Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Aug 7.
Published in final edited form as: Pediatr Cardiol. 2009 Dec 5;31(3):342–348. doi: 10.1007/s00246-009-9606-z

The Importance of Wnt Signaling in Cardiovascular Development

Ying Tian 1, Ethan David Cohen 2, Edward E Morrisey 3,4,5,6,
PMCID: PMC3736804  NIHMSID: NIHMS455748  PMID: 19967349

Abstract

Cardiac development is comprised of a series of morphological events tightly controlled both spatially and temporally. The molecular pathways controlling early cardiac differentiation are poorly understood, but Wnt signaling is emerging as a critical pathway for multiple aspects of early cardiovascular development. The Wnt pathway plays multiple roles in regulating cellular behavior including proliferation, differentiation, cell migration, and cell polarity. Recent data have demonstrated that Wnt activity is important for early precardiac mesoderm differentiation but must be inhibited in subsequent steps for cardiomyocyte differentiation to proceed. Given the important role that Wnt signaling plays in both the differentiation of cardiomyocytes from pluripotential stem cells and tissue regeneration in general, an increased understanding of this pathway is likely to enhance our knowledge about both cardiovascular development and reparative mechanisms.

Keywords: Cardiac development, Progenitor, Wnt

Wnt Signaling Overview

Wnt proteins are homologs of the Drosophila wingless gene and have been show to play important roles in regulating cell differentiation, proliferation, and polarity (reviewed in [7, 53, 64]). Wnt proteins are cysteine-rich secreted glycoproteins that signal through several possible pathways. The best understood of these, commonly called the canonical pathway, involves binding of Wnt proteins to Frizzled cell surface receptors, which inhibits GSK-3β phosphorylation of β-catenin. Hypophosphorylated β-catenin is translocated to the nucleus, where it binds to members of the lymphoid enhancement factor/T-cell factor (LEF/TCF) family of transcription factors. Binding of β-catenin converts LEF/TCF factors from repressors to activators, thereby switching on cell-specific gene transcription.

Wnt proteins also can signal through a poorly understood network of noncanonical effectors to control subtle aspects of cell behavior such as cell polarization, adhesion, and motility [22, 35, 67, 68, 71, 87]. Activation of these effectors can inhibit canonical Wnt signaling. Moreover, it has been shown that a single Wnt ligand, such as Wnt5a, can signal through both canonical and noncanonical pathways in a cell type-dependent context [47, 77]. These issues have led some to promote the idea that Wnt signaling is more a network of interacting factors that regulate many aspects of cell biology than a simple linear signaling pathway [78]. This notion has much merit and likely will garner increased support as the diversity of cellular responses due to Wnt signaling increases (Table 1)

Table 1.

Components of the Wnt pathway and their role in cardiovascular development

Gene Species Pathway Phenotype References
Wnt ligands
  Wnt2 Mouse Canonical Placental vascular failure, inflow tract
  development (E. Morrisey, unpublished observations)
[49]
  Wnt5a Mouse Noncanonical Outflow tract septation [45, 59]
  Wnt7a Mouse Canonical Cerebral vascular development (with Wnt7b) [14, 66]
  Wnt7b Mouse Canonical Pulmonary vascular smooth muscle development,
  cerebral vascular development (with Wnt7a)
[13, 14, 62, 66, 86]
  Wnt9a Mouse Unknown Epicardial development [46]
  Wnt11 Mouse, zebra fish,
  Xenopus
Noncanonical Outflow tract development, cardiac
  myocyte differentiation
[15, 17, 60, 69, 91]
Wnt receptors
  Fzd4 Mouse Unknown Retinal vascular development (with norrin) [70, 88]
  Fzd5 Mouse Unknown Placental vascular development [21]
  Norrin Mouse, human Canonical Retinal vascular development (with Fzd4) [88]
  Lrp5 Mouse, human Canonical Retinal vascular development [24, 70, 86]
  Lrp6 Mouse Canonical Vascular smooth muscle proliferation and survival [85]
Signaling components
  Beta-catenin Mouse, zebra fish Canonical SHF progenitor proliferation, cardiac valve
  development, adult hypertrophic growth
[2, 10, 12, 20, 29, 3739,
  50, 51, 76, 79, 84]
  Dvl1 Mouse Unknown Myofibroblast proliferation [9]
  Dvl2 Mouse Unknown Outflow tract development [18]
  Gsk3β Mouse Canonical Adult cardiac hypertrophic growth [10, 72, 73]
  Vangl2 Mouse Noncanonical Outflow tract development [54]
  Apc Zebra fish Canonical Cardiac valve development [20]

SHF second heart field

Noncanonical Wnt effectors are loosely organized into two pathways, the Wnt/RhoA and Wnt/Ca2+ pathways. In Wnt/RhoA signaling, Wnt proteins bind to Frizzled receptors, which mediate both canonical and noncanonical Wnt signaling, to activate Rho-family small GTPases and their downstream effectors such as Rho-associated kinase [22, 35, 67, 68]. This pathway is similar to the Drosophila planar cell polarity pathway, which is required for proper fly wing hair orientation, cell polarization during convergence and extension movements in gastrulation, and orientation of steriocilia in the cochlea [16, 19, 25, 28, 43, 55, 80, 81, 83, 92]. In Wnt/Ca2+ signaling, Wnt proteins induce intracellular Ca2+ release and activate the Ca2+-dependent protein kinases PKC and CamKII via the G-protein-dependent activity of Frizzled receptors [3234, 42, 58, 61, 63].

Wnt signaling has been shown to play an essential role in brain, limb, mammary, skin, and, more recently, cardiovascular development [3, 4, 12, 30, 37, 39, 44, 52, 56, 57, 65, 76]. There are 19 Wnt proteins, 10 Fzd receptors, and 2 LRP co-receptors in mammals. Such complexity suggests multifarious possibilities of Wnt-signaling outputs. Recent studies indicate that individual Wnt ligands can activate canonical or noncanonical Wnt signaling in a cell context-dependent manner [47, 74]. Therefore, the pathway likely is regulated in a spatial and temporal context related to the expression patterns of specific Wnt ligands and receptors.

Early Cardiovascular Development and Wnt Signaling

The heart arises from two different but overlapping sources of mesoderm called the first heart field (FHF) and the second heart field (SHF). The SHF lies peripherally in relation to the FHF in the early cardiac crescent, and both migrate to the midline to form the simple cardiac tube. SHF progenitors continue to contribute to the growing and septating heart, playing a critical role in the formation of the right ventricle, and atria, as well as the outflow and inflow tracts [26, 27]. At the molecular level, the distinctions between FHF and SHF progenitors are poorly understood, and both fields likely overlap extensively. However, recent data point to a unique sensitivity of SHF progenitor differentiation and proliferation to Wnt/β-catenin signaling [31, 76].

The FHF and SHF and their derivatives are marked by expression of specific transcription factors. Myocyte enhancer 2c (Mef2c), GATA-binding protein 4 (Gata4), and NK2 transcription factor related 5 (Nkx2.5) mark both the FHF and SHF [36, 40, 41, 48]. Tbx5 expression is observed in a graded pattern, with the highest levels in the left ventricle and the formation of the left ventricle is preferentially affected in loss of function mutants, supporting the notion that Tbx5 is a reasonable marker of the FHF [5, 6]. The LIM-homeodomain gene islet 1 (Isl1) is expressed primarily in the SHF and marks this group of cardiac progenitors as a distinct cell population [8].

All these factors are critical determinants of cardiomyocyte specification and development. In mice, Gata4 and Gata6 are required for specification of FHF progenitors but not SHF progenitors [90]. Loss of both Gata4 and Gata6 leads to heart agenesis, with a loss in Tbx5 expression (FHF marker) but continued Isl1 expression (SHF marker). Despite this important finding, specification of FHF and SHF cardiac progenitors is likely to rely on a complex network of interactions between multiple families of transcription factors. Moreover, the activity of several signaling networks including BMP, TGFβ, and Wnt are important in the regulation of these transcription factor interactions at the earliest stages of cardiac specification and differentiation.

The activity of the Wnt/β-catenin pathway has been explored during cardiac development using several LEF/TCF transgenic reporter lines. Although activity is observed in the pericardium, the endocardial cushions, the adjacent cardiac mesoderm, and the early outflow tract, little to no activity is observed in the developing ventricular myocardium [12, 39]. This is surprising given the high expression level of several Wnt ligands in the early heart including Wnt2, Wnt2b, Wnt11, and Wnt8a [15, 23, 49, 89]. These data could indicate that these ligands act primarily through the noncanonical portion of the Wnt network or that the LEF/TCF transgenic lines are limited in their ability to report Wnt/β-catenin signaling accurately in the developing heart.

Wnt signaling has been shown to regulate the development of the heart’s anterior portion including the outflow tract and the right ventricle. Loss of β-catenin in the SHF leads to decreased numbers of anterior Isl1+ progenitors, whereas stabilization of β-catenin expands the numbers of Isl1+ cells [2, 12, 37, 39]. The deletion of β-catenin in the SHF causes a dramatic reduction in both the levels of Isl1 expression and the numbers of cells that express Isl1 [12, 39]. Moreover, chromatin immunoprecipitation and in vitro reporter assays indicate that β-catenin directly binds to and regulates the Isl1 promoter [39].

These data strongly suggest that canonical Wnt/β-catenin signaling plays a role in the initiation of Isl1 expression and in the specification of Isl1+ cardiac progenitors. Isl1+ progenitors also are found in the posterior region of the developing cardiac mesoderm. Our recent studies have shown that Wnt2, a ligand expressed specifically in the posterior pole of the developing heart, is essential for the development and differentiation of posterior structures including the atria, pulmonary veins and atrioventricular canal (Y. Tian and E. Morrisey, manuscript submitted). Wnt2 regulates the proliferation and differentiation of Isl1 + progenitors and early cardiomyocytes within the posterior pole of the heart and does so through the β-catenin-dependent canonical Wnt pathway. Thus, Wnt/β-catenin signaling is important for both outflow and inflow tract development via its regulation of Isl1+ SHF progenitor cell expansion and subsequent myocyte differentiation.

Wnt Signaling During Cardiac Early Differentiation: More Than One Way of Looking at It

Studies on chick and frog embryos suggest that the initial specification of cardiac tissue is governed by the balanced expression of canonical Wnt activators and repressors both within and outside the early mesoderm. Canonical Wnt signaling, through Wnt1 and Wnt3a expression in the anterior mesoderm, inhibits the expression of early cardiac genes in the cardiac crescent, including Nkx2.5 and GATA4 [44, 60]. Secreted Wnt antagonists, including crescent and Dikkopf (Dkk), are expressed in the endoderm underlying the cardiac mesoderm [60]. Moreover, Wnt expression in the dorsal neural tube blocks cardiogenesis in the adjacent paraxial mesoderm [75]. Supporting this balance in Wnt activity are data showing that forced expression of crescent or Dkk in the noncardiac posterior mesoderm induces cardiac gene expression and the appearance of beating cardiomyoctes [44, 60].

Additional data from experiments with Xenopus suggest that Wnt signaling inhibits cardiogenesis in some contexts. In Xenopus, findings have shown that Wnt11 expression is required for heart specification [15, 17]. Wnt11 is thought to promote cardiogenesis through its ability to inhibit β-catenin-dependent canonical signaling [11, 15, 17, 52]. Blocking Wnt11 signaling in the anterior mesoderm of Xenopus embryos blocks the expression of early cardiac genes including Nkx2.5, GATA4, and Tbx5, whereas expressing Wnt11 in the posterior mesoderm of frog and chick embryos induces ectopic expression of these markers as well as the appearance of beating cardiomyocytes [15, 60].

In Xenopus animal pole explants, which normally take on a neuro-ectodermal fate, Wnt11 induces cardiac tissues without inducing the expression of pan-mesodermal markers, suggesting that the effect of Wnt11 on cardiac specification is direct and not the result of increased mesoderm induction [52]. Wnt11 expression similarly coincides with the onset of cardiac gene expression in differentiating embryonic stem cells, and treating these cells with recombinant Wnt11 increases the specification of cardiac progenitors, indicating that Wnt11 also plays an essential role in murine heart induction [76].

Recent data have demonstrated a link between the important cardiac transcription factors Gata4, Gata6, and the Wnt-signaling pathway. These data show that Gata4 and Gata6 are required for specification of FHF progenitors in mice [90]. Studies in Xenopus have shown that the cardiac-promoting abilities of Wnt11 require Gata4 and Gata6 function, whereas β-catenin simultaneously inhibits Gata4 and Gata6 expression in early frog development [1].

Recently, our lab found that Wnt2 works collaboratively with Gata6 in a positive feed-forward loop to promote the proper differentiation and proliferation of posterior pole Isl1 + cardiac progenitors and early cardiac development (Y. Tian and E. Morrisey, manuscript submitted). The discrepant results highlight the differences in Wnt-signaling activity in FHF versus SHF progenitors and their derivatives. Moreover, these data highlight the differences in Wnt-signaling activity during cardiovascular development obtained with different model organisms, which is a critical point to consider in the interpretation of such data. Together, these data suggest that Wnt/β-catenin signaling plays an important role in the expansion and early differentiation of SHF Isl1 + progenitors but inhibits further differentiation of either SHF or FHF progenitors.

As described earlier, much of the discrepant roles for Wnt signaling in cardiogenesis could be reconciled by imposing a biphasic role for the pathway in which Wnt is procardiogenic in early precardiac mesoderm and inhibitory to cardiogenesis during the later stages of cardiac differentiation. This model is supported by data from multiple systems including embryonic stem cells and the developing zebra fish. The expression of canonical Wnt ligands and the activity of Wnt reporters are transiently increased in differentiating embryonic stem cells just before the expression of cardiac genes such as Nkx2.5 and GATA4 [50, 76]. Blocking canonical Wnt signaling during this early period of differentiation inhibits the expression of early cardiac markers and the appearance of beating cardiomyocytes [50]. In contrast, slightly later activation of Wnt/β-catenin leads to inhibition of cardiac differentiation [50].

In zebra fish, heat- and shock-inducible expression of both activators (wnt8) and inhibitors (dkk1) show that the temporal difference between Wnt activity promoting cardiogenesis and inhibiting it is as little as 1 h [76]. The possibility that canonical Wnt signaling plays an early positive role in cardiac induction is especially interesting in light of recent data that Wnt2 is required for cardiac differentiation in embryonic stem cells. Wnt2 has been shown to activate canonical Wnt signaling in several contexts and is expressed in the early cardiac crescent [49]. Wnt2-deficient embryonic stem cells exhibit enhanced hematopoietic differentiation but decreased cardiac and endothelial cell differentiation [82]. Thus, Wnt2 and possibly its homolog Wnt2b may play an important role in the specification of cardiac cell types from the early mesoderm [23, 49, 89].

These data suggest that canonical Wnt signaling plays a biphasic role in mouse cardiac induction, positively regulating cardiac gene expression early and inhibiting cardiac differentiation later. Such temporal specificity also helps to explain the discrepant activities of Wnt in cardiogenesis in different model systems. Thus, the precise temporal activity of Wnt/β-catenin activity would have to be controlled carefully if this pathway is ever to be harnessed for cardiovascular regenerative therapies.

Noncanonical Wnt Signaling in Cardiac Development

Growing evidence shows that noncanonical or β-cateninindependent Wnt signaling plays a role in SHF development. Two noncanonical Wnt ligands, Wnt5a and Wnt11, are expressed at the anterior pole of the heart as SHF cells migrate through on their way to the right ventricle, and mice homozygous for mutations in Wnt5a or Wnt11 have outflow tract defects consistent with those caused by disruption of the SHF. Both Wnt5a and Wnt11 promote cardiac differentiation in embryonic and adult stem cells through noncanonical pathways and may be necessary to balance β-catenin-dependent SHF proliferation in the outflow tract. However, our current understanding of the molecular and cellular mechanisms behind the effects of Wnt5a and Wnt11 on cardiac development is far from complete. The Wnt5a and Wnt11 alleles result in global loss of function, making it difficult to conclude that the effects of these mutations on the SHF are direct and not caused by the loss of paracrine signaling to adjacent cell types. This is especially important to consider in the interpretation of Wnt5a and Wnt11 mutants as the outflow tract defects observed are similar to those observed upon disruption of proper SHF development.

Furthermore, evidence suggesting that Wnt5a and Wnt11 act noncanonically in the SHF is largely composed of data from in vitro experiments, and this model remains to be tested genetically. Experiments using conditional alleles of noncanonical Wnt pathway components including Wnt5a and Wnt11 in the context of Wnt transgenic reporters are required to advance our understanding of noncanonical Wnt signaling in SHF development.

Other pathways including jun N-terminal kinase (JNK) and protein kinase C (PKC) are known to act downstream of some β-catenin-independent Wnt signaling. Inhibiting either JNK or PKC signaling blocks the ability of Wnt11 to induce cardiac specification, whereas coactivating JNK and PKC induces cardiac specification, suggesting that both the RhoA/JNK and Ca2+/PKC pathways mediate Wnt11 signaling [52]. These data indicate that the activation of noncanonical Wnt signaling by Wnt11 is required for the induction of cardiac tissues through JNK and PKC signaling.

Summary

A plethora of recent data has shown that Wnt/β-catenin signaling plays an important role in many stages of cardiovascular development including progenitor proliferation and myocyte differentiation. The use of multiple model systems has resulted in contradictory data in some cases, but recent studies in the pluripotential stem cell field and with zebra fish have helped to resolve these discrepancies and have highlighted a model in which Wnt/β-catenin is procardiogenic in the early precardiac mesoderm and inhibitory later in cardiac differentiation. Given the importance of Wnt/β-catenin signaling in promoting stem/progenitor cells from various sources, it will be important to determine whether regulation of this pathway can help to generate sufficient cardiac and vascular progenitors and their derivatives for therapeutic use in the future.

Contributor Information

Ying Tian, Department of Medicine, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA.

Ethan David Cohen, Department of Medicine, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA.

Edward E. Morrisey, Email: emorrise@mail.med.upenn.edu, Department of Medicine, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA; Cardiovascular Institute, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, 956 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104, USA.

References

  • 1.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. doi: 10.1242/dev.026443. [DOI] [PubMed] [Google Scholar]
  • 2.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 USA. 2007;104:9319–9324. doi: 10.1073/pnas.0701212104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Baker JC, Beddington RS, Harland RM. Wnt signaling in Xenopus embryos inhibits bmp4 expression and activates neural development. Genes Dev. 1999;13:3149–3159. doi: 10.1101/gad.13.23.3149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bradley RS, Brown AM. A soluble form of Wnt-1 protein with mitogenic activity on mammary epithelial cells. Mol Cell Biol. 1995;15:4616–4622. doi: 10.1128/mcb.15.8.4616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Bruneau BG, Logan M, Davis N, Levi T, Tabin CJ, Seidman JG, Seidman CE. Chamber-specific cardiac expression of Tbx5 and heart defects in Holt-Oram syndrome. Dev Biol. 1999;211:100–108. doi: 10.1006/dbio.1999.9298. [DOI] [PubMed] [Google Scholar]
  • 6.Bruneau BG, Nemer G, Schmitt JP, Charron F, Robitaille L, Caron S, Conner DA, Gessler M, Nemer M, Seidman CE, Seidman JG. A murine model of Holt-Oram syndrome defines roles of the T-box transcription factor Tbx5 in cardiogenesis and disease. Cell. 2001;106:709–721. doi: 10.1016/s0092-8674(01)00493-7. [DOI] [PubMed] [Google Scholar]
  • 7.Cadigan KM, Nusse R. Wnt signaling: a common theme in animal development. Genes Dev. 1997;11:3286–3305. doi: 10.1101/gad.11.24.3286. [DOI] [PubMed] [Google Scholar]
  • 8.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. doi: 10.1016/s1534-5807(03)00363-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen L, Wu Q, Guo F, Xia B, Zuo J. Expression of Dishevelled-1 in wound healing after acute myocardial infarction: possible involvement in myofibroblast proliferation and migration. J Cell Mol Med. 2004;8:257–264. doi: 10.1111/j.1582-4934.2004.tb00281.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Chen X, Shevtsov SP, Hsich E, Cui L, Haq S, Aronovitz M, Kerkela R, Molkentin JD, Liao R, Salomon RN, Patten R, Force T. The beta-catenin/T-cell factor/lymphocyte enhancer factor signaling pathway is required for normal and stressinduced cardiac hypertrophy. Mol Cell Biol. 2006;26:4462–4473. doi: 10.1128/MCB.02157-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Christiansen JH, Monkley SJ, Wainwright BJ. Murine WNT11 is a secreted glycoprotein that morphologically transforms mammary epithelial cells. Oncogene. 1996;12:2705–2711. [PubMed] [Google Scholar]
  • 12.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. doi: 10.1172/JCI31731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cohen ED, Ihida-Stansbury K, Lu MM, Panettieri RA, Jones PL, Morrisey EE. Wnt signaling regulates smooth muscle precursor development in the mouse lung via a tenascin C/PDGFR pathway. J Clin Invest. 2009;119:2538–2549. doi: 10.1172/JCI38079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Daneman R, Agalliu D, Zhou L, Kuhnert F, Kuo CJ, Barres BA. Wnt/beta-catenin signaling is required for CNS, but not non-CNS, angiogenesis. Proc Natl Acad Sci USA. 2009;106:641–646. doi: 10.1073/pnas.0805165106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Eisenberg CA, Eisenberg LM. WNT11 promotes cardiac tissue formation of early mesoderm. Dev Dyn. 1999;216:45–58. doi: 10.1002/(SICI)1097-0177(199909)216:1<45::AID-DVDY7>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 16.Etheridge SL, Ray S, Li S, Hamblet NS, Lijam N, Tsang M, Greer J, Kardos N, Wang J, Sussman DJ, Chen P, Wynshaw-Boris A. Murine dishevelled 3 functions in redundant pathways with dishevelled 1 and 2 in normal cardiac outflow tract, cochlea, and neural tube development. PLoS Genet. 2008;4:e1000259. doi: 10.1371/journal.pgen.1000259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.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. doi: 10.1016/j.ydbio.2004.12.013. [DOI] [PubMed] [Google Scholar]
  • 18.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. doi: 10.1242/dev.00164. [DOI] [PubMed] [Google Scholar]
  • 19.Heisenberg CP, Tada M, Rauch GJ, Saude L, Concha ML, Geisler R, Stemple DL, Smith JC, Wilson SW. Silberblick/Wnt11 mediates convergent extension movements during zebra-fish gastrulation. Nature. 2000;405:76–81. doi: 10.1038/35011068. [DOI] [PubMed] [Google Scholar]
  • 20.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. doi: 10.1038/nature02028. [DOI] [PubMed] [Google Scholar]
  • 21.Ishikawa T, Tamai Y, Zorn AM, Yoshida H, Seldin MF, Nishikawa S, Taketo MM. Mouse Wnt receptor gene Fzd5 is essential for yolk sac and placental angiogenesis. Development. 2001;128:25–33. doi: 10.1242/dev.128.1.25. [DOI] [PubMed] [Google Scholar]
  • 22.James RG, Conrad WH, Moon RT. Beta-catenin-independent Wnt pathways: signals, core proteins, and effectors. Methods Mol Biol. 2008;468:131–144. doi: 10.1007/978-1-59745-249-6_10. [DOI] [PubMed] [Google Scholar]
  • 23.Jaspard B, Couffinhal T, Dufourcq P, Moreau C, Duplaa C. Expression pattern of mouse sFRP-1 and mWnt-8 gene during heart morphogenesis. Mech Dev. 2000;90:263–267. doi: 10.1016/s0925-4773(99)00236-1. [DOI] [PubMed] [Google Scholar]
  • 24.Kato M, Patel MS, Levasseur R, Lobov I, Chang BH, Glass DA, II, Hartmann C, Li L, Hwang TH, Brayton CF, Lang RA, Karsenty G, Chan L. Cbfa1-independent decrease in osteoblast proliferation, osteopenia, and persistent embryonic eye vascularization in mice deficient in Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–314. doi: 10.1083/jcb.200201089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Kelly M, Chen P. Shaping the mammalian auditory sensory organ by the planar cell polarity pathway. Int J Dev Biol. 2007;51:535–547. doi: 10.1387/ijdb.072344mk. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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. doi: 10.1016/s1534-5807(01)00040-5. [DOI] [PubMed] [Google Scholar]
  • 27.Kelly RG, Buckingham ME. The anterior heart-forming field: voyage to the arterial pole of the heart. Trends Genet. 2002;18:210–216. doi: 10.1016/s0168-9525(02)02642-2. [DOI] [PubMed] [Google Scholar]
  • 28.Kilian B, Mansukoski H, Barbosa FC, Ulrich F, Tada M, Heisenberg CP. The role of Ppt/Wnt5 in regulating cell shape and movement during zebrafish gastrulation. Mech Dev. 2003;120:467–476. doi: 10.1016/s0925-4773(03)00004-2. [DOI] [PubMed] [Google Scholar]
  • 29.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-&gt; Pitx2 pathway mediating cell-type-specific proliferation during development. Cell. 2002;111:673–685. doi: 10.1016/s0092-8674(02)01084-x. [DOI] [PubMed] [Google Scholar]
  • 30.Kispert A, Vainio S, McMahon AP. Wnt-4 is a mesenchymal signal for epithelial transformation of metanephric mesenchyme in the developing kidney. Development. 1998;125:4225–4234. doi: 10.1242/dev.125.21.4225. [DOI] [PubMed] [Google Scholar]
  • 31.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. doi: 10.1073/pnas.0703113104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kohn AD, Moon RT. Wnt and calcium signaling: betacatenin-independent pathways. Cell Calcium. 2005;38:439–446. doi: 10.1016/j.ceca.2005.06.022. [DOI] [PubMed] [Google Scholar]
  • 33.Kuhl M, Sheldahl LC, Malbon CC, Moon RT. Ca(2 +)/calmodulin-dependent protein kinase II is stimulated by Wnt and Frizzled homologs and promotes ventral cell fates in Xenopus. J Biol Chem. 2000;275:12701–12711. doi: 10.1074/jbc.275.17.12701. [DOI] [PubMed] [Google Scholar]
  • 34.Kuhl M, Sheldahl LC, Park M, Miller JR, Moon RT. The Wnt/Ca2 + pathway: a new vertebrate Wnt-signaling pathway takes shape. Trends Genet. 2000;16:279–283. doi: 10.1016/s0168-9525(00)02028-x. [DOI] [PubMed] [Google Scholar]
  • 35.Kuhl M. Noncanonical Wnt signaling in Xenopus: regulation of axis formation and gastrulation. Semin Cell Dev Biol. 2002;13:243–249. doi: 10.1016/s1084-9521(02)00050-2. [DOI] [PubMed] [Google Scholar]
  • 36.Kuo CT, Morrisey EE, Anandappa R, Sigrist K, Lu MM, Parmacek MS, Soudais C, Leiden JM. GATA4 transcription factor is required for ventral morphogenesis and heart tube formation. Genes Dev. 1997;11:1048–1060. doi: 10.1101/gad.11.8.1048. [DOI] [PubMed] [Google Scholar]
  • 37.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 USA. 2007;104:10894–10899. doi: 10.1073/pnas.0704044104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.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. doi: 10.1083/jcb.200403050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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 U S A. 2007;104:9313–9318. doi: 10.1073/pnas.0700923104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lin Q, Schwarz J, Bucana C, Olson EN. Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science. 1997;276:1404–1407. doi: 10.1126/science.276.5317.1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lyons I, Parsons LM, Hartley L, Li R, Andrews JE, Robb L, Harvey RP. Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeobox gene Nkx2-5. Genes Dev. 1995;9:1654–1666. doi: 10.1101/gad.9.13.1654. [DOI] [PubMed] [Google Scholar]
  • 42.Malbon CC, Wang H, Moon RT. Wnt signaling and heterotrimeric G-proteins: strange bedfellows or a classic romance? Biochem Biophys Res Commun. 2001;287:589–593. doi: 10.1006/bbrc.2001.5630. [DOI] [PubMed] [Google Scholar]
  • 43.Marlow F, Topczewski J, Sepich D, Solnica-Krezel L. Zebrafish Rho kinase 2 acts downstream of Wnt11 to mediate cell polarity and effective convergence and extension movements. Curr Biol. 2002;12:876–884. doi: 10.1016/s0960-9822(02)00864-3. [DOI] [PubMed] [Google Scholar]
  • 44.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. doi: 10.1101/gad.855501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Masckauchan TN, Agalliu D, Vorontchikhina M, Ahn A, Parmalee NL, Li CM, Khoo A, Tycko B, Brown AM, Kitajewski J. Wnt5a signaling induces proliferation and survival of endothelial cells in vitro and expression of MMP-1 and Tie-2. Mol Biol Cell. 2006;17:5163–5172. doi: 10.1091/mbc.E06-04-0320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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. doi: 10.1073/pnas.0504343102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mikels AJ, Nusse R. Purified Wnt5a protein activates or inhibits beta-catenin-TCF signaling depending on receptor context. PLoS Biol. 2006;4:e115. doi: 10.1371/journal.pbio.0040115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Molkentin JD, Lin Q, Duncan SA, Olson EN. Requirement of the transcription factor GATA4 for heart tube formation and ventral morphogenesis. Genes Dev. 1997;11:1061–1072. doi: 10.1101/gad.11.8.1061. [DOI] [PubMed] [Google Scholar]
  • 49.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. doi: 10.1242/dev.122.11.3343. [DOI] [PubMed] [Google Scholar]
  • 50.Naito AT, Shiojima I, Akazawa H, Hidaka K, Morisaki T, Kikuchi A, Komuro I. Developmental stage-specific biphasic roles of Wnt/beta-catenin signaling in cardiomyogenesis and hematopoiesis. Proc Natl Acad Sci U S A. 2006;103:19812–19817. doi: 10.1073/pnas.0605768103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Nakamura T, Sano M, Songyang Z, Schneider MD. A Wnt- and beta-catenin-dependent pathway for mammalian cardiac myogenesis. Proc Natl Acad Sci U S A. 2003;100:5834–5839. doi: 10.1073/pnas.0935626100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.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. doi: 10.1038/nature00921. [DOI] [PubMed] [Google Scholar]
  • 53.Parr BA, McMahon AP. Wnt genes and vertebrate development. Curr Opin Genet Dev. 1994;4:523–528. doi: 10.1016/0959-437x(94)90067-d. [DOI] [PubMed] [Google Scholar]
  • 54.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. doi: 10.1161/01.RES.0000154912.08695.88. [DOI] [PubMed] [Google Scholar]
  • 55.Qian D, Jones C, Rzadzinska A, Mark S, Zhang X, Steel KP, Dai X, Chen P. Wnt5a functions in planar cell polarity regulation in mice. Dev Biol. 2007;306:121–133. doi: 10.1016/j.ydbio.2007.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Qyang Y, Martin-Puig S, Chiravuri M, Chen S, Xu H, Bu L, Jiang X, 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. doi: 10.1016/j.stem.2007.05.018. [DOI] [PubMed] [Google Scholar]
  • 57.Reya T, O’Riordan M, Okamura R, Devaney E, Willert K, Nusse R, Grosschedl R. Wnt signaling regulates B lymphocyte proliferation through a LEF-1 dependent-mechanism. Immunity. 2000;13:15–24. doi: 10.1016/s1074-7613(00)00004-2. [DOI] [PubMed] [Google Scholar]
  • 58.Saneyoshi T, Kume S, Amasaki Y, Mikoshiba K. The Wnt/calcium pathway activates NF-AT and promotes ventral cell fate in Xenopus embryos. Nature. 2002;417:295–299. doi: 10.1038/417295a. [DOI] [PubMed] [Google Scholar]
  • 59.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. doi: 10.1203/pdr.0b013e3180323810. [DOI] [PubMed] [Google Scholar]
  • 60.Schneider VA, Mercola M. Wnt antagonism initiates cardiogenesis in Xenopus laevis. Genes Dev. 2001;15:304–315. doi: 10.1101/gad.855601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Sheldahl LC, Slusarski DC, Pandur P, Miller JR, Kuhl M, Moon RT. Dishevelled activates Ca2+ flux, PKC, and CamKII in vertebrate embryos. J Cell Biol. 2003;161:769–777. doi: 10.1083/jcb.200211094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Shu W, Jiang YQ, Lu MM, Morrisey EE. Wnt7b regulates mesenchymal proliferation and vascular development in the lung. Development. 2002;129:4831–4842. doi: 10.1242/dev.129.20.4831. [DOI] [PubMed] [Google Scholar]
  • 63.Slusarski DC, Corces VG, Moon RT. Interaction of Wnt and a Frizzled homologue triggers G-protein-linked phosphatidylinositol signalling. Nature. 1997;390:410–413. doi: 10.1038/37138. [DOI] [PubMed] [Google Scholar]
  • 64.Smalley MJ, Dale TC. Wnt signalling in mammalian development and cancer. Cancer Metastasis Rev. 1999;18:215–230. doi: 10.1023/a:1006369223282. [DOI] [PubMed] [Google Scholar]
  • 65.Stark K, Vainio S, Vassileva G, McMahon AP. Epithelial transformation of metanephric mesenchyme in the developing kidney regulated by Wnt-4. Nature. 1994;372:679–683. doi: 10.1038/372679a0. [DOI] [PubMed] [Google Scholar]
  • 66.Stenman JM, Rajagopal J, Carroll TJ, Ishibashi M, McMahon J, McMahon AP. Canonical Wnt signaling regulates organspecific assembly and differentiation of CNS vasculature. Science. 2008;322:1247–1250. doi: 10.1126/science.1164594. [DOI] [PubMed] [Google Scholar]
  • 67.Strutt D. Frizzled signalling and cell polarisation in Drosophila and vertebrates. Development. 2003;130:4501–4513. doi: 10.1242/dev.00695. [DOI] [PubMed] [Google Scholar]
  • 68.Tada M, Concha ML, Heisenberg CP. Noncanonical Wnt signalling and regulation of gastrulation movements. Semin Cell Dev Biol. 2002;13:251–260. doi: 10.1016/s1084-9521(02)00052-6. [DOI] [PubMed] [Google Scholar]
  • 69.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. doi: 10.1016/j.bbrc.2004.10.103. [DOI] [PubMed] [Google Scholar]
  • 70.Toomes C, Bottomley HM, Jackson RM, Towns KV, Scott S, Mackey DA, Craig JE, Jiang L, Yang Z, Trembath R, Woodruff G, Gregory-Evans CY, Gregory-Evans K, Parker MJ, Black GC, Downey LM, Zhang K, Inglehearn CF. Mutations in LRP5 or FZD4 underlie the common familial exudative vitreoretinopathy locus on chromosome 11q. Am J Hum Genet. 2004;74:721–730. doi: 10.1086/383202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Topol L, Jiang X, Choi H, Garrett-Beal L, Carolan PJ, Yang Y. Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent beta-catenin degradation. J Cell Biol. 2003;162:899–908. doi: 10.1083/jcb.200303158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Trivedi CM, Luo Y, Yin Z, Zhang M, Zhu W, Wang T, Floss T, Goettlicher M, Noppinger PR, Wurst W, Ferrari VA, Abrams CS, Gruber PJ, Epstein JA. Hdac2 regulates the cardiac hypertrophic response by modulating Gsk3 beta activity. Nat Med. 2007;13:324–331. doi: 10.1038/nm1552. [DOI] [PubMed] [Google Scholar]
  • 73.Tseng AS, Engel FB, Keating MT. The GSK-3 inhibitor BIO promotes proliferation in mammalian cardiomyocytes. Chem Biol. 2006;13:957–963. doi: 10.1016/j.chembiol.2006.08.004. [DOI] [PubMed] [Google Scholar]
  • 74.Tu X, Joeng KS, Nakayama KI, Nakayama K, Rajagopal J, Carroll TJ, McMahon AP, Long F. Noncanonical Wnt signaling through G protein-linked PKCdelta activation promotes bone formation. Dev Cell. 2007;12:113–127. doi: 10.1016/j.devcel.2006.11.00. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tzahor E, Lassar AB. Wnt signals from the neural tube block ectopic cardiogenesis. Genes Dev. 2001;15:255–260. doi: 10.1101/gad.871501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.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 zebra fish and embryonic stem cells. Proc Natl Acad Sci U S A. 2007;104:9685–9690. doi: 10.1073/pnas.0702859104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.van Amerongen R, Mikels A, Nusse R. Alternative wnt signaling is initiated by distinct receptors. Sci Signal. 2008;1:re9. doi: 10.1126/scisignal.135re9. [DOI] [PubMed] [Google Scholar]
  • 78.van Amerongen R, Nusse R. Towards an integrated view of Wnt signaling in development. Development. 2009;136:3205–3214. doi: 10.1242/dev.033910. [DOI] [PubMed] [Google Scholar]
  • 79.van de Schans VA, van den Borne SW, Strzelecka AE, Janssen BJ, van der Velden JL, Langen RC, Wynshaw-Boris A, Smits JF, Blankesteijn WM. Interruption of Wnt signaling attenuates the onset of pressure overload-induced cardiac hypertrophy. Hypertension. 2007;49:473–480. doi: 10.1161/01.HYP.0000255946.55091.24. [DOI] [PubMed] [Google Scholar]
  • 80.Wallingford JB, Rowning BA, Vogeli KM, Rothbacher U, Fraser SE, Harland RM. Dishevelled controls cell polarity during Xenopus gastrulation. Nature. 2000;405:81–85. doi: 10.1038/35011077. [DOI] [PubMed] [Google Scholar]
  • 81.Wallingford JB, Fraser SE, Harland RM. Convergent extension: the molecular control of polarized cell movement during embryonic development. Dev Cell. 2002;2:695–706. doi: 10.1016/s1534-5807(02)00197-1. [DOI] [PubMed] [Google Scholar]
  • 82.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. doi: 10.1074/jbc.M606610200. [DOI] [PubMed] [Google Scholar]
  • 83.Wang J, Mark S, Zhang X, Qian D, Yoo SJ, Radde-Gallwitz K, Zhang Y, Lin X, Collazo A, Wynshaw-Boris A, Chen P. Regulation of polarized extension and planar cell polarity in the cochlea by the vertebrate PCP pathway. Nat Genet. 2005;37:980–985. doi: 10.1038/ng1622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang X, Xiao Y, Mou Y, Zhao Y, Blankesteijn WM, Hall JL. A role for the beta-catenin/T-cell factor signaling cascade in vascular remodeling. Circ Res. 2002;90:340–347. doi: 10.1161/hh0302.104466. [DOI] [PubMed] [Google Scholar]
  • 85.Wang X, Adhikari N, Li Q, Hall JL. LDL receptor-related protein LRP6 regulates proliferation and survival through the Wnt cascade in vascular smooth muscle cells. Am J Physiol Heart Circ Physiol. 2004;287:H2376–H2383. doi: 10.1152/ajpheart.01173.2003. [DOI] [PubMed] [Google Scholar]
  • 86.Wang Z, Shu W, Lu MM, Morrisey EE. Wnt7b activates canonical signaling in epithelial and vascular smooth muscle cells through interactions with Fzd1, Fzd10, and LRP5. Mol Cell Biol. 2005;25:5022–5030. doi: 10.1128/MCB.25.12.5022-5030.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Westfall TA, Brimeyer R, Twedt J, Gladon J, Olberding A, Furutani-Seiki M, Slusarski DC. Wnt-5/pipetail functions in vertebrate axis formation as a negative regulator of Wnt/betacatenin activity. J Cell Biol. 2003;162:889–898. doi: 10.1083/jcb.200303107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Xu Q, Wang Y, Dabdoub A, Smallwood PM, Williams J, Woods C, Kelley MW, Jiang L, Tasman W, Zhang K, Nathans J. Vascular development in the retina and inner ear: control by Norrin and Frizzled-4, a high-affinity ligand-receptor pair. Cell. 2004;116:883–895. doi: 10.1016/s0092-8674(04)00216-8. [DOI] [PubMed] [Google Scholar]
  • 89.Zakin LD, Mazan S, Maury M, Martin N, Guenet JL, Brulet P. Structure and expression of Wnt13, a novel mouse Wnt2 related gene. Mech Dev. 1998;73:107–116. doi: 10.1016/s0925-4773(98)00040-9. [DOI] [PubMed] [Google Scholar]
  • 90.Zhao R, Watt AJ, Battle MA, Li J, Bondow BJ, Duncan SA. Loss of both GATA4 and GATA6 blocks cardiac myocyte differentiation and results in acardia in mice. Dev Biol. 2008;317:614–619. doi: 10.1016/j.ydbio.2008.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.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. doi: 10.1038/ng2112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhu S, Liu L, Korzh V, Gong Z, Low BC. RhoA acts downstream of Wnt5 and Wnt11 to regulate convergence and extension movements by involving effectors Rho kinase and Diaphanous: use of zebra fish as an in vivo model for GTPase signaling. Cell Signal. 2006;18:359–372. doi: 10.1016/j.cellsig.2005.05.019. [DOI] [PubMed] [Google Scholar]

RESOURCES