (Jensen et al.
2013b). Deeper further insights into septum for-
mation of lower vertebrates like snakes, lizards
and turtles could also help to understand signal-
ling networks of human congenital heart diseases.
Maybe in the future will be possible to engineer a
reptile with four chambered heart, thus leading to
better understanding of cardiac regeneration pro-
cess and allowing to develop new therapeutic
Table 2 Homeobox genes having less important role in heart development and diseases
Class Subclass Gene Development and disease Reference
ANTP HOXL MEOX1
and
MEOX2
Control of vascular endothelial cells proliferation in
mice. Dysregulation might be associated with heart
disease in mouse.
Lu et al. (2018), Douville
et al. (2011)
ANTP NKL Msx1 and
Msx2
Regulate survival of secondary heart eld precursors
and post-migratory proliferation of cardiac neural crest
in the outow tract
Chen et al. (2007)
ANTP NKL Lbx1 Specication of a subpopulation of cardiac neural crest
necessary for normal heart development.
Schäfer et al. (2003)
ANTP NKL Nkx2-6 NKX2-6 mutation predisposes to familial atrial
brillation.
Wang et al. (2014)
PRD Prrx1
and
Prrx2
Formation of cardiovascular system and connective
tissues of the heart and in the great arteries and veins.
Bergwerff et al. (2000)
PRD Pax3,
Pax7
Involved in neural crest and cardiac development JA (1996)
ZF
(TALE)
ZFHX3 Genetic polymorphisms in are associated with atrial
brillation in a Chinese Han population.
Liu et al. (2014)
PBC
(TALE)
Pbx1 Patterning of the great arteries and cardiac outow
tract.
Stankunas et al. (2008)
and Arrington et al.
(2012), Chang et al.
(2008)
Pbx acts with Hand2 in early myocardial
differentiation in zebrash. Non-synonymous variants
in PBX genes are associated with congenital heart
defects.
TGIF
(TALE)
Tgif1 and
Tgif2
Left-right asymmetry formation and embryonic heart
looping.
Powers et al. (2010)
SIX/SO Six2 Six2 marks a dynamic subset of second heart eld
progenitors.
Zhou et al. (2017)
R. Miksiunas et al.
strategies for human cardiac congenital and other
types of diseases.
Acknowledgements The study is funded by the Lithua-
nian Research council, project No. S-MIP-17-13.
Ethics Approval and Consent to Participate Not
applicable.
Consent for Publication All authors agree to the publi-
cation of this manuscript.
Availability of Data and Material Not applicable.
Competing Interests The authors declare that they have
no competing interests.
AuthorsContributions RM wrote the manuscript draft.
DB revised the manuscript. AM read, corrected and
approved the nal manuscript.
Funding The study is funded by the Lithuanian Research
council, project No. S-MIP-17-13.
References
Akazawa H, Komuro I (2005) Cardiac transcription factor
Csx/Nkx2-5: its role in cardiac development and
diseases. Pharmacol Ther 107:252268. https://doi.
org/10.1016/j.pharmthera.2005.03.005
Alig J, Marger L, Mesirca P, Ehmke H, Mangoni ME,
Isbrandt D (2009) Control of heart rate by cAMP
sensitivity of HCN channels. Proc Natl Acad Sci U S
A 106:1218912194. https://doi.org/10.1073/pnas.
0810332106
Allen BG, Allen-Brady K, Weeks DL (2006) Reduction of
XNkx2-10 expression leads to anterior defects and
malformation of the embryonic heart. Mech Dev
123:719729. https://doi.org/10.1016/j.mod.2006.07.
008
Anderson DJ, Kaplan DI, Bell KM, Koutsis K, Haynes
JM, Mills RJ, Phelan DG, Qian EL, Leitoguinho AR,
Arasaratnam D, Labonne T, Ng ES, Davis RP,
Casini S, Passier R, Hudson JE, Porrello ER, Costa
MW, Rai A, Curl CL, Delbridge LM et al (2018)
NKX2-5 regulates human cardiomyogenesis via a
HEY2 dependent transcriptional network. Nat
Commun 9:113. https://doi.org/10.1038/s41467-
018-03714-x
Arrington CB, Dowse BR, Bleyl SB, Bowles NE (2012)
Non-synonymous variants in pre-B cell leukemia
homeobox (PBX) genes are associated with congenital
heart defects. Eur J Med Genet 55:235237. https://
doi.org/10.1016/j.ejmg.2012.02.002.Non-
synonymous
Azcoitia V, Aracil M, Martínez-A C, Torres M (2005) The
homeodomain protein Meis1 is essential for denitive
hematopoiesis and vascular patterning in the mouse
embryo. Dev Biol 280:307320. https://doi.org/10.
1016/j.ydbio.2005.01.004
Bao ZZ, Bruneau BG, Seidman JG, Seidman CE, Cepko
CL (1999) Regulation of chamber-specic gene
expression in the developing heart by irx4. Science
283:11611164
Bedford FK, Ashworth A, Enver T, Wiedemann LM
(1993) HEX: a novel homeobox gene expressed during
haematopoiesis and conserved between mouse and
human. Nucleic Acids Res 21:12451249
Belaguli NS, Sepulveda JL, Nigam V, Charron F,
Nemer M, Schwartz RJ (2000) Cardiac tissue enriched
factors serum response factor and GATA-4 are mutual
coregulators. Mol Cell Biol 20:75507558
Benchabane H, Wrana JL (2003) GATA- and Smad1-
dependent enhancers in the Smad7 gene differentially
interpret bone morphogenetic protein concentrations.
Mol Cell Biol 23:66466661
Benson DW (2002) The genetics of congenital heart dis-
ease: a point in the revolution. Cardiol Clin
20:385394
Bergwerff M, Gittenberger-de Groot AC, Wisse LJ,
DeRuiter MC, Wessels A, Martin JF, Olson EN, Kern
MJ (2000) Loss of function of the Prx1 and Prx2
homeobox genes alters architecture of the great elastic
arteries and ductus arteriosus. Virchows Arch 436:12
19
Bertrand N, Roux M, Ryckebüsch L, Niederreither K,
Dollé P, Moon A, Capecchi M, Zaffran S (2011) Hox
genes dene distinct progenitor sub-domains within
the second heart eld. Dev Biol 353:266274. https://
doi.org/10.1016/j.ydbio.2011.02.029
Blaschke RJ, Hahurij ND, Kuijper S, Just S, Wisse LJ,
Deissler K, Maxelon T, Anastassiadis K, Spitzer J,
Hardt SE, Schöler H, Feitsma H, Rottbauer W,
Blum M, Meijlink F, Rappold G, Gittenberger-de
Groot AC (2007) Targeted mutation reveals essential
functions of the homeodomain transcription factor
Shox2 in sinoatrial and pacemaking development. Cir-
culation 115:18301838. https://doi.org/10.1161/
CIRCULATIONAHA.106.637819
Bosley TM, Alorainy IA, Salih MA, Aldhalaan HM,
Abu-Amero KK, Oystreck DT, Tischeld MA, Engle
EC, Erickson RP (2008) The clinical spectrum of
homozygous HOXA1 mutations. Am J Med Genet A
146:12351240. https://doi.org/10.1002/ajmg.a.32262
Brand T, Andrée B, Schneider A, Buchberger A, Arnold H
(1997) Chicken NKx2-8, a novel homeobox gene
expressed during early heart and foregut development.
Mech Dev 64:5359
Bruneau BG, Nemer G, Schmitt JP, Charron F,
Robitaille L, Caron S, Conner DA, Gessler M,
Nemer M, Seidman CE, Seidman JG (2001a) A murine
model of Holt-Oram syndrome denes roles of the
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
T-box transcription factor Tbx5 in cardiogenesis and
disease. Cell 106:709721
Bruneau BG, Bao ZZ, Fatkin D, Xavier-Neto JGD,
Maguire CT, Berul CI, Kass DA, Kuroski-de Bold
ML, de Bold AJ, Conner DA, Rosenthal N, Cepko
CL, Seidman CE, Seidman JG (2001b) Cardiomyopa-
thy in irx4-decient mice is preceded by abnormal
ventricular gene expression. Mol Cell Biol
21:17301736. https://doi.org/10.1128/MCB.21.5.
1730-1736.2001
Buckingham M, Meilhac S, Zaffran S (2005) Building the
mammalian heart from two sources of myocardial
cells. Nat Rev Genet 6:826837. https://doi.org/10.
1038/nrg1710
Bürglin TR, Affolter M (2016) Homeodomain proteins: an
update. Chromosoma 125:497521. https://doi.org/10.
1007/s00412-015-0543-8
Busser BW, Lin Y, Yang Y et al (2015) An orthologous
epigenetic gene expression signature derived from
differentiating embryonic stem cells identies
regulators of cardiogenesis. PLoS One 10:e0141066.
https://doi.org/10.1371/journal.pone.0141066
Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, Evans
S (2003) Isl1 identies a cardiac progenitor population
that proliferates prior to differentiation and contributes
a majority of cells to the heart. Dev Cell 5:877889
Camacho P, Fan H, Liu Z, He J (2016) Small mammalian
animal models of heart disease. Am J Cardiovasc Dis
6:7080
Cambier L, Plate M, Sucov HM, Pashmforoush M (2014)
Nkx2-5 regulates cardiac growth through modulation
of Wnt signaling by R-spondin3. Development
141:29592971. https://doi.org/10.1242/dev.103416
Campione M, Steinbeisser H, Schweickert A, Deissler K,
van Bebber F, Lowe LA, Nowotschin S, Viebahn C,
Haffter P, Kuehn MR, Blum M (1999) The homeobox
gene Pitx2: mediator of asymmetric left-right signaling
in vertebrate heart and gut looping. Development
126:12251234
Cavodeassi F, Modolell J, Gómez-Skarmeta JL (2001) The
Iroquois family of genes: from body building to neural
patterning. Development 128:28472855
Chang CP, Stankunas K, Shang C, Kao SC, Twu KY,
Cleary ML (2008) Pbx1 functions in distinct regulatory
networks to pattern the great arteries and cardiac out-
ow tract. Development 135:35773586. https://doi.
org/10.1242/dev.022350
Chen F, Kook H, Milewski R, Gitler AD, Lu MM, Li J,
Nazarian R, Schnepp R, Jen K, Biben C, Runke G,
Mackay JP, Novotny J, Schwartz RJ, Harvey RP,
Mullins MC, Epstein JA (2002) Hop is an unusual
homeobox gene that modulates cardiac development.
Cell 110:713723
Chen YH, Ishii M, Sun J, Sucov HM, Maxson RE Jr
(2007) Msx1 and Msx2 regulate survival of secondary
heart eld precursors and post-migratory proliferation
of cardiac neural crest in the outow tract. Dev Biol
308:421437. https://doi.org/10.1016/j.ydbio.2007.05.
037
Chen Y, Yang L, Cui T, Pacyna-Gengelbach M, Petersen I
(2015) Hopx is methylated and exerts tumour-
suppressive function through ras-induced senescence
in human lung cancer. J Pathol 235:397407. https://
doi.org/10.1002/path.4469
Cheng CW, Chow RL, Lebel M, Sakuma R, Cheung HO,
Thanabalasingham V, Zhang X, Bruneau BG, Birch
DG, Hui CC, McInnes RR, Cheng S (2005) The Iro-
quois homeobox gene, Irx5, is required for retinal cone
bipolar cell development. Dev Biol 287:4860. https://
doi.org/10.1016/j.ydbio.2005.08.029
Cheng Z, Wang J, Su D, Pan H, Huang G, Li X, Li Z,
Shen A, Xie X, Wang B, Ma X (2011) Two novel
mutations of the IRX4 gene in patients with congenital
heart disease. Hum Genet 130:657662. https://doi.
org/10.1007/s00439-011-0996-7
Christoffels VM, Keijser AG, Houweling AC, Clout DE,
Moorman AFM (2000) Patterning the embryonic heart:
identication of ve mouse Iroquois homeobox genes
in the developing heart. Dev Biol 224:263274. https://
doi.org/10.1006/dbio.2000.9801
Costantini DL, Arruda EP, Agarwal P, Kim KH, Zhu Y,
Zhu W, Lebel M, Cheng CW, Park CY, Pierce SA,
Guerchicoff A, Pollevick GD, Chan TY, Kabir MG,
Cheng SH, Husain M, Antzelevitch C, Srivastava D,
Gross GJ, Hui CC, Backx PH, Bruneau BG (2005) The
homeodomain transcription factor Irx5 establishes the
mouse cardiac ventricular repolarization gradient. Cell
123:347358. https://doi.org/10.1016/j.cell.2005.08.
004
Date Y, Hasegawa S, Yamada T, Inoue Y, Mizutani H,
Nakata S, Akamatsu H (2013) Major amino acids in
collagen hydrolysate regulate the differentiation of
mouse embryoid bodies. J Biosci Bioeng
116:386390. https://doi.org/10.1016/j.jbiosc.2013.
03.014
Dorn T, Goedel A, Lam JT, Haas J, Tian Q, Herrmann F,
Bundschu K, Dobreva G, Schiemann M,
Dirschinger R, Guo Y, Kühl SJ, Sinnecker D, Lipp P,
Laugwitz K-L, Kühl M, Moretti A (2015) Direct
Nkx2-5 transcriptional repression of isl1 controls
cardiomyocyte subtype identity. Stem Cells
33:11131129. https://doi.org/10.1002/stem.1923
Douville JM, Cheung DY, Herbert KL, Moffatt T, Wigle
JT (2011) Mechanisms of MEOX1 and MEOX2 regu-
lation of the cyclin dependent kinase inhibitors p21 and
p16 in vascular endothelial cells. PLoS One 6:e29099.
https://doi.org/10.1371/journal.pone.0029099
Dupays L, Shang C, Wilson R, Kotecha S, Wood S,
Towers N, Mohun T (2015) Sequential binding of
MEIS1 and NKX2-5 on the Popdc2 gene: a mechanism
for spatiotemporal regulation of enhancers during
cardiogenesis. Cell Rep 13:183195. https://doi.org/
10.1016/j.celrep.2015.08.065
Dyer LA, Kirby ML (2009) The role of secondary heart
eld in cardiac development. Dev Biol 336:137144.
https://doi.org/10.1016/j.ydbio.2009.10.009
Elsir T, Smits A, Lindström MS, Nister M (2012) Tran-
scription factor PROX1: its role in development and
R. Miksiunas et al.
cancer. Cancer Metastasis Rev 31:793805. https://doi.
org/10.1007/s10555-012-9390-8
Espinoza-Lewis RA, Yu L, He F, Liu H, Tang R, Shi J,
Sun X, Martin JF, Wang D, Yang J, Chen Y (2009)
Shox2 is essential for the differentiation of cardiac
pacemaker cells by repressing Nkx2-5. Dev Biol
327:376385. https://doi.org/10.1021/nl061786n.
Core-Shell
Espinoza-Lewis RA, Liu H, Sun C, Chen C, Jiao K, Chen
Y (2011) Ectopic expression of Nkx2.5 suppresses the
formation of the sinoatrial node in mice. Dev Biol
356:359369. https://doi.org/10.1016/j.ydbio.2011.
05.663
Epstein JA (1996) Pax3, neural crest and cardiovascular
development. Trends Cardiovasc Med 6:255260.
https://doi.org/10.1016/S1050-1738(96)00110-7
Evans AL, Gage PJ (2005) Expression of the homeobox
gene Pitx2 in neural crest is required for optic stalk and
ocular anterior segment development. Hum Mol Genet
14:33473359. https://doi.org/10.1093/hmg/ddi365
Feng Y, Yang P, Luo S, Zhang Z, Li H, Zhu P, Song Z
(2016) Shox2 inuences mesenchymal stem cell fate in
a co-culture model in vitro. Mol Med Rep 14:637642.
https://doi.org/10.3892/mmr.2016.5306
Franco D, Sedmera D, Lozano-Velasco E (2017) Multiple
roles of Pitx2 in cardiac development and disease. J
Cardiovasc Dev Dis 4:16. https://doi.org/10.3390/
jcdd4040016
Gao XR, Tan YZ, Wang HJ (2011) Overexpression of
Csx/Nkx2.5 and GATA-4 enhances the efcacy of
mesenchymal stem cell transplantation after
myocardial infarction. Circ J 75:26832691. https://
doi.org/10.1253/circj.CJ-11-0238
Garavelli L, Mainardi PC (2007) Mowat-Wilson syn-
drome. Orphanet J Rare Dis 12:112. https://doi.org/
10.1186/1750-1172-2-42
Garavelli L, Ivanovski I, CarafSG, Santodirocco D,
Pollazzon M, Cordelli DM, Abdalla E, Accorsi P,
Adam MP, Baldo C, Bayat A, Belligni E,
Bonvicini F, Breckpot J, Callewaert B, Cocchi G,
Cuturilo G, Devriendt K, Dinulos MB, Djuric O et al
(2017) Neuroimaging ndings in Mowat Wilson
syndrome: a study of 54 patients. Genet Med
19:691700. https://doi.org/10.1038/gim.2016.176
Gheldof A, Hulpiau P, van Roy F, De Craene B, Berx G
(2012) Evolutionary functional analysis and molecular
regulation of the ZEB transcription factors. Cell Mol
Life Sci 69:25272541. https://doi.org/10.1007/
s00018-012-0935-3
Gill HK, Parsons SR, Spalluto C, Davies AF, Knorz VJ,
Burlinson CE, Ng B, Carter NP, Ogilvie CM, Wilson
DI, Roberts RG (2009) Separation of the PROX1 gene
from upstream conserved elements in a complex inver-
sion/translocation patient with hypoplastic left heart.
Eur J Hum Genet 17:14231431. https://doi.org/10.
1038/ejhg.2009.91
Gómez-Skarmeta JL, Modolell J (2002) Iroquois genes:
genomic organization and function in vertebrate neural
development. Curr Opin Genet Dev 12:403408
Gong LG, Qiu GR, Jiang H, Xu XY, Zhu HY, Sun KL
(2005) Analysis of single nucleotide polymorphisms
and haplotypes in HOXC gene cluster within suscepti-
ble region 12q13 of simple congenital heart disease.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi 22:497501
Gu S, Wei N, Yu L, Fei J, Chen Y (2008) Shox2-
deciency leads to dysplasia and ankylosis of the tem-
poromandibular joint in mice. Mech Dev
125:729742. https://doi.org/10.1016/j.mod.2008.04.
003
Guddati AK, Otero JJ, Kessler E, Aistrup G, Wasserstrom
JA, Han X, Lomasney JW, Kessler JA (2009) Embry-
onic stem cells overexpressing Pitx2 engraft in
infarcted myocardium and improve cardiac function.
Int Heart J 50:783799
Güleç Ç, Abac{N, Bayrak F, Kömürcü Bayrak E,
Kahveci G, Güven C, Ünaltuna NE (2014) Association
between non-coding polymorphisms of HOPX gene
and syncope in hypertrophic cardiomyopathy. Anadolu
Kardiyol Derg 14:617624. https://doi.org/10.5152/
akd.2014.4972
Guo C, Wang Q, Wang Y, Yang L, Luo H, Cao XF, An L,
Qiu Y, Du M, Ma X, Hui L, Lu C (2017) Exome
sequencing reveals novel IRXI mutation in congenital
heart disease. Mol Med Rep 15:31933197. https://doi.
org/10.3892/mmr.2017.6410
Haas J, Frese KS, Park YJ, Keller A, Vogel B, Lindroth
AM, Weichenhan D, Franke J, Fischer S, Bauer A,
Marquart S, Sedaghat-Hamedani FKE, Köhler D,
Wolf NM, Hassel S, Nietsch R, Wieland T,
Ehlermann P, Schultz JH, Dösch A, Mereles D,
Hardt S, Backs J, Hoheisel JD, Plass C, Katus HA,
Meder B (2013) Alterations in cardiac DNA methyla-
tion in human dilated cardiomyopathy. EMBO Mol
Med 5:413429. https://doi.org/10.1002/emmm.
201201553
Hallaq H, Pinter E, Enciso J et al (1998) A null mutation of
Hhex results in abnormal cardiac development, defec-
tive vasculogenesis and elevated Vegfa levels. Devel-
opment 131:51975209. https://doi.org/10.1242/dev.
01393
Harvey RP (1996) NK-2 homeobox genes and heart devel-
opment. Dev Biol 178:203216
Harvey RP (2002) Patterning the vertebrate heart. Nat Rev
Genet 3:544556. https://doi.org/10.1038/nrg843
Hatcher CJ, Diman NY, McDermott DA, Basson CT
(2003) Transcription factor cascades in congenital
heart malformation. Trends Mol Med 9:512515
Hegarty SV, Sullivan AM, Keeffe GWO (2015) Progress
in Neurobiology Zeb2: a multifunctional regulator of
nervous system development. Prog Neurobiol
132:8195. https://doi.org/10.1016/j.pneurobio.2015.
07.001
Hiroi Y, Kudoh S, Monzen K, Ikeda Y, Yazaki Y,
Nagai R, Komuro I (2001) Tbx5 associates with
Nkx2-5 and synergistically promotes cardiomyocyte
differentiation. Nat Genet 28:276280. https://doi.org/
10.1038/90123
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
Hisa T, Spence SE, Rachel RA, Fujita M, Nakamura T,
Ward JM, Devor-Henneman DE, Saiki Y, Kutsuna H,
Tessarollo L, Jenkins NA, Copeland NG (2004)
Hematopoietic, angiogenic and eye defects in Meis1
mutant animals. EMBO J 23:450459. https://doi.org/
10.1038/sj.emboj.7600038
Hoffmann S, Clauss S, Berger IM, Weiß B,
Montalbano A, Röth R, Bucher M, Klier I, Wakili R,
Seitz H, Schulze-Bahr E, Katus HA, Flachsbart F,
Nebel A, Guenther SP, Bagaev E, Rottbauer W,
Kääb S, Just S, Rappold G (2016) Coding and
non-coding variants in the SHOX2 gene in patients
with early-onset atrial brillation. Basic Res Cardiol
111:36. https://doi.org/10.1007/s00395-016-0557-2
Holland PWH, Booth HAF, Bruford EA (2007) Classi-
cation and nomenclature of all human homeobox
genes. BMC Biol 5:129. https://doi.org/10.1186/
1741-7007-5-47
Innis JW (1997) Role of HOX genes in human develop-
ment. Curr Opin Pediatr 9:617622
Ionta V, Liang W, Kim EH, Rae R, Giacomello A,
Marbán E, Cho C (2015) SHOX2 overexpression
favors differentiation of embryonic stem cells into
cardiac pacemaker cells, improving biological pacing
ability. Stem Cell Reports 4:129142. https://doi.org/
10.1016/j.stemcr.2014.11.004
Ismat FA, Zhang M, Kook H, Huang B, Zhou R, Ferrari
VA, Epstein JA, Patel VV (2005) Homeobox protein
Hop functions in the adult cardiac conduction system.
Circ Res 96:898903. https://doi.org/10.1161/01.RES.
0000163108.47258.f3
JA E (1996) Pax3, neural crest and cardiovascular devel-
opment. Trends Cardiovasc Med 6:25560. https://doi.
org/10.1016/S1050-1738(96)00110-7
Jain R, Li D, Gupta M, Mandereld LJ, Ifkovits JL,
Wang Q, Liu F, Liu Y, Poleshko A, Padmanabhan A,
Raum JC, Li L, Morrisey EE, Lu MM, Won KJ,
Epstein JA (2015) Integration of Bmp and Wnt signal-
ing by Hopx species commitment of cardiomyoblasts.
Science 348:aaa6071. https://doi.org/10.1016/j.joca.
2015.05.020.Osteoarthritic
Jensen B, Wang T, Christoffels VM, Moorman AFM
(2013a) Evolution and development of the building
plan of the vertebrate heart. Biochim Biophys Acta
1833:783794. https://doi.org/10.1016/j.bbamcr.2012.
10.004
Jensen B, van den Berg G, van den Doel R, Oostra RJ,
Wang T, Moorman AFM (2013b) Development of the
hearts of lizards and snakes and perspectives to cardiac
evolution. PLoS One 8:e63651. https://doi.org/10.
1371/journal.pone.0063651
Johansson S, Berland S, Gradek GA, Bongers E, de
Leeuw N, Pfundt R, Fannemel M, Rødningen O,
Brendehaug A, Haukanes BI, Hovland R, Helland G,
Houge G (2014) Haploinsufciency of MEIS2 is
associated with orofacial clefting and learning disabil-
ity. Am J Med Genet A 164A:16221626. https://doi.
org/10.1002/ajmg.a.36498
Jorgensen JS, Gao L (2005) Irx3 is differentially
up-regulated in female gonads during sex determina-
tion. Gene Expr Pattern 5:756762. https://doi.org/10.
1016/j.modgep.2005.04.011
Kang KW, Lee MJ, Song JA, Jeong JY, Kim YK, Lee C,
Kim TH, Kwak KB, Kim OJ, An HJ (2014)
Overexpression of goosecoid homeobox is associated
with chemoresistance and poor prognosis in ovarian
carcinoma. Oncol Rep 32:189198. https://doi.org/10.
3892/or.2014.3203
Karamboulas C, Swedani A, Ward C, Al-Madhoun AS,
Wilton S, Boisvenue S, Ridgeway AG, Skerjanc IS
(2006) HDAC activity regulates entry of mesoderm
cells into the cardiac muscle lineage. J Cell Sci
119:43054314. https://doi.org/10.1242/jcs.03185
Karns R, Succop P, Zhang G, Sun G, Indugula SR, Havas-
Augustin D, Novokmet N, Durakovic Z, Milanovic
SM, Missoni S, Vuletic S, Chakraborty R, Rudan P,
Deka R (2013) Modeling metabolic syndrome through
structural equations of metabolic traits, comorbid
diseases, and GWAS variants. Obesity 21:745754.
https://doi.org/10.1002/oby.20445
Kasahara H, Ueyama T, Wakimoto H, Liu MK, Maguire
CT, Converso KL, Kang PM, Manning WJ, Lawitts J,
Paul DL, Berul CI, Izumo S (2003) Nkx2.5
homeoprotein regulates expression of gap junction
protein connexin 43 and sarcomere organization in
postnatal cardiomyocytes. J Mol Cell Cardiol
35:243256. https://doi.org/10.1016/S0022-2828(03)
00002-6
Kathiresan S, Srivastava D (2012) Genetics of human
cardiovascular disease. Cell 148:12421257. https://
doi.org/10.1016/j.cell.2012.03.001
Kawamura T, Ono K, Morimoto T et al (2005) Acetylation
of GATA-4 is involved in the differentiation of embry-
onic stem cells into cardiac myocytes. J Biol Chem
280:1968219688. https://doi.org/10.1074/jbc.
M412428200
Kelliny C, Ekelund U, Andersen LB, Brage S, Loos RJ,
Wareham NJ, Langenberg C (2009) Common genetic
determinants of glucose homeostasis in healthy chil-
dren: the European Youth Heart Study. Diabetes
58:29392945. https://doi.org/10.2337/db09-0374
Kitajima S, Takagi A, Inoue T, Saga Y (2000) MesP1 and
MesP2 are essential for the development of cardiac
mesoderm. Development 127:32153226
Koizumi A, Sasano T, Kimura W, Miyamoto Y, Aiba T,
Ishikawa T, Nogami A, Fukamizu S, Sakurada H,
Takahashi Y, Nakamura H, Ishikura T, Koseki H,
Arimura T, Kimura A, Hirao K, Isobe M,
Shimizu W, Miura N, Furukawa T (2016) Genetic
defects in a His-Purkinje system transcription factor,
IRX3, cause lethal cardiac arrhythmias. Eur Heart J
37:14691475. https://doi.org/10.1093/eurheartj/
ehv449
Kook H, Yung WW, Simpson RJ, Kee HJ, Shin S, Lowry
JA, Loughlin FE, Yin Z, Epstein JA, Mackay J (2006)
Analysis of the structure and function of the
R. Miksiunas et al.
transcriptional coregulator HOP. Biochemistry
45:1058410590. https://doi.org/10.1021/bi060641s
Lage K, Møllgård K, Greenway S, Wakimoto H, Gorham
JM, Workman CT, Bendsen E, Hansen NT, Rigina O,
Roque FS, Wiese C, Christoffels VM, Roberts AE,
Smoot LB, Pu WT, Donahoe PK, Tommerup N,
Brunak S, Seidman CE, Seidman JG, Larsen LA
(2010) Dissecting spatio-temporal protein networks
driving human heart development and related
disorders. Mol Syst Biol 6:381. https://doi.org/10.
1038/msb.2010.36
Larroux C, Fahey B, Degnan SM, Adamski M, Rokhsar
DS, Degnan BM (2007) The NK homeobox gene
cluster predates the origin of Hox genes. Curr Biol
17:706710. https://doi.org/10.1016/j.cub.2007.03.
008
Laugwitz KL, Moretti A, Caron L, Nakano A, Chien KR
(2007) Islet1 cardiovascular progenitors: a single
source for heart lineages. Development 135:193205.
https://doi.org/10.1242/dev.001883
Lebel M, Agarwal P, Cheng CW, Kabir MG, Chan TY,
Thanabalasingham V, Zhang X, Cohen DR, Husain M,
Cheng SH, Bruneau BG, Cheng SH (2003) The Iro-
quois homeobox gene irx2 is not essential for normal
development of the heart and midbrain-hindbrain
boundary in mice. Mol Cell Biol 23:82168225
Li J, Cao Y, Wu Y, Chen W, Yuan Y, Ma X, Huang G
(2015) The expression prole analysis of NKX2-5
knock-out embryonic mice to explore the pathogenesis
of congenital heart disease. J Cardiol 66:527531.
https://doi.org/10.1016/j.jjcc.2014.12.022
Liang X, Wang G, Lin L, Lowe J, Zhang Q, Bu L, Chen Y,
Chen J, Sun Y, Evans SM (2013) HCN4 dynamically
marks the rst heart eld and conduction system
precursors. Circ Res 113:399407. https://doi.org/10.
1161/CIRCRESAHA.113.301588
Liang X, Evans SM, Sun Y (2017) Development of the
cardiac pacemaker. Cell Mol Life Sci 74:12471259.
https://doi.org/10.1007/s00018-016-2400-1
Lin Q, Schwarz J, Bucana C, Olson EN (1997) Control of
mouse cardiac morphogenesis and myogenesis by tran-
scription factor MEF2C. Science 276:14041407
Lin CR, Kioussi C, OConnell S et al (1999) Pitx2
regulates lung asymmetry, cardiac positioning and
pituitary and tooth morphogenesis. Nature
401:279282. https://doi.org/10.1038/45803
Lints TJ, Parsons LM, Hartley L, Lyons I, Harvey RP
(1993) Nkx-2.5: a novel murine homeobox gene
expressed in early heart progenitor cells and their myo-
genic descendants. Development 119:419431
Liu CC, Liu WW, Palie JJ et al (2002) Pitx2c patterns
anterior myocardium and aortic arch vessels and is
required for local cell movement into atrioventricular
cushions. Development 129:50815091. https://doi.
org/10.1242/dev.00173
Liu H, Chen CH, Espinoza-Lewis RA, Jiao Z, Sheu I,
Hu X, Lin M, Zhang Y, Chen Y (2011) Functional
redundancy between human SHOX and mouse Shox2
genes in the regulation of sinoatrial node formation and
Pacemaking function *. J Biol Chem
286:1702917038. https://doi.org/10.1074/jbc.M111.
234252
Liu Y, Kaneda R, Leja TW, Subkhankulova T,
Tolmachov O, Minchiotti G, Schwartz RJ,
Barahona M, Schneider M (2014) Hhex and Cer1
mediate the Sox17 pathway for cardiac mesoderm for-
mation in embryonic stem. Stem Cells 32:15151526.
https://doi.org/10.1002/stem.1695
Liu Y, Ni B, Lin Y, Chen XG, Fang Z, Zhao L, Hu Z,
Zhang F, Ai X (2014) Genetic polymorphisms in
ZFHX3 are associated with atrial brillation in a Chi-
nese Han population. PLoS One 9:e101318. https://
doi.org/10.1371/journal.pone.0101318
Louw JJ, Corveleyn A, Jia Y, Hens G, Gewillig M,
Devriendt K (2015) MEIS2 involvement in cardiac
development, cleft palate, and intellectual disability.
Am J Med Genet A 167A:11421146. https://doi.org/
10.1002/ajmg.a.36989
Lozano-Velasco E, Chinchilla A, Martínez-Fernández S
et al (2011) Pitx2c modulates cardiac-specic tran-
scription factors networks in differentiating
cardiomyocytes from murine embryonic stem cells.
Cells Tissues Organs 194:349362. https://doi.org/10.
1159/000323533
Lu D, Wang J, Li J, Guan F, Zhang X, Dong W, Liu N,
Gao S, Zhang L (2018) Meox1 accelerates myocardial
hypertrophic decompensation through Gata4.
Cardiovasc Res 114:300311. https://doi.org/10.
1093/cvr/cvx222
Luo ZL, Sun H, Yang ZQ, Ma YH, Gu Y, He YQ, Wei D,
Xia LB, Yang BH, Guo T (2014) Genetic variations of
ISL1 associated with human congenital heart disease in
Chinese Han people. Genet Mol Res 13:13291338.
https://doi.org/10.4238/2014.February.28.5
Luxán G, DAmato G, de la Pompa JL (2016) Endocardial
notch signaling in cardiac development and disease.
Circ Res 118:118. https://doi.org/10.1161/
CIRCRESAHA.115.305350
Lyons I, Parsons LM, Hartley L, Li R, Andrews JE,
Robb L, Harvey RP (1995) Myogenic and morphoge-
netic defects in the heart tubes of murine embryos
lacking the homeo box gene Nkx25. Genes Dev
9:16541666
Machon O, Masek J, Machonova O, Krauss S, Kozmik Z
(2015) Meis2 is essential for cranial and cardiac neural
crest development. BMC Dev Biol 15:40. https://doi.
org/10.1186/s12861-015-0093-6
Mariotto A, Pavlova O, Park HS, Huber M, Sadek HA
(2013) Meis1 regulates postnatal cardiomyocyte cell
cycle arrest. Nature 497:249253. https://doi.org/10.
1038/jid.2014.371
Mariotto A, Pavlova O, Park HS et al (2016) HOPX: the
unusual homeodomain-containing protein. J Invest
Dermatol 136:905911. https://doi.org/10.1016/j.jid.
2016.01.032
Mathieu ML, Demily C, Chantot-Bastaraud S, Afenjar A,
Mignot C, Andrieux J, Gerard M, Catala-Mora J, Jouk
PS, Labalme A, Edery P, Sanlaville D, Rossi M (2017)
Clinical and molecular cytogenetic characterization of
four unrelated patients carrying 2p14 microdeletions.
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
Am J Med Genet A 173:22682274. https://doi.org/10.
1002/ajmg.a.38307
Maves L, Tyler A, Moens CB, Tapscott SJ (2009) Pbx acts
with Hand2 in early myocardial differentiation. Dev
Biol 333:409418. https://doi.org/10.1016/j.ydbio.
2009.07.004
McCulley DJ, Black BL (2012) Transcription factor
pathways and congenital heart disease. Curr Top Dev
Biol 100:253277. https://doi.org/10.1016/B978-0-12-
387786-4.00008-7.Transcription
McDonald LA, Gerrelli D, Fok Y, Hurst LD, Tickle C
(2010) Comparison of Iroquois gene expression in
limbs/ns of vertebrate embryos. J Anat
216:683691. https://doi.org/10.1111/j.1469-7580.
2010.01233.x
McElhinney DB, Geiger E, Blinder J, Benson DW,
Goldmuntz E (2003) NKX2.5 mutations in patients
with congenital heart disease. J Am Coll Cardiol
42:16501655
Moorman A, Webb S, Brown NA et al (2003) Develop-
ment of the heart: (1) formation of the cardiac
chambers and arterial trunks. Heart 89:806814
Nayor M, Vasan RS (2015) Preventing heart failure: the
role of physical activity. Curr Opin Cardiol
30:543550. https://doi.org/10.1097/HCO.
0000000000000206
Nemer M (2008) Genetic insights into normal and abnor-
mal heart development. Cardiovasc Pathol 17:4854.
https://doi.org/10.1016/j.carpath.2007.06.005
Newman CS, Krieg PA (1998) Tinman-related genes
expressed during heart development in Xenopus. Dev
Genet 22:230238. https://doi.org/10.1002/(SICI)
1520-6408(1998)22:3<230::AID-DVG5>3.0.CO;2-7
OToole TE, Conklin DJ, Bhatnagar A (2008) Environ-
mental risk factors for heart disease. Rev Environ
Health 23:167202. https://doi.org/10.1038/nrcardio.
2015.152
Olson EN (2006) Gene regulatory networks in the evolu-
tion and development of the heart. Science
313:19221927. https://doi.org/10.1126/science.
1132292
Pearson JC, Lemons D, McGinnis W (2005) Modulating
Hox gene functions during animal body patterning. Nat
Rev Genet 6:893904. https://doi.org/10.1038/
nrg1726
Pechlivanis S, Scherag A, Mühleisen TW, Möhlenkamp S,
Horsthemke B, Boes T, Bröcker-Preuss M, Mann K,
Erbel R, Jöckel KH, Nöthen MM, Moebus S (2010)
Coronary artery calcication and its relationship to
validated genetic variants for diabetes mellitus
assessed in the Heinz Nixdorf recall cohort.
Arterioscler Thromb Vasc Biol 30:18671872.
https://doi.org/10.1161/ATVBAHA.110.208496
Petchey LK, Risebro CA, Vieira JM, Roberts T, Bryson
JB, Greensmith L, Lythgoe MF, Riley PR (2014) Loss
of Prox1 in striated muscle causes slow to fast skeletal
muscle ber conversion and dilated cardiomyopathy.
Proc Natl Acad Sci U S A 111:95159520. https://doi.
org/10.1073/pnas.1406191111
Powers SE, Taniguchi K, Yen W, Melhuish TA, Shen J,
Walsh CA, Sutherland AE, Wotton D (2010) Tgif1 and
Tgif2 regulate Nodal signaling and are required for
gastrulation. Development 137:249259. https://doi.
org/10.1242/dev.040782
Pradhan L, Gopal S, Li S, Ashur S, Suryanarayanan S,
Kasahara H, Nam H (2016) Intermolecular interactions
of cardiac transcription factors NKX2.5 and TBX5.
Biochemistry 55:17021710. https://doi.org/10.1021/
acs.biochem.6b00171
Prall OW, Menon MK, Solloway MJ, Watanabe Y,
Zaffran S, Bajolle F, Biben C, McBride JJ, Robertson
BR, Chaulet H, Stennard FA, Wise N, Schaft D,
Wolstein O, Furtado MB, Shiratori H, Chien KR,
Hamada H, Black BL, Saga Y, Robertson EJ,
Buckingham ME, Harvey RP (2007) An Nkx2-5/
Bmp2/Smad1 negative feedback loop controls heart
progenitor specication and proliferation. Cell
128:947959
Puskaric S, Schmitteckert S, Mori AD, Glaser A,
Schneider KU, Bruneau BG, Blaschke RJ,
Steinbeisser H, Rappold G (2010) Shox2 mediates
Tbx5 activity by regulating Bmp4 in the pacemaker
region of the developing heart. Hum Mol Genet
19:46254633. https://doi.org/10.1093/hmg/ddq393
Ragvin A, Moro E, Fredman D, Navratilova P, Drivenes
Ø, Engström PG, Alonso ME, de la Calle Mustienes E,
Gómez Skarmeta JL, Tavares MJ, Casares F,
Manzanares M, van Heyningen V, Molven A, Njølstad
PR, Argenton F, Lenhard B, Becker TS (2010) Long-
range gene regulation links genomic type 2 diabetes
and obesity risk regions to HHEX, SOX4, and IRX3.
Proc Natl Acad Sci U S A 107:775780. https://doi.
org/10.1073/pnas.0911591107
Reamon-Buettner SM, Hecker H, Spanel-Borowski K,
Craatz S, Kuenzel E, Borlak J (2004) Novel NKX2-5
mutations in diseased heart tissues of patients with
cardiac malformations. Am J Pathol 164:21172125.
https://doi.org/10.1016/S0002-9440(10)63770-4
Risebro CA, Searles RG, Melville A, Athalie AD et al
(2009) Prox1 maintains muscle structure and growth in
the developing heart. Development 136:495505.
https://doi.org/10.1242/dev.030007
Risebro CA, Petchey LK, Smart N et al (2012) Epistatic
rescue of Nkx2.5 adult cardiac conduction disease
phenotypes by prospero-related homeobox protein
1 and HDAC3. Circ Res 111:e19-31. https://doi.org/
10.1161/CIRCRESAHA.111.260695
Roux M, Laforest B, Capecchi M, Bertrand N, Zaffran S
(2015) Hoxb1 regulates proliferation and differentia-
tion of second heart eld progenitors in pharyngeal
mesoderm and genetically interacts with Hoxa1 during
cardiac outow tract development. Dev Biol
406:247258. https://doi.org/10.1016/j.ydbio.2015.
08.015
Santini MP, Forte E, Harvey RP, Kovacic JC (2016)
Developmental origin and lineage plasticity of endog-
enous cardiac stem cells. Development 4:12421258.
https://doi.org/10.1242/dev.111591
R. Miksiunas et al.
Schäfer K, Neuhaus P, Kruse J, Braun T (2003) The
homeobox gene Lbx1 species a subpopulation of
cardiac neural crest necessary for normal heart devel-
opment. Circ Res 92:7380
Schneider MD, Baker AH, Riley P (2015) Hopx and the
cardiomyocyte parentage. Mol Ther 23:14201422.
https://doi.org/10.1038/mt.2015.140
Schwab K, Hartman HA, Liang HC, Aronow BJ, Patterson
LT, Potter SS (2006) Comprehensive microarray anal-
ysis of hoxa11/hoxd11 mutant kidney development.
Dev Biol 293:540554. https://doi.org/10.1016/j.
ydbio.2006.02.023
Seifert A, Werheid DF, Knapp SM, Tobiasch E (2015)
Role of Hox genes in stem cell differentiation. World J
Stem Cells 7:583595. https://doi.org/10.4252/wjsc.
v7.i3.583
Sepulveda JL, Belaguli N, Nigam V, Chen CY, Nemer M,
Schwartz RJ (1998) GATA-4 and Nkx-2.5 coactivate
Nkx-2 DNA binding targets: role for regulating early
cardiac gene expression. Mol Cell Biol 18:34053415
Shashikant CS, Utset MF, Violette SM, Wise TL, Einat P,
Einat MPJ, Schughart K, Ruddle FH (1991) Homeo-
box genes in mouse development. Crit Rev Eukaryot
Gene Expr 1:207245
Shiratori H, Yashiro K, Shen MM, Hamada H (2006)
Conserved regulation and role of Pitx2 in situs-specic
morphogenesis of visceral organs. Development
133:30153025. https://doi.org/10.1242/dev.02470
Skerjanc IS, Petropoulos H, Ridgeway AG, Wilton S
(1998) Myocyte enhancer factor 2C and Nkx2-5
up-regulate each others expression and initiate
cardiomyogenesis in P19 cells. J Biol Chem
273:3490434910. https://doi.org/10.1074/jbc.273.52.
34904
Smith JG, Newton-Cheh C (2015) Genome-wide associa-
tion studies of late-onset cardiovascular disease. J Mol
Cell Cardiol 83:131141. https://doi.org/10.1016/j.
yjmcc.2015.04.004
Soibam B, Benham A, Kim J, Weng KC, Yang L, Xu X,
Robertson M, Azares A, Cooney AJ, Schwartz RJ, Liu
Y (2015) Genome-wide identication of MESP1
targets demonstrates primary regulation over. Stem
Cells 33:32543265. https://doi.org/10.1002/stem.
2111
Stankunas K, Shang C, Twu KY, Kao SC, Jenkins NA,
Copeland NG, Sanyal M, Selleri L, Cleary ML, Chang
C (2008) Pbx/Meis deciencies demonstrate
multigenetic origins of congenital heart disease. Circ
Res 103:702709. https://doi.org/10.1161/
CIRCRESAHA.108.175489
Stevens KN, Hakonarson H, Kim CE, Doevendans PA,
Koeleman BP, Mital S, Raue J, Glessner JT, Coles JG,
Moreno V, Granger A, Gruber SB, Gruber PJ (2010)
Common variation in ISL1 confers genetic susceptibil-
ity for human congenital heart disease. PLoS One 26:
e10855. https://doi.org/10.1371/journal.pone.0010855
Sun R, Liu M, Lu L, Zheng Y, Zhang P (2015) Congenital
heart disease: causes, diagnosis, symptoms, and
treatments. Cell Biochem Biophys 72:857860.
https://doi.org/10.1007/s12013-015-0551-6
Tanaka M, Chen Z, Bartunkova S, Yamasaki N, Izumo S
(1999) The cardiac homeobox gene Csx/Nkx2.5 lies
genetically upstream of multiple genes essential for
heart development. Development 126:12691280
Tanaka M, Yamasaki N, Izumo S (2000) Phenotypic char-
acterization of the murine Nkx2.6 homeobox gene by
gene targeting. Mol Cell Biol 20:28742879
Trivedi CM, Zhu W, Wang Q et al (2010) Hopx and
Hdac2 interact to modulate Gata4 acetylation and
embryonic cardiac myocyte proliferation. Dev Cell
19:450459. https://doi.org/10.1016/j.devcel.2010.08.
012
Trivedi CM, Cappola TP, Margulies KB, Epstein JA
(2011) Homeodomain only protein x is down-
regulated in human heart failure. J Mol Cell Cardiol
50:10561058. https://doi.org/10.1021/nl061786n.
Core-Shell
Tu CT, Yang TC, Tsai HJ (2009) Nkx2.7 and Nkx2.5
function redundantly and are required for cardiac mor-
phogenesis of zebrash embryos. PLoS One 4:e4249.
https://doi.org/10.1371/journal.pone.0004249
van der Harst P, van Setten J, Verweij N, Vogler G,
Franke L, Maurano MT, Wang X, Mateo Leach I,
Eijgelsheim M, Sotoodehnia N, Hayward C,
Sorice R, Meirelles O, Lyytikäinen LP, Polašek O,
Tanaka T, Arking DE, Ulivi S, Trompet S et al
(2016) 52 genetic loci inuencing myocardial mass. J
Am Coll Cardiol 68:14351448. https://doi.org/10.
1016/j.jacc.2016.07.729
van Tuyl M, Liu J, Groenman F, Ridsdale R, Han RN,
Venkatesh V, Tibboel D, Post M (2006) Iroquois genes
inuence proximo-distal morphogenesis during rat
lung development. Am J Physiol Lung Cell Mol
Physiol 290:L777L789. https://doi.org/10.1152/
ajplung.00293.2005
van Weerd JH, Christoffels VM (2016) The formation and
function of the cardiac conduction system. Develop-
ment 143:197210. https://doi.org/10.1242/dev.
124883
Vandewalle C, Van Roy F, Berx G (2009) The role of the
ZEB family of transcription factors in development and
disease. Cell Mol Life Sci 66:773787. https://doi.org/
10.1007/s00018-008-8465-8
Wang J, Klysik E, Sood S, Johnson RL, Wehrens XH,
Martin JF (2010) Pitx2 prevents susceptibility to atrial
arrhythmias by inhibiting left-sided pacemaker speci-
cation. Proc Natl Acad Sci U S A 107:97539758.
https://doi.org/10.1073/pnas.0912585107
Wang J, Greene SB, Martin JF (2011) BMP signaling in
congenital heart disease: new developments and future
directions. Birth Defects Res A Clin Mol Teratol
91:441448. https://doi.org/10.1002/bdra.20785
Wang J, Xin YF, Xu WJ, Liu ZM, Qiu XB, Qu XK, Xu L,
Li X, Yang Y (2013) Prevalence and spectrum of
PITX2c mutations associated with congenital heart
disease. DNA Cell Biol 32:708716. https://doi.org/
10.1089/dna.2013.2185
Wang J, Zhang DF, Sun YM, Li RG, Qiu XB, Qu XK,
Liu X, Fang WY, Yang YQ (2014) NKX2-6 mutation
predisposes to familial atrial brillation. Int J Mol Med
Homeobox Genes and Homeodomain Proteins: New Insights into Cardiac...
34:15811590. https://doi.org/10.3892/ijmm.2014.
1971
Wang Y, Li Y, Guo C, Lu Q, Wang W, Jia Z, Chen P,
Ma K, Reinberg D, Zhou C (2016) ISL1 and JMJD3
synergistically control cardiac differentiation of
embryonic stem cells. Nucl Acids Res 44:67416755.
https://doi.org/10.1093/nar/gkw301
Waraya M, Yamashita K, Katoh H, Ooki A, Kawamata H,
Nishimiya HNK, Ema A, Watanabe M (2012) Cancer
specic promoter CpG Islands hypermethylation of
HOP homeobox (HOPX) gene and its potential tumor
suppressive role in pancreatic carcinogenesis. BMC
Cancer 12:397. https://doi.org/10.1186/1471-2407-
12-397
Wei D, Gong XH, Qiu G, Wang J, Yang Y (2014) Novel
PITX2c loss-of-function mutations associated with
complex congenital heart disease. Int J Mol Med
33:12011208. https://doi.org/10.3892/ijmm.2014.
1689
Witzel HR, Jungblut B, Choe CP, Crump JG, Braun T,
Crump JG (2012) The LIM protein Ajuba restricts the
second heart eld progenitor pool by regulating Isl1
activity. Dev Cell 23:5870. https://doi.org/10.1016/j.
devcel.2012.06.005
Wu YH, Zhao H, Zhou LP, Zhao CX, Wu YF, Zhen LX,
Li J, Ge DX, Xu L, Lin L, Liu Y, Liang DD, Chen Y
(2015) miR-134 modulates the proliferation of human
cardiomyocyte progenitor cells by targeting Meis2. Int
J Mol Sci 6224:2519925213. https://doi.org/10.3390/
ijms161025199
Xu H, Baldini A (2007) Genetic pathways to mammalian
heart development: recent progress from manipulation
of the mouse genome. Semin Cell Dev Biol 18:7783.
https://doi.org/10.1016/j.semcdb.2006.11.011
Xu H, Yi Q, Yang C, Wang Y, Tian J, Zhu J (2016)
Histone modications interact with DNA methylation
at the GATA4 promoter during differentiation of mes-
enchymal stem cells into cardiomyocyte-like cells. Cell
Prolif 49:315329. https://doi.org/10.1111/cpr.12253
Yamagishi H, Yamagishi C, Nakagawa O, Harvey RP,
Olson EN, Srivastava D (2001) The combinatorial
activities of Nkx2.5 and dHAND are essential for
cardiac ventricle formation. Dev Biol 239:190203.
https://doi.org/10.1006/dbio.2001.0417
Yap LF, Lai SL, Patmanathan SN et al (2016) HOPX
functions as a tumour suppressor in head and neck
cancer. Sci Rep 6:38758. https://doi.org/10.1038/
srep38758
Yousef MS, Matthews BW (2005) Structural basis of
Prospero-DNA interaction: implications for transcrip-
tion regulation in developing cells. Structure
13:601607. https://doi.org/10.1016/j.str.2005.01.023
Yu X, St Amand TR, Wang S, Li G, Zhang Y, Hu YP,
Nguyen L, Qiu MS, Chen Y (2001) Differential
expression and functional analysis of Pitx2 isoforms
in regulation of heart looping in the chick. Develop-
ment 1013:10051013
Yu Z, Kong J, Pan B, Sun H, Lv T, Zhu J, Huang G, Tian J
(2013) Islet-1 may function as an assistant factor for
histone acetylation and regulation of cardiac
development-related transcription factor Mef2c
expression. PLoS One 8:e77690. https://doi.org/10.
1371/journal.pone.0077690
Zakariyah AF, Rajgara RF, Veinot JP, Skerjanc IS,
Burgon PG (2017) Congenital heart defect causing
mutation in Nkx2.5 displays in vivo functional decit.
J Mol Cell Cardiol 105:8998. https://doi.org/10.1016/
j.yjmcc.2017.03.003
Zhang SS, Kim KH, Rosen A, Smyth JW, Sakuma R,
Delgado-Olguín P, Davis M, Chi NC, Puviindran V,
Gaborit N, Sukonnik T, Wylie JN, Brand-Arzamendi-
K, Farman GP, Kim J, Rose RA, Marsden PA, Zhu Y,
Zhou YQ, Miquerol L, Henkelman RM, Stainier DY,
Shaw RM, Hui CC, Bruneau BG, Backx PH (2011)
Iroquois homeobox gene 3 establishes fast conduction
in the cardiac His-Purkinje network. Proc Natl Acad
Sci U S A 108:1357613581. https://doi.org/10.1073/
pnas.1106911108
Zhang Y, Si Y, Ma N, Mei J (2015) The RNA-binding
protein PCBP2 inhibits Ang II-induced hypertrophy of
cardiomyocytes though promoting GPR56 mRNA
degeneration. Biochem Biophys Res Commun
464:679684. https://doi.org/10.1016/j.bbrc.2015.06.139
Zhang Y, Si Y, Ma N (2016) Meis1 promotes poly (rC)-
binding protein 2 expression and inhibits angiotensin
II-induced cardiomyocyte hypertrophy. IUBMB Life
68:1322. https://doi.org/10.1002/iub.1456
Zhou Z, Wang J, Guo C, Chang W, Zhuang J, Zhu P, Li X
(2017) Temporally distinct Six2-positive second heart
eld progenitors regulate mammalian heart develop-
ment and disease. Cell Rep 18:10191032. https://doi.
org/10.1016/j.celrep.2017.01.002
Zhu CC, Yamada G, Nakamura S, Terashi T,
Schweickert A, Blum M (1998) Malformation of tra-
chea and pelvic region in goosecoid mutant mice. Dev
Dyn 211:374381. https://doi.org/10.1002/(SICI)
1097-0177(199804)211:4<374::AID-AJA8>3.0.
CO;2-E
Zhuang S, Zhang Q, Zhuang T, Evans SM, Liang X, Sun
Y (2013) Expression of Isl1 during mouse develop-
ment. Gene Expr Patterns 13:407412. https://doi.org/
10.1016/j.gep.2013.07.001.Expression
R. Miksiunas et al.
... The region-specific enrichment of genes leads to region-specific congenital defects in case of mutations or dysregulation during cardiac regionalization processes. The IFT development, for example, is dependent on Tbx5, Osr1, Foxf1, Bmp4, Wnt2, and Gata4 [27•]; known ventricle-specific genes are Hey2, Irx4, Kcne1, and Myl2; and atrial markers comprise Nr2f2, Myl7, Gja5, and Pitx2 [28]. It is important to note here that some of the genes are not specific for the mentioned heart region but might mark a particular subpopulation at a given developmental stage but not in other stages. ...
... Additionally, the right atrium signaling is disrupted what impacts the anterior-posterior patterning of the cardiac progenitors. The right ventricle myocardium can be identified based on the expression of Irx4 and Plagl1, while the OFT myocardium is specified by Hand2, Tead2, and Arid3b [3,28]. Mouse Hand2-null embryos displayed dysregulated genes in transcriptomes from the anterior SHF, OFT, and right ventricle before the occurrence of a visible morphological defect [3]. ...
Article
Full-text available
Numerous human syndromes are characterized by congenital heart defects and craniofacial anomalies. This is because the development of the head and heart are closely related. Several studies revealed that specific regions of the myocardium are closer related to specific head muscle groups than to other myocardial regions. The gene regulatory network underlying the specification and differentiation of cardiac and branchiomeric musculature from a common progenitor field, the cardiopharyngeal field, is just starting to get revealed. However, every single new insight into the evolution and development of the head and heart tissues contributes to our understanding of complex human syndromes. Here, I summarize the most recent finding regarding the underlying mechanisms for the proper head and heart musculature development and indicate syndromes, e.g., DiGeorge syndrome and Axenfeld-Rieger syndrome, that might be better understood due to those new insights.
... HOXB7 belongs to a family of transcription factors that can maintain adult tissue homeostasis and regulate growth and differentiation during embryonic development. 34 HOXB7 is of10 abnormally expressed and acts as a tumor promoter via modulating cell behaviors in many cancers, such as gastric, 35 prostate, 36 colorectal, 36 and breast 37 cancers. Previous research showed that HOXB7 upregulation might facilitate CSCC tumor progression. ...
Article
Full-text available
Background Circular RNAs (circRNAs) are often dysregulated in cancers and closely related to cancer progression, including cutaneous squamous cell carcinoma (CSCC). However, the role and mechanism of circ_0068631 in CSCC progression have not been reported. Methods The expression of circ_0068631, microRNA‐139‐5p (miR‐139‐5p), and homeobox B7 (HOXB7) was measured by real‐time quantitative polymerase chain reaction (RT‐qPCR). Cell counting kit‐8 (CCK‐8) assay, 5‐ethynyl‐2′‐deoxyuridine (EdU) assay, and colony formation assay were used to measure cell proliferation. Cell apoptosis was assessed by flow cytometry. Cell migration was detected by transwell assay. The interaction between miR‐139‐5p and circ_0068631 or HOXB7 was confirmed by dual‐luciferase reporter assay. A xenograft tumor model was established to confirm the function of circ_0068631 in vivo. Results Circ_0068631 was upregulated in CSCC tissues and cells, and its silencing could inhibit CSCC cell proliferation and metastasis while promoting apoptosis in vitro, as well as restrain CSCC tumor growth in vivo. Circ_0068631 acted as a sponge of miR‐139‐5p, and miR‐139‐5p inhibition reversed the repressive effect of circ_0068631 knockdown on CSCC cell progression. Furthermore, HOXB7 was a target of miR‐139‐5p, and miR‐139‐5p inhibited the malignant behaviors by downregulating HOXB7 expression in CSCC cells. Further, circ_0068631 sponged miR‐139‐5p to regulate HOXB7 expression. Conclusion Circ_0068631 functioned as a novel oncogene in CSCC progression by regulating miR‐139‐5p/HOXB7 axis, suggesting that circ_0068631 may be a potential target for CSCC treatment. Highlights Circ_0068631 was overexpressed in CSCC tissues and cells. Circ_0068631 downregulation suppressed CSCC progression via miR‐139‐5p. Circ_0068631 regulated HOXB7 via sponging miR‐139‐5p.
... Appropriately inhibiting the expression of AP-1 can effectively alleviate the cardiovascular toxicity caused by various factors, such as intimal hyperplasia, myocardial hypertrophy, hypertension, and myocardial fibrosis [37][38][39]. Homeobox proteins are a group of phylogenetically conserved transcription factors that regulate tissue growth and development, and homeobox genes are the main genes that control development and play a key role in the regulation of animal organogenesis and cell differentiation [40,41]. There is no Meox2 expression in the early stage of heart development and only in the relatively late stage of heart development; that is, when the proliferation of cardiac cells begins to decline, Meox2 begins to be expressed [42]. ...
Article
Full-text available
Pirarubicin (THP) is one of the classic chemotherapy drugs for cancer treatment. It is often clinically limited because of its cardiotoxicity. The occurrence and development of THP-mediated chemotherapy-related cardiotoxicity (CRC) may be reversed by RING finger protein 10 (RNF10). This study was performed with the aim of evaluating the inhibitory effect of RNF10 on THP-mediated CRC and its molecular mechanism. In vivo, we found that the expression of RNF10 decreased in THP-induced CRC rats, accompanied by Meox2 inhibition and AP-1 activation, resulting in increased cardiomyocyte apoptosis. After small interfering RNA (siRNA) and lentivirus transfection (Lv) of RNF10 in vitro, the expression of RNF10, Meox2, and AP-1 proteins and the degree of cardiomyocyte apoptosis were detected. We found that overexpression of RNF10 in H9C2 cardiomyocytes significantly promoted Meox2 and inhibited AP-1, alleviated apoptosis, and showed further inhibitory activity on THP-induced cardiomyocyte toxicity. Silencing RNF10 showed the opposite result. Our study showed that RNF10 inhibited THP-induced CRC through the activity of Meox2 and AP-1 proteins. RNF10 may be the next drug target for the treatment of CRC and other related cardiovascular diseases.
... Following the Hugo gene nomenclature committee, the deleted segment encompassed 36 genes, 19 of whom were referenced in the OMIM database, and among which only IGF1R, NR2F2, CHD2 and MEF2A, were consistent with the described patient's phenotype [55,56]. Following the authors' opinion, 15q26 monosomy should be taken in consideration in the differential diagnosis process when growth retardation is associated with congenital heart defect, mainly atrioventricular septal defects and /or aortic arch anomaly [57]. ...
Article
Full-text available
In this review, we report the developments across pediatric subspecialties that have been published in the Italian Journal of Pediatrics in 2021. We highlight advances in allergy and immunology, critical care, endocrinology, gastroenterology, genetics, hematology, infectious diseases, neonatology, neurology, nutrition, palliative care, respiratory tract illnesses and telemedicine.
... PDE_07199 amino acid sequence analysis using the Simple Modular Architecture Research Tool (SMART) (Letunic et al., 2021) showed the presence of a homeodomain. Homeodomaincontaining proteins have been proved to function as transcription factors and play important roles in animals, fungi, and plants during some developmental processes, such as development and differentiation (Burglin, 2011;Miksiunas et al., 2020). In fungi, homeodomain transcriptional factors (Htfs) play a crucial role in regulating developmental processes. ...
Article
Full-text available
Homeodomain-containing transcription factors (Htfs) play important roles in animals, fungi, and plants during some developmental processes. Here, a homeodomain-containing transcription factor PoHtf1 was functionally characterized in the cellulase-producing fungi Penicillium oxalicum 114-2. PoHtf1 was shown to participate in colony growth and conidiation through regulating the expression of its downstream transcription factor BrlA, the key regulator of conidiation in P. oxalicum 114-2. Additionally, PoHtf1 inhibited the expression of the major cellulase genes by coordinated regulation of cellulolytic regulators CreA, AmyR, ClrB, and XlnR. Furthermore, transcriptome analysis showed that PoHtf1 participated in the secondary metabolism including the pathway synthesizing conidial yellow pigment. These data show that PoHtf1 mediates the complex transcriptional-regulatory network cascade between developmental processes and cellulolytic gene expression in P. oxalicum 114-2. Our results should assist the development of strategies for the metabolic engineering of mutants for applications in the enzymatic hydrolysis for biochemical production.
Article
Placental insufficiency is one of the major causes of fetal growth restriction (FGR), a significant pregnancy disorder in which the fetus fails to achieve its full growth potential in utero. As well as the acute consequences of being born too small, affected offspring are at increased risk of cardiovascular disease, diabetes and other chronic diseases in later life. The placenta and heart develop concurrently, therefore placental maldevelopment and function in FGR may have profound effect on the growth and differentiation of many organ systems, including the heart. Hence, understanding the key molecular players that are synergistically linked in the development of the placenta and heart is critical. This review highlights the key growth factors, angiogenic molecules and transcription factors that are common causes of defective placental and cardiovascular development.
Article
Full-text available
Homeobox transcription factors are key regulators of morphogenesis and development in both animals and plants. In plants, the WUSCHEL-related homeobox (WOX) family of transcription factors function as central organizers of several developmental programs ranging from embryo patterning to meristematic stem-cell maintenance through transcriptional activation and repression mechanisms. The Medicago truncatula STENOFOLIA ( STF ) gene is a master regulator of leaf-blade lateral development. Here, the crystal structure of the homeodomain (HD) of STF (STF-HD) in complex with its promoter DNA is reported at 2.1 Å resolution. STF-HD binds DNA as a tetramer, enclosing nearly the entire bound DNA surface. The STF-HD tetramer is partially stabilized by docking of the C-terminal tail of one protomer onto a conserved hydrophobic surface on the head of another protomer in a head-to-tail manner. STF-HD specifically binds TGA motifs, although the promoter sequence also contains TAAT motifs. Helix α3 not only serves a canonical role as a base reader in the major groove, but also provides DNA binding in the minor groove through basic residues located at its C-terminus. The structural and functional data in planta reported here provide new insights into the DNA-binding mechanisms of plant-specific HDs from the WOX family of transcription factors.
Article
Full-text available
Circular ribonucleic acids (circRNAs) serve a vital role in the pathological processes of a number of diseases. Previous microarray results of circRNA expression revealed that hsa_circ_0070933 and hsa_circ_0070934, two circRNAs associated with the La ribonucleoprotein 1B gene, were highly expressed in cutaneous squamous cell carcinoma (CSCC). The present study aimed to explore the specific role of these circRNAs in CSCC. Through reverse transcription‑quantitative PCR, hsa_circ_0070933 and hsa_circ_0070934 expression levels in CSCC cell lines and a human keratinocyte cell line were detected. Additionally, direct interactions between miR‑1236‑3p and HOXB7 or circ‑0070934 were identified using RNA binding protein immunoprecipitation assays and dual‑luciferase reporter assays. Cell Counting Kit‑8, 5‑ethynyl‑2'‑deoxyuridine, Transwell invasion and flow cytometry assays were used to assess the roles of miR‑1236‑3p or circ‑0070934 in cell invasion, proliferation and apoptosis. Subsequently, in vivo tumor formation assays were used to verify the role of circ‑0070934 in CSCC. The results demonstrated that the expression of circ‑0070934 was stably upregulated in a number of CSCC cell lines compared with that in normal human keratinocytes. Knockdown of circ‑0070934 inhibited the invasive and proliferative potential of CSCC cells and promoted apoptosis both in vivo and in vitro. In addition, circ‑0070934 modulated HOXB7 expression through competitive binding with miR‑1236‑3p. In conclusion, the results of the present study demonstrated the effects of the circ‑0070934/miR‑1236‑3p/HOXB7 regulatory axis on CSCC and provided a novel insight for the pathogenesis of CSCC.
Article
Full-text available
Congenital heart defects can be caused by mutations in genes that guide cardiac lineage formation. Here, we show deletion of NKX2-5, a critical component of the cardiac gene regulatory network, in human embryonic stem cells (hESCs), results in impaired cardiomyogenesis, failure to activate VCAM1 and to downregulate the progenitor marker PDGFRα. Furthermore, NKX2-5 null cardiomyocytes have abnormal physiology, with asynchronous contractions and altered action potentials. Molecular profiling and genetic rescue experiments demonstrate that the bHLH protein HEY2 is a key mediator of NKX2-5 function during human cardiomyogenesis. These findings identify HEY2 as a novel component of the NKX2-5 cardiac transcriptional network, providing tangible evidence that hESC models can decipher the complex pathways that regulate early stage human heart development. These data provide a human context for the evaluation of pathogenic mutations in congenital heart disease.
Article
Full-text available
Cardiac development is a complex morphogenetic process initiated as bilateral cardiogenic mesoderm is specified at both sides of the gastrulating embryo. Soon thereafter, these cardiogenic cells fuse at the embryonic midline configuring a symmetrical linear cardiac tube. Left/right bilateral asymmetry is first detected in the forming heart as the cardiac tube bends to the right, and subsequently, atrial and ventricular chambers develop. Molecular signals emanating from the node confer distinct left/right signalling pathways that ultimately lead to activation of the homeobox transcription factor Pitx2 in the left side of distinct embryonic organ anlagen, including the developing heart. Asymmetric expression of Pitx2 has therefore been reported during different cardiac developmental stages, and genetic deletion of Pitx2 provided evidence of key regulatory roles of this transcription factor during cardiogenesis and thus congenital heart diseases. More recently, impaired Pitx2 function has also been linked to arrhythmogenic processes, providing novel roles in the adult heart. In this manuscript, we provide a state-of-the-art review of the fundamental roles of Pitx2 during cardiogenesis, arrhythmogenesis and its contribution to congenital heart diseases.
Article
Full-text available
The embryonic process of forming a complex structure such as the heart remains poorly understood. Here, we show that Six2 marks a dynamic subset of second heart field progenitors. Six2-positive (Six2⁺) progenitors are rapidly recruited and assigned, and their descendants are allocated successively to regions of the heart from the right ventricle (RV) to the pulmonary trunk. Global ablation of Six2⁺ progenitors resulted in RV hypoplasia and pulmonary atresia. An early stage-specific ablation of a small subset of Six2⁺ progenitors did not cause any apparent structural defect at birth but rather resulted in adult-onset cardiac hypertrophy and dysfunction. Furthermore, Six2 expression depends in part on Shh signaling, and Shh deletion resulted in severe deficiency of Six2⁺ progenitors. Collectively, these findings unveil the chronological features of cardiogenesis, in which the mammalian heart is built sequentially by temporally distinct populations of cardiac progenitors, and provide insights into late-onset congenital heart disease.
Article
Full-text available
Head and neck squamous cell carcinoma (HNSCC) is generalized term that encompasses a diverse group of cancers that includes tumours of the oral cavity (OSCC), oropharynx (OPSCC) and nasopharynx (NPC). Genetic alterations that are common to all HNSCC types are likely to be important for squamous carcinogenesis. In this study, we have investigated the role of the homeodomain-only homeobox gene, HOPX, in the pathogenesis of HNSCC. We show that HOPX mRNA levels are reduced in OSCC and NPC cell lines and tissues and there is a general reduction of HOPX protein expression in these tumours and OPSCCs. HOPX promoter methylation was observed in a subset of HNSCCs and was associated with a worse overall survival in HPV negative tumours. RNAseq analysis of OSCC cells transfected with HOPX revealed a widespread deregulation of the transcription of genes related to epithelial homeostasis and ectopic over-expression of HOPX in OSCC and NPC cells inhibited cell proliferation, plating efficiency and migration, and enhanced sensitivity to UVA-induced apoptosis. Our results demonstrate that HOPX functions as a tumour suppressor in HNSCC and suggest a central role for HOPX in suppressing epithelial carcinogenesis.
Article
Aims: Pathological hypertrophy is the result of gene network regulation, which ultimately leads to adverse cardiac remodelling and heart failure (HF), and is accompanied by the reactivation of a 'foetal gene programme'. The Mesenchyme homeobox 1 (Meox1) gene is one of the foetal programme genes. Meox1 may play a role in embryonic development, but its regulation of pathological hypertrophy is not known. Therefore, this study investigated the effect of Meox1 on pathological hypertrophy, including familial and pressure overload-induced hypertrophy, and its potential mechanism of action. Methods and results: Meox1 expression was markedly down-regulated in the wild-type adult mouse heart with age, and expression was up-regulated in heart tissues from familial dilated cardiomyopathy (FDCM) mice of the cTnTR141W strain, familial hypertrophic cardiomyopathy (FHCM) mice of the cTnTR92Q strain, pressure overload-induced HF mice, and hypertrophic cardiomyopathy (HCM) patients. Echocardiography, histopathology, and hypertrophic molecular markers consistently demonstrated that Meox1 overexpression exacerbated the phenotypes in FHCM and in mice with thoracic aorta constriction (TAC), and that Meox1 knockdown improved the pathological changes. Gata4 was identified as a potential downstream target of Meox1 using digital gene expression (DGE) profiling, real-time PCR, and bioinformatics analysis. Promoter activity data and chromatin immunoprecipitation (ChIP) and Gata4 knockdown analyses indicated that Meox1 acted via activation of Gata4 transcription. Conclusion: Meox1 accelerated decompensation via the downstream target Gata4, at least in part directly. Meox1 and other foetal programme genes form a highly interconnected network, which offers multiple therapeutic entry points to dampen the aberrant expression of foetal genes and pathological hypertrophy.
Article
We report the clinical and molecular cytogenetic characterization of four unrelated patients from France and Spain, carrying 2p14 microdeletions and presenting with intellectual disability and dysmorphisms. 2p14 microdeletions are very rare. Seven patients have been reported so far harboring deletions including 2p14p15 and encompassing OTX1, whose haploinsufficiency is frequently associated with genitourinary defects. To date, only one patient has been reported carrying a more proximal 2p14 microdeletion which does not include OTX1. Here, we report three further patients carrying proximal 2p14 microdeletions not including OTX1 and one patient carrying a more distal 2p14p15 microdeletion including this gene, providing new insights into the associated phenotypic spectrum. First, our study and a review of the literature showed that 3/4 patients carrying proximal 2p14 microdeletions had sensorineural hearing loss, suggesting the presence of a previously unreported deafness-causing gene in this chromosomal region. Second, one patient developed a progressive cardiomyopathy, suggesting that a cardiac follow-up should be systematically warranted even in the absence of congenital heart disease. We speculate that ACTR2 and MEIS1 might respectively play a role in the pathogenesis of the observed deafness and cardiomyopathy. Third, we observed other previously unreported features such as glaucoma, retinopathy, and mild midline abnormalities including short corpus callosum, hypospadias and anteriorly placed anus. Finally, the patient carrying a 2p14p15 deletion including OTX1 had normal kidneys and genitalia, thus confirming that OTX1 haploinsufficiency is not invariably associated with genitourinary defects. In conclusion, our study contributes significantly to delineate the phenotypic spectrum of 2p14 microdeletions.
Article
Genetic variation in specific transcription factors during heart formation may lead to congenital heart disease (CHD) or even miscarriage. The aim of the present study was to identify CHD?associated genes using next generation sequencing (NGS). The whole exome DNA sequence was obtained from a stillborn fetus diagnosed with tricuspid atresia and complete transposition of the great arteries using high?throughput sequencing methods. Subsequently, genetic variants of CHD?associated genes were selected and verified in 215 non?syndromic CHD patients and 249 healthy control subjects using polymerase chain reaction combined with Sanger sequencing. Genetic variants of previously reported CHD?inducing genes, such as cysteine rich with EGF like domains 1 and cbp/p300?interacting transactivator with Glu/Asp rich carboxy?terminal domain 2, were discovered through the NGS analysis. In addition, a novel non?synonymous mutation of the iroquois homeobox 1 (IRX1) gene (p.Gln240Glu) was identified. A total of three non?synonymous mutations (p.Gln240Glu, p.Ser298Asn and p.Ala381Glu) of the IRX1 gene were verified in 215 non?syndromic CHD patients, but not in 249 healthy volunteers. The results demonstrated that NGS is a powerful tool to study the etiology of CHD. In addition, the results suggest that genetic variants of the IRX1 gene may contribute to the pathogenesis of CHD.
Article
The Nkx2.5 gene encodes a transcription factor that plays a critical role in heart development. In humans, heterozygous mutations in NKX2.5 result in congenital heart defects (CHDs). However, the molecular mechanisms by which these mutations cause the disease remain unknown. NKX2.5-R142C is a mutation that was reported to be associated with atrial septal defect (ASD) and atrioventricular (AV) block in 13-patients from one family. The R142C mutation is located within both the DNA-binding domain and the nuclear localization sequence of NKX2.5 protein. The pathogenesis of CHDs in humans with R142C point mutation is not well understood. To examine the functional deficit associated with this mutation in vivo, we generated and characterized a knock-in mouse that harbours the human mutation R142C. Systematic structural and functional examination of the embryonic, newborn, and adult mice revealed that the homozygous embryos Nkx2.5R141C/R141C are developmentally arrested around E10.5 with delayed heart morphogenesis and downregulation of Nkx2.5 target genes, Anf, Mlc2v, Actc1 and Cx40. Histological examination of Nkx2.5R141C/+ newborn hearts showed that 36% displayed ASD, with at least 80% 0f adult heterozygotes displaying a septal defect. Moreover, heterozygous Nkx2.5R141C/+ newborn mice have downregulation of ion channel genes with 11/12 adult mice manifesting a prolonged PR interval that is indicative of 1st degree AV block. Collectively, the present study demonstrates that mice with the R141C point mutation in the Nkx2.5 allele phenocopies humans with the NKX2.5 R142C point mutation.
Article
Purpose: Mowat-Wilson syndrome (MWS) is a genetic disease characterized by distinctive facial features, moderate to severe intellectual disability, and congenital malformations, including Hirschsprung disease, genital and eye anomalies, and congenital heart defects, caused by haploinsufficiency of the ZEB2 gene. To date, no characteristic pattern of brain dysmorphology in MWS has been defined. Methods: Through brain magnetic resonance imaging (MRI) analysis, we delineated a neuroimaging phenotype in 54 MWS patients with a proven ZEB2 defect, compared it with the features identified in a thorough review of published cases, and evaluated genotype-phenotype correlations. Results: Ninety-six percent of patients had abnormal MRI results. The most common features were anomalies of corpus callosum (79.6% of cases), hippocampal abnormalities (77.8%), enlargement of cerebral ventricles (68.5%), and white matter abnormalities (reduction of thickness 40.7%, localized signal alterations 22.2%). Other consistent findings were large basal ganglia, cortical, and cerebellar malformations. Most features were underrepresented in the literature. We also found ZEB2 variations leading to synthesis of a defective protein to be favorable for psychomotor development and some epilepsy features but also associated with corpus callosum agenesis. Conclusion: This study delineated the spectrum of brain anomalies in MWS and provided new insights into the role of ZEB2 in neurodevelopment.Genet Med advance online publication 10 November 2016Genetics in Medicine (2016); doi:10.1038/gim.2016.176.