Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

download Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

of 7

Transcript of Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    1/14

    Cardiogenesis and the Regulation of Cardiac-Specific

    Gene Expression

    Jau-Nian Chen, PhDa , Douglas B. Cowan, PhD b, John D. Mably, PhDc,*

    a University of California Los Angeles, Los Angeles, CA, USA b

    Children’s Hospital Boston, Boston, MA, USAc Massachusetts General Hospital, Boston, MA, USA

    Development and maturation of the embryonic

    vertebrate heart is an exquisitely conducted ensemble

    of cell movements, interactions, and morphologic

    transformations. The genetic orchestration of these

    events is precise and can be reduced to dissonance by

    the loss of a single component in this process. Proper 

    heart morphogenesis is important to normal embry-

    onic development because the heart is the first organ

    formed. All subsequent events depend on the ability

    of the heart to supply oxygen and nutrients to fulfill

    the metabolic requirements of the organism. Abnor-

    malities in the formation of the heart often lead to

    abnormal function and embryonic lethality or may

    manifest later in life, causing severe health issues.

    Cardiac defects are among the most common birth

    defects, estimated at an incidence of 6 in 1000 live

     births, with an even higher frequency in spontane-

    ously aborted pregnancies   [1].   Much of our under-

    standing of the mechanisms and pathways regulating

    cardiogenesis evolved from studies in model systems,

    notably the mouse, chick, fly, frog, and zebrafish.

    This article outlines the molecular events and mecha-nisms regulating heart formation, focusing on

    recently identified members of the cardiogenic reper-

    toire [2–5].

    Early morphogenesis

    Adult vertebrate hearts may vary greatly in their 

    overall structures, but the morphogenic processes that 

    shape the embryonic hearts of vertebrate species are

    shared. In brief, vertebrate heart formation begins

    when a bilaterally symmetric population of meso-

    dermal cells in the anterior lateral plate becomes

    committed to a cardiac fate in response to inductive

    signals from the adjacent endoderm [6]. Migration of 

    these cardiogenic cells to the midline of the embryo

    results in the formation of a linear heart tube (the

     primitive heart tube). Cardiomyocytes adopt an atrial

    or a ventricular cell fate during differentiation of the

     primitive heart tube along the anterior-posterior axis,

    although studies in zebrafish suggest this specifica-

    tion occurs earlier, prior even to heart tube formation

    [7].  The atrium and ventricle then undergo a right-

    ward looping (cardiac looping) that is essential for 

    alignment of the inflow and outflow tracts and for 

    orienting the atrial and ventricular chambers [8]. Data

    accumulated from studies over the last decadesuggest that in addition to these morphogenic events,

    the genetic circuits critical for heart development also

    are conserved across species [9].

    This complex crosstalk between tissues is essen-

    tial for cardiogenesis, in part contingent on the ex-

     pression of an extensive assemblage of transcription

    factors. Through precise temporal and spatial regu-

    lation, the expression of these factors leads uncom-

    mitted cells to enter the cardiac lineage. In the fruit 

    fly,   Drosophila melanogaster , the activity of the

    homeobox gene tinman  is required for the formation

    1551-7136/05/$ – see front matter  D 2005 Elsevier Inc. All rights reserved.

    doi:10.1016/j.hfc.2005.03.002   heartfailure.theclinics.com

    * Corresponding author. Cardiovascular Research Cen-

    ter, Massachusetts General Hospital and the Department 

    of Medicine, Harvard Medical School, 149 13th Street,

    Boston, MA 02129.

     E-mail address:   [email protected] (J.D. Mably).

    Heart Failure Clin 1 (2005) 157 – 170

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    2/14

    of the primitive heart. Loss of function of   tinman

     blocks the formation of cardiac tissues in developing

    fly embryos [10,11]. To confine tinman  expression in

    the cardiac lineage and to maintain its expression at 

    critical level requires signals from adjacent tissues.

    Endoderm-derived   dpp   expression (the   Drosophilaortholog of the bone morphogenic protein [bmp]

    family) promotes formation of the cardiac lineage

    [12],   whereas   wingless   proteins (orthologs of the

    vertebrate   wnt   proteins) have   positive and negative

    roles in this process   [13,14].   In vertebrates, this

     pathway is conserved, as shown in the chick, where

     BMP2  can induce  Nkx2.5  (the vertebrate ortholog of 

    tinman) in the anterior mesoderm. Studies in chick 

    and frog have revealed further that  wnt3a  and wnt8c,

    signaling through the canonical   wnt   pathway, sup-

     press the cardiac lineage (and promote the differen-tiation of hematopoeitic lineage)   [15–17],   whereas

    wnt11   promotes cardiac differentiation through the

    noncanonical  wnt -signaling pathway [18].

    In vertebrates,   Nkx2.5   is expressed in cardiac

    muscle cells from the onset of embryonic heart 

    formation until adulthood. Although overexpression

    of  Nkx2.5  induces an expansion of cardiac tissues in

    the frog and zebrafish  [19,20], the role of   Nkx2.5   in

    establishment of cardiac lineage in vertebrates is not 

    as clear as the role of  tinman  in the fly. The fact that 

    mice lacking   Nkx2.5   develop a heart suggests that 

     Nkx2.5  is dispensable for establishment of the cardiaclineage   [21].   Although it is possible that   Nkx2.5

    serves a different function than  tinman  in the mouse,

    it is more likely that other genes have redundant 

    functions in the specification of cardiac cell fate. This

    notion is supported by the observations that other 

     Nkx genes, such as  Nkx2.3,  Nkx2.6 , and  Nkx2.7 , are

    expressed in cardiac tissues and that overexpression

    of dominant negative forms of either   Nkx2.5   and

     Nkx2.3  in the frog blocks cardiogenesis  [22,23].

    In contrast to skeletal muscle, where a single

    transcription factor, MyoD, is sufficient to activate thefull program of muscle differentiation, tinman/  Nkx2.5

    cooperates with other transcription factors to com-

     plete the program of cardiac gene expression. For 

    example,  Nkx2.5  and  GATA4  have synergistic effects

    on the activation of downstream cardiac genes in fly

    embryos and in cultured mammalian cells   [24–26].

    In addition, myocardin and serum response factor 

    (SRF) cooperate with   Nkx2.5   in controlling the

    expression of muscle structure genes [27].

     Migration of the bilateral cardiac precursors

    In fly and vertebrates, the cardiac precursors

    initially are positioned bilaterally in the embryo. As

    development proceeds, the bilaterally positioned car-

    diac mesoderm migrates to and fuses at the midline to

    form the primitive heart tube. In the fly,  fgf   signaling

    has an instrumental role in guiding cardiac precursors

    to the midline   [28–30].   Although the role of   fgf  

    in the migration of precardiac cells to the midlineneeds to be explored further in the vertebrate, genetic

    studies in the zebrafish demonstrate that   Fgf8   is

    expressed in and required for development of the

    zebrafish heart precursors,   especially at the onset of 

    cardiac gene expression   [31].   Because studies in

    zebrafish have been instrumental in defining many

    features of early cardiogenesis, this article discusses

    zebrafish heart development and several of the

    mutants identified from the large-scale ethyl nitro-

    sourea mutagenesis screens.

    The zebrafish heart is a primitive vertebrate heart consisting of two major cell types (myocardium and

    endocardium) and two chambers (atrium and ven-

    tricle), and its development is fast compared with

    other vertebrate model organisms. The bilateral

    cardiac primordia visualized at 14 somites (approx-

    imately 16 hours postfertilization [hpf]) by  Nkx2.5   in

    situ staining (Fig. 1)   fuse at the midline in the

    zebrafish by 20 somites (19 hpf). The fused heart 

    soon grows into a long tubular structure, known as

    the primitive heart tube, and can ensure circulation

    throughout the body by 24 hpf. Within 2 days of de-

    velopment, cardiomyocytes assume chamber-specificfates and cardiac looping is completed, placing the

    ventricle to the right of the atrium  [32].

    The mechanisms underlying the fusion of the

    cardiac mesoderm have been well described through

    analysis of zebrafish mutations that block this pro-

    cess and lead to the development of two hearts in

    the affected embryos, a phenotype known as cardia

     bifida   [33–35].   Positional cloning of the   natter 

    (nat ) and   miles apart   (mil ) mutations demonstrated

    that  fibronectin   (the gene defective in   nat )   [36]  and

     sphingosine-1-phosphate receptor    (mil )   [37]   areindispensable for the fusion of cardiac mesoderm

    and revealed a significant role for the extracellular 

    matrix (ECM) in precardiac cell migration. The

    essential role of endodermal signals in the migration

    of the cardiac precursors was revealed through the

    identification of the gene defects associated with the

    bonnie and clyde   (bon),   casanova   (cas),   hands off   

    (han), and   faust   ( fau) mutations.  bon  encodes a Mix

    family homeobox gene that regulates the generation

    of endoderm precursors [38], whereas  cas  encodes a

     sox -related protein that is sufficient to induce

    endoderm differentiation [39]. The  fau  gene encodesthe zebrafish homolog of  gata5 [40] and  han  encodes

    dHAND  ( Hand2) [41], each of which is expressed in

    chen  et al158

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    3/14

    endoderm and cardiomyocytes. Loss of function of bon,   cas,   han,   and   fau   not only prevents cardiac

     precursors from fusing at the midline, but also affects

    the number and the differentiation of cardiac pre-

    cursors, indicating an instrumental role of endoderm

    in early cardiac patterning   [40,42].   Similarly, mice

    lacking   GATA4  exhibit cardia bifida in addition to a

    reduction of cardiomyocytes  [43,44].

    The association of cardia bifida with defects in

    cardiac patterning and cardiomyocyte differentiation

    had led to a model in which the fusion of the bilateral

    cardiac primordia at the midline is essential for subsequent cardiac development and morphogenesis.

    This notion has been challenged recently by a study

    on mice lacking   Foxp4, however   [45].   Foxp4   is a

    member of the Fox gene family whose expression is

    abundant in the anterior foregut endoderm but cannot 

     be detected in the cardiomyoctyes.   Foxp4   mutants

    exhibit cell death–mediated loss of anterior foregut 

    mesoderm and cardia bifida, but each cardiac

     primordium successfully differentiates into a four-

    chambered heart with proper alignment according to

    the left-right axis. Thus, although many factors that 

    regulate fusion of the cardiac primordia also playessential roles in the maturation of the heart, the

     processes are not inseparable. These results hint at a

    greater level of preprogramming in the bilateral precursors than previously appreciated.

     Heart tube formation

    In contrast to the extensive studies on early

    cardiac patterning, molecular and cellular mecha-

    nisms involved in the morphogenesis of the primi-

    tive heart tube, which occurs immediately after the

     bilateral cardiac primordia fuse at the midline, are

    largely unknown. The survival of mammalian em-

     bryos depends on proper cardiac function for circu-lation. Therefore, mutations affecting early cardiac

     patterning decisions such as primitive heart tube

    morphogenesis often cause embryonic lethality at 

    early developmental stages, making such studies

    difficult in traditional models such as the mouse.

    Another attribute of the zebrafish that has facilitated

    such studies is the ability of embryos to survive

    through early development without an active circu-

    lation. In particular, two zebrafish mutations affect-

    ing primitive heart formation,   heart and soul   (has)

    [46,47]   and   heart and mind   (had )   [48],   provide

     points of entry to understanding the molecular andcellular mechanisms guiding primitive heart tube

    morphogenesis. In   has   and   had   mutants, the bi-

    Fig. 1. Expression pattern of  Nkx2.5  defines the heart field. ( A) Wholemount in situ analysis of  Nkx2.5  expression in zebrafish

    embryos stains the two bilateral heart fields at 14 somites (approximately 16 hpf ), indicated by the two arrowheads. Arrow

    indicates midline expression of no tail (ntl , zebrafish T brachyury homolog)  [149]. ( B) By 20 somites (approximately 19 hpf)

    these two populations of cells fuse at the midline (arrowhead ). Arrow indicates staining by midline marker  ntl . (C ) The fused

    heart field develops into the primitive heart tube by 24 hpf, highlighted by  Nkx2.5  in situ staining, and the embryonic onset of 

    circulation begins with contraction of the heart at this time. ( D) By 2 days of development, cardiomyocytes have assumed

    chamber-specific fates and cardiac looping is complet ed, placing the ventricle to the right of the atrium [32]. Both chambers are

    visible by staining with the myosin antibody MF20  [7].

    cardiogenesis   &   gene expression   159

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    4/14

    lateral myocardial precursors migrate to the midline

    normally, but myocardial precursors of both mutants

    remain at the midline as a clump of cells. As a

    result, the   had   mutants develop a small heart with

    severe defects in the differentiation and contractility

    of cardiomyocytes  [48], and the   has   mutants have amispatterned heart with the ventricular   cells encom-

     passed by a single layer of atrial cells   [46,47].

    Identification of the genetic lesions responsible

    for these defects has revealed roles for the genes

    encoding atypical protein kinase C  l  (aPKC  l,  has)

    [46,47] and the a1B1 isoform of Na,K-ATPase (had )

    [48].   The identification of these genes in the

    regulation of primitive heart tube formation suggests

    that changes in myocardial cellular architecture

    during heart development can have striking effects

    on this process. aPKC   l   and Na,K-ATPase are po-larized proteins. In the chick, Na,K-ATPase   a1 as-

    sumes a lateral position in precardiomyocytes   [49],

    and mislocalization of Na,K-ATPase has been asso-

    ciated with Type 4 Long-QT cardiac arrhythmia in

    humans   [50],   indicating the importance of Na,K-

    ATPase polarity in maintaining cardiac function. On

    the other hand, aPKC is part of the evolutionarily

    conserved Par3/Par6/aPKC protein complex, a key

     player in the establishment of cell polarity. In the

    zebrafish, the medial myocardial precursors transform

    from a cuboidal to columnar shape upon the fusion of 

    the bilateral primordia, and this change in cell shapeis accompanied by the transition of aPKC cellular 

    localization from cell-cell contacts to the apicolateral

    domain [36]. Overexpression of a dominant negative

    form of aPKC in MDCK cells abolishes polarized

    distribution of Na,K-ATPase  [51]   and prevents the

    initial spotlike adherens junction complexes from

    maturing into beltlike adherens junctions   [52,53].

    These data suggest that aPKC and Na,K-ATPase may

    function in a common pathway for the establishment 

    or maintenance of cell polarity.

    Chamber maturation

    Growth of the myocardium

    The regulation of normal myocardial development 

    is exquisitely sensitive and can be disrupted by

    mutations within many different genes   [2]. The

    growth and thickening of the ventricular myocardium

    is essential to vertebrate development because of the

    vigorous contraction required to maintain blood

    circulation and pressure. The control of myocardialdevelopment continues to be elucidated through

    analyses of gene disruption in model systems such

    as fly, zebrafish, and mouse. The serum response

    factor (SRF) is one protein that has been implicated

    as a controller of multiple signaling pathways essen-

    tial for cardiac development, including the regulation

    of immediate-early   response genes and muscle-

    specific genes   [54].   Generation of a conditionalmouse transgenic mutant that deleted the cardiac-

    specific expression of SRF resulted in embryonic

    lethality associated with an abnormally thin myocar-

    dium and dilated chambers, as well as a loss of nor-

    mal trabeculation. In addition, several genes essential

    for normal heart development also were downregu-

    lated, suggesting a role for SRF as a global regulator 

    essential for cardiac maturation. Similarly, inactiva-

    tion of the homeodomain  Hop  in mice and zebrafish,

    which physically interacts with SRF and inhibits its

    activity, results in a thin ventricular myocardium withfewer cells than normal [55,56].

    The role of the ligand neuregulin and its co-

    receptors ErbB2/ErbB4 in the trabeculation of the

    ventricle was revealed through knockout studies in

    mice   [57–59].   Loss of any of these three pathway

    members resulted in embryonic lethality between E10

    and E12. All were associated with a failure of the

    ventricle to develop normally, although atrial devel-

    opment was not affected severely   [57–59].   The

    endocardial cushions also were reduced, with fewer 

    mesenchymal cells forming these valvular prescursor 

    structures   [57–59].   Although ErbB2 and ErbB4 areexpressed on the myocardial surface, their ligand,

    neuregulin, is expressed in the endocardium, impli-

    cating crosstalk between these cell types for myo-

    cardial development. In addition, the clustering of 

    endocardial-derived mesenchymal cells at the sites of 

    valve formation was disrupted by loss of the two myo-

    cardial receptors ErbB2/4. These results established

    the paradigm for signaling between the adjacent 

    endocardial and myocardial layers of the heart.

    Another aspect of ventricular development that 

    has received less attention is the orientation of cellgrowth in a concentric direction to produce a thick,

    multicell-layered myocardium. This type of oriented

    cell growth may contribute to normal chamber 

    formation by thickening at appropriate positions

    along the axis of the early heart tube. Studies in

    zebrafish [60] and mouse [61] have revealed roles for 

    this process in normal heart morphogenesis. Charac-

    terization of a zebrafish mutant with a single-cell– 

    layered myocardium has revealed a novel endocardial

    transmembrane protein, heart of glass  (heg ), which is

    essential for the concentric thickening of the ven-

    tricle. Although the hearts of these mutants developwith the normal complement of myocardial cells, they

    are not added along the axis from the endocardium

    chen  et al160

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    5/14

    to myocardium, but instead spread over the circum-

    ference of the hear t, resulting in two massive cham-

     bers (Fig. 2)   [60].   Reminiscent of the neuregulin/ 

    ErbB2/ErbB4 mice knockouts, loss of the endocardial

    gene   heg   also results in a failure to develop endo-

    cardial cushions, further implicating this molecule inthe communication between the two adjacent cell

    layers of the heart.

    The analysis of patterns of clonal cell organization

    in the mouse heart supports the importance of 

    directionality of cell growth in proper heart forma-

    tion. Using a construct containing the   lacZ   reporter 

    gene targeted to an allele of the endogenous  a-cardiac

    actin gene   [62],   Meilhac and colleagues  [61]   dem-

    onstrate expansion in clonal cells in a concentric

    direction within the ventricle (along the axis from

    endocardium to myocardium). This proliferation of cells correlated with the thickening within regions of 

    the ventricle during development of this chamber.

    This process of oriented cell growth may facilitate

    regional increases in the thickness of the myocardium

    along the axis of the heart tube, thereby playing a

     prominent role in chamber morphogenesis.

    The secondary heart field 

    The contribution by the secondary (or anterior)

    heart field to development of the myocardium and

    outflow tract of the heart has emerged as an important 

    and potentially clinically significant aspect of cardio-

    genesis   [63–65].   (Although these field names often

    are used interchangeably, there are distinctions

     between them as a result of the experimental ap-

     proaches taken for their definition   [66].) The con-tribution of the heart-forming fields at the venous

    Fig. 2. Endocardial-myocardial signaling is necessary for normal myocardial development. ( A) During normal heart development 

    in vertebrates, the myocardium thickens by proliferation of cells in a concentric direction, along the axis from the lumen

    outwards. This process results in a thickened myocardial wall by 48 hpf of zebrafish development. ( B) In a class of zebrafish

    mutants, including the mutant  heart of glass   (heg ), this process is interrupted. The result is a single-cell–layered myocardium

    and a failure of the myocardium to thicken. The chambers dilate resulting in a massively enlarged heart. The endocardial

    cushions, which normally develop as swellings at the atrial-ventricular junction, also fail to develop in this mutant.

    cardiogenesis   &   gene expression   161

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    6/14

     pole of the heart to normal myocardial growth is well

    established, but the secondary heart field at the

    arterial pole of the heart is less well characterized.

    Many cardiac defects are associated with a failure

    of the outflow tract to develop normally, including

    atrioventricular and ventricular septal defects andtetralogy of Fallot, and together may account for as

    much as   one third of all congenital cardiovascular 

    disease   [67]. Therefore, understanding the mecha-

    nisms regulating the proper formation and migration

    of the secondary heart field may provide insights into

    the pathogenesis of these conditions.

    DiGeorge syndrome (DGS) is one relatively com-

    mon disorder associated with severe outflow tract 

    defects and anomalies in the glands and facial struc-

    tures. Mice with a null mutation in the gene   Tbx1,

    which is also within the deleted interval associatedwith the human disease, exhibit cardiac outflow tract 

    defects, consistent with a role for this transcription

    factor in outflow tract formation and the pathology of 

    DGS [68,69]. Further analysis of the requirement for 

    Tbx1 in outflow tract formation has implicated it in

    the maintenance of cell proliferation within the

    secondary heart field, consistent with the outflow

    tract defects associated with the mouse knockout 

    and DGS   [70]. Expression of   Tbx1   within   Nkx2.5 – 

     positive cells is essential for septation of the heart,

    and in part may be mediated through the action of 

     Fgf10  [70]. These results provide a firm link betweenthe secondary heart field and a congenital disorder of 

    the heart.

     Neural crest cells are also essential for outflow

    tract formation, and experiments in which neural crest 

    cells were ablated in chick embryos led to a shortened

    outflow tract and abnormal heart looping   [71].   The

    molecular interactions between these migrating neu-

    ral crest cells and those within the secondary heart 

    field seem essential to the normal formation of the

    outflow tract myocardium. Loss of the transcription

    factors Hand1  (dHand ) [72], Nkx2.5  [73], and  Mef2c[74],  expressed in these cells, exhibit outflow tract 

    abnormalities. Also, mice lacking the transcription

    factor  Foxh1  can form a primitive heart tube but fail

    to form normal outflow tract and right ventricular 

    myocardium [75]. The similarity of the phenotype of 

    the   Foxh1/  and   Mef2c/  mice is consistent 

    with the ability of   Foxh1, through interaction with

     Nkx2.5, to directly target the regulation of   Mef2c

    [75]. Through a single transforming growth factor– b

    (TGF-b) response element in   Mef2c, von Both and

    colleagues [75] demonstrate that   Foxh1  can mediate

    Smad-dependent activation of  Mef2c, and targets ex- pression of a transgene to the anterior heart field, and

    subsequently to the outflow tract and right ventricle.

    Islet-1 (Isl1), a LIM-homeodomain protein, is best 

    known for its important role in the embryogenesis of 

    the pancreatic islets of Langerhans [76]. Examination

    of the hearts of mice lacking Isl1, however, reveals

    a complete loss of the   outf low tract, right ventricle,

    and much of the atria   [77].  Expression analysis andlineage tracing of Isl1-positive progenitors have con-

    firmed that this gene is a marker for a set of undiffer-

    entiated cardiac progenitors, loss of which correlates

    with t he loss of cardiac structures in the   Isl1   mutant 

    mice   [77].   An enhancer within   Mef2c   contains two

    consensus binding sites for   Isl1   that are essential to

    its transcriptional regulation [78], further evidence of 

    the interplay between the genes expressed within this

    heart field.

    Valve formation

    The endocardial cushions are the precursors to the

    valves that eventually will complete the segmentation

    of the chambers comprising the mature heart. They

    develop as swellings of the cardiac jelly, the ECM

    layered between the endocardium and the myocar-

    dium at the chamber junctions. These regions then are

     populated by a specific subset of endocardial cells

    that undergo an epithelial-mesenchymal transition

    (EMT) and delaminate from the surface of the endo-

    cardial lining to migrate into the cardiac jelly   [79].Once localized to the site of the future valves, these

    cells proliferate to form the swellings that partition

    the chambers. These structures then undergo exten-

    sive remodeling to form the mature, tapered leaflet 

    structures characteristic of mature valves. The devel-

    opment of these structures is perturbed easily, and not 

    only are defects in valve formation associated with

    many congenital heart defects, but adult valvular 

    disease is a significant cause of morbidity and

    mortality  [67].   The coordinated expression of many

    genes is essential for proper valve formation, and wellestablished roles for the connexins and members of 

    the  notch,  wnt , and TGF-b  signaling pathways have

     been defined [80,81].

    Proper valve formation depends on the precise

    communication between the myocardial and endo-

    cardial layers of the heart through the ECM or cardiac

     jelly that separates these layers. Vascular endothelial

    growth factor (VEGF) is a key regulator of endothe-

    lial cell proliferation and is essential for the EMT

    required for valve development   [82]:   increases and

    decreases in VEGF levels adversely affect valve

    development. Loss of a single VEGF allele in mice issufficient to impair endocardial cushion development 

    [83],   but defects in EMT and valve formation are

    chen  et al162

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    7/14

    observed when myocardial   VEGF   levels are stimu-

    lated artificially at E9.5   [84,85].   Normally, upon

    completion of EMT by E10.5, VEGF levels increase

    dramatically, suggesting that high VEGF levels are

    required to terminate this process as well, whereas

    lower levels at   earlier stages are essential for itsinitiation [84,85].

    Recently, the role of VEGF in valve formation has

     been linked with nuclear factor of activated T-cells

    (NFATc1),   long known to be required for this pro-

    cess   [86].   NFATc1 is a transcription factor ex-

     pressed only in the endothelial cells of the heart,

     but it is not essential for the EMT associated with the

    early stages of endocardial cushion formation   [87].

    Inactivation of this gene in mice results in embryonic

    lethality associated with the selective absence of the

    aortic and pulmonary valves   [87,88].   The normaldevelopment of the tricuspid and mitral valve is of 

    note, however, because these two structures are

    derived entirely from the endocardial cushion tissue

    [89], whereas the final stages of development of the

    aortic and pulmonary valves are less well defined and

    include contributions from neural crest cells  [90]. At 

    E9.0 to E9.5 in mice hearts, calcineurin stimulates

    expression of the other NFATc isoforms, NFATc2, c3,

    and c4 in the myocardium and dampens VEGF

    expression in the endocardium, facilitating EMT [86].

    Subsequently, by E10.5, endocardial NFATc1 is

    activated in the endocardium, which is essential for valve maturation [86].

    These results are especially interesting when

    examined in the context of the endocardial cushion-

    derived defects associated with Down syndrome

     because the gene   DSCR1  (also known as ‘‘regulator 

    of calcineurin 1’’) is located in the minimal candidate

    region for the Down syndrome phenotype and in-

    hibits calcineurin activity [91]. The gene is expressed

    in heart tissue and overexpressed in the brain of 

    Down syndrome fetuses   [91].   Further linking these

    molecules in a single regulatory network is theobservation that   DSCR1   is induced by VEGF and

    that expression of   DSCR1   in endothelial cells can

     block dephosphorylation, nuclear translocation, and

    activity of NFATc1, presumably through loss of 

    calcineurin signaling   [92]. In mice,   DSCR1   is

    expressed in the endocardium of the developing

    atrioventricular and semilunar valves, as well as in

    the interventricular septum and the ventricular myo-

    cardium   [93].   In the outflow tract of the   Nfatc1/ 

    mice,   DSCR1   also is decreased, consistent with the

    transactivation by NFATc1 of an intragenic element 

    within DSCR1  [93]. Thus, as previously described for VEGF, precise regulation of   DSCR1   and associated

    calcineurin activity is essential for valve formation.

    VEGF also exerts its influence on valve formation

    through other signaling conduits, including the

    regulation of   b-catenin activity. Phosphorylation of 

    b-catenin and subsequent association with  PECAM-1

    (CD31) is increased by VEGF signaling, modulating

    b-catenin localization and signaling   [94].   The   wnt / b-catenin signaling pathway also is activated con-

    stitutively by truncation of the tumor suppr essor 

     protein adenomatous polyposis coli (Apc)   [95].

    Although mice embryos with truncated Apc do not 

    complete gastrulation, a zebrafish mutant expressing

    a truncated form of Apc completes gastrulation but 

    develops defects in heart morphology, including

    failure of the hearts to loop and excessive endocardial

    cushion formation   [96].  Although   b-catenin is upre-

    gulated normally only in valve-forming cells, there is

    accumulation of nuclear  b-catenin in all cardiac cellsif the zebrafish mutant, correlating with the expansion

    in cushion formation and concomitant increase in

    valve marker expression [96]. Thus, cell proliferation

    and EMT, normally restricted to the site of the endo-

    cardial cushions, occur throughout the endocardium.

    In contrast, endocardial cushion formation is inhib-

    ited by overexpression of full-length Apc  or  Dickkopf  

    1   ( Dkk1), a secreted  wnt   inhibitor  [96].

    The signaling required for normal valve formation

     between and within endocardial and myocardial cell

    layers is facilitated by the cardiac jelly, the ECM of 

    the heart. The role of one component of this complex,hyaluronic acid (HA), has been characterized particu-

    larly well for its role in mediating signaling, such

    as ErbB2/4 activity   [81].   HA is a glycosaminogly-

    can comprised of alternating glucoronic acid and

     N-acetylglucosamine (NAG) residues, with no pro-

    tein backbone   [97].   HA is synthesized by HA

    synthases (HAS) at the plasma membrane, and is

    released to the outside of the cell where it forms a gel

    that occupies the extracellular space. Through inter-

    actions with other matrix components, such as

    versican [98], HA can regulate ligand activity withinthe ECM. In mammals, three HAS genes exist:  has1,

    has2, and   has3. Loss of   has2   in mice results in

    embryonic lethality by E9.5, and is characterized by a

    complete loss of cardiac jelly, severe pericardial

    edema, and abnormal vessel development. The im-

     portance of adequate matrix formation also has been

    revealed by analysis of the zebrafish valve mutant 

     jekyll , characterized by a lack of endocardial cushions

    with toggling of blood back and forth between

    chambers and pericardial edema [99]. Mutations were

    defined within the gene   50-diphosphate (UDP) – 

     glucose dehydrogenase   (Ugdh), an enzyme requiredfor the conversion of UDP-glucose into UDP-

    glucoronic acid, and the subsequent production of 

    cardiogenesis   &   gene expression   163

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    8/14

    the matrix com ponents HA, chondroitin sulfate, and

    heparin sulfate [100]. Clearly, alterations in the com-

     position of the ECM can have profound effects on

    valve development, and may affect further other attri-

     butes of cardiac growth, such as myocardial growth

    and trabeculation.

    Development of the conduction system

    There has also been substantial progress in

    understanding early development of the cardiac con-

    duction system. Central to this understanding are

    questions regarding the origin of the cells involved

    in generating and conducting electrical impulses to

    the atrial and ventricular chambers. The finding

    that some cells of the conduction system expressneural markers, such as neurofilament  [101], HNK-1

    [102], and EAP-300   [103,104],   and more recent 

    experiments that show the contribution of neural

    crest-derived cells to the maturation of conductive

     phenotypes and the development of autonomic

    innervation   [105,106],   suggest that cardiac neural

    crest-derived cell populations are recruited to form

    the conduction system. Several recent studies, how-

    ever, have demonstrated that cells of the central and

     peripheral conduction tissues emanate from a multi-

     potent cardiomyogenic lineage, rather than from

    migratory neurogenic populations or outgrowth froma specified pool of myogenic precursors   [107,108].

    The recruitment of these cells seems continuous

    throughout the early stages of heart development 

    [109]. Moreover, the ability to record coordinated

    depolarization of distinct cardiac compartments

     before the arrival of neural crest-derived or epithe-

    lial-derived cells lends credibility to the argument that 

    even before different components of the conduction

    system can be identified morphologically, the basic

    electrical configuration of the heart already has been

    established [110].Although the entire conduction system shares

    some common transcriptional mechanisms that drive

    specification and differentiation of recruited cells, the

    functional differences between the central and

     peripheral conductive regions point toward a complex

    genetic program [111].   For example, loss of   Nkx2.5

    function causes hypoplastic development throughout 

    the conduction system, which contributes to func-

    tional defects in the postnatal heart   [112].   This in-

    dicates that   Nkx2.5   is either important in the

    recruitment of cells to the conduction system in early

    cardiac development or in the retention of those cellsthrough the prevention of apoptosis  [113,114].   The

    T-box transcription factor   Tbx-3   similarly delineates

    most of the conduction system, including what seems

    to be a remnant of the embryonic slow-conducting

    regions that contribute to the ventricular conduction

    tissue [115,116]. In this instance,  Tbx-3  (and possibly

    Tbx-2, which is also expressed transiently in the

    conduction system) may govern development of the conduction system by preventing expression of 

    chamber-specification genes such as atrial natriuretic

    factor  [116,117]. In contrast, the Tbx-5  protein seems

    to function in specification   and   patterning of the

    central conduction system  [118].   Demonstration of 

    the interaction of   Nkx2.5   and  Tbx   proteins indicates

    cooperation of these transcription factors in the

    development of specific cardiac compartments. Other 

    transcription factors such as   GATA6 ,  HF-1b,  MyoD,

    and   Msx-2   have been implied to be associated with

    the development of sections of the conduction sys-tem, but the precise role of these proteins in trig-

    gering a conduction specific genetic program remains

    enigmatic [109,119,120].

    The proximity of the Purkinje fiber network with

     branching coronary arteries led to speculation that 

     progressive conscription of cardiomyogenic cells to-

    ward a conductive fate was the result of autocrine and

     paracrine interactions. In fact, induction of cells in the

     peripheral conduction system seems dependent on

    vascular-derived endothelin-1 (ET-1), Neuregulin-1

    (NG-1), and their receptors, despite some spatial and

    temporal inconsistencies likely caused by variationacross species   [121–123].   Exposure to ET-1 can

    confer pacemaker activity upon cultured embryonic

    stem cells   [124]   and the regulated expression of 

    endothelin-converting enzyme may provide an addi-

    tional layer of control over the specification of con-

    duction tissues in early development   [101,125]. In

    addition, treatment of embryonic cardiomyocytes

    isolated from looped, tubular hearts with ET-1 re-

    sulted in increased expression of   wnt7a   and   wnt11,

    suggesting a role for these proteins in patterning

    the conduction system [126]. Because ET-1 is respon-sive to hemodynamic forces such as shear stress

    and stretch, blood flow and pressure in the develop-

    ing heart may serve as a fundamental inductive

    signal for the formation of the conduction system

    [123,127,128].

    Aberrant development of the conduction system

    has been implicated in arrhythmias and other con-

    genital abnormalities of cardiac activation  [109,129].

    Perturbations in cellular excitability, intercellular 

    communication, and tissue architecture are associated

    with a slowing of conduction velocity and, as a result,

    increase the likelihood for the development of life-threatening arrhythmias   [130,131].   In particular,

    inadequate electrical coupling of cardiomyocytes

    chen  et al164

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    9/14

    through gap junctions seems central to arrhythmo-

    genesis. Null mutations have been made in genes

    encoding   each of t  he principal cardiac connexin

     proteins   [132–138]. Although   Cx43- and   Cx45-

    deficient mice die prematurely from congenital

    deformations of the cardiovascular system, mice that completely lack  Cx40  are viable. The Cx40-knockout 

    mice show partial atrioventricular (AV)– bundle

     branch block that results in a greater susceptibility

    to arrhythmia formation. In addition, the   Cx40-null

    mice exhibit some characteristics reminiscent of 

    human congenital heart defects. These include myo-

    cardial hypertrophy, an AV canal defect, incomplete

    formation of the mesenchymal cap of the atrial sep-

    tum, and various ventricular septal defects   [139].

    Heterozygous   Cx45-deficient mice have conduction

     blockade through the AV canal and contractions inthe outflow track that are not coordinated with those

    of the ventricle. The  Cx45-null embryos also exhibit 

    an endocardial cushion defect believed to arise from a

    failure of cells in the endocardium to undergo the

    requisite epithelial-mesenchymal transformation. The

    combination of conduction block and congenital

    abnormalities results in   Cx45-knockout mice dying

    of heart failure at embryonic day 10.  Cx43-knockout 

    mice die from a pulmonary outflow tract obstruction

    shortly after birth and conditionally deleted mice

    have slow ventricular conduction velocities and

     propensity for ventricular fibrillation [140,141]. Micedeficient for  Cx40   and  Cx43  have validated the idea

    that loss of either isoform causes reduction in

     propagation velocity and leads to congenital defor-

    mations. Similarly, experiments that involve ablation

    or swapping of connexin gene combinations show

    defects in conduction and deformations in other 

    cardiovascular structures  [142–144].

    Summary

    This article summarizes some recent contributions

    to our understanding of cardiogenesis, although these

    new findings build on the framework of earlier 

    studies. One paradigm that has emerged is the im-

     portance of signaling between the endocardium and

    myocardium for normal heart morphology. A require-

    ment for endothelial cells in organogenesis has been

    shown in mice, zebrafish, and frog, which have

    demonstrated that kidney   [145]   and pancreas for-

    mation [146] are contingent on the presence of these

    cells. Similarly, studies in mouse and zebrafish

    describe the loss of genes expressed in either cardiaccell type that have profound effects on the develop-

    ment of the adjacent cell layer. The endocardium of 

    the heart seems to be a vertebrate-specific develop-

    ment because the primitive chordate heart has no such

    structure [2]. Consistent with this observation, much

    of normal vertebrate cardiogenesis is predicated on

    the sophisticated interweaving of signals between the

    endocardial and myocardial layers, as well as signal-ing within these layers. As our understanding of the

    genetic control of cardiogenesis evolves, new regu-

    latory models will emerge and others will progress,

    including the role of the endocardium in mediating

    signals from the circulation. In fact, a role for flow

    itself already has been defined for formation of the

    zebrafish kidney  [147]   and heart   [148].   Inevitably,

    the principles derived from these studies will frame

    the foundation upon which the heart is built.

    References

    [1] Hoffman JI, Kaplan S. The incidence of congenital

    heart disease. J Am Coll Cardiol 2002;39(12):

    1890–900.

    [2] Fishman MC, Chien KR. Fashioning the vertebrate

    heart: earliest embryonic decisions. Development 

    1997;124(11):2099–117.

    [3] Harvey RP. Patterning the vertebrate heart. Nat Rev

    Genet 2002;3(7):544–56.

    [4] Zaffran S, Frasch M. Early signals in cardiac

    development. Circ Res 2002;91(6):457–69.

    [5] Olson EN. A decade of discoveries in cardiac biology.

     Nat Med 2004;10(5):467– 74.

    [6] Schultheis TM, Xydas S, Lassar AB. Induction of 

    avian cardiac myogenesis by anterior endoderm.

    Development 1995;121(12):4203– 14.

    [7] Yelon D, Horne SA, Stainier DY. Restricted expres-

    sion of cardiac myosin genes reveals regulated

    aspects of heart tube assembly in zebrafish. Dev Biol

    1999;214(1):23–37.

    [8] Srivastava D, Olson EN. A genetic blueprint for 

    cardiac development. Nature 2000;407(6801):221–6.

    [9] Chien KR. Genomic circuits and the integrative

     biology of cardiac diseases. Nature 2000;407(6801):227–32.

    [10] Azpiazu N, Frasch M. tinman and bagpipe: two

    homeo box genes that determine cell fates in the

    dorsal mesoderm of Drosophila. Genes Dev 1993;

    7(7B):1325–40.

    [11] Bodmer R. The gene tinman is required for specifi-

    cation of the heart and visceral muscles in Drosophila.

    Development 1993;118(3):719–29.

    [12] Frasch M. Induction of visceral and cardiac meso-

    derm by ectodermal Dpp in the early Drosophila

    embryo. Nature 1995;374(6521):464– 7.

    [13] Park M, Wu X, Golden K, et al. The wingless

    signaling pathway is directly involved in Drosophilaheart development. Dev Biol 1996;177(1):104–16.

    [14] Wu X, Golden K, Bodmer R. Heart development in

    cardiogenesis   &   gene expression   165

    http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-http://-/?-

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    10/14

    Drosophila requires the segment polarity gene wing-

    less. Dev Biol 1995;169(2):619–28.

    [15] Schneider VA, Mercola M. Wnt antagonism initiates

    cardiogenesis in Xenopus laevis. Genes Dev 2001;

    15(3):304–15.

    [16] Tzahor E, Lassar AB. Wnt signals from the neuraltube block ectopic cardiogenesis. Genes Dev 2001;

    15(3):255–60.

    [17] Marvin MJ, Di Rocco G, Gardiner A, et al. Inhibition

    of Wnt activity induces heart formation from poste-

    rior mesoderm. Genes Dev 2001;15(3):316–27.

    [18] Eisenberg CA, Eisenberg LM. WNT11 promotes

    cardiac tissue formation of early mesoderm. Dev

    Dyn 1999;216(1):45–58.

    [19] Chen JN, Fishman MC. Zebrafish tinman homo-

    log demarcates the heart field and initiates myo-

    cardial differentiation. Development 1996;122(12):

    3809–16.

    [20] Cleaver OB, Patterson KD, Krieg PA. Overexpressionof the tinman-related genes XNkx-2.5 and XNkx-2.3

    in Xenopus embryos results in myocardial hyper-

     plasia. Development 1996;122(11):3549 – 56.

    [21] Tanaka M, Chen Z, Bartunkova S, et al. The cardiac

    homeobox gene Csx/Nkx2.5 lies genetically upstream

    of multiple genes essential for heart development.

    Development 1999;126(6):1269 – 80.

    [22] Tanaka M, Kasahara H, Bartunkova S, et al. Verte-

     brate homologs of tinman and bagpipe: roles of the

    homeobox genes in cardiovascular development. Dev

    Genet 1998;22(3):239– 49.

    [23] Grow MW, Krieg PA. Tinman function is essential for 

    vertebrate heart development: elimination of cardiac

    differentiation by dominant inhibitory mutants of the

    tinman-related genes, XNkx2–3 and XNkx2–5. Dev

    Biol 1998;204(1):187– 96.

    [24] Lien CL, Wu C, Mercer B, et al. Control of early

    cardiac-specific transcription of Nkx2– 5 by a

    GATA-dependent enhancer. Development 1999;

    126(1):75–84.

    [25] Sepulveda JL, Vlahopoulos S, Iyer D, et al. Combi-

    natorial expression of GATA4, Nkx2–5, and serum

    response factor directs early cardiac gene activity.

    J Biol Chem 2002;277(28):25775–82.

    [26] Brown III CO, Chi X, Garcia-Gras E, et al. Thecardiac determination factor, Nkx2– 5, is activated by

    mutual cofactors GATA-4 and Smad1/4 via a novel

    upstream enhancer. J Biol Chem 2004;279(11):

    10659–69.

    [27] Wang D, Chang PS, Wang Z, et al. Activation of 

    cardiac gene expression by myocardin, a transcrip-

    tional cofactor for serum response factor. Cell

    2001;105(7):851–62.

    [28] Gisselbrecht S, Skeath JB, Doe CQ, et al. Heartless

    encodes a fibroblast growth factor receptor (DFR1/ 

    DFGF-R2) involved in the directional migration of 

    early mesodermal cells in the Drosophila embryo.

    Genes Dev 1996;10(23):3003 – 17.[29] Beiman M, Shilo BZ, Volk T. Heartless, a Drosophila

    FGF receptor homolog, is essential for cell migration

    and establishment of several mesodermal lineages.

    Genes Dev 1996;10(23):2993 – 3002.

    [30] Shishido E, Ono N, Kojima T, et al. Requirements of 

    DFR1/Heartless, a mesoderm-specific Drosophila

    FGF-receptor, for the formation of heart, visceral

    and somatic muscles, and ensheathing of longitudinalaxon tracts in CNS. Development 1997;124(11):

    2119–28.

    [31] Reifers F, Walsh EC, Leger S, et al. Induction and

    differentiation of the zebrafish heart requires fibro-

     blast growth factor 8 (fgf8/acerebellar). Development 

    2000;127(2):225–35.

    [32] Chen JN, Fishman MC. Genetics of heart develop-

    ment. Trends Genet 2000;16(9):383–8.

    [33] Stainier DY, Fouquet B, Chen JN, et al. Mutations

    affecting the formation and function of the cardio-

    vascular system in the zebrafish embryo. Develop-

    ment 1996;123:285–92.

    [34] Chen JN, Haffter P, Odenthal J, et al. Mutationsaffecting the cardiovascular system and other inter-

    nal organs in zebrafish. Development 1996;123:

    293–302.

    [35] Alexander J, Stainier DY, Yelon D. Screening mosaic

    F1 females for mutations affecting zebrafish heart 

    induction and patterning. Dev Genet 1998;22(3):

    288–99.

    [36] Trinh LA, Stainier DY. Fibronectin regulates epithe-

    lial organization during myocardial migration in

    zebrafish. Dev Cell 2004;6(3):371–82.

    [37] Kupperman E, An S, Osborne N, et al. A sphingo-

    sine-1-phosphate receptor regulates cell migration

    during vertebrate heart development. Nature

    2000;406(6792):192– 5.

    [38] Kikuchi Y, Trinh LA, Reiter JF, et al. The zebrafish

     bonnie and clyde gene encodes a Mix family

    homeodomain protein that regulates the generation

    of endodermal precursors. Genes Dev 2000;14(10):

    1279–89.

    [39] Kikuchi Y, Agathon A, Alexander J, et al. casanova

    encodes a novel Sox-related protein necessary and

    sufficient for early endoderm formation in zebrafish.

    Genes Dev 2001;15(12):1493 – 505.

    [40] Reiter JF, Alexander J, Rodaway A, et al. Gata5 is

    required for the development of the heart andendoderm in zebrafish. Genes Dev 1999;13(22):

    2983–95.

    [41] Yelon D, Ticho B, Halpern ME, et al. The bHLH

    transcription factor hand2 plays parallel roles in

    zebrafish heart and pectoral fin development. Devel-

    opment 2000;127(12):2573– 82.

    [42] Alexander J, Stainier DY. A molecular pathway

    leading to endoderm formation in zebrafish. Curr 

    Biol 1999;9(20):1147 – 57.

    [43] Kuo CT, Morrisey EE, Anandappa R, et al. GATA4

    transcription factor is required for ventral morpho-

    genesis and heart tube formation. Genes Dev 1997;

    11(8):1048–60.[44] Molkentin JD, Lin Q, Duncan SA, et al. Requirement 

    of the transcription factor GATA4 for heart tube

    chen  et al166

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    11/14

    formation and ventral morphogenesis. Genes Dev

    1997;11(8):1061–72.

    [45] Li S, Zhou D, Lu MM, et al. Advanced cardiac

    morphogenesis does not require heart tube fusion.

    Science 2004;305(5690):1619– 22.

    [46] Horne-Badovinac S, Lin D, Waldron S, et al.Positional cloning of heart and soul reveals multiple

    roles for PKC lambda in zebrafish organogenesis.

    Curr Biol 2001;11(19):1492– 502.

    [47] Peterson RT, Mably JD, Chen JN, et al. Convergence

    of distinct pathways to heart patterning revealed by

    the small molecule concentramide and the mutation

    heart-and-soul. Curr Biol 2001;11(19):1481– 91.

    [48] Shu X, et al. Na,K-ATPase is essential for embryonic

    heart development in the zebrafish. Development 

    2003;130(25):6165– 73.

    [49] Linask KK. N-cadherin localization in early heart 

    development and polar expression of Na+, K(+)-

    ATPase, and integrin during pericardial coelomformation and epithelialization of the differentiating

    myocardium. Dev Biol 1992;151(1):213– 24.

    [50] Mohler PJ, Schott JJ, Gramolini AO, et al. Ankyrin-B

    mutation causes type 4 long-QT cardiac arrhythmia

    and sudden cardiac death. Nature 2003;421(6923):

    634–9.

    [51] Suzuki A, Yamanaka T, Hirose T, et al. Atypical pro-

    tein kinase C is involved in the evolutionarily con-

    served par protein complex and plays a critical role

    in establishing epithelia-specific junctional structures.

    J Cell Biol 2001;152(6):1183–96.

    [52] Suzuki A, Hirata M, Kamimura K, et al. aPKC acts

    upstream of PAR-1b in both the establishment and

    maintenance of mammalian epithelial polarity. Curr 

    Biol 2004;14(16):1425 – 35.

    [53] Crackower MA, Oudit GY, Kozieradzki I, et al.

    Regulation of myocardial contractility and cell size by

    distinct PI3K-PTEN signaling pathways. Cell 2002;

    110(6):737–49.

    [54] Wang D, Passier R, Liu ZP, et al. Regulation of 

    cardiac growth and development by SRF and its co-

    factors. Cold Spring Harb Symp Quant Biol 2002;

    67:97–105.

    [55] Chen F, Kook H, Milewski R, et al. Hop is an un-

    usual homeobox gene that modulates cardiac devel-opment. Cell 2002;110(6):713 – 23.

    [56] Shin CH, Liu ZP, Passier R, et al. Modulation of 

    cardiac growth and development by HOP, an unusual

    homeodomain protein. Cell 2002;110(6):725– 35.

    [57] Meyer D, Birchmeier C. Multiple essential func-

    tions of neuregulin in development. Nature 1995;

    378(6555):386–90.

    [58] Gassmann M, Casagrande F, Orioli D, et al. Aber-

    rant neural and cardiac development in mice lack-

    ing the ErbB4 neuregulin receptor. Nature 1995;

    378(6555):390–4.

    [59] Lee KF, Simon H, Chen H, et al. Requirement for 

    neuregulin receptor erbB2 in neural and cardiac de-velopment. Nature 1995;378(6555):394– 8.

    [60] Mably JD, Mohideen M-APK, Burns CG, et al. heart 

    of glass regulates the concentric growth of the heart in

    zebrafish. Curr Biol 2003;13(24):2138– 47.

    [61] Meilhac SM, Esner M, Kerszberg M, et al. Oriented

    clonal cell growth in the developing mouse myocar-

    dium underlies cardiac morphogenesis. J Cell Biol

    2004;164(1):97–109.[62] Sassoon DA, Garner I, Buckingham M. Transcripts

    of alpha-cardiac and alpha-skeletal actins are early

    markers for myogenesis in the mouse embryo.

    Development 1988;104(1):155 – 64.

    [63] Mjaatvedt CH, Nakaoka T, Moreno-Rodriguez R,

    et al. The outflow tract of the heart is recruited from

    a novel heart-forming field. Dev Biol 2001;238(1):

    97–109.

    [64] Waldo KL, Kumiski DH, Wallis KT, et al. Cono-

    truncal myocardium arises from a secondary heart 

    field. Development 2001;128(16):3179– 88.

    [65] Kelly RG, Brown NA, Buckingham ME. The arterial

     pole of the mouse heart forms from Fgf10-expressingcells in pharyngeal mesoderm. Dev Cell 2001;

    1(3):435–40.

    [66] Abu-Issa R, Waldo K, Kirby ML. Heart fields: one,

    two or more? Dev Biol 2004;272(2):281–5.

    [67] Loffredo CA. Epidemiology of cardiovascular mal-

    formations: prevalence and risk factors. Am J Med

    Genet 2000;97(4):319– 25.

    [68] Chieffo C, Garvey N, Gong W, et al. Isolation and

    characterization of a gene from the DiGeorge chro-

    mosomal region homologous to the mouse Tbx1

    gene. Genomics 1997;43(3):267– 77.

    [69] Jerome LA, Papaioannou VE. DiGeorge syndrome

     phenotype in mice mutant for the T-box gene, Tbx1.

     Nat Genet 2001;27(3):286– 91.

    [70] Xu H, Morishima M, Wylie JN, et al. Tbx1 has a

    dual role in the morphogenesis of the cardiac outflow

    tract. Development 2004;131(13):3217– 27.

    [71] Yelbuz TM, Waldo KL, Kumiski DH, et al. Shortened

    outflow tract leads to altered cardiac looping after neu-

    ral crest ablation. Circulation 2002;106(4):504–10.

    [72] Srivastava D, Thomas T, Lin Q, et al. Regulation of 

    cardiac mesodermal and neural crest development by

    the bHLH transcription factor, dHAND. Nat Genet 

    1997;16(2):154–60.

    [73] Lyons I, Parsons LM, Hartley L, et al. Myogenic andmorphogenetic defects in the heart tubes of murine

    embryos lacking the homeo box gene Nkx2– 5.

    Genes Dev 1995;9(13):1654–66.

    [74] Lin Q, Schwartz J, Bucana C, et al. Control of mouse

    cardiac morphogenesis and myogenesis by tran-

    scription factor MEF2C. Science 1997;276(5317):

    1404–7.

    [75] von Both I, Silvestri C, Erdemir T, et al. Foxh1 is

    essential for development of the anterior heart field.

    Dev Cell 2004;7(3):331–45.

    [76] Ahlgren U, Pfaff SL, Jessell TM, et al. Independent 

    requirement for ISL1 in formation of pancreatic

    mesenchyme and islet cells. Nature 1997;385(6613):257–60.

    [77] Cai CL, Liang X, Shi Y, et al. Isl1 identifies a car-

    cardiogenesis   &   gene expression   167

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    12/14

    diac progenitor population that proliferates prior to

    differentiation and contributes a majority of cells to

    the heart. Dev Cell 2003;5(6):877–89.

    [78] Dodou E, Verzi MP, Anderson JP, et al. Mef2c is a

    direct transcriptional target of ISL1 and GATA factors

    in the anterior heart field during mouse embryonicdevelopment. Development 2004;131(16):3931–42.

    [79] Markwald RR, Fitzharris TP, Manasek FJ. Structural

    development of endocardial cushions. Am J Anat 

    1977;148(1):85– 119.

    [80] Armstrong EJ, Bischoff J. Heart valve development:

    endothelial cell signaling and differentiation. Circ Res

    2004;95(5):459–70.

    [81] Schroeder JA, Jackson LF, Lee DC, et al. Form and

    function of developing heart valves: coordination

     by extracellular matrix and growth factor signaling.

    J Mol Med 2003;81(7):392–403.

    [82] Lambrechts D, Carmeliet P. Genetics in zebrafish,

    mice, and humans to dissect congenital heart disease:insights in the role of VEGF. Curr Top Dev Biol

    2004;62:189–224.

    [83] Stalmans I, Lambrechts D, De Smet F, et al. VEGF:

    a modifier of the del22q11 (DiGeorge) syndrome?

     Nat Med 2003;9(2):173– 82.

    [84] Dor Y, Klewer SE, McDonald JA, et al. VEGF modu-

    lates early heart valve formation. Anat Rec 2003;

    271A(1):202–8.

    [85] Miquerol L, Langille BL, Nagy A. Embryonic

    development is disrupted by modest increases in

    vascular endothelial growth factor gene expression.

    Development 2000;127(18):3941– 6.

    [86] Chang CP, Neilson JR, Bayle JH, et al. A field of 

    myocardial-endocardial NFAT signaling underlies

    heart valve morphogenesis. Cell 2004;118(5):649–63.

    [87] Ranger AM, Grusby MJ, Hodge MR, et al. The tran-

    scription factor NF-ATc is essential for cardiac valve

    formation. Nature 1998;392(6672):186– 90.

    [88] de la Pompa JL, Timmerman LA, Takimoto H, et al.

    Role of the NF-ATc transcription factor in morpho-

    genesis of cardiac valves and septum. Nature 1998;

    392(6672):182–6.

    [89] Wessels A, Markman MW, Vermeulin JL, et al. The

    development of the atrioventricular junction in the

    human heart. Circ Res 1996;78(1):110–7.[90] Kirby ML, Gale TF, Stewart DE. Neural crest cells

    contribute to normal aorticopulmonary septation.

    Science 1983;220(4601):1059– 61.

    [91] Fuentes JJ, Genesca L, Kingsbury TJ, et al. DSCR1,

    overexpressed in Down syndrome, is an inhibitor of 

    calcineurin-mediated signaling pathways. Hum Mol

    Genet 2000;9(11):1681 – 90.

    [92] Hesser BA, Liang XH, Camenisch G, et al. Down

    syndrome critical region protein 1 (DSCR1), a novel

    VEGF target gene that regulates expression of in-

    flammatory markers on activated endothelial cells.

    Blood 2004;104(1):149 – 58.

    [93] Lange AW, Molkentin JD, Yutzey KE. DSCR1 geneexpression is dependent on NFATc1 during cardiac

    valve formation and colocalizes with anomalous or-

    gan development in trisomy 16 mice. Dev Biol 2004;

    266(2):346–60.

    [94] Ilan N, Mahooti S, Kimm DL, et al. PECAM-1

    (CD31) functions as a reservoir for and a modulator 

    of tyrosine-phosphorylated beta-catenin. J Cell Sci

    1999;112(Pt 18):3005– 14.[95] Fodde R, Smits R, Clevers H. APC, signal trans-

    duction and genetic instability in colorectal cancer.

     Nat Rev Cancer 2001;1(1):55– 67.

    [96] Hurlstone AF, Haramis AP, Weinholds E, et al. The

    Wnt/beta-catenin pathway regulates cardiac valve

    formation. Nature 2003;425(6958):633– 7.

    [97] Fraser JR, Laurent TC, Laurent UB. Hyaluronan: its

    nature, distribution, functions and turnover. J Intern

    Med 1997;242(1):27–33.

    [98] Day AJ, Prestwich GD. Hyaluronan-binding proteins:

    tying up the giant. J Biol Chem 2002;277(7):4585– 8.

    [99] Walsh EC, Stainier DY. UDP-glucose dehydrogenase

    required for cardiac valve formation in zebrafish.Science 2001;293(5535):1670– 3.

    [100] Lander AD, Selleck SB. The elusive functions of 

     proteoglycans: in vivo veritas. J Cell Biol 2000;

    148(2):227–32.

    [101] Takebayashi-Suzuki K, Pauliks LB, Eltsefon Y, et al.

    Purkinje fibers of the avian heart express a myogenic

    transcription factor program distinct from cardiac and

    skeletal muscle. Dev Biol 2001;234(2):390–401.

    [102] Gorza L, Schiaffino S, Vitadello M. Heart conduction

    system: a neural crest derivative? Brain Res 1988;

    457(2):360–6.

    [103] Kelly MM, Phanhthourath C, Brees DK, et al. Mo-

    lecular characterization of EAP-300: a high molecular 

    weight, embryonic polypeptide containing an amino

    acid repeat comprised of multiple leucine-zipper 

    motifs. Brain Res Dev Brain Res 1995;85(1):31–47.

    [104] McCabe CF, Gourdie RG, Thompson RP, et al. De-

    velopmentally regulated neural protein EAP-300 is

    expressed by myocardium and cardiac neural crest 

    during chick embryogenesis. Dev Dyn 1995;203(1):

    51–60.

    [105] Poelmann RE, Lie-Venema H, Gittenberger-de Groot 

    AC. The role of the epicardium and neural crest as

    extracardiac contributors to coronary vascular devel-

    opment. Tex Heart Inst J 2002;29(4):255–61.[106] Poelmann RE, Jongbloed MR, Molin DG, et al.

    The neural crest is contiguous with the cardiac con-

    duction system in the mouse embryo: a role in induc-

    tion? Anat Embryol (Berl) 2004;208(5):389–93.

    [107] Gourdie RG, Mima T, Thompson RP, et al. Terminal

    diversification of the myocyte lineage generates Pur-

    kinje fibers of the cardiac conduction system. Devel-

    opment 1995;121(5):1423 – 31.

    [108] Cheng G, Litchenberg WH, Cole GJ, et al. Develop-

    ment of the cardiac conduction system involves

    recruitment within a multipotent cardiomyogenic

    lineage. Development 1999;126(22):5041– 9.

    [109] Nguyen-Tran VT, Kubalak SW, Minamisawa S, et al.A novel genetic pathway for sudden cardiac death via

    defects in the transition between ventricular and

    chen  et al168

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    13/14

    conduction system cell lineages. Cell 2000;102(5):

    671–82.

    [110] Moorman AF, Christoffels VM. Cardiac chamber 

    formation: development, genes, and evolution. Phys-

    iol Rev 2003;83(4):1223–67.

    [111] Myers DC, Fishman GI. Toward an understandingof the genetics of murine cardiac pacemaking and

    conduction system development. Anat Rec 2004;

    280A(2):1018–21.

    [112] Jay PY, Harris BS, Maguire CT, et al. Nkx2 – 5 mu-

    tation causes anatomic hypoplasia of the cardiac con-

    duction system. J Clin Invest 2004;113(8):1130–7.

    [113] Jay PY, Harris BS, Buerger A, et al. Function follows

    form: cardiac conduction system defects in Nkx2–5

    mutation. Anat Rec 2004;280A(2):966–72.

    [114] Cheng G, Wessels A, Gourdie RG, et al. Spatiotem-

     poral and tissue specific distribution of apoptosis in

    the developing chick heart. Dev Dyn 2002;223(1):

    119–33.[115] Hoogaars WM, Tessari A, Moorman AF, et al. The

    transcriptional repressor Tbx3 delineates the devel-

    oping central conduction system of the heart.

    Cardiovasc Res 2004;62(3):489–99.

    [116] Franco D. Unveiling the transcriptional control of the

    developing cardiac conduction system. Cardiovasc

    Res 2004;62(3):444–6.

    [117] Moorman AF, Soufan AT, Hagoort J, et al. Devel-

    opment of the building plan of the heart. Ann N Y

    Acad Sci 2004;1015:171–81.

    [118] Moskowitz IP, Pizard A, Pafel W, et al. The T-Box

    transcription factor Tbx5 is required for the patterning

    and maturation of the murine cardiac conduction

    system. Development 2004;131(16):4107–16.

    [119] Robbins J, Dorn II GW. Listening for hoof beats in

    heart beats. Nat Med 2000;6(9):968–70.

    [120] Harris BS, Jay PY, Rackley MS, et al. Transcriptional

    regulation of cardiac conduction system develop-

    ment: 2004 FASEB cardiac conduction system mini-

    meeting, Washington, DC. Anat Rec 2004;280A(2):

    1036–45.

    [121] Hyer J, Johansen M, Prasad A, et al. Induction of Pur-

    kinje fiber differentiation by coronary arterialization.

    Proc Natl Acad Sci USA 1999;96(23):13214–8.

    [122] Rentschler S, Zander J, Meyers K, et al. Neuregulin-1 promotes formation of the murine cardiac conduc-

    tion system. Proc Natl Acad Sci USA 2002;99(16):

    10464–9.

    [123] Hall CE, Hurtado R, Hewett KW, et al. Hemodynamic-

    dependent patterning of endothelin converting enzyme

    1 expression and differentiation of impulse-conducting

    Purkinje fibers in the embryonic heart. Development 

    2004;131(3):581–92.

    [124] Gassanov N, Zagidullin N, Hoppe UC. Endothelin

    induces differentiation of ANP-EGFP expressing em-

     bryonic stem cells towards a pacemaker phenotype.

    FASEB J 2004;18(14):1710 – 2.

    [125] Adamo RF, Guay CL, Edwards AV, et al. GATA-6gene enhancer contains nested regulatory modules for 

     primary myocardium and the embedded nascent 

    atrioventricular conduction system. Anat Rec 2004;

    280A(2):1062–71.

    [126] Bond J, Sedmera D, Jourdan J, et al. Wnt11 and

    Wnt7a are up-regulated in association with differ-

    entiation of cardiac conduction cells in vitro and in

    vivo. Dev Dyn 2003;227(4):536–43.[127] Reckova M, Rosengarten C, deAlmeida A, et al.

    Hemodynamics is a key epigenetic factor in develop-

    ment of the cardiac conduction system. Circ Res

    2003;93(1):77–85.

    [128] Cowan DB, Lye SJ, Langille BL. Regulation of 

    vascular connexin43 gene expression by mechanical

    loads. Circ Res 1998;82(7):786–93.

    [129] Schott JJ, Benson DW, Basson CT, et al. Congenital

    heart disease caused by mutations in the transcription

    factor NKX2–5. Science 1998;281(5373):108–11.

    [130] Spach MS, Heidlage JF, Dolber PC, et al. Electro-

     physiological effects of remodeling cardiac gap junc-

    tions and cell size: experimental and model studiesof normal cardiac growth. Circ Res 2000;86(3):

    302–11.

    [131] Tamaddon HS, Vaidya D, Simon AM, et al. High-

    resolution optical mapping of the right bundle branch

    in connexin40 knockout mice reveals slow conduc-

    tion in the specialized conduction system. Circ Res

    2000;87(10):929–36.

    [132] Reaume AG, de Sousa PA, Kulkarni S, et al. Cardiac

    malformation in neonatal mice lacking connexin43.

    Science 1995;267(5205):1831 – 4.

    [133] Kirchhoff S, Nelles E, Hagendorff A, et al. Reduced

    cardiac conduction velocity and predisposition to ar-

    rhythmias in connexin40-deficient mice. Curr Biol

    1998;8(5):299–302.

    [134] Simon AM, Goodenough DA, Paul DL. Mice lacking

    connexin40 have cardiac conduction abnormalities

    characteristic of atrioventricular block and bundle

     branch block. Curr Biol 1998;8(5):295– 8.

    [135] Kumai M, Nishii K, Nakamura K, et al. Loss of 

    connexin45 causes a cushion defect in early cardio-

    genesis. Development 2000;127(16):3501– 12.

    [136] Kruger O, Plum A, Kim JS, et al. Defective vascular 

    development in connexin 45-deficient mice. Devel-

    opment 2000;127(19):4179– 93.

    [137] Kirchhoff S, Kim JS, Hagendorff A, et al. Abnor-mal cardiac conduction and morphogenesis in con-

    nexin40 and connexin43 double-deficient mice. Circ

    Res 2000;87(5):399–405.

    [138] Gros D, Dupays L, Alcolea S, et al. Genetically

    modified mice: tools to decode the functions of 

    connexins in the heart-new models for cardiovascular 

    research. Cardiovasc Res 2004;62(2):299– 308.

    [139] Lo CW. Role of gap junctions in cardiac conduction

    and development: insights from the connexin knock-

    out mice. Circ Res 2000;87(5):346–8.

    [140] Gutstein DE, Morley GE, Tamaddon H, et al. Con-

    duction slowing and sudden arrhythmic death in mice

    with cardiac-restricted inactivation of connexin43.Circ Res 2001;88(3):333–9.

    [141] van Rijen HV, Eckardt D, Degen J, et al. Slow con-

    cardiogenesis   &   gene expression   169

  • 8/20/2019 Cardiogenesis and the Regulation of Cardiac-Specific Gene Expression

    14/14

    duction and enhanced anisotropy increase the pro-

     pensity for ventricular tachyarrhythmias in adult mice

    with induced deletion of connexin43. Circulation

    2004;109(8):1048 – 55.

    [142] Plum A, Hallas G, Magin T, et al. Unique and shared

    functions of different connexins in mice. Curr Biol2000;10(18):1083 – 91.

    [143] Alcolea S, Jarry-Guichard T, de Bakker J, et al.

    Replacement of connexin40 by connexin45 in the

    mouse: impact on cardiac electrical conduction. Circ

    Res 2004;94(1):100–9.

    [144] Simon AM, McWhorter AR, Dones JA, et al. Heart 

    and head defects in mice lacking pairs of connexins.

    Dev Biol 2004;265(2):369–83.

    [145] Majumdar A, Drummond IA. Podocyte differentia-

    tion in the absence of endothelial cells as revealed in

    the zebrafish avascular mutant, cloche. Dev Genet 

    1999;24(3–4):220–9.

    [146] Lammert E, Cleaver O, Melton D. Induction of 

     pancreatic differentiation by signals from blood

    vessels. Science 2001;294(5542):564– 7.

    [147] Serluca FC, Drummond IA, Fishman MC. Endothe-lial signaling in kidney morphogenesis: a role for 

    hemodynamic forces. Curr Biol 2002;12(6):492–7.

    [148] Hove JR, Koster RW, Forouhar AS, et al. Intra-

    cardiac fluid forces are an essential epigenetic factor 

    for embryonic cardiogenesis. Nature 2003;421(6919):

    172–7.

    [149] Schulte-Merker S, van Eeden FJ, Halpern ME, et al.

    no tail (ntl) is the zebrafish homologue of the mouse

    T (Brachyury) gene. Development 1994;120(4):

    1009–15.

    chen  et al170