The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and Development

Similar documents
Development of the Heart

When you see this diagram, remember that you are looking at the embryo from above, through the amniotic cavity, where the epiblast appears as an oval

Organogenesis Part 2. V. Lateral Plate Mesoderm VI. Endoderm VII. Development of the Tetrapod Limb VIII. Sex Determination. V. Lateral Plate Mesoderm

"Lecture Index. 1) Heart Progenitors. 2) Cardiac Tube Formation. 3) Valvulogenesis and Chamber Formation. 4) Epicardium Development.

Molecular and genetic basis of congenital conotruncal heart defects Rana, M.S.

Heart Development. Origins of congenital heart defects Properties of cardiac progenitor cells. Robert G. Kelly

Heart Development. Robert G. Kelly Developmental Biology Institute of Marseilles - Luminy

Heart Development and Congenital Heart Disease

CARDIAC DEVELOPMENT CARDIAC DEVELOPMENT

DEVELOPMENT OF THE CIRCULATORY SYSTEM L E C T U R E 5

W.S. O The University of Hong Kong

Supplementary Figure S1 Enlarged coronary artery branches in Edn1-knockout mice. a-d, Coronary angiography by ink injection in wild-type (a, b) and

CHAPTER 8 MOLECULAR AND CELLULAR DEVELOPMENT OF THE HEART

Supplementary Figure 1. A microarray screen of organizers compared to non-organizer tissue reveals a putative organizer gene set.

The cardiomyocyte is the fundamental work unit of the. Myocardial Lineage Development

CHAPTER 6 SUMMARIZING DISCUSSION

Development of the heart

Development of the Heart *

Maoqing Ye, Yan Yin, Kazumi Fukatsu, and Paul Grossfeld

W.S. O. School of Biomedical Sciences, University of Hong Kong

Axis Formation and Mesoderm Induction

Since Robert Gross inaugurated pediatric cardiac surgery. Congenital Heart Disease Compendium. Cardiac Regeneration. Lessons From Development

Circulatory system. Lecture #2

6. HEART AND CIRCULATORY SYSTEM I

Heart & vascular system I. Dawei Dong

Pregestational Diabetes and Congenital Heart Defects: Role of Reactive Oxygen Species

The Cardiovascular System (Part I) 黃敏銓 解剖學暨細胞生物學研究所

human anatomy 2016 lecture thirteen Dr meethak ali ahmed neurosurgeon

Cardiovascular System

CARDIOVASCULAR SYSTEM

Polarity and Segmentation. Chapter Two

Congenital Heart Defects

Heart. Heart 2-Tunica media: middle layer (media ='middle') muscle fibers (smooth or cardiac).

CV Anatomy Quiz. Dr Ella Kim Dr Pip Green

The role of Wnt signalling in cardiac development and tissue remodelling in the mature heart

The HEART. What is it???? Pericardium. Heart Facts. This muscle never stops working It works when you are asleep

Middle mediastinum---- heart & pericardium. Dep. of Human Anatomy Zhou Hongying

LECTURE 5. Anatomy of the heart

Ch.15 Cardiovascular System Pgs {15-12} {15-13}

Co-ordinating Notch, BMP, and TGFβ Signalling During Heart Valve Development

the Cardiovascular System I

2. right heart = pulmonary pump takes blood to lungs to pick up oxygen and get rid of carbon dioxide

The Heart. Happy Friday! #takeoutyournotes #testnotgradedyet

4. The two inferior chambers of the heart are known as the atria. the superior and inferior vena cava, which empty into the left atrium.

Cardiovascular System Notes: Physiology of the Heart

Development and teratology of cardiovascular and lymphatic systems. Repetition: Muscle tissue

The heart & Cardiovascular system

Signaling Vascular Morphogenesis and Maintenance

Embryology of the Heart

Heart 3: Organogenesis, CHD, prenatal circulation

The cardiovascular system

6. Development of circulatory system II. Cardiac looping. Septation of atria and ventricles. Common heart malformations.

Major Function of the Cardiovascular System. Transportation. Structures of the Cardiovascular System. Heart - muscular pump

Lab Activity 23. Cardiac Anatomy. Portland Community College BI 232

Cardiac Conduction System

Cardiovascular Physiology. Heart Physiology. Introduction. The heart. Electrophysiology of the heart

The Heart. The Heart A muscular double pump. The Pulmonary and Systemic Circuits

THE HEART OBJECTIVES: LOCATION OF THE HEART IN THE THORACIC CAVITY CARDIOVASCULAR SYSTEM

Review. Endocardial and Epicardial Epithelial to Mesenchymal Transitions in Heart Development and Disease

10. Thick deposits of lipids on the walls of blood vessels, called, can lead to serious circulatory issues. A. aneurysm B. atherosclerosis C.

The Cardiovascular and Lymphatic Systems Cardiovascular System Blood Vessels Blood Vessels Arteries Arteries Arteries

Chapter 14. The Cardiovascular System

ULTRASOUND OF THE FETAL HEART

Chapter 13 The Cardiovascular System: Cardiac Function

Pearson's Comprehensive Medical Assisting Administrative and Clinical Competencies

Wnt signaling: an essential regulator of cardiovascular differentiation, morphogenesis and progenitor self-renewal

The Cardiovascular System

Chapter 20 (1) The Heart

Chapter 18 - Heart. I. Heart Anatomy: size of your fist; located in mediastinum (medial cavity)

Segmental Analysis. Gautam K. Singh, M.D. Washington University School of Medicine St. Louis

All About the Heart. Structures of the heart. Layers. Chambers

The Circulatory System. Lesson Overview. Lesson Overview The Circulatory System

Chapter 6. Villous Growth

Notes: 1)Membranous part contribute in the formation of small portion in the septal cusp.

Anatomy lab -1- Imp note: papillary muscle Trabeculae Carneae chordae tendineae

Mr. Epithelium s Anatomy and Physiology Test SSSS

Chapter 20: Cardiovascular System: The Heart

The cardiovascular system is composed of the heart and blood vessels that carry blood to and from the body s organs. There are 2 major circuits:

37 1 The Circulatory System

C. elegans Embryonic Development

THE HEART. A. The Pericardium - a double sac of serous membrane surrounding the heart

1 General Introduction

BME 5742 Bio-Systems Modeling and Control. Lecture 41 Heart & Blood Circulation Heart Function Basics

11/10/2014. Muscular pump Two atria Two ventricles. In mediastinum of thoracic cavity 2/3 of heart's mass lies left of midline of sternum

The Cardiovascular System (Heart)

Read Me. covering the Heart Anatomy. Labs. textbook. use. car: you

CIRCULATION & GAS EXCHANGE

Cardiac Cycle. Each heartbeat is called a cardiac cycle. First the two atria contract at the same time.

Ch 19: Cardiovascular System - The Heart -

Vasculature and innervation of the heart. A. Bendelic Human Anatomy Department

Anatomy of the Heart. Figure 20 2c

Introduction to Anatomy. Dr. Maher Hadidi. Bayan Yanes. April/9 th /2013

Class XI Chapter 18 Body Fluids and Circulation Biology

Collin County Community College. ! BIOL Anatomy & Physiology! WEEK 5. The Heart

WNT2 and WNT7B Cooperative Signaling in Lung Development

The Cardiovascular System

Section 5.1 The heart and heart disease

The Cardiovascular and Lymphatic Systems

MODULE 2: CARDIOVASCULAR SYSTEM ANTOMY An Introduction to the Anatomy of the Heart and Blood vessels

THE VESSELS OF THE HEART

Transcription:

This Review is the introduction of a new thematic series on Wnts in Cardiovascular Development and Disease, which includes the following articles: An Updated Overview on Wnt Signaling Pathways: A Prelude for More [2010;106:1798 1806] The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and Development Wnt Signaling in Vascular Progenitor Cells and Angiogenesis Wnt Signaling in Cardiac Hypertrophy and Remodelling Wnt Signaling in Heart Failure and Aging Wnt Signaling and Stem Cells Michael Kühl, Guest Editor The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and Development Susanne Gessert, Michael Kühl Abstract: Understanding heart development on a molecular level is a prerequisite for uncovering the causes of congenital heart diseases. Therapeutic approaches that try to enhance cardiac regeneration or that involve the differentiation of resident cardiac progenitor cells or patient-specific induced pluripotent stem cells will also benefit tremendously from this knowledge. Wnt proteins have been shown to play multiple roles during cardiac differentiation and development. They are extracellular growth factors that activate different intracellular signaling branches. Here, we summarize our current understanding of how these factors affect different aspects of cardiogenesis, starting from early specification of cardiac progenitors and continuing on to later developmental steps, such as morphogenetic processes, valve formation, and establishment of the conduction system. (Circ Res. 2010;107:186-199.) Key Words: Wnt cardiac differentiation cardiac development Wnt proteins are growth factors that function during embryonic development and in the adult organism by regulating diverse cellular processes such as gene transcription and cell proliferation, migration, polarity, or division. 1 Not surprisingly, Wnt proteins are also involved in cardiac development and differentiation. To understand the function of Wnts during cardiogenesis, a basic knowledge of how Wnt proteins transduce their signal is a prerequisite. Consistently, we start with a short introduction into Wnt signaling; for a more in-depth discussion of Wnt signal transduction pathways, more sophisticated recent reviews have been published. 2 4 Wnt Signaling Pathways: A Short Synopsis On a molecular level, members of the Wnt family can, in principle, activate different intracellular signaling pathways. Recent data suggest that these pathways are highly interconnected and, in fact, represent one complex signaling network. 2,3 The classic canonical Wnt signaling pathway involves the multifunctional protein -catenin. -Catenin is well known for its function in cell adhesion through interaction with transmembrane proteins of the cadherin family but functions here as a signaling mediator. In absence of Wnt, cytosolic -catenin is interacting with the destruction complex composed of axin, APC, and GSK3, and thereby is phosphorylated and subsequently degraded by the proteasome. In the presence of a Wnt ligand, the destruction complex is disassembled, and -catenin is stabilized, enters the nucleus, and regulates target gene transcription through interaction with TCF/LEF transcription factors. Through particular target genes such as c-myc and cyclind1, the Original received April 6, 2010; revision received May 28, 2010; accepted June 15, 2010. In May 2010, the average time from submission to first decision for all original research papers submitted to Circulation Research was 14.6 days. From the Institute for Biochemistry and Molecular Biology, Ulm University, Germany. Correspondence to Michael Kühl, Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, D-89081 Ulm, Germany. E-mail michael.kuehl@uni-ulm.de 2010 American Heart Association, Inc. Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.110.221531 186

Gessert and Kühl Wnt in Cardiac Differentiation 187 canonical Wnt/ -catenin pathway is also linked to cell proliferation control. Well-known extracellular inhibitors of the Wnt/ -catenin signaling branch are the secreted proteins Dkk1 and Crescent. In contrast, noncanonical Wnt signaling branches are independent of -catenin and involve diverse intracellular mediators. In the Wnt/JNK pathway, GTPases of the rho family are upstream of jun-n-terminal kinase (JNK), whereas in the Wnt/Ca 2 pathway, an intracellular release of calcium ions results in the activation of calcium-sensitive enzymes such as protein kinase (PK)C, calcium/calmodulindependent kinase (CaMK)II, or calcineurin (CaCN). Noncanonical Wnt pathways have often been linked to cell polarity and cell migration caused by cytoskeletal rearrangements, although examples of an effect on gene expression are also known and discussed below. Of note, noncanonical Wnt signaling has been shown to be inhibitory for canonical Wnt signaling through multiple mechanisms. 5 Figure 1 provides an overview of Wnt signaling pathways as required for this review. In the past, individual Wnt ligands and their receptors of the Frizzled family have been linked to either canonical or noncanonical Wnt signaling pathways, eg, Wnt3a mostly couples to -catenin signaling, whereas Wnt5a and Wnt11 mainly trigger noncanonical Wnt signaling. Recently, however, exceptions to this rule have been described. 2 4 An analysis of Wnt function in a particular context therefore should also aim to uncover the underlying molecular mechanism. Below, we focus on the role of Wnt signaling during different aspects of cardiac development. Wherever known, the intracellular downstream effectors are also highlighted. Non-standard Abbreviations and Acronyms APS AV Bry CaCN CaMKII CCAVC CNCC CPC E ES FHF IFT JNK MV OFT PCP PKC SHF TV aorticopulmonary septum atrioventricular Brachyury calcineurin calcium/calmodulin-dependent kinase II complete common atrioventricular canal cardiac neural crest cell cardiac progenitor cell embryonic day embryonic stem first heart field inflow tract, sinus venosus jun N-terminal kinase mitral valve outflow tract planar cell polarity protein kinase C second heart field tricuspid valve Early Heart Development: From a Common Progenitor to Different Lineages The mammalian heart is one of the first functional organs that develops during embryogenesis to already provide the growing embryo with sufficient nutrients and oxygen. Formation Figure 1. A synopsis of Wnt signaling pathways. A, Canonical Wnt or Wnt/ -Catenin pathway. In the absence of Wnt, -catenin is binding to the destruction complex consisting of axin, APC, and GSK3 and is thereby phosphorylated. This leads to the degradation of -catenin by the proteasome. In the presence of Wnt, Wnt interacts with its receptor complex Fz and Lrp6. This results in the activation of dsh and in the ablation of axin from the destruction complex, which thereby disintegrates. -Catenin is accumulating in the cytosol and activating the expression of target genes through its interaction with the TCF/LEF transcription factors. Dkk1 and Crescent are extracellular inhibitors of Wnt/ -catenin signaling. B, Noncanonical Wnt/JNK pathway. The interaction of Wnt with Fz leads to the activation of dsh in the cell. dsh can activate the small GTPases rhoa and rac, which thereby activate Rho kinase and JNK. This results in a rearrangement of the cytoskeleton. JNK can also activate AP1, which participates in the regulation of gene transcription. C, Wnt/ Ca 2 pathway. In this pathway, the binding of Wnt to the Fz receptor results in an intracellular Ca 2 increase. Ca 2 thereby can activate CaMKII, PKC, and CaCN. CaMKII itself is regulating TAK1 and NLK, whereas CaCN is regulating NF-AT, which plays a role in transcriptional gene regulation.

188 Circulation Research July 23, 2010 of the vertebrate heart can be subdivided into distinct but partially overlapping phases such as specification of cardiac progenitors and formation of the linear heart tube by cell migration and morphogenetic movements, followed by cardiac looping, chamber formation, septation, and maturation. During all of these phases, Wnt ligands, Frizzled receptors, or extracellular Wnt inhibitors are expressed in relevant tissues (see Table 1). Note that, as a prerequisite for intracellular signal transduction, the main intracellular mediators of Wnt signaling are virtually expressed ubiquitously in all cells. Our current view of early cardiac development is based on gene expression studies in different organisms, cell tracking in genetically modified mice, and classic lineage-labeling experiments in chicken, mice, and frogs (Figure 2). In mice, the cardiac progenitor cells that give rise to the linear heart tube derive from the cardiac crescent at embryonic day (E)7.5 of development, also called primary heart field or first heart field (FHF). These cells will mainly contribute to the left ventricle. To form a mature heart, the linear heart tube subsequently needs to expand. This is achieved by 2 mechanisms, cell proliferation and recruitment of additional cells. The latter was shown by lineage labeling experiments in avian and mouse embryos where cells are added to the arterial and venous poles of the linear heart tube. 6 9 These cells originate in the second heart field (SHF), a cardiac precursor cell population distinct from the first heart field. In mice, different Cre-driver lines using regulatory elements of important cardiac genes such as Islet1, FGF10, and Mef2C were used to describe this second lineage of cardiac precursor cells. 10 13 An Isl1 Cre-driver line indicated that cells of the SHF will mainly contribute to the outflow tract (OFT) and the right ventricle, as well as to both atria. 10 Interestingly, by use of some regulatory elements of the Mef2C gene only cells of the OFT and the right ventricle can be labeled indicating that the SHF field can be further subdivided into diverse subpopulations. 13 Furthermore, it should be noted that the use of these different Cre-driver lines to manipulate heart development might result in slightly different results thereby reflecting the timing, tissue specificity and penetrance of a given Cre driver. Also, in chicken, it has been shown that cells of the SHF populate the right ventricle. 14 In frogs, that have just one ventricle, cells of the SHF exclusively end up in the OFT. 15 The LIM homeodomain transcription factor Islet (Isl)1 has become a popular molecular marker demarcating the SHF. By gene expression, cells of the SHF are located more medial and posterior to the FHF at E7.5, and later are found more dorsally to the heart tube at E8.0 in the mouse embryo (Figure 2). 10,16 FHF progenitors in contrast express the T-box transcription factor Tbx5. The homeobox transcription factor Nkx2.5 is found in both populations. 15 Retrospective clonal analyzes indicated that cells of both heart fields derive from one common cardiac progenitor cell population (CPC) and rather represent different cardiogenic lineages than unbridgeable cell populations. According to this model, both lineages differ with respect to the onset of terminal differentiation, 17,18 with cells of the second heart field lineage entering terminal differentiation later than those of the first heart field lineage. Consistently, cells of the second lineage express Isl1 longer than those of the first Table 1. Expression of Wnt Ligands and Extracellular Wnt Inhibitors at Different Steps of Cardiogenesis Gene Expression Wnt2 Mouse: cardiac crescent (FHF/SHF?) Mouse EB: Flk1 subpopulation 46 Wnt2b Mouse: cardiac crescent (FHF/SHF?), primordium of OFT, atrioventricular region of linear heart tube Chicken: tubular heart Mouse EB: Flk1 subpopulation 69 Wnt3a Mouse: primitive streak Chicken: primitive streak Wnt4 Chicken: primitive streak Mouse EB: Flk1 subpopulation 46 Wnt5a Mouse: node, primitive streak, primordium of the OFT, OFT, common ventricle Chicken: Hensen s Node, tubular heart Xenopus: maternally stored Mouse EB: Flk1 subpopulation, 69,129 Mesp1 subpopulation 44 Wnt6 Chicken: atrioventricular region Xenopus: ectoderm overlying developing heart, endocardium of heart tube, OFT and atriventricular region 87 Wnt7a Chicken: tubular heart, primitive conduction system Wnt8, Wnt8a Mouse: primitive streak, tubular heart Chicken: primitive streak Xenopus: marginal zone Wnt9, Wnt9a chicken: tubular heart, OFT, atrioventricular canal Wnt11 Mouse: node, cardiac crescent (FHF/SHF?), linear heart tube, OFT myocardium, ventricle 76 Chicken: Hensen s Node, precardiac mesoderm, primitive conduction system Xenopus: maternally stored, Spemann Organizer, marginal zone, precardiac mesoderm; Wnt11-R: FHF, linear heart tube 15,88 Mouse EB: upregulated during cardiac differentiation, 32,60 Flk1 subpopulation Dkk1 Chicken: Hensen s Node, primitive streak Xenopus: Spemann organizer, Endoderm adjacent to the anterior SHF Mouse EB: Flk1 subpopulation, 69 Mesp1 subpopulation 44,50 Fz2 Mouse: CNCC Fz3 Xenopus: CNCC Fz7 Xenopus: common cardiac progenitor Fz8 Xenopus: first heart field Fz9 Mouse: adult heart Crescent Xenopus: Spemann Organizer Sfrp1 Mouse EB: Flk1 subpopulation 46 Sfrp2 Chicken: common ventricle Xenopus: Spemann Organizer Sfrp3 (FrzB) Chicken: precardiac mesoderm, OFT, atrioventricular region Xenopus: Spemann Organizer, cardiac cushions Sfrp5 Mouse EB: Flk1 subpopulation 46 For detailed references and more information concerning expression domains, see also the recent reviews. 126 128 This list is not exclusive because, for some genes, a potential cardiac expression has not been analyzed. The main intracellular mediators of Wnt signaling are present in virtually all cells and therefore are not listed here. EB indicates embryoid body.

Gessert and Kühl Wnt in Cardiac Differentiation 189 Figure 2. Schematic overview of murine cardiac development. A through E, Anterior is at the top; posterior at the bottom. A, 7.0. The cardiac progenitor cells migrate after their ingression through the primitive streak toward the anterior side of the embryo, where they form 2 populations of cardiac progenitor cells (blue). B, E7.5. The heart-forming cell populations can be subdivided into 2 different heart field lineages, a first heart field (FHF) (also called cardiac crescent at this embryonic day; blue) and a second heart field (SHF) (red). C, Cells of the FHF form the primary heart tube (PHT) (blue). The arterious pole is at the top; the venous pole at the bottom. D, Lateral view of a mouse embryo at E8.0. Cells of the FHF form the primary heart tube (PHT), cells of the SHF migrate into the PHT from both sides of the heart to allow a ballooning of the heart. E, Looping process of the linear heart tube at E8.5. The heart tube loops to the right (black arrows). The outflow tract (OFT) arises at the arterious pole; the inflow tract (IFT) at the venous pole. F and G, Sections through hearts at E9.5 (F) and E10.5 (G). The heart is looped to the right and consists of the epicard (blue), the myocard (green), and endocard, as well as cardiac jelly (orange). Cells of the cardiac jelly are produced by the endocard undergoing epithelial mesenchymal transition (EMT) and represent a thick extracellular layer between the endocard and the myocard. The ventricular chambers start to develop trabeculae. The endocard and the cardiac jelly extends into the heart lumen forming the cardiac cushions and later on the cardiac valves (see I). The separations of the atrial, as well as ventricular chambers, starts by forming the primary atrial septum (PAS) and the interventricular septum (IVS). H, Function of the cardiac neural crest during heart development shown at E12.5. Dorsal is at the top; anterior to the left. The cardiac neural crest originates at the dorsal neural tube between the middle portion of the otic vesicle (O) and the third somite (S3) as part of the cranial neural crest. Afterward these cells migrate to the ventral side through the pharyngeal arches 3, 4, and 6 toward the OFT. In the heart, the cardiac neural crest cells are responsible for the OFT septation in pulmonary trunk (P) and aorta (A). I, Development of the cardiac pacemaking and conduction system. Transverse sections through a heart at E14.5. The 4 heart chambers are completely separated by the atria, as well as ventricular septum (AS and VS) and the valves (TV and MV). The contraction impulse originates in the sinuatrial node (SA node) and is then transmitted to the atrioventricular (AV) node, both located in the right atrium. Afterward, the impulse is sent to the ventricular myocardium by the His bundles, bundle branches and Purkinje fibers with a slight delay to allow a coordinate beating of the different cardiac chambers. A indicates aorta; AA, aortic arch arteries; AS, atrial septum; APS, aorticopulmonary septum; LA, left atrium; LV, left ventricle; MV, mitral valve; NT, neural tube; O, otic vesicle; P, pulmonary trunk; RA, right atrium; RV, right ventricle; S, somite; SA, sinuatrial; TV, tricuspid valve; VS, ventricular septum. lineage. Experiments in differentiating embryonic stem (ES) cell cultures revealed that this common progenitor likely is positive for Isl1, the homeobox transcription factor Nkx2.5, and the VEGF receptor tyrosine kinase Flk1 (Figure 3). Immunohistochemistry studies indicated the presence of these CPCs also in the developing mouse embryo. Clonal analyzes demonstrated that these CPCs give rise to different cell types of the mature heart, including cardiomyocytes, smooth muscle cells, and endothelial cells (Figure 3). 19 21 Specification of these cardiac progenitor cells starts at the onset of gastrulation in the mesodermal germ layer. In chicken and mouse, this happens during the bilateral ingression of cells through the anterior portion of the primitive streak in close proximity of the node. The earliest cardiacspecific marker genes are the transcription factors Mesp1 and Mesp2, that are also required for heart development. 22 These cells migrate to the anterior part of the embryo and contribute to cells of both cardiogenic lineages (Figure 2). Similar in Xenopus, cardiac progenitor cells are specified bilaterally at either side of the Spemann organizer, the equivalent structure to the node in chicken and mice. During gastrulation, these cells migrate toward the anterior and ventral side of the embryo and form there a crescent-shaped population of cardiac progenitor cells for both lineages. 15 Explant experiments in Xenopus indicated that cardiac tissue is induced by signals from the Spemann organizer and the underlying endoderm. 23 25 The size of the Xenopus embryo and the knowledge of the spatial origin of cardiac inducing activities allows to analyze the identity of these molecules in sophisticated explant studies to which we refer further below. 26 Wnt Signaling During Specification of the Common Cardiac Progenitor Initial Experiments, Contradicting Results The first data concerning the function of Wnt/ -catenin signaling during cardiac development in vertebrates were contradictory and confusing. The positive involvement of Wnt/ -catenin signaling in cardiogenesis was known for

190 Circulation Research July 23, 2010 Figure 3. The multiple faces of Wnt signaling during early cardiac development. A, Wnt signaling is required for early cardiogenic development. The canonical Wnt pathway is highlighted in red, noncanonical Wnt pathways in blue. The diverse cell types are depicted by gray circles; genes active or inactive in these cells are described. For the detailed description of the different steps, see the text. Candidates for the inducing factors delivered by endodermal cells are nodals, FGFs, and cerberus. B, The 4 main phases in early cardiac development. The actions of canonical (red) and noncanonical (blue) Wnt pathways are depicted. A indicates atrium; V, ventricle; WT1, Wilms tumor 1. some time in Drosophila. 27 It was therefore quite surprising to find that activation of canonical Wnt signaling in anterior mesoderm suppresses cardiac differentiation in chicken and Xenopus embryos whereas inhibition of the pathway through inhibitors such as Crescent or Dkk1 favors cardiac development. 28,29 Note that Crescent, a member of the Sfrp family of Wnt inhibitors, is not present in mammals. The endoderm specific knockout of -catenin in the mouse even results in the formation of multiple, ectopic hearts. 30 In contrast, cell culture based experiments suggested a positive role for canonical Wnts in early cardiac specification. In P19 teratocarcinoma stem cells interference with canonical Wnt signaling leads to a decrease in cardiac specific gene expression. 31 This apparent conflict of diverse roles of Wnt/ -catenin signaling has been resolved by the observation that Wnt signaling can have different effects depending on the time of action. An example is a recently published study in heat shock inducible transgenic zebrafish embryos indicating that an application of Wnt8 before gastrulation results in more, whereas an application after gastrulation results in less cardiomyocytes. A reverse effect could be achieved by the timed delivery of the Wnt inhibitor Dkk1. 32 Recent publications confirmed these observations in mouse ES cells. In this system, the expression of some canonical Wnt ligands starts slightly earlier than the expression of cardiac genes. 32,33 Inhibition of canonical Wnts at this early time point leads to an inhibition of cardiac differentiation and a reduction in contractile areas within embryoid bodies at later time points. 33 These findings raised a model in which Wnt/ catenin signaling was suggested to have a biphasic function during cardiogenesis. 32 34 In a variation, a Wnt/ -catenin signaling gradient in time was recently suggested with the highest requirement for -catenin signaling early during development. 35 More novel findings indicated that even this biphasic model is still too simple. A more advanced model has to integrate different cardiac progenitor cell populations as described by their molecular signature of expressed genes and the time of their appearance during development as well as the different cellular effects of Wnt signaling such as differentiation, proliferation, and migration. Indeed, specification of the common cardiac progenitor, which is characterized by the expression of Isl1, Nkx2.5 and Flk1, is a multistep process that involves generation of mesodermal progenitor cell and subsequent intermediate cell populations (Figure 3). Furthermore, cell-autonomous and nonautonomous effects have to be considered as many experiments were done in ES cell cultures harboring different but communicating cell types or by non tissue-specific gain or loss-of-function experiments in frogs or chicken. Here, isolation and analysis of defined progenitor cell populations as defined by specific markers or tissue specific modulation of Wnt signaling by use of trans-

Gessert and Kühl Wnt in Cardiac Differentiation 191 genic mice are the gold standard for further investigations. Based on such experiments, a complex picture of Wnt signaling during cardiac differentiation emerges. Still, the main challenge here is to integrate data from different model systems such as ES cells, Xenopus, zebrafish and genetically modified mice into a common and consistent model. Induction of the Mesodermal Germlayer: Requirement for Wnt/ -Catenin Signaling Cardiac progenitor cells are specified in the mesodermal germ layer early during development. Many studies have shown that the formation of mesoderm is dependent on canonical Wnt signaling. The knockout mouse of -catenin, the central player of canonical Wnt signaling, fails to generate mesodermal tissue. 36 Also the loss of Wnt3a function is accompanied by an absence of mesoderm-specific marker genes. 37 In murine ES cells, the early inhibition of the canonical Wnt pathway blocks the expression of mesodermspecific marker genes. 38 The T-box transcription factor Brachyury (Bry) is a panmesodermal marker gene and required for posterior mesoderm formation. In different organisms, it has been shown that mesoderm formation depends on Wnt/ -catenin signaling using Bry expression as a marker. 39,40 This regulation is likely direct because the authors could demonstrate a binding site for TCF in the Bry promotor. These data indicate a strong requirement of canonical Wnt signaling during mesoderm induction in the early embryo, a prerequisite for the formation of cardiac progenitor cells. Cardiac Specification: Negative Influences of Canonical Wnt Signaling The earliest marker gene for the cardiovascular lineage is the basic helix loop helix (bhlh) transcription factor Mesp1. Functional studies could demonstrate an important function of Mesp1 during cardiac development. 41,42 An in-depth analysis of Mesp1-regulated genes indicated that Mesp1 is on top of the cardiovascular lineage upstream of other important markers for the common cardiac progenitor such as Flk1 or Isl1. 43,44 At the same time, Mesp1 represses essential genes for the hematopoietic cell lineage. The receptor tyrosine kinase Flk1 is an important marker for early mesoderm development. Two phases of Flk1 expression have been defined in ES cell culture experiments. 21,45 Those cells expressing Flk1 early (Bry /Flk /Mesp1 ) are fated to become the hemangioblast that further develops into endothelial cells and the blood lineage. A second wave of Flk1 expression slightly later results in a progenitor cell for the cardiac lineage (Bry /Flk1 /Mesp1 ) (see also Figure 3). Consistent with the above-mentioned data concerning the function of Wnt/ -catenin signaling during mesoderm formation, depletion of Wnt/ -catenin signaling results in a loss of Mesp1 along with a downregulation of Flk1 expression. 38 Recently, it was shown that Notch signaling redirects Flk1 cells fated to become hemangioblasts toward Flk1 cells of the cardiogenic lineage. 46 An in-depth analysis of this observation indicated that Notch4 activation in Bry /Flk cells results in an upregulation of molecules inhibiting Wnt/ -catenin signaling such as the secreted Frizzled-related proteins Sfrp1 or Sfrp5. Indeed, the addition of Wnt3a but not of Wnt5a completely abolished this effect of Notch4. This diverse behavior of Wnt3a and Wnt5a, together with the knowledge of Wnt3a activating -catenin during mesoderm formation, makes it likely that Wnt5a acts through -catenin independent noncanonical Wnt signaling branches in this context, an argument that is further substantiated below. The interplay between Notch and -catenin signaling was also analyzed independently by the Srivastava laboratory. 47 Here, the authors overexpressed a stabilized variant of -catenin in Isl1 CPCs, resulting in a downregulation of several important cardiac genes such as Isl1, myocardin, shh, or Smyd1 at E9.0 of mouse development. Similarly, a cardiac-specific loss of Notch1 resulted in severely affected right ventricular structures. It should be kept in mind that this requirement for Notch signaling and the negative influence of -catenin signaling on differentiation is significantly later during development than described in the study of the Keller laboratory. 46 Both studies, however, indicate that Notch mediated inhibition of canonical Wnt signaling is a recurrent motive during cardiomyocyte differentiation. In line with these data is the observation that Flk1 /CXCR4 /VE-cadherin cells isolated from an OP9 coculture system can give rise to cardiomyocytes under certain conditions. In this setting, inhibition of Wnt/ -catenin signaling favors the formation of cardiomyocytes. Wnt3a inhibits this process. 48 In accordance with the inhibitory role of Wnt/ -catenin signaling during cardiac development, it has recently been shown that -catenin negatively regulates the expression of GATA6, one of the major early cardiac transcription factors. 49 Taken together, all of these data show that during specification of cardiac precursor cells the downregulation of Wnt/ -catenin signaling is essential. These findings are furthermore in line with the observation that the canonical Wnt inhibitor Dkk1 can improve the differentiation of Mesp1-induced cardiomyogenesis. 44 The question of whether Dkk1 is regulated directly by Mesp1 is controversial. In the search for Mesp1 target genes, Dkk1 was identified by chromatin immunoprecipitation experiments and a direct binding of Mesp1 to the Dkk1 promotor has been shown. 50 In ES cells overexpressing Mesp1, Dkk1 is upregulated, whereas in Mesp1 / /Mesp2 / embryos, Dkk1 expression was dramatically downregulated. 50 However, these findings have recently been challenged. 44 Dkk1 was found to be upregulated by Mesp1 but this was not evident before 24 hours after Mesp1 activation, suggesting a more indirect effect of Mesp1 on Dkk1 expression. 44 Moreover, a direct activation of Dkk1 by Mesp1 was not confirmed in a third study. 43 These discrepancies might be explained by different technical approaches or levels of Mesp1 overexpression in ES cells and require further investigations. Interestingly, Dkk1 has been shown to have more functions than just inhibiting Wnt/ -catenin signaling, in particular, activating JNK signaling 51,52 or a so-far unknown signaling pathway. 53 Non Cell-Autonomous Roles of Wnt/ -Catenin Signaling During Precardiac Specification: A Role for Endodermal Tissue Explant techniques in Xenopus suggested a role for the Spemann organizer and the underlying anterior endoderm for

192 Circulation Research July 23, 2010 cardiac induction in vivo. 23 25 This has also been analyzed recently in greater detail by the help of sophisticated combinations of anterior endoderm explants with animal caps representing a population of pluripotent progenitor cell populations. 26 In this assay, cardiac induction by anterior endoderm was independent of -catenin signaling. These data are in line with previous data indicating that the cardiac inducing activity of Dkk1 in Xenopus is attributable to the generation of Hex1-positive endodermal cells. 54 In murine embryonic stem cells, -catenin signaling has been shown to be essential for Sox17 expression. Sox17 expression itself is required for cardiogenesis in a non cell-autonomous role, thus likely being required in endodermal tissue. Consistently, loss of Sox17 results in the downregulation of Hex1 and the endodermal marker genes Foxa1 and Foxa2. 55 Thus, also in the endoderm, positive and negative roles of Wnt/ -catenin signaling have been described: a positive role for the formation of Sox17 positive progenitor cells; a negative for the formation of likely Sox17 /Hex1 cells (Figure 3). The endoderm then releases factors initiating cardiogenesis in the mesodermal tissue (FGF, nodal, Cerberus) that can act in parallel. 26,56 Interestingly, Sox17 knockout mice do not show a severe cardiac phenotype. 57 Of note, cardiac differentiation was only examined by analyzing GATA4 expression that is independent of Sox17 function. 55 More detailed molecular analysis of Sox17 knockout mice, in particular taking into account the novel concept of 2 cardiac lineages, might therefore be of interest. In addition, it might well be that other factors can compensate the loss of Sox17 during cardiac induction in the embryo. Cardiac Specification: Positive Influence of Noncanonical Wnt Signaling Noncanonical Wnt signaling also contributes to correct cardiac specification in the mesodermal germ layer. The best investigated factor to this end is Wnt11. In Xenopus, Wnt11 is maternally expressed in the presumptive dorsal mesoderm and in the dorsal marginal zone during gastrulation. Loss-offunction experiments demonstrated that Wnt11 is required for normal heart development and cardiac marker gene expression. 49,51 Overexpression of Wnt11 in Xenopus explants is sufficient to induce contractile tissue formation. 51 This cardiac promoting activity of Wnt11 has also been shown in other model systems such as quail mesoderm, 58,59 murine ES cells, 32,60 endothelial progenitor cells 61 and mesenchymal or bone marrow derived stem cells. 62 64 The cardiac supporting activity of Wnt11 could be linked to noncanonical Wnt signaling branches. 51,61,62 In Bry /Flk1 cardiogenic progenitor cells noncanonical Wnt ligands are upregulated. In particular, Wnt5a functions in synergy with BMP6 and Sfrp1 to promote formation of troponin-positive cells, likely through noncanonical Wnt signaling activities, 46 whereas treatment with either single factor did not result in significant cardiomyocyte formation. This indicates that Wnt5a might be supportive but not required for cardiogenesis and might explain why Wnt5a knock mice do not display a cardiac differentiation phenotype. Nevertheless, this finding is of particular interest because Wnt5a is upregulated by Mesp1 44 and has also been suggested to function during cardiogenesis in an independent ES cell based approach. 65 Like in the ES cells system, in endothelial progenitor cells (EPCs), a cardiogenic program can also be initiated by activating Notch signaling through overexpression of the intracellular notch domain (NICD). Interestingly, this is accompanied by an upregulation of Wnt5a. Cardiogenesis in EPCs can also be triggered by cocultivation with neonatal rat cardiomyocytes and this effect was sensitive to -secretase inhibition thereby downregulating Notch signaling. 66 Thus, upregulation of noncanonical Wnt ligands in response to either Notch signaling or Mesp1 activation might be a common theme during cardiac development. The downstream signaling activity of these 2 Wnt ligands, Wnt5a and Wnt11, has been analyzed by different groups in more detail. The ability of Wnt11 to induce cardiac tissue in Xenopus is impaired by inhibiting JNK or PKC. Vice versa, coactivation of JNK and PKC in embryonic explants activates cardiac marker gene expression. 51 Similar in endothelial progenitor cells, mesenchymal stem cells or bone marrow stem cells, triggering a cardiogenic program by Wnt5a/Wnt11 involves activation of PKC. 61,62,64 In a recent study, the type of PKC likely involved has been determined. In endothelial progenitor cells, activation of a cardiogenic program is increased on treatment with the PKC activator phorbol-12-myristate-13- acetate (PMA). This excludes the involvement of atypical PKCs that are PMA-independent. By use of isoform-specific PKC inhibitors, the involvement of PKC but not PKC downstream of Wnt5a has been demonstrated. 67 It might be appropriate here to discuss the requirement of Wnt2b for cardiogenesis. 68 Wnt2b was initially found to be highly expressed in Flk1 progenitor cells 69 and subsequent generation of Wnt2b / ES cells indicated a requirement for Wnt2b for the cardiogenic but not for hematopoietic lineage. 68 More detailed analyses of these cells, however, indicated that the loss of Wnt2b is accompanied by an accelerated expression of Bry and Flk1. This is an interesting observation given the positive role of Wnt/ -catenin signaling for Bry expression, as discussed above. Furthermore, activating Wnt/ -catenin signaling during early differentiation of ES cells has also been linked to accelerated differentiation of ES cells by others. 70 Because, in this respect, the loss of Wnt2b function has an identical phenotype to the Wnt3a gain of function, this strongly argues for the fact that Wnt2b might activate a noncanonical Wnt pathway in this context. Of course, this awaits further analyses. Interestingly, Wnt2b and Wnt11 are both direct target genes of GATA6, 49,71 and both are coexpressed in early mouse 72,73 but not in Xenopus embryos. 74,75 This putative functional redundancy might explain why a loss of Wnt11 results in an early cardiac phenotype in Xenopus 49,51 but not in the mouse with later phenotypes. 76,77 Slightly later, however, it might be Wnt2b that regulates proliferation of the common cardiac progenitor through a Wnt/ -catenin pathway. 78 Lineage-Specific Requirements of Wnt Signaling SHF-Specific Manipulations of -Catenin Signaling At E8.0 of mouse development, cells of the SHF lineage are found dorsally to the primarily formed tubular heart and can

Gessert and Kühl Wnt in Cardiac Differentiation 193 enter the heart tube either from the anterior site giving rise to the right ventricle and the OFT or they can enter from the posterior site contributing to the atria and the inflow tract region. Several studies could show a specific role for the canonical Wnt signaling pathway in the second heart field lineage and SHF-derived structures. The tissue-specific downregulation of -catenin in the SHF using different Cre mouse lines leads to a reduction of Isl1-positive cells in comparison to wild-type mice as well as a loss of the SHF-derived OFT and right ventricle. 79 83 Moreover, it has been shown by chromatin immunoprecipitation experiments that -catenin is directly binding and positively regulating the Isl1 promotor 83 whereas others reported a negative regulation. 47 Also, FGF ligands were supposed to be downstream of -catenin signaling. 81 The overexpression of -catenin, however, results in an expansion of SHF-derived structures. 79,82 These phenotypes were clearly linked to an effect of Wnt/ catenin signaling on cell proliferation. Also in the posterior part of the SHF Wnt signaling regulates cell proliferation. Here Wnt2b has been identified as a ligand. Wnt2b / embryos display smaller and thinner atria, a loss of the primary atrial septum and defects in the AV cushion resembling a human congenital condition called complete common atrioventricular canal (CCAVC) or atrial septum defects. Also, the pulmonary vein is affected. 78 Taken together, all of these data indicate a positive effect of the canonical Wnt signaling pathway in the development of SHF-derived structures in the mature heart. Nkx2.5-Cre Specific Manipulation of -Catenin Signaling A specific depletion of -catenin in the FHF lineage has not been performed so far. The best evidence for a role of -catenin signaling in this lineage has been gained from mice depleting -catenin function with a specific Nkx2.5 enhancer that is not active in the cardiac crescent at E7.5 but later is expressed in both ventricular chambers. This resulted in smaller ventricles and reduced proliferation. In contrast, overexpressing a stabilized version of -catenin under the control of the same enhancer resulted in enlarged ventricular chambers, more cells, and upregulated expression of cyclind2. 82 Because both chambers were affected, these phenotypes strongly argue that in both cardiac lineages Wnt/ catenin signaling supports the expansion of committed cardiac progenitor cells. In contrast, deleting -catenin using a Mesp1 driver mainly affected the SHF but not the FHF. This would suggest that the FHF lineage is not requiring -catenin signaling. 80 The differences are likely explained, however, by the different timing and tissue specific expression in which the different Cre drivers are active. Wnt Signaling During Terminal Differentiation Different evidences indicate that Wnt/ -catenin signaling inhibits terminal differentiation of cardiomyocytes. This has been well documented in ES cell based systems in which the late addition of Wnt3a during embryoid body differentiation resulted in a reduced number of cardiomyocytes. 32,33 Also, differentiation of Isl1 cardiac progenitors was inhibited by the addition of Wnt3a-conditioned media. 84 Interestingly, the above-mentioned overexpression of constitutively active -catenin by use of a SHF-specific enhancer of the Mef2c gene resulted in massive accumulation of Isl1-positive progenitor cells attributable not only to increased proliferation but also to a massive inhibition of terminal differentiation. 84 A novel extracellular inhibitor of Wnt/ -catenin signaling was identified recently as IGFBP-4 (insulin-like growth factors binding protein). In Xenopus, it was shown that IGFBP-4 binds to Frizzled and LRP6, thereby preventing binding of a Wnt ligand. Interestingly, this inhibitor of Wnt/ -catenin signaling is required for terminal differentiation. 85 In line, the intracellular Wnt/ -catenin inhibitor Dbf4 is also required for cardiogenesis. 86 A natural in vivo ligand for this repressive effect of Wnt/ -catenin signaling on cardiogenesis could be Wnt6. 87 Overexpression of Wnt6 in transgenic Xenopus embryos using a heat shock inducible promotor during stages of terminal differentiation resulted in decreased cardiac structures. In contrast, depleting Wnt6 by use of antisense morpholino oligonucleotides resulted in enlarged cardiac structures. Thus, Wnt6 could be required to restrict the cardiogenic field during embryogenesis. A Wnt ligand that shows similar properties in the mammalian systems awaits its discovery. Also, noncanonical Wnt signaling plays important roles during terminal differentiation. In murine ES cells, terminal differentiation of cardiomyocytes is accompanied by a marked increase in Wnt11 expression and adding Wnt11 to differentiating ES cell cultures increases formation of beating foci. 32 In Xenopus 2 Wnt11 homologs have been identified: A maternally expressed Wnt11 (also called Wnt11b) and the later expressed Wnt11-R (also called Wnt11a). 88 As described above, the maternal Wnt11 is involved in the specification of cardiac progenitor cells. In contrast, Wnt11-R expression starts at the onset of terminal differentiation. 15,88 Depletion of Wnt11-R by use of antisense morpholino oligonucleotides resulted in a downregulation of marker genes for terminal differentiation. 89 Interestingly, the cell adhesion molecule DM-GRASP could be identified as a potential noncanonical Wnt target gene in this context. Similar to Wnt11-R, a loss of this cell adhesion molecule also led to defects in terminal differentiation. The phenotype on Wnt11-R depletion could be partially reverted by adding back DM-GRASP into the system indicating that proper cell adhesion is required for terminal differentiation. 89 A Preliminary Model for Wnt Action During Cardiac Specification and Differentiation Taken together, these data draw a complex picture of Wnt signaling during early cardiac specification and differentiation (Figure 3A). Based on these findings, one can describe the involvement of Wnt signaling during cardiac specification and differentiation with 4 phases (see Figure 3B). (1) Wnt/ -catenin signaling is required for mesoderm (and endoderm) formation. Wnt3a is the prime candidate for this function in mice. (2) Wnt/ -catenin signaling needs to be low for cardiac specification and generation of multipotent cardiovascular progenitor cells. Sfrp1 and Sfrp5 might be involved in downregulating Wnt/ -catenin signaling. Noncanonical signaling supports this step. Wnt5a and Wnt11 are good candi-

194 Circulation Research July 23, 2010 dates. (3) Wnt/ -catenin signaling is required for proliferation and expansion of several specified cardiac progenitors. Wnt2b is a good candidate to regulate proliferation of early common progenitor cells and the posterior SHF progenitor. (4) Finally, for terminal differentiation, Wnt/ -catenin signaling has to be low again. Noncanonical Wnt signaling is supportive and again Wnt11 is the prime candidate. From the Tubular Heart Tube to the Four Chambers Wnt Signaling Regulates Cell Adhesion of Cardiomyocytes The process of cardiac morphogenesis includes different processes such as heart tube formation, cardiac looping, chamber formation, and maturation of the heart. For all of these processes, the adhesion of cardiac cells is an important prerequisite. In this context, N-cadherin, a calcium-dependent cell cell adhesion molecule, plays an essential function. On loss N-cadherin, cardiomyocytes develop but cells do not form a heart tube because of defects in cell cell contact formation. 90 The function of N-cadherin depends on its intracellular interaction with - and -catenins. This interaction is also essential for heart development. 91 Noncanonical Wnt signaling has been known for quite some time to regulate cadherin mediated cell adhesion. 92 Wnt11 has been shown to regulate N-cadherin expression in the quail mesodermal progenitor cell line QCE6, 59 and N-cadherin expression and function in neonatal rat cardiomyocytes is regulated by Wnt/Fz2 signaling. 93 Wnt11 knockout embryos display defects in the OFT (see below) 77 but also thinner ventricular walls and less trabeculae. A detailed analysis indicated defects in N-cadherin/ -catenin mediated cell adhesion as well. 76 Similarly, Wnt5a promotes N-cadherin mediated cell adhesion in cardiac myocytes. 94 Besides N-cadherin, the cell adhesion molecule DM-GRASP also has been shown to be regulated by noncanonical Wnt signaling in Xenopus. 89 In Xenopus, loss of Wnt11-R is accompanied by reduced cell adhesion in the heart tube. 88 In addition, we could recently show that a loss of the potential Wnt11-R target gene DM-GRASP also results in reduced cell cell contacts among the myocardial cells followed by cardiac looping defects. 89 Consistently, loss of Wnt11 or Wnt11-R leads to malformed linear heart tubes or later morphogenetic defects in Xenopus. 51,89 In zebrafish, the loss of Wnt11, Wnt5a, and Wnt4 results in cardia bifida and subsequent defects in heart tube formation. 95 These data highlight the functional requirement of noncanonical Wnt signaling for proper cell adhesion, tube formation, looping, and chamber maturation of the heart. Septation and Cardiac Valve Formation Cardiac looping is followed by the formation of the 4 chambers of the heart, which requires different morphogenetic processes: ballooning of the atria and ventricles and remodeling of the inner heart curvature, including trabeculation of the ventricles, chamber septation, and formation of valves. Chamber septation involves the formation of the primary atrial septum (PAS) segregating both atria, as well as the interventricular septum (IVS) dividing the right from the left ventricle. To our knowledge, no data are available indicating any function of Wnt signaling during intraventricular septum formation. In Wnt2b knockout embryos, however, the primary atrial septum does not form. 78 During valve formation, cells of the endocardium located between the atrial and ventricular chambers, the atrioventricular canal, delaminate, undergo an epithelial-mesenchymal transition (EMT), migrate into the cardiac jelly located between endocardium and myocardium, proliferate and form the so-called cardiac cushions. During further development, these cardiac cushions build the cardiac valves, the tricuspid valve (TV), separating the right atrium from the right ventricle and the mitral valve (MV) dividing the left chambers. During valve formation, the canonical Wnt pathway possesses important functions. 96 Analyses for molecular markers of endocardial cushion development identified active Wnt/ catenin signaling as one feature. 97 In zebrafish, loss of APC function, which leads to an accumulation of signaling active -catenin, results in unlooped hearts and excessive endocardial cushions based on an increased proliferation and EMT of endocardial cells. In contrast, overexpression of APC or Dkk1 inhibits cardiac cushion formation. 98,99 In line with the data in fish, Wnt9a and the Wnt inhibitor Frzb1 are expressed in AV endocardial cushions in chicken. Whereas Wnt9a transcripts can exclusively be detected in endocardial cells, Frzb1 expression can also be found in cells undergoing EMT. Wnt9a supports endocardial cell proliferation by activating Wnt/ -catenin signaling, whereas Frzb1 inhibits Wnt9a function. 100 Interestingly, Wnt9b has also been shown to activate Wnt/JNK signaling during kidney development. 101 Thus, it is an attractive hypothesis to assume that Wnt9b might also regulate EMT and the migratory behavior of endocardial cells. Also, in Xenopus, Frzb1 is expressed in cardiac cushions suggesting a conserved function among distinct species. 102 The Neural Crest and the Development of the Outflow Tract Cardiac neural crest cells (CNCCs) are a subpopulation of the cranial neural crest cells and originate between the otic vesicle and the third somite (Figure 2H). CNCCs are specified at the dorsal neural tube and migrate through the pharyngeal arches 3, 4 and 6 along the aortic arch arteries toward the heart. They are required for the septation of the OFT into the pulmonary trunk and the aorta by forming the aorticopulmonary septum (APS). Defects during CNCCs migration result in congenital heart malformations such as persistent truncus arteriosus (PTA), double outlet right ventricle (DORV), tetralogy of Fallot and DiGeorge syndrome. Furthermore, cell-labeling approaches indicated that CNCCs also differentiate into smooth muscle cells of the aortic arch arteries. Wnt signaling pathways have been shown to have a role in both development of CNCC and the OFT itself. The requirement of canonical Wnt signaling in CNCC development has been shown in multiple studies. Wnt1 and Wnt3a are expressed in CNCCs of the neural tube. In mice, loss of

Gessert and Kühl Wnt in Cardiac Differentiation 195 -catenin in Wnt1 positive cells by use of a Wnt1-Cre line results in reduced proliferation of CNCCs. 103 Downregulation of the Wnt coreceptor Lrp6 leads to a reduction of CNCCs in the dorsal neural tube and in the pharyngeal arches. Furthermore, the embryos display DORV, VSD (ventricular septal defect) and OFT cushion hypoplasia. Furthermore, depletion of dsh2 causes defects in OFT development such as PTA and DORV, as well as a downregulation of CNCC-specific marker genes. 104 Tissue-specific loss of -catenin in the CNCCs also leads to OFT malformations and in addition to an inhibited proliferation of CNCCs. 105 As a consequence of dsh2 and -catenin downregulation, the expression of the bicoid-related transcription factor Pitx2 is reduced. Furthermore, the authors could show cyclind2 as a Pitx2 target gene involved in cell cycle. These results match with the observation that Pitx2 depletion in mice results in a similar phenotype, PTA and DORV. These results strongly support the hypothesis that canonical Wnt signaling is essential for cell cycle regulation of CNCC development. On the other hand, noncanonical Wnt signaling also plays an important role during neural crest development. In Xenopus, noncanonical Wnt signaling activated by Wnt4, Wnt11 or Wnt11-R are required for neural crest migration. 106 108 For Wnt11-R, this has been linked to the activation of a noncanonical Wnt pathway involving calcium signaling and activation of CaMKII. 108 In mice, Wnt5a and Wnt11 are expressed in the myocardium of the OFT. Loss of Wnt5a results in OFT septation defects resulting in PTA and DORV. In this context Wnt5a signals to immigrated CNCC and functions via the noncanonical Wnt/Ca 2 pathway. This leads to a downregulation of CNCC marker genes and deficits during aorticopulmonary septum formation. 109 Wnt11 is another Wnt ligand expressed in the OFT and activates a noncanonical Wnt pathway regulating the expression of TGF 2. 77 Furthermore, Wnt5a and Wnt11 are required for cytoskeletal rearrangements during morphogenesis of the OFT. Others could show a requirement for components of the planar cell polarity (PCP) pathway in OFT formation. Investigation of the surface transmembrane protein Vangl2 (also known as strabismus or Van Gogh in Drosophila), a component of PCP signaling, reveals a role for this gene in lamellipodia and filopodia formation in the OFT presumably caused by defects in cell-cell and cell ECM interactions. 110,111 A Third Cardiac Lineage: The Epicardium The epicardium represents the outer layer of the heart enveloping the myocardium and endocardium. The epicardium spreads over the myocardium during looping stages and originates mostly from the proepicardium. A minor part of the epicardium covering the outflow tract region originates from splanchnic mesoderm of the ventral pharynx. It has been thought for a long time that the proepicardium represents an extracardiac population of cells; however, more recent molecular data indicated that the epicardium derives from an early Isl1 proepicardial progenitor cell population. 112,113 These proepicardial progenitors likely derive from Isl1 / Nkx2.5 cardiac progenitor cells. BMP and FGF signaling pathways play an important role in the separation of epicardial and myocardial lineages. 114 Epicardial cells are characterized by the expression of Tbx5, WT1, and Tbx18 but are finally negative for Isl1. The epicardium can further differentiate into different cell types. Based on Tbx18:lacZ mice, it has been suggested that epicardial cells can contribute to the myocardium. 115 In support of the first hypothesis, it could be demonstrated that also WT1 epicardial cells can give rise to heart muscles. 112 These findings have been challenged by others. 116 Furthermore, it is current textbook knowledge that the epicardium forms the coronary vessels. A very recent study indicated in contrast that the coronary arteries form by developmental reprogramming of venous cells. 117 Given this, it is tempting to assume that the epicardium either contributes to a small part of coronary arteries or supports artery formation rather indirect by determining the microenvironment. Besides this, the epicardium forms cardiac fibroblasts. Two studies demonstrate a function of Wnts during epicard development. Depletion of -catenin in the proepicardium using a specific GATA5-Cre line resulted in defects in coronary artery formation that finally is lethal. 118 A second study investigated the function of retinoic acid in epicardium development. 119 The loss of retinoic X receptor (RXR) in the epicardium indicated that RXR is required for EMT of epicardial cells, resulting in damaged myocardial growth and coronary artery formations. The authors indicated Wnt9b to be downstream of RXR, which in turn regulates FGF2 expression in the myocardium. The Conduction System A further major step in cardiac development is the establishment of the conduction system that is essential for the coordinated beating of the different cardiac chambers. This process starts by forming the so-called cardiac pacemaking region in the developing right atrium at E8.0 of mouse development (Figure 2I). Later, the sinoatrial node is formed in which the contractile impulse originates. Afterward this impulse is transduced to the other parts of the conduction system, the atrioventricular (AV) node, the His bundles, bundle branches and the Purkinje fibers. The conduction system derives from an Isl1-positive progenitor cell. 120 The role of Wnts for this process is not well investigated. Bond and colleagues revealed a specific expression of Wnt7a as well as Wnt11 in the developing conduction system. Additionally, the treatment with endothelin-1 led to an increased expression of Wnt7a and Wnt11. However, functional data are missing. 121 Outlook As outlined, Wnt signaling plays multiple roles throughout whole cardiac development (see Table 2 for a referenced summary). Future work will have to describe precisely in which cell type Wnt signals emerge and in which cell type these signals elicit their function. A detailed analysis of intracellular mediators used by these Wnt ligands will have to identify the Wnt signaling branch involved. This detailed knowledge of how Wnt proteins influence cardiac development will help to improve protocols for directed differentiation of ES cells into different cardiac lineages. Together with the establish-

196 Circulation Research July 23, 2010 Table 2. Wnt Signaling Pathways During Cardiac Specification, Differentiation, and Development Developmental Process Wnt/ -Catenin Signaling Noncanonical Wnt Signaling Mesoderm formation Required (mouse, ES cells, Xenopus) 36 40 Not known Specification of cardiac progenitor cells Negative effect (ES cells, chicken, Xenopus) 28,29,44,46 50 Promoting (Xenopus, chicken, ES cells) 32,46,49,51,58 60 Migration of cardiac progenitor cells Not known Required during gastrulation Multipotent cardiac progenitor cells Supports proliferation 78 83 Not known Cardiac neural crest cells Required for specification, delamination, proliferation Essential for migration and differentiation Terminal differentiation Inhibits differentiation (Xenopus, ES cells) 32,33,84 87 Required (Xenopus, quail) 59,89 ; supportive (ES cells, EPC, diverse cell types) 32,61 64,66 Linear heart tube formation -catenin required for cell-cell contact Supports cell adhesion 59,77,88,89,92,93 ; required for morphogenesis 87,89,95 OFT morphogenesis Required for septation (CNCCs) 103 105 Required for cytoskeleton formation and cell cell interactions 77,106 111 Cardiac looping Required (zebrafish) 98 Not known Development of the epicardium Required 118,119 Not known Heart valve formation Required (zebrafish, chicken) 96 100 Not known Development of conduction system Not known Expression but no functional data 121 The role of different aspects of the Wnt signaling network during cardiac differentiation and development are given as discussed in the text. ment of safe protocols to generate human, patient-specific induced pluripotent stem cells, this opens the window of opportunity to develop novel therapeutic approaches. This knowledge might also help to activate recently described resident stem cells in the heart on heart damage. 122 125 Sources of Funding Work in our laboratory is supported by the Deutsche Forschungsgemeinschaft. None. Disclosures References 1. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781 810. 2. Kestler HA, Kuhl M. From individual Wnt pathways towards a Wnt signalling network. Philos Trans R Soc Lond B Biol Sci. 2008;363: 1333 1347. 3. van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205 3214. 4. Rao TP, Kuhl M. An updated overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010;107. 5. Maye P, Zheng J, Li L, Wu D. Multiple mechanisms for Wnt11- mediated repression of the canonical Wnt signaling pathway. J Biol Chem. 2004;279:24659 24665. 6. Buckingham M, Meilhac S, Zaffran S. Building the mammalian heart from two sources of myocardial cells. Nat Rev Genet. 2005;6:826 835. 7. Moorman AF, Christoffels VM, Anderson RH, van den Hoff MJ. The heart-forming fields: one or multiple? Philos Trans R Soc Lond B Biol Sci. 2007;362:1257 1265. 8. Waldo KL, Kumiski DH, Wallis KT, Stadt HA, Hutson MR, Platt DH, Kirby ML. Conotruncal myocardium arises from a secondary heart field. Development. 2001;128:3179 3188. 9. Mjaatvedt CH, Nakaoka T, Moreno-Rodriguez R, Norris RA, Kern MJ, Eisenberg CA, Turner D, Markwald RR. The outflow tract of the heart is recruited from a novel heart-forming field. Dev Biol. 2001;238: 97 109. 10. Cai CL, Liang X, Shi Y, Chu PH, Pfaff SL, Chen J, Evans S. Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart. Dev Cell. 2003;5:877 889. 11. Kelly RG, Brown NA, Buckingham ME. The arterial pole of the mouse heart forms from Fgf10-expressing cells in pharyngeal mesoderm. Dev Cell. 2001;1:435 440. 12. Zaffran S, Kelly RG, Meilhac SM, Buckingham ME, Brown NA. Right ventricular myocardium derives from the anterior heart field. Circ Res. 2004;95:261 268. 13. Verzi MP, McCulley DJ, De Val S, Dodou E, Black BL. The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field. Dev Biol. 2005;287:134 145. 14. Rana MS, Horsten NC, Tesink-Taekema S, Lamers WH, Moorman AF, van den Hoff MJ. Trabeculated right ventricular free wall in the chicken heart forms by ventricularization of the myocardium initially forming the outflow tract. Circ Res. 2007;100:1000 1007. 15. Gessert S, Kuhl M. Comparative gene expression analysis and fate mapping studies suggest an early segregation of cardiogenic lineages in Xenopus laevis. Dev Biol. 2009;334:395 408. 16. Laugwitz KL, Moretti A, Caron L, Nakano A, Chien KR. Islet1 cardiovascular progenitors: a single source for heart lineages? Development. 2008;135:193 205. 17. Meilhac SM, Esner M, Kelly RG, Nicolas JF, Buckingham ME. The clonal origin of myocardial cells in different regions of the embryonic mouse heart. Dev Cell. 2004;6:685 698. 18. Abu-Issa R, Kirby ML. Heart field: from mesoderm to heart tube. Annu Rev Cell Dev Biol. 2007;23:45 68. 19. Wu SM, Fujiwara Y, Cibulsky SM, Clapham DE, Lien CL, Schultheiss TM, Orkin SH. Developmental origin of a bipotential myocardial and smooth muscle cell precursor in the mammalian heart. Cell. 2006;127: 1137 1150. 20. Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, Qyang Y, Bu L, Sasaki M, Martin-Puig S, Sun Y, Evans SM, Laugwitz KL, Chien KR. Multipotent embryonic isl1 progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell. 2006;127: 1151 1165. 21. Kattman SJ, Huber TL, Keller GM. Multipotent flk-1 cardiovascular progenitor cells give rise to the cardiomyocyte, endothelial, and vascular smooth muscle lineages. Dev Cell. 2006;11:723 732. 22. Kitajima S, Takagi A, Inoue T, Saga Y. MesP1 and MesP2 are essential for the development of cardiac mesoderm. Development. 2000;127: 3215 3226. 23. Sater AK, Jacobson AG. The specification of heart mesoderm occurs during gastrulation in Xenopus laevis. Development. 1989;105: 821 830. 24. Sater AK, Jacobson AG. The restriction of the heart morphogenetic field in Xenopus laevis. Dev Biol. 1990;140:328 336. 25. Sater AK, Jacobson AG. The role of the dorsal lip in the induction of heart mesoderm in Xenopus laevis. Development. 1990;108:461 470. 26. Samuel LJ, Latinkic BV. Early activation of FGF and nodal pathways mediates cardiac specification independently of Wnt/beta-catenin signaling. PLoS One. 2009;4:e7650.

Gessert and Kühl Wnt in Cardiac Differentiation 197 27. Park M, Wu X, Golden K, Axelrod JD, Bodmer R. The wingless signaling pathway is directly involved in Drosophila heart development. Dev Biol. 1996;177:104 116. 28. Marvin MJ, Di Rocco G, Gardiner A, Bush SM, Lassar AB. Inhibition of Wnt activity induces heart formation from posterior mesoderm. Genes Dev. 2001;15:316 327. 29. Schneider VA, Mercola M. Wnt antagonism initiates cardiogenesis in Xenopus laevis. Genes Dev. 2001;15:304 315. 30. Lickert H, Kutsch S, Kanzler B, Tamai Y, Taketo MM, Kemler R. Formation of multiple hearts in mice following deletion of beta-catenin in the embryonic endoderm. Dev Cell. 2002;3:171 181. 31. Nakamura T, Sano M, Songyang Z, Schneider MD. A Wnt- and betacatenin-dependent pathway for mammalian cardiac myogenesis. Proc Natl Acad Sci U S A. 2003;100:5834 5839. 32. Ueno S, Weidinger G, Osugi T, Kohn AD, Golob JL, Pabon L, Reinecke H, Moon RT, Murry CE. Biphasic role for Wnt/beta-catenin signaling in cardiac specification in zebrafish and embryonic stem cells. Proc Natl Acad Sci U S A. 2007;104:9685 9690. 33. Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A, Komuro I. Developmental stage-specific biphasic roles of Wnt/betacatenin signaling in cardiomyogenesis and hematopoiesis. Proc Natl Acad Sci U S A. 2006;103:19812 19817. 34. Tzahor E. Wnt/beta-catenin signaling and cardiogenesis: timing does matter. Dev Cell. 2007;13:10 13. 35. Wu SM. Mesp1 at the heart of mesoderm lineage specification. Cell Stem Cell. 2008;3:1 2. 36. Huelsken J, Vogel R, Brinkmann V, Erdmann B, Birchmeier C, Birchmeier W. Requirement for beta-catenin in anterior-posterior axis formation in mice. J Cell Biol. 2000;148:567 578. 37. Liu P, Wakamiya M, Shea MJ, Albrecht U, Behringer RR, Bradley A. Requirement for Wnt3 in vertebrate axis formation. Nat Genet. 1999; 22:361 365. 38. Lindsley RC, Gill JG, Kyba M, Murphy TL, Murphy KM. Canonical Wnt signaling is required for development of embryonic stem cellderived mesoderm. Development. 2006;133:3787 3796. 39. Yamaguchi TP, Takada S, Yoshikawa Y, Wu N, McMahon AP. T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification. Genes Dev. 1999;13:3185 3190. 40. Vonica A, Gumbiner BM. Zygotic Wnt activity is required for Brachyury expression in the early Xenopus laevis embryo. Dev Biol. 2002;250:112 127. 41. Saga Y, Kitajima S, Miyagawa-Tomita S. Mesp1 expression is the earliest sign of cardiovascular development. Trends Cardiovasc Med. 2000;10:345 352. 42. Saga Y, Miyagawa-Tomita S, Takagi A, Kitajima S, Miyazaki J, Inoue T. MesP1 is expressed in the heart precursor cells and required for the formation of a single heart tube. Development. 1999;126:3437 3447. 43. Bondue A, Lapouge G, Paulissen C, Semeraro C, Iacovino M, Kyba M, Blanpain C. Mesp1 acts as a master regulator of multipotent cardiovascular progenitor specification. Cell Stem Cell. 2008;3:69 84. 44. Lindsley RC, Gill JG, Murphy TL, Langer EM, Cai M, Mashayekhi M, Wang W, Niwa N, Nerbonne JM, Kyba M, Murphy KM. Mesp1 coordinately regulates cardiovascular fate restriction and epithelialmesenchymal transition in differentiating ESCs. Cell Stem Cell. 2008; 3:55 68. 45. Kouskoff V, Lacaud G, Schwantz S, Fehling HJ, Keller G. Sequential development of hematopoietic and cardiac mesoderm during embryonic stem cell differentiation. Proc Natl Acad Sci U S A. 2005;102: 13170 13175. 46. Chen VC, Stull R, Joo D, Cheng X, Keller G. Notch signaling respecifies the hemangioblast to a cardiac fate. Nat Biotechnol. 2008; 26:1169 1178. 47. Kwon C, Qian L, Cheng P, Nigam V, Arnold J, Srivastava D. A regulatory pathway involving Notch1/beta-catenin/Isl1 determines cardiac progenitor cell fate. Nat Cell Biol. 2009;11:951 957. 48. Yamashita JK, Takano M, Hiraoka-Kanie M, Shimazu C, Peishi Y, Yanagi K, Nakano A, Inoue E, Kita F, Nishikawa S. Prospective identification of cardiac progenitors by a novel single cell-based cardiomyocyte induction. FASEB J. 2005;19:1534 1536. 49. Afouda BA, Martin J, Liu F, Ciau-Uitz A, Patient R, Hoppler S. GATA transcription factors integrate Wnt signalling during heart development. Development. 2008;135:3185 3190. 50. David R, Brenner C, Stieber J, Schwarz F, Brunner S, Vollmer M, Mentele E, Muller-Hocker J, Kitajima S, Lickert H, Rupp R, Franz WM. MesP1 drives vertebrate cardiovascular differentiation through Dkk-1- mediated blockade of Wnt-signalling. Nat Cell Biol. 2008;10:338 345. 51. Pandur P, Lasche M, Eisenberg LM, Kuhl M. Wnt-11 activation of a non-canonical Wnt signalling pathway is required for cardiogenesis. Nature. 2002;418:636 641. 52. Caneparo L, Huang YL, Staudt N, Tada M, Ahrendt R, Kazanskaya O, Niehrs C, Houart C. Dickkopf-1 regulates gastrulation movements by coordinated modulation of Wnt/beta catenin and Wnt/PCP activities, through interaction with the Dally-like homolog Knypek. Genes Dev. 2007;21:465 480. 53. Korol O, Gupta RW, Mercola M. A novel activity of the Dickkopf-1 amino terminal domain promotes axial and heart development independently of canonical Wnt inhibition. Dev Biol. 2008;324:131 138. 54. Foley AC, Mercola M. Heart induction by Wnt antagonists depends on the homeodomain transcription factor Hex. Genes Dev. 2005;19: 387 396. 55. Liu Y, Asakura M, Inoue H, Nakamura T, Sano M, Niu Z, Chen M, Schwartz RJ, Schneider MD. Sox17 is essential for the specification of cardiac mesoderm in embryonic stem cells. Proc Natl Acad Sci U S A. 2007;104:3859 3864. 56. Foley AC, Korol O, Timmer AM, Mercola M. Multiple functions of Cerberus cooperate to induce heart downstream of Nodal. Dev Biol. 2007;303:57 65. 57. Kanai-Azuma M, Kanai Y, Gad JM, Tajima Y, Taya C, Kurohmaru M, Sanai Y, Yonekawa H, Yazaki K, Tam PP, Hayashi Y. Depletion of definitive gut endoderm in Sox17-null mutant mice. Development. 2002; 129:2367 2379. 58. Eisenberg CA, Eisenberg LM. WNT11 promotes cardiac tissue formation of early mesoderm. Dev Dyn. 1999;216:45 58. 59. Eisenberg CA, Gourdie RG, Eisenberg LM. Wnt-11 is expressed in early avian mesoderm and required for the differentiation of the quail mesoderm cell line QCE-6. Development. 1997;124:525 536. 60. Terami H, Hidaka K, Katsumata T, Iio A, Morisaki T. Wnt11 facilitates embryonic stem cell differentiation to Nkx2.5-positive cardiomyocytes. Biochem Biophys Res Commun. 2004;325:968 975. 61. Koyanagi M, Haendeler J, Badorff C, Brandes RP, Hoffmann J, Pandur P, Zeiher AM, Kuhl M, Dimmeler S. Non-canonical Wnt signaling enhances differentiation of human circulating progenitor cells to cardiomyogenic cells. J Biol Chem. 2005;280:16838 16842. 62. Belema Bedada F, Technau A, Ebelt H, Schulze M, Braun T. Activation of myogenic differentiation pathways in adult bone marrow-derived stem cells. Mol Cell Biol. 2005;25:9509 9519. 63. Flaherty MP, Dawn B. Noncanonical Wnt11 signaling and cardiomyogenic differentiation. Trends Cardiovasc Med. 2008;18:260 268. 64. Schulze M, Belema-Bedada F, Technau A, Braun T. Mesenchymal stem cells are recruited to striated muscle by NFAT/IL-4-mediated cell fusion. Genes Dev. 2005;19:1787 1798. 65. Fraidenraich D, Stillwell E, Romero E, Wilkes D, Manova K, Basson CT, Benezra R. Rescue of cardiac defects in id knockout embryos by injection of embryonic stem cells. Science. 2004;306:247 252. 66. Koyanagi M, Bushoven P, Iwasaki M, Urbich C, Zeiher AM, Dimmeler S. Notch signaling contributes to the expression of cardiac markers in human circulating progenitor cells. Circ Res. 2007;101:1139 1145. 67. Koyanagi M, Iwasaki M, Haendeler J, Leitges M, Zeiher AM, Dimmeler S. Wnt5a increases cardiac gene expressions of cultured human circulating progenitor cells via a PKC delta activation. PLoS One. 2009;4: e5765. 68. Wang H, Gilner JB, Bautch VL, Wang DZ, Wainwright BJ, Kirby SL, Patterson C. Wnt2 coordinates the commitment of mesoderm to hematopoietic, endothelial, and cardiac lineages in embryoid bodies. J Biol Chem. 2007;282:782 791. 69. Wang H, Charles PC, Wu Y, Ren R, Pi X, Moser M, Barshishat-Kupper M, Rubin JS, Perou C, Bautch V, Patterson C. Gene expression profile signatures indicate a role for Wnt signaling in endothelial commitment from embryonic stem cells. Circ Res. 2006;98:1331 1339. 70. Anton R, Kestler HA, Kuhl M. Beta-catenin signaling contributes to stemness and regulates early differentiation in murine embryonic stem cells. FEBS Lett. 2007;581:5247 5254. 71. Alexandrovich A, Arno M, Patient RK, Shah AM, Pizzey JA, Brewer AC. Wnt2 is a direct downstream target of GATA6 during early cardiogenesis. Mech Dev. 2006;123:297 311. 72. Monkley SJ, Delaney SJ, Pennisi DJ, Christiansen JH, Wainwright BJ. Targeted disruption of the Wnt2 gene results in placentation defects. Development. 1996;122:3343 3353.

198 Circulation Research July 23, 2010 73. Kispert A, Vainio S, Shen L, Rowitch DH, McMahon AP. Proteoglycans are required for maintenance of Wnt-11 expression in the ureter tips. Development. 1996;122:3627 3637. 74. Ku M, Melton DA. Xwnt-11: a maternally expressed Xenopus wnt gene. Development. 1993;119:1161 1173. 75. Landesman Y, Sokol SY. Xwnt-2b is a novel axis-inducing Xenopus Wnt, which is expressed in embryonic brain. Mech Dev. 1997;63: 199 209. 76. Nagy II, Railo A, Rapila R, Hast T, Sormunen R, Tavi P, Rasanen J, Vainio SJ. Wnt-11 signalling controls ventricular myocardium development by patterning N-cadherin and beta-catenin expression. Cardiovasc Res. 85:100 109. 77. Zhou W, Lin L, Majumdar A, Li X, Zhang X, Liu W, Etheridge L, Shi Y, Martin J, Van de Ven W, Kaartinen V, Wynshaw-Boris A, McMahon AP, Rosenfeld MG, Evans SM. Modulation of morphogenesis by noncanonical Wnt signaling requires ATF/CREB family-mediated transcriptional activation of TGFbeta2. Nat Genet. 2007;39:1225 1234. 78. Tian Y, Yuan L, Goss AM, Wang T, Yang J, Lepore JJ, Zhou D, Schwartz RJ, Patel V, Cohen ED, Morrisey EE. Characterization and in vivo pharmacological rescue of a Wnt2-Gata6 pathway required for cardiac inflow tract development. Dev Cell. 18:275 287. 79. Ai D, Fu X, Wang J, Lu MF, Chen L, Baldini A, Klein WH, Martin JF. Canonical Wnt signaling functions in second heart field to promote right ventricular growth. Proc Natl Acad Sci U S A. 2007;104:9319 9324. 80. Klaus A, Saga Y, Taketo MM, Tzahor E, Birchmeier W. Distinct roles of Wnt/beta-catenin and Bmp signaling during early cardiogenesis. Proc Natl Acad Sci U S A. 2007;104:18531 18536. 81. Cohen ED, Wang Z, Lepore JJ, Lu MM, Taketo MM, Epstein DJ, Morrisey EE. Wnt/beta-catenin signaling promotes expansion of Isl-1- positive cardiac progenitor cells through regulation of FGF signaling. J Clin Invest. 2007;117:1794 1804. 82. Kwon C, Arnold J, Hsiao EC, Taketo MM, Conklin BR, Srivastava D. Canonical Wnt signaling is a positive regulator of mammalian cardiac progenitors. Proc Natl Acad Sci U S A. 2007;104:10894 10899. 83. Lin L, Cui L, Zhou W, Dufort D, Zhang X, Cai CL, Bu L, Yang L, Martin J, Kemler R, Rosenfeld MG, Chen J, Evans SM. Beta-catenin directly regulates Islet1 expression in cardiovascular progenitors and is required for multiple aspects of cardiogenesis. Proc Natl Acad Sci USA. 2007;104:9313 9318. 84. Qyang Y, Martin-Puig S, Chiravuri M, Chen S, Xu H, Bu L, Jiang X, Lin L, Granger A, Moretti A, Caron L, Wu X, Clarke J, Taketo MM, Laugwitz KL, Moon RT, Gruber P, Evans SM, Ding S, Chien KR. The renewal and differentiation of Isl1 cardiovascular progenitors are controlled by a Wnt/beta-catenin pathway. Cell Stem Cell. 2007;1: 165 179. 85. Zhu W, Shiojima I, Ito Y, Li Z, Ikeda H, Yoshida M, Naito AT, Nishi J, Ueno H, Umezawa A, Minamino T, Nagai T, Kikuchi A, Asashima M, Komuro I. IGFBP-4 is an inhibitor of canonical Wnt signalling required for cardiogenesis. Nature. 2008;454:345 349. 86. Brott BK, Sokol SY. A vertebrate homolog of the cell cycle regulator Dbf4 is an inhibitor of Wnt signaling required for heart development. Dev Cell. 2005;8:703 715. 87. Lavery DL, Martin J, Turnbull YD, Hoppler S. Wnt6 signaling regulates heart muscle development during organogenesis. Dev Biol. 2008;323: 177 188. 88. Garriock RJ, D Agostino SL, Pilcher KC, Krieg PA. Wnt11-R, a protein closely related to mammalian Wnt11, is required for heart morphogenesis in Xenopus. Dev Biol. 2005;279:179 192. 89. Gessert S, Maurus D, Brade T, Walther P, Pandur P, Kuhl M. DM-GRASP/ALCAM/CD166 is required for cardiac morphogenesis and maintenance of cardiac identity in first heart field derived cells. Dev Biol. 2008;321:150 161. 90. Radice GL, Rayburn H, Matsunami H, Knudsen KA, Takeichi M, Hynes RO. Developmental defects in mouse embryos lacking N-cadherin. Dev Biol. 1997;181:64 78. 91. Linask KK, Knudsen KA, Gui Y-H. N-cadherin-catenin interaction: necessary component of cardiac cell compartmentalization during early vertebrate heart development. Dev Biol. 1997;185:148 164. 92. Torres MA, Yang-Snyder JA, Purcell SM, DeMarais AA, McGrew LL, Moon RT. Activities of the Wnt-1 class of secreted signaling factors are antagonized by the Wnt-5A class and by a dominant negative cadherin in early Xenopus development. J Cell Biol. 1996;133:1123 1137. 93. Toyofuku T, Hong Z, Kuzuya T, Tada M, Hori M. Wnt/frizzled-2 signaling induces aggregation and adhesion among cardiac myocytes by increased cadherin-beta-catenin complex. J Cell Biol. 2000;150: 225 241. 94. Fujio Y, Matsuda T, Oshima Y, Maeda M, Mohri T, Ito T, Takatani T, Hirata M, Nakaoka Y, Kimura R, Kishimoto T, Azuma J. Signals through gp130 upregulate Wnt5a and contribute to cell adhesion in cardiac myocytes. FEBS Lett. 2004;573:202 206. 95. Matsui T, Raya A, Kawakami Y, Callol-Massot C, Capdevila J, Rodriguez-Esteban C, Izpisua Belmonte JC. Noncanonical Wnt signaling regulates midline convergence of organ primordia during zebrafish development. Genes Dev. 2005;19:164 175. 96. Alfieri CM, Cheek J, Chakraborty S, Yutzey KE. Wnt signaling in heart valve development and osteogenic gene induction. Dev Biol. 338: 127 135. 97. Gitler AD, Lu MM, Jiang YQ, Epstein JA, Gruber PJ. Molecular markers of cardiac endocardial cushion development. Dev Dyn. 2003; 228:643 650. 98. Hurlstone AF, Haramis AP, Wienholds E, Begthel H, Korving J, Van Eeden F, Cuppen E, Zivkovic D, Plasterk RH, Clevers H. The Wnt/ beta-catenin pathway regulates cardiac valve formation. Nature. 2003; 425:633 637. 99. Liebner S, Cattelino A, Gallini R, Rudini N, Iurlaro M, Piccolo S, Dejana E. Beta-catenin is required for endothelial-mesenchymal transformation during heart cushion development in the mouse. J Cell Biol. 2004;166:359 367. 100. Person AD, Garriock RJ, Krieg PA, Runyan RB, Klewer SE. Frzb modulates Wnt-9a-mediated beta-catenin signaling during avian atrioventricular cardiac cushion development. Dev Biol. 2005;278:35 48. 101. Karner CM, Chirumamilla R, Aoki S, Igarashi P, Wallingford JB, Carroll TJ. Wnt9b signaling regulates planar cell polarity and kidney tubule morphogenesis. Nat Genet. 2009;41:793 799. 102. Lee YH, Saint-Jeannet JP. Characterization of molecular markers to assess cardiac cushions formation in Xenopus. Dev Dyn. 2009;238: 3257 3265. 103. Brault V, Moore R, Kutsch S, Ishibashi M, Rowitch DH, McMahon AP, Sommer L, Boussadia O, Kemler R. Inactivation of the beta-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development. Development. 2001;128: 1253 1264. 104. Hamblet NS, Lijam N, Ruiz-Lozano P, Wang J, Yang Y, Luo Z, Mei L, Chien KR, Sussman DJ, Wynshaw-Boris A. Dishevelled 2 is essential for cardiac outflow tract development, somite segmentation and neural tube closure. Development. 2002;129:5827 5838. 105. Kioussi C, Briata P, Baek SH, Rose DW, Hamblet NS, Herman T, Ohgi KA, Lin C, Gleiberman A, Wang J, Brault V, Ruiz-Lozano P, Nguyen HD, Kemler R, Glass CK, Wynshaw-Boris A, Rosenfeld MG. Identification of a Wnt/Dvl/beta-Catenin Pitx2 pathway mediating celltype-specific proliferation during development. Cell. 2002;111: 673 685. 106. Gessert S, Maurus D, Rossner A, Kuhl M. Pescadillo is required for Xenopus laevis eye development and neural crest migration. Dev Biol. 2007;310:99 112. 107. De Calisto J, Araya C, Marchant L, Riaz CF, Mayor R. Essential role of non-canonical Wnt signalling in neural crest migration. Development. 2005;132:2587 2597. 108. Garriock RJ, Krieg PA. Wnt11-R signaling regulates a calcium sensitive EMT event essential for dorsal fin development of Xenopus. Dev Biol. 2007;304:127 140. 109. Schleiffarth JR, Person AD, Martinsen BJ, Sukovich DJ, Neumann A, Baker CV, Lohr JL, Cornfield DN, Ekker SC, Petryk A. Wnt5a is required for cardiac outflow tract septation in mice. Pediatr Res. 2007; 61:386 391. 110. Phillips HM, Murdoch JN, Chaudhry B, Copp AJ, Henderson DJ. Vangl2 acts via RhoA signaling to regulate polarized cell movements during development of the proximal outflow tract. Circ Res. 2005;96: 292 299. 111. Henderson DJ, Phillips HM, Chaudhry B. Vang-like 2 and noncanonical Wnt signaling in outflow tract development. Trends Cardiovasc Med. 2006;16:38 45. 112. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J, Jiang D, von Gise A, Ikeda S, Chien KR, Pu WT. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature. 2008;454:109 113. 113. Zhou B, von Gise A, Ma Q, Rivera-Feliciano J, Pu WT. Nkx2 5- and Isl1-expressing cardiac progenitors contribute to proepicardium. Biochem Biophys Res Commun. 2008;375:450 453.

Gessert and Kühl Wnt in Cardiac Differentiation 199 114. van Wijk B, van den Berg G, Abu-Issa R, Barnett P, van der Velden S, Schmidt M, Ruijter JM, Kirby ML, Moorman AF, van den Hoff MJ. Epicardium and myocardium separate from a common precursor pool by crosstalk between bone morphogenetic protein- and fibroblast growth factor-signaling pathways. Circ Res. 2009;105: 431 441. 115. Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K, Yang L, Bu L, Liang X, Zhang X, Stallcup WB, Denton CP, McCulloch A, Chen J, Evans SM. A myocardial lineage derives from Tbx18 epicardial cells. Nature. 2008;454:104 108. 116. Christoffels VM, Grieskamp T, Norden J, Mommersteeg MT, Rudat C, Kispert A Tbx18 and the fate of epicardial progenitors. Nature. 2009; 458:E8 E9; discussion E9 E10. 117. Red-Horse K, Ueno H, Weissman IL, Krasnow MA. Coronary arteries form by developmental reprogramming of venous cells. Nature. 2010; 464:549 553. 118. Zamora M, Manner J, Ruiz-Lozano P. Epicardium-derived progenitor cells require beta-catenin for coronary artery formation. Proc Natl Acad Sci U S A. 2007;104:18109 18114. 119. Merki E, Zamora M, Raya A, Kawakami Y, Wang J, Zhang X, Burch J, Kubalak SW, Kaliman P, Belmonte JC, Chien KR, Ruiz-Lozano P. Epicardial retinoid X receptor alpha is required for myocardial growth and coronary artery formation. Proc Natl Acad Sci U S A. 2005;102: 18455 18460. 120. Sun Y, Liang X, Najafi N, Cass M, Lin L, Cai CL, Chen J, Evans SM. Islet 1 is expressed in distinct cardiovascular lineages, including pacemaker and coronary vascular cells. Dev Biol. 2007;304:286 296. 121. Bond J, Sedmera D, Jourdan J, Zhang Y, Eisenberg CA, Eisenberg LM, Gourdie RG. Wnt11 and Wnt7a are up-regulated in association with differentiation of cardiac conduction cells in vitro and in vivo. Dev Dyn. 2003;227:536 543. 122. Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, Lin LZ, Cai CL, Lu MM, Reth M, Platoshyn O, Yuan JX, Evans S, Chien KR. Postnatal isl1 cardioblasts enter fully differentiated cardiomyocyte lineages. Nature. 2005;433:647 653. 123. Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael LH, Behringer RR, Garry DJ, Entman ML, Schneider MD. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003; 100:12313 12318. 124. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal- Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763 776. 125. Martin CM, Meeson AP, Robertson SM, Hawke TJ, Richardson JA, Bates S, Goetsch SC, Gallardo TD, Garry DJ. Persistent expression of the ATP-binding cassette transporter, Abcg2, identifies cardiac SP cells in the developing and adult heart. Dev Biol. 2004;265:262 275. 126. Brade T, Manner J, Kuhl M. The role of Wnt signalling in cardiac development and tissue remodelling in the mature heart. Cardiovasc Res. 2006;72:198 209. 127. Cohen ED, Tian Y, Morrisey EE. Wnt signaling: an essential regulator of cardiovascular differentiation, morphogenesis and progenitor selfrenewal. Development. 2008;135:789 798. 128. Eisenberg LM, Eisenberg CA. Wnt signal transduction and the formation of the myocardium. Dev Biol. 2006;293:305 315.

The Multiple Phases and Faces of Wnt Signaling During Cardiac Differentiation and Development Susanne Gessert and Michael Kühl Circ Res. 2010;107:186-199 doi: 10.1161/CIRCRESAHA.110.221531 Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright 2010 American Heart Association, Inc. All rights reserved. Print ISSN: 0009-7330. Online ISSN: 1524-4571 The online version of this article, along with updated information and services, is located on the World Wide Web at: http://circres.ahajournals.org/content/107/2/186 Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Circulation Research is online at: http://circres.ahajournals.org//subscriptions/