Top Banner
© 2014. Published by The Company of Biologists Ltd | Development (2014) 141, 1-11 doi:10.1242/dev.100495 1 ABSTRACT The VEGFC/VEGFR3 signaling pathway is essential for lymphangiogenesis (the formation of lymphatic vessels from pre- existing vasculature) during embryonic development, tissue regeneration and tumor progression. The recently identified secreted protein CCBE1 is indispensible for lymphangiogenesis during development. The role of CCBE1 orthologs is highly conserved in zebrafish, mice and humans with mutations in CCBE1 causing generalized lymphatic dysplasia and lymphedema (Hennekam syndrome). To date, the mechanism by which CCBE1 acts remains unknown. Here, we find that ccbe1 genetically interacts with both vegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven by increased Vegfc are suppressed in the absence of Ccbe1, and Vegfc- driven sprouting is enhanced by local Ccbe1 overexpression. Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired in the absence of Ccbe1. Finally, CCBE1 is capable of upregulating the levels of fully processed, mature VEGFC in vitro and the overexpression of mature VEGFC rescues ccbe1 loss-of-function phenotypes in zebrafish. Taken together, these data identify Ccbe1 as a crucial component of the Vegfc/Vegfr3 pathway in the embryo. KEY WORDS: Angiogenesis, Lymphangiogenesis, Lymphedema, Vasculature, Zebrafish INTRODUCTION The lymphatic vasculature develops from pre-existing vessels in a dynamic process called lymphangiogenesis, and is necessary to preserve tissue fluid homeostasis, for fat absorption and normal immune function. The lymphatic vascular network originates in the mouse embryo from the cardinal vein, where lymphatic endothelial precursor cells first bud and migrate dorsolaterally away from the vein under the control of Vegfc and its receptor Vegfr3 (Flt4 – Mouse Genome Informatics). In mice, the overexpression of Vegfc in the skin results in dramatic hyperplasia of lymphatic vessels (Jeltsch et al., 1997), whereas Vegfc knockout mice develop lymphatic hypoplasia and lymphedema (Karkkainen et al., 2004). In developing Vegfc knockout embryos, endothelial cells are still specified to the lymphatic RESEARCH ARTICLE 1 Division of Molecular Genetics and Development, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4073, Australia. 2 Hubrecht Institute – KNAW & UMC Utrecht, 3584 CT Utrecht, The Netherlands. 3 Tumour Angiogenesis Programme, Peter MacCallum Cancer Centre, Locked Bag 1, A’Beckett Street, Melbourne, VIC 8006, Australia. 4 Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3010, Australia. 5 EZO, WUR, 6708LX Wageningen, The Netherlands. *Present address: U1046, Université Montpellier 1, Université Montpellier 2, 34967 CEDEX 2 Montpellier, France. These authors contributed equally to this work § Author for correspondence ([email protected]) Received 26 June 2013; Accepted 30 December 2013 lineage, and express Prox1, but fail to migrate from the cardinal vein (Karkkainen et al., 2004; Hägerling et al., 2013). Reduced development, or impaired function of lymphatics, leads to tissue fluid accumulation and lymphedema in humans, which can be inherited as a result of mutations in key developmental genes (Karkkainen et al., 2000; Irrthum et al., 2003; Alders et al., 2009; Connell et al., 2010). A mutation in VEGFC has recently been shown to be responsible for inherited lymphedema (Gordon et al., 2013). Vegfr3-deficient mice die early during gestation (embryonic day 9.5) owing to cardiovascular failure (Dumont et al., 1998), consistent with the expression of Vegfr3 in blood vascular endothelium in the early embryo. However, after midgestation, Vegfr3 expression becomes enriched in the developing lymphatic vasculature (Kaipainen et al., 1995) and Vegfr3 signaling is sufficient to initiate lymphangiogenesis (Veikkola et al., 2001). Importantly, patients with mutations in VEGFR3 (FLT4 – Human Gene Nomenclature Database) develop Milroy’s disease, characterized by reduced lymphatic drainage and lymphedema in the lower limbs (Karkkainen et al., 2000). These studies, and many more, have shown that the precise modulation of the Vegfc/Vegfr3 signaling pathway is crucial during lymphatic vascular development. In zebrafish, lymphatic vascular development initiates from 32 hours post-fertilization (hpf), when precursor cells first migrate dorsally from the cardinal vein to colonize the horizontal myoseptum, generating a population of parachordal lymphangioblasts (PLs) (Küchler et al., 2006; Yaniv et al., 2006; Hogan et al., 2009a; Isogai et al., 2009). These PLs subsequently migrate alongside arteries, both dorsally and ventrally (from ~60 hpf), and remodel into the major trunk lymphatic vessels (Bussmann et al., 2010; Cha et al., 2012), forming the thoracic duct, intersegmental lymphatic vessels and dorsal longitudinal lymphatic vessels (for reviews, see Koltowska et al., 2013; van Impel and Schulte-Merker, 2014). In addition, a complex craniofacial lymphatic network is formed (Okuda et al., 2012). Zebrafish vegfr3 (flt4 – Zebrafish Information Network) and vegfc function is essential for all secondary (venous) angiogenesis, including the sprouting of lymphatic precursors from the cardinal vein and subsequent formation of PLs (Isogai et al., 2003; Küchler et al., 2006; Yaniv et al., 2006; Hogan et al., 2009a). We have previously reported two zebrafish mutants with an absence of lymphatic (but not blood vascular) development (Hogan et al., 2009a; Hogan et al., 2009b). These phenotypes were caused by mutations in vegfr3 and the previously uncharacterized gene collagen and calcium-binding EGF domains-1 (ccbe1) (Hogan et al., 2009a; Hogan et al., 2009b). ccbe1 was shown to act at stages identical to vegfr3 and vegfc (Hogan et al., 2009a). ccbe1 encodes an extracellular matrix (ECM) protein, and is composed of N- terminal calcium-binding epidermal growth factor (EGF)-like and EGF domains, and two collagen-repeat domains towards the C Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signaling during embryonic lymphangiogenesis Ludovic Le Guen 1, *, Terhi Karpanen 2 , Dörte Schulte 2 , Nicole C. Harris 3,4 , Katarzyna Koltowska 1 , Guy Roukens 2 , Neil I. Bower 1 , Andreas van Impel 2 , Steven A. Stacker 3,4 , Marc G. Achen 3,4 , Stefan Schulte-Merker 2,5,‡ and Benjamin M. Hogan 1,‡,§ Development Development ePress. Posted online 12 February 2014 http://dev.biologists.org/lookup/doi/10.1242/dev.100495 Access the most recent version at First posted online on 12 February 2014 as 10.1242/dev.100495
11

Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

Jun 06, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

© 2014. Published by The Company of Biologists Ltd | Development (2014) 141, 1-11 doi:10.1242/dev.100495

1

ABSTRACTThe VEGFC/VEGFR3 signaling pathway is essential forlymphangiogenesis (the formation of lymphatic vessels from pre-existing vasculature) during embryonic development, tissueregeneration and tumor progression. The recently identified secretedprotein CCBE1 is indispensible for lymphangiogenesis duringdevelopment. The role of CCBE1 orthologs is highly conserved inzebrafish, mice and humans with mutations in CCBE1 causinggeneralized lymphatic dysplasia and lymphedema (Hennekamsyndrome). To date, the mechanism by which CCBE1 acts remainsunknown. Here, we find that ccbe1 genetically interacts with bothvegfc and vegfr3 in zebrafish. In the embryo, phenotypes driven byincreased Vegfc are suppressed in the absence of Ccbe1, and Vegfc-driven sprouting is enhanced by local Ccbe1 overexpression.Moreover, Vegfc- and Vegfr3-dependent Erk signaling is impaired inthe absence of Ccbe1. Finally, CCBE1 is capable of upregulating thelevels of fully processed, mature VEGFC in vitro and theoverexpression of mature VEGFC rescues ccbe1 loss-of-functionphenotypes in zebrafish. Taken together, these data identify Ccbe1as a crucial component of the Vegfc/Vegfr3 pathway in the embryo.

KEY WORDS: Angiogenesis, Lymphangiogenesis, Lymphedema,Vasculature, Zebrafish

INTRODUCTIONThe lymphatic vasculature develops from pre-existing vessels in adynamic process called lymphangiogenesis, and is necessary topreserve tissue fluid homeostasis, for fat absorption and normalimmune function. The lymphatic vascular network originates in themouse embryo from the cardinal vein, where lymphatic endothelialprecursor cells first bud and migrate dorsolaterally away from the veinunder the control of Vegfc and its receptor Vegfr3 (Flt4 – MouseGenome Informatics). In mice, the overexpression of Vegfc in the skinresults in dramatic hyperplasia of lymphatic vessels (Jeltsch et al.,1997), whereas Vegfc knockout mice develop lymphatic hypoplasiaand lymphedema (Karkkainen et al., 2004). In developing Vegfcknockout embryos, endothelial cells are still specified to the lymphatic

RESEARCH ARTICLE

1Division of Molecular Genetics and Development, Institute for MolecularBioscience, The University of Queensland, St Lucia, QLD 4073, Australia.2Hubrecht Institute – KNAW & UMC Utrecht, 3584 CT Utrecht, The Netherlands.3Tumour Angiogenesis Programme, Peter MacCallum Cancer Centre, Locked Bag1, A’Beckett Street, Melbourne, VIC 8006, Australia. 4Sir Peter MacCallumDepartment of Oncology, University of Melbourne, Parkville, VIC 3010, Australia.5EZO, WUR, 6708LX Wageningen, The Netherlands. *Present address: U1046, Université Montpellier 1, Université Montpellier 2, 34967CEDEX 2 Montpellier, France.‡These authors contributed equally to this work

§Author for correspondence ([email protected])

Received 26 June 2013; Accepted 30 December 2013

lineage, and express Prox1, but fail to migrate from the cardinal vein(Karkkainen et al., 2004; Hägerling et al., 2013). Reduceddevelopment, or impaired function of lymphatics, leads to tissue fluidaccumulation and lymphedema in humans, which can be inherited asa result of mutations in key developmental genes (Karkkainen et al.,2000; Irrthum et al., 2003; Alders et al., 2009; Connell et al., 2010).A mutation in VEGFC has recently been shown to be responsible forinherited lymphedema (Gordon et al., 2013).

Vegfr3-deficient mice die early during gestation (embryonic day9.5) owing to cardiovascular failure (Dumont et al., 1998),consistent with the expression of Vegfr3 in blood vascularendothelium in the early embryo. However, after midgestation,Vegfr3 expression becomes enriched in the developing lymphaticvasculature (Kaipainen et al., 1995) and Vegfr3 signaling issufficient to initiate lymphangiogenesis (Veikkola et al., 2001).Importantly, patients with mutations in VEGFR3 (FLT4 – HumanGene Nomenclature Database) develop Milroy’s disease,characterized by reduced lymphatic drainage and lymphedema in thelower limbs (Karkkainen et al., 2000). These studies, and manymore, have shown that the precise modulation of the Vegfc/Vegfr3signaling pathway is crucial during lymphatic vascular development.

In zebrafish, lymphatic vascular development initiates from32 hours post-fertilization (hpf), when precursor cells first migratedorsally from the cardinal vein to colonize the horizontalmyoseptum, generating a population of parachordallymphangioblasts (PLs) (Küchler et al., 2006; Yaniv et al., 2006;Hogan et al., 2009a; Isogai et al., 2009). These PLs subsequentlymigrate alongside arteries, both dorsally and ventrally (from ~60hpf), and remodel into the major trunk lymphatic vessels (Bussmannet al., 2010; Cha et al., 2012), forming the thoracic duct,intersegmental lymphatic vessels and dorsal longitudinal lymphaticvessels (for reviews, see Koltowska et al., 2013; van Impel andSchulte-Merker, 2014). In addition, a complex craniofaciallymphatic network is formed (Okuda et al., 2012). Zebrafish vegfr3(flt4 – Zebrafish Information Network) and vegfc function isessential for all secondary (venous) angiogenesis, including thesprouting of lymphatic precursors from the cardinal vein andsubsequent formation of PLs (Isogai et al., 2003; Küchler et al.,2006; Yaniv et al., 2006; Hogan et al., 2009a).

We have previously reported two zebrafish mutants with anabsence of lymphatic (but not blood vascular) development (Hoganet al., 2009a; Hogan et al., 2009b). These phenotypes were causedby mutations in vegfr3 and the previously uncharacterized genecollagen and calcium-binding EGF domains-1 (ccbe1) (Hogan etal., 2009a; Hogan et al., 2009b). ccbe1 was shown to act at stagesidentical to vegfr3 and vegfc (Hogan et al., 2009a). ccbe1 encodesan extracellular matrix (ECM) protein, and is composed of N-terminal calcium-binding epidermal growth factor (EGF)-like andEGF domains, and two collagen-repeat domains towards the C

Ccbe1 regulates Vegfc-mediated induction of Vegfr3 signalingduring embryonic lymphangiogenesisLudovic Le Guen1,*, Terhi Karpanen2, Dörte Schulte2, Nicole C. Harris3,4, Katarzyna Koltowska1, Guy Roukens2, Neil I. Bower1, Andreas van Impel2, Steven A. Stacker3,4, Marc G. Achen3,4, Stefan Schulte-Merker2,5,‡ and Benjamin M. Hogan1,‡,§

Dev

elop

men

t

Development ePress. Posted online 12 February 2014http://dev.biologists.org/lookup/doi/10.1242/dev.100495Access the most recent version at First posted online on 12 February 2014 as 10.1242/dev.100495

Page 2: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

2

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 1. See next page for legend. Dev

elop

men

t

Page 3: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

terminus of the protein. The protein localizes to secretory vesicles(Alders et al., 2009), and is secreted to the ECM, where it bindsproteins such as collagens or vitronectin (Bos et al., 2011).Consistently, Ccbe1 functions non-cell-autonomously in zebrafish(Hogan et al., 2009a). There are no described receptors or pathwaysknown to interact with Ccbe1, with progress limited at least in partby the fact that full-length CCBE1 protein is insoluble in manystandard assays (S.S.-M. and B.M.H., unpublished observations).

Importantly, mice deficient for Ccbe1 lack lymphatic vasculature(Bos et al., 2011) and mutations in human CCBE1 are causative forgeneralized lymphedema and lymphangiectasia in Hennekamsyndrome (Alders et al., 2009; Connell et al., 2010). Interestingly,treatment with Ccbe1 protein (a truncated soluble form) increasedVegfc-induced lymphangiogenesis in a cornea micropocket assay (Boset al., 2011). Furthermore, mice heterozygous for both Vegfc andCcbe1 show a genetic interaction in lymphatic development(Hägerling et al., 2013). These results were suggestive that Ccbe1 andVegfc can function together in lymphangiogenesis, but mechanisticinsights into the molecular function of Ccbe1 have remained elusive.

Here, we have investigated the relationship between Ccbe1 and theVegfc/Vegfr3 signaling pathways. Using genetic epistasis and analysisof signaling events in vivo, we show that the embryo requires Ccbe1for normal Vegfc/Vegfr3/Erk signaling. CCBE1 is capable ofincreasing the levels of mature, processed VEGFC in vitro, which canrescue ccbe1 loss-of-function phenotypes when overexpressed in vivo.Together, our findings suggest that Ccbe1 activates Vegfc through itsprocessing and release from the ECM in order to regulatelymphangiogenesis and venous sprouting in the embryo. Thesefindings suggest common mechanisms in development and vascularpathologies (e.g. lymphedema) and further suggest Ccbe1 as a newtherapeutic entry point in treating pathological lymphangiogenesis.

RESULTSIdentification of four zebrafish vegfc mutant allelesIn a forward genetic screen for zebrafish lymphatic vascularmutants, we identified several mutants that were indistinguishable

from vegfr3 and ccbe1 mutants at the level of gross embryomorphology (supplementary material Fig. S1A-D), and thatdisplayed a severe reduction in secondary angiogenesis and block inlymphatic precursor sprouting from the cardinal vein (Fig. 1A-D;Fig. 2A-H; supplementary material Fig. S1E-K). At 5 days post-fertilization (dpf), the hu5055, hu6124, hu5142 and hu6410 mutantshave a grossly normal, functional blood vasculature, but thelymphatic vasculature is absent (Fig. 1B,D). Using a positionalcloning approach, we found that these mutants were linked tochromosome 1, containing the vegfc gene (Fig. 1E; data not shown).Sequencing of vegfc in hu5055 mutants revealed a missensemutation changing a cysteine into an arginine residue (C365R) inthe highly conserved C-terminal propeptide (Fig. 1F,G). Additionalmutations identified caused a premature stop in hu6410 (L107X), aphenylalanine to cysteine change in the highly conserved VHD-VEGF homology domain (F138C) and another cysteine-to-serinesubstitution in the C-terminal propeptide of Vegfc (C339S)(Fig. 1F,G). All mutations were predicted to be damaging byPolyPhen-2 (Adzhubei et al., 2010). These vegfc mutationssegregate with the thoracic duct (TD) deficiency phenotype andphenocopy MO-vegfc knockdown or soluble Vegfr3 ligand trap-induced phenotypes (Küchler et al., 2006; Yaniv et al., 2006; Hoganet al., 2009a; Hogan et al., 2009b). Furthermore, these phenotypesare consistent with recently described phenotypes in a Vegfctruncation mutant (Villefranc et al., 2013).

Interestingly, scoring both TD extent and PL number, we notedvariable penetrance of different alleles and haplo-insufficientphenotypes for the more penetrant alleles (Fig. 1I,J). The hu5055allele segregated in a Mendelian, autosomal recessive manner and wasthus used in subsequent assays. Crossing hu5055 mutant carriers tothe previously described expando/vegfr3 mutant (Hogan et al., 2009b),we found an increased frequency of TD defects consistent withgenetic interaction in the same pathway (Fig. 1K; Fig. 2I).

ccbe1, vegfr3 and vegfc zebrafish mutants geneticallyinteractThe ccbe1hu3613, vegfr3hu4602 and vegfchu5055 mutants display acomplete loss of the lymphatic vasculature when homozygous(Fig. 2A-H). Previous, non-quantitative observations (Hogan et al.,2009a; Hogan et al., 2009b) indicated that partial loss of ccbe1 orvegfr3 led to phenotypically wild-type lymphatic development. Wetook advantage of these mutants to determine more rigorously ifcombined heterozygous mutations led to enhanced lymphaticphenotypes. We crossed heterozygous carriers and quantified theextent of the TD across ten body segments in the trunks ofindividual embryos. Subsequent genotyping first confirmed thegenetic interaction in vegfchu5055/+, vegfr3hu4602/+ doubleheterozygous animals (Fig. 2I). We found that ~28% of scoredembryos displayed ≤50% TD extent; within this population, 71% ofthe embryos were double heterozygotes for vegfc and vegfr3, asignificant enrichment from total double heterozygosity of 27%(Fig. 2I). Importantly, similar robust interactions of vegfr3 and vegfcwere observed with ccbe1. The population of embryos displaying≤50% TD extent from a ccbe1hu3613/+, vegfr3hu4602/+ cross wasenriched (54%) for double heterozygotes, as were embryos from accbe1hu3613/+, vegfchu5055/+ cross (61%) (Fig. 2J,K). This geneticinteraction seems to be selective to the ccbe1hu3613, vegfr3hu4602 andvegfchu5055 mutants because in crosses to two other, as yet geneticallyuncharacterized lymphatic mutants, we did not observe anyinteraction (L.L.G. and B.M.H., unpublished observations). Finally,we generated triple heterozygote embryos and found that thepopulation developing ≤50% TD extent contained most of the triple

3

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 1. Zebrafish vegfc mutants lack lymphatic vessels, but appearotherwise normal. (A-D) Confocal projections of Tg(fli1a:EGFP) showunaltered overall morphology and blood vasculature in vegfchu5055 mutants(B) compared with wild type (A). The TD (C, arrows) is absent in vegfchu5055

(D, asterisks). (E) Positional cloning linked alleles hu6124 and hu5055 tolarge regions of chromosome 1, containing the vegfc gene. For hu6124, 13recombinant embryos identified at z9394 were reduced to four recombinantembryos common with z5508, identifying linkage in a large region containingvegfc. For hu5055, eight recombinant embryos at z2691 and 33 differentrecombinant embryos at z4694 flanked a large region of chromosome 1, alsocontaining vegfc. (F) Sequencing of vegfc alleles. hu6410 encodes an earlystop allele leading to a predicted truncated protein lacking the core (VHD)region. hu5142, hu6124 and hu5055 encode missense mutations, all inhighly conserved regions of the protein (G) and predicted to be damaging byPolyPhen-2 (Adzhubei et al., 2010) with scores of 1 out of 1. (G) Alleleshu5055, hu6410, hu6124 and hu5142 all encode mutations in regions thatare conserved across multiple species. (H) Alleles hu5055 and hu6410 fail tocomplement, with trans-heterozygote embryos displaying a lack of lymphaticstructures; phenotype scored as percentage extent of thoracic duct over tensomites (total number of embryos scored n=212). (I,J) The penetrance ofvegfc mutant phenotypes varies, as shown by the variable number of PLs (I)or TD extent (J). The hu6410 allele (L107X) shows the most severe defectsin heterozygous and homozygous mutants, whereas hu5055 does not showhaplo-insufficiency phenotypes. (K) vegfchu5055 genetically interacts withvegfr3hu4602 in trans-heterozygous embryos. Confocal projections ofTg(fli1a:EGFP; kdr-l:mCherry) showing examples of the heterogeneityobserved during TD development at 5 dpf in vegfc+/−; vegfr3+/− embryos.Arrows indicate the thoracic duct, asterisks indicate absence of thoracic duct.

Dev

elop

men

t

Page 4: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

4

heterozygote embryos, demonstrating a strong interaction (Fig. 2L).This synergistic genetic interaction shows that lymphaticdevelopment is sensitive to the level of activation of the Vegfc/Vegfr3 pathway, and that altering ccbe1 dosage modifies phenotypesin this pathway. Combined with the fact that ccbe1, vegfc and vegfr3act at the same stage during the cellular progression oflymphangiogenesis and that these mutants present the most selectiveloss of lymphatic vascular structures that we have observed, weconsidered this a strong indication that Ccbe1 could be a novelcomponent of the Vegfc/Vegfr3 signaling pathway.

ccbe1 mutation suppresses phenotypes driven by ectopicVegfc/Vegfr3 signaling in embryonic arteriesPrevious studies have shown that the knockdown of dll4 leads to anexcessive intersegmental artery (aISV) angiogenesis phenotype by 72hpf (Leslie et al., 2007; Siekmann and Lawson, 2007; Hogan et al.,2009b). This phenotype occurs because in wild-type arteries, theVegfc/Vegfr3 pathway is inhibited by Dll4. Depletion of Dll4 leads toincreased activation of Vegfc/Vegfr3 signaling in aISVs, resulting inaISV hyperbranching (Leslie et al., 2007; Siekmann and Lawson,

2007; Hogan et al., 2009b). We took advantage of the hyperbranchingphenotype to investigate ccbe1 function. In clutches of embryosderived from incrosses of ccbe1 heterozygous carriers, we activatedthe Vegfc/Vegfr3 pathway in arteries by knocking down dll4.Embryos were first sorted at 72 hpf according to the severity ofarterial hyperbranching, and then subsequently genotyped. Thepopulation containing animals with vastly reduced or nohyperbranching (Fig. 3Aiii, graph) was significantly enriched inccbe1hu3613 mutants (81%; P<0.0001) whereas the category containingsevere aISV hyperbranching (Fig. 3Aii) was enriched in wild-type andheterozygote embryos (83%; P=0.0019; Fig. 3A, graph). Hence,ccbe1 mutation suppressed the dll4 loss-of-function phenotype.

To validate this observation further, we used a second phenotypicassay that has been previously described (Hogan et al., 2009b). Whenvegfc is overexpressed by mRNA injection and dll4 is simultaneouslydepleted, we observe ectopic turning of aISVs in the trunk from asearly as 30 hpf (compare Fig. 3Bi with 3Bii) that is not seen in singletreatment controls (supplementary material Fig. S2). aISVs deficientfor dll4 are more responsive to vegfc RNA injection (Hogan et al.,2009b) and phenotypes appear the same as those observed when vegfc

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 2. ccbe1, vegfc and vegfr3 genetically interact in double and triple heterozygous animals. (A-H) Confocal projections of Tg(fli1a:EGFP; kdr-l:mCherry) show grossly unaltered overall morphology and blood vasculature in ccbe1hu3613 (B), vegfr3hu4602 (E) and vegfchu5055 (F) mutants compared with wildtype (A). The TD (C, arrows) is absent in ccbe1hu3613 (D), vegfr3 hu4602 (G) and vegfc hu5055 (H) mutants (asterisks). (I-K) ccbe1, vegfc and vegfr3 geneticallyinteract in double heterozygote embryos, which display lymphatic defects. Offspring from vegfc+/− and vegfr3+/− carriers give rise to 28% of embryos (n=28/99)with a TD length of ≤50%. This population is significantly enriched (71%; n=20/28; P<0.0001) in double heterozygotes (I). Similarly in ccbe1+/− and vegfr3+/−

crosses, 24% (n=24/100) embryos develop ≤50% of their TD, and again this population is significantly enriched (54%; n=13/24; P=0.0098) in doubleheterozygotes (J). In ccbe1+/− and vegfc+/− crosses, 14% (n=18/126) of embryos develop ≤50% of their TD, this population being significantly enriched (61%;n=11/18; P=0.0096) in double heterozygotes (K). (L) Crossing double heterozygous ccbe1+/−; vegfr3+/− animals to vegfc+/− animals, results in 21% (n=25/121) ofembryos with ≤50% of TD: within this population, 40% (n=10/25; P=0.0004) of the embryos were triple heterozygotes, a significant enrichment from thestatistical triple heterozygosity rate of 15%.

Dev

elop

men

t

Page 5: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

is expressed from the hsp70l promoter (Nicoli et al., 2012). In clutchesof embryos derived from ccbe1 heterozygous incrosses, weectopically activated the Vegfc/Vegfr3 pathway by injecting vegfcmRNA and a dll4 morpholino. Animals were sorted at 30 hpfaccording to the severity of aISV phenotype, and then genotyped. Ofthe embryos displaying a weak phenotype, or no phenotype at all(Fig. 3Biii), 70% were ccbe1 mutants (Fig. 3B, graph), a highlysignificant (P<0.0001) enrichment. The population displaying severephenotypes was significantly enriched in wild-type and heterozygoteanimals (83%; P=0.0006; Fig. 3B, graph). Hence, ccbe1 mutationsuppressed the dll4 loss-of-function, vegfc overexpression phenotype.

Ccbe1 knockdown suppresses phenotypes driven by ectopicVegfc/Vegfr3 signaling in embryonic veinsWe next generated a new zebrafish transgenic line(hsp70l:GAL4;UAS:vegfc), which ubiquitously expresses full-lengthvegfc under the control of the heat shock (hsp70l) promoter(supplementary material Fig. S3A,B) (Scheer et al., 2001). In theseembryos (following two consecutive heat shocks at 28 and 56 hpf),veins sprout ectopically, resulting in hyperbranched intersegmentalvessels (vISVs) as visualized in fli1a:GFP, flt1:tomato doubletransgenic animals (Fig. 3Ciii). ccbe1 morpholino injection rescuedthis phenotype, blocking excessive venous sprouting (Fig. 3Civ,graph; P<0.0001).

Taken together, we found in two blind, genotype-basedexperiments, that Vegfc- and Vegfr3-driven arterial phenotypes are

suppressed in ccbe1hu3613 mutants, and additionally that a Vegfc- andVegfr3-driven venous phenotype is rescued in ccbe1 morphantembryos. These findings suggest that Ccbe1 is necessary for thefunction of the Vegfc/Vegfr3 signaling pathway during embryonicdevelopment.

Vegfr3-dependent Erk activation in embryonic veinsrequires ccbe1It is well established that VEGFR3 signals via intracellular kinasesthat include ERK (reviewed by Bahram and Claesson-Welsh, 2010).We decided to further investigate downstream signaling pathwaysto determine if Ccbe1-deficient embryos display a signaling blockin venous endothelium. We used whole-mount immunofluorescenceto examine phospho-Erk in the context of the embryonicvasculature. The signal observed was specific as it was reduced inembryos treated with the MEK inhibitor PD98059 (which inhibitsMek activation and hence Erk phosphorylation) and was ectopicallyinduced in the ventral posterior cardinal vein (PCV) in the contextof vegfc overexpression (Fig. 4A). In wild-type embryos, we foundthat Erk activity was broadly detected in the neural tube, muscle andepithelia, but also in the endothelium of the PCV. In PCVendothelium, Erk activation was segmented and dorsally enrichedin individual cells at 32 hpf, concomitant with the induction ofsecondary sprouting. Importantly, this patterned activation of Erkwas markedly reduced in both MO-vegfr3- and MO-ccbe1-injectedembryos compared with wild-type controls (Fig. 4B,C). These

5

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 3. Phenotypes driven by ectopic Vegfc/Vegfr3 signaling are suppressed in ccbe1-deficient embryos. (A) At 72 hpf, dll4 morphants display anarterial hyperbranching phenotype (arrow) driven by increased Vegfc/Vegfr3 signaling in the transgenic Tg(fli1a:EGFP) line. This phenotype was suppressed inccbe1hu3613 mutants. Eighty-one per cent of MO-dll4 injected embryos displaying wild-type or mild phenotypes were ccbe1 mutants (n=17/21), whereas thepopulation displaying the most severe phenotype was mainly composed of wild-type or heterozygous siblings (83%; n=139/168). (B) In dll4 morphants, arteriesare sensitized to increased vegfc expression during primary sprouting. Arteries in MO-dll4, vegfc mRNA-injected embryos display aberrant, ectopic turning(arrow) as early as 30 hpf. Embryos from ccbe1 carrier incrosses, injected with 100 ng vegfc mRNA and 5 ng MO-dll4, were sorted into the phenotypiccategories ‘wild type’ and ‘severe’. Genotyping revealed that 70% of the embryos displaying wild-type morphology were ccbe1 mutants (n=19/27). By contrast,the population affected by the most severe phenotype was composed of 83% wild-type or heterozygous siblings (n=122/147). (C) Confocal projections ofTg(fli1a:EGFP; flt1:tomato; hsp70l:Gal;4XUAS:vegfc) embryos show that endothelial cells in heat-shocked embryos display aberrant ectopic branching at 72hpf. The ectopic endothelial cells are venous derived (flt1:tomato negative, arrow in Ciii). Heat-shocked embryos that were injected with 2.5 ng of MO-ccbe1 donot show this phenotype (asterisks). Scoring of the number of aberrant vISVs per heat-shocked embryo showed a significant rescue (0.12 in MO-ccbe1injected n=22, versus 4.76 in uninjected controls n=25; P<0.0001) of the phenotype.

Dev

elop

men

t

Page 6: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

6

findings confirm the presence of Vegfc- and Vegfr3-dependent Erksignaling in embryonic veins and demonstrate that Ccbe1 isnecessary for activation of this pathway.

We also examined the activation status of Vegfr3 by western blotof embryonic lysates. The phosphorylation of Tyr1063/1068 inhuman VEGFR3 reflects the activation status of VEGFR3,promoting downstream signaling essential for lymphangiogenesis(Dixelius et al., 2003; Salameh et al., 2005). Antibodies against

zebrafish Vegfr3 are unavailable, but we found two commercialantibodies directed against conserved regions of human VEGFR3that cross-reacted with zebrafish Vegfr3. One of these was directedagainst the phosphorylated residues Tyr1063/1068 (human) orTyr1071/1076 in zebrafish (supplementary material Fig. S4). Usingthis phospho-VEGFR3 antibody for western blotting we detectedVegfr3 in zebrafish lysates immunoprecipitated with the sameantibody at 32 hpf, during initiation of secondary sprouting. The

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 4. Vegfr3-dependent Erk signaling requires ccbe1 during the induction of secondary sprouting in zebrafish. (A) Analysis of phospho-Erk (P-Erk)expression in 32 hpf embryos. P-Erk (green) and fli1a:EGFP (white) images (lateral view) show P-Erk detected broadly in whole-mount and cross-sectioned(right-hand panels, merge upper, P-Erk lower) control embryos. Signal was increased in Vegfc-induced (dll4 MO + vegfc mRNA-injected) embryos in theposterior cardinal vein (n=8/8; Vegfc-induced embryos all showed ectopic expression in the ventral wall of the PCV). Cross section merged channel imagesshown in a and b, P-Erk only in c and d. Treatment with the Erk inhibitor PD98059 led to a reduction in all P-Erk staining. Arrows indicate P-Erk expression inthe dorsal PCV. DA (dorsal aorta) and PCV (posterior cardinal vein) are indicated. (B) Comparison of P-Erk staining in control uninjected (left), MO-vegfr3 andMO-ccbe1 embryos. Upper panels are merged images and lower P-Erk only, viewed laterally (left) and cross-sectioned (right). Cross sections (right) are fromseparate embryos. Arrows indicate P-Erk expression in the dorsal PCV. DA and PCV are indicated. (C) Quantification of P-Erk-positive cells in the cardinal veinlocated in the dorsal compared with ventral wall (left-hand graph). Scores through individual sections of z-stack images from 12 control embryos, scoredlaterally across three somites in the trunk. Quantification of P-Erk-positive cells in the cardinal vein in control and MO-injected conditions (right-hand graph)(scores from n=10 control embryos, n=13 MO-vegfr3-injected and n=15 MO-ccbe1-injected embryos). (D) Immunoprecipitation (IP) and western blot (IB)detection of phosphorylated Vegfr3 at 32 hpf in wild type and in ccbe1, vegfr3, vegfc morphant and vegfc mRNA-injected embryos. The level of phosphorylatedVegfr3 is markedly reduced in ccbe1, vegfr3 and vegfc morphants, but is increased in vegfc-mRNA injected (500 ng) embryos compared with wild type (D,upper blot, IP for phospho-Vegfr3 and IB detection with phospho-Vegfr3). Loading controls were: the IgG light chain [IgG(l)] present in all blots after IP (D,middle blot), and Myosin to monitor protein input in IPs (D, lower blot). Quantification of Vegfr3 phosphorylation (relative to the loading control) based on threeindependent experiments is shown in right-hand panel. The decrease in MO-ccbe1 compared with uninjected controls is statistically significant (P<0.05).(E) qPCR analysis of the expression of ccbe1, vegfr3, vegfc, kdr and kdrl in uninjected control and MO-ccbe1-, MO-vegfc-, and MO-vegfr3-injected embryos.Error bars represent s.d. (C) or s.e.m. (D,E).

Dev

elop

men

t

Page 7: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

signal was increased in vegfc mRNA-injected embryos, but wasreduced in MO-vegfr3- and MO-vegfc-injected embryos. A similarreduction in phospho-Vegfr3 was observed in MO-ccbe1-injectedembryos quantified and normalized over three independentexperiments (Fig. 4D). We were not able to precipitate Vegfr3 usinganother cross-reacting antibody directed against VEGFR3intracellular domains. However, we were able to validate thespecificity of the bands and result observed by blotting lysatesimmunoprecipitated by the phospho-Vegfr3 antibody with theVEGFR3 antibody directed against Vegfr3 intracellular domains. Wedetected the bands of the same molecular weight and intensities(supplementary material Fig. S5). Importantly, Ccbe1, Vegfc andVegfr3 do not cross-regulate each other transcriptionally (Fig. 4E),indicating that Vegfr3 expression is unchanged in this assay. This isconsistent with immunofluorescence-based observations in theCcbe1 knockout mouse (Hägerling et al., 2013). We also detectednormal expression of all other signaling Vegfr family members inthe morphants examined (Fig. 4E).

Ccbe1 enhances Vegfc-driven sproutingTo characterize further the function of Ccbe1 in Vegfc/Vegfr3signaling, we generated two transgenic lines that express ccbe1 orvegfc from the shh promoter in the floor plate (FP) from as early as24 hpf (supplementary material Fig. S3C,D). At 32 hpf, vegfc- orccbe1-overexpressing animals display no phenotype. However,when these two lines were crossed, we found that double transgenicanimals display aberrant ectopic turning of ISVs at 32 hpf (Fig. 5A).

At 48 hpf, ccbe1-overexpressing animals still show no phenotype,whereas vegfc-overexpressing animals display vastly increasedendothelial cell accumulation at the horizontal myoseptum, as wellas dramatic ectopic sprouting of the ISVs (Fig. 5A), as also observedin the Tg(hsp70l:Gal4;UAS:vegfc) line (Fig. 3Cii). Interestingly, at48 hpf in double ccbe1/vegfc-overexpressing animals, we observe adramatic accumulation of endothelial cells in dorsal aspects of theembryo (Fig. 5A), combined with ectopic ISV sprouting. Theaccumulation of endothelial cells (arrow) occurs at the approximatelevel of the FP in double Tg(shh:ccbe1;shh:vegfc), but not in singleTg(shh:vegfc) or Tg(shh:ccbe1) animals. This could be taken toindicate that Ccbe1 expression is capable of locally enhancingVegfc-driven sprouting in the embryo.

We also examined the activity of the Tg(shh:vegfc) transgenic linein ccbe1hu3613 mutants. ccbe1hu3613 mutant embryos showed asuppression of the Vegfc-driven hyperbranching of ISVs, which wasconsistent with our findings that ccbe1 is needed for Vegfc-drivenhyperbranching in the hsp70l:GAL4;UAS:vegfc line and dll4depletion models. When these animals were scored at 3.5 dpf forendothelial cells at the horizontal myoseptum or 5 dpf for thepresence of the TD, we found that overexpression of full-lengthVegfc could partially rescue ccbe1hu3613 mutant lymphaticphenotypes (supplementary material Fig. S6A,B). However, the shh-driven vegfc expression did not rescue the MO-ccbe1 lymphaticphenotype and, furthermore, we did not see any rescue of lymphaticdevelopment in MO-ccbe1-injected, hsp70l:GAL4;UAS:vegfcembryos (supplementary material Fig. S6C-E). We take this to

7

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 5. Ccbe1 enhances Vegfc-drivensprouting and regulates levels ofbioactive VEGFC in vitro. (A) Confocalprojections at 32 hpf of Tg(shh:ccbe1),Tg(shh:vegfc) and Tg(shh:ccbe1;shh:vegfc)in a Tg(fli1a:EGFP) background. Co-overexpression of ccbe1 and vegfc in thefloorplate leads to aberrant ectopic turning ofthe ISVs at 32 hpf (upper panels; n=32/36;P<0.0001). At 48 hpf, ccbe1 overexpressionin the floorplate does not result in anyphenotype, whereas vegfc-overexpressinganimals display hyperbranching of the ISVs,and enhanced endothelial cell accumulationat the horizontal myoseptum (arrowhead).ccbe1 and vegfc co-overexpression in thefloorplate also leads to hyperbranching ISVs,and to a marked accumulation of endothelialcells at dorsal aspects of the embryo (arrow).(B) Western blot of 293EBNA-1 cells (stablyexpressing VEGFC) indicate that CCBE1 isdetected in the lysate of cells transfected withCCBE1 plasmid, but not in controls. (C) Anincrease in the levels of all forms of VEGFCis detected in the medium of CCBE1-transfected cells, compared with control cells.The mature form of VEGFC (detected at ~23kDa) is predominant. (C′) Relative intensity(split axis) of the different processed forms ofVEGFC presented in C based on multipleexposures. Note the saturation of the matureform in C. (D) qPCR showing that CCBE1transfection does not affect VEGFC mRNAlevels in vitro in 293EBNA-1 cells stablyexpressing VEGF-C (D, left panel).Consistent with this, in zebrafish embryos theinjection of vegfc or ccbe1 mRNA does notaffect the endogenous levels of the other (D,right panel). Error bars represent s.e.m. D

evel

opm

ent

Page 8: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

8

indicate that partial rescue exclusively in the ccbe1hu3613 mutantoccurs as a result of some retained capability of the ccbe1hu3613

D162E (hypomorphic) mutant protein.

CCBE1 overexpression leads to enhanced levels ofprocessed, soluble VEGFC in vitroVEGFC is produced as a secreted precursor protein requiringproteolytic cleavage of N- and C-terminal propeptides to produce thehighly bioactive mature form, consisting of dimers of the VEGFhomology domain (VHD), which potently activates VEGFR3signaling (Joukov et al., 1997). Although proteases that can promoteprocessing of VEGFC have been identified (McColl et al., 2003;Siegfried et al., 2003), other factors that might regulate the function ofVEGFC in the developing embryo remain to be described. Todetermine if the capability of CCBE1 to modulate VEGFC activity invivo may be at the level of VEGFC production, release from the cellsurface/ECM or proteolytic processing, we turned to in vitro cell-basedmodels. 293EBNA-1 cells stably harboring an expression constructencoding full-length human VEGFC allow analysis of secretedVEGFC polypeptides in conditioned cell culture medium. These cellswere transfected with an expression vector encoding human CCBE1(Fig. 5B). In control (empty vector) or mock transfected cells lackingdetectable CCBE1, mature VEGFC was readily detected inconditioned cell culture media, with partially processed forms detectedat lower levels (Fig. 5C,C′). CCBE1 transfection resulted in asignificant enrichment of all forms of VEGFC in the conditioned cellculture media but in particular the relative abundance of the matureform was increased compared with other forms of VEGFC(Fig. 5C,C′). This indicates that although release of VEGFC from thecell surface or ECM is enhanced, processing to the mature form isparticularly increased in the presence of CCBE1.

We confirmed the enhanced processing of VEGFC in anindependent cell line, HEK293T cells, using transient transfections ofCCBE1 and VEGFC plasmids; note the increased abundance ofmature VEGFC in the cell culture media in response to CCBE1 insupplementary material Fig. S7B,C. However, the release of VEGFCwas not generally enhanced in this system as the abundance of otherforms of VEGFC did not increase. In both cell systems, we alsoexamined the levels of VEGFC present in the cell lysates and foundthat in 293EBNA-1 cells we could detect partially processed VEGFC(i.e. the form containing the N-terminal propeptide and VEGFhomology domain) (supplementary material Fig. S7A) whereas inHEK293T cells we detected both full-length and partially processedforms (supplementary material Fig. S7C). The relative levels ofdifferent forms of VEGFC in cell lysates were not markedly alteredin the presence of CCBE1 (supplementary material Fig. S7A,C). Wetake this to indicate that the cell-associated VEGFC may be far moreabundant than VEGFC in the medium, hence a small decrease in theproportion of cell-associated VEGFC, due to release from the cellsurface or ECM, could lead to a significant increase in the relativeabundance of VEGFC detected in the medium on western blots.Importantly, CCBE1 overexpression had no effect on VEGFC mRNAlevels in vitro or in vivo in zebrafish embryos (Fig. 5D).

In the in vitro assays above, VEGFC and CCBE1 were producedand secreted from the same cells. To test if CCBE1 needs to beexpressed in the same cell as VEGFC to promote VEGFC processing,we transfected two different populations of HEK293T cells withCCBE1 and/or VEGFC and mixed them before detection of VEGFCin the culture medium. We found that CCBE1 was indeed capable ofenhancing VEGFC processing in trans (supplementary material Fig.S7B), indicating that CCBE1 can exert its effects on VEGFC outsidethe cell. These data, taken together, indicate that in these cell-based

systems CCBE1 regulates VEGFC at a post-transcriptional level, thatthis regulation occurs extracellularly and that it is capable ofincreasing the release and processing of VEGFC. The degree to whicheach of these two mechanisms apply may be dependent on the celltype, given that the effect on VEGFC release was not observed in theHEK293T model system.

Constitutively secreted, processed VEGFC rescues ccbe1loss-of-function phenotypesTo build on these in vitro findings, and given the observation that full-length Vegfc overexpression cannot rescue the MO-ccbe1 loss-of-function phenotype (supplementary material Fig. S5), we decided toinvestigate whether a constitutively secreted, mature form of VEGFCwas sufficient to recue ccbe1 loss of function. To do this, wegenerated a truncated form of human VEGFC (∆N∆CVEGFC),missing the N- and C-terminal domains but retaining the signalpeptide for secretion, and placed it under the control of the hsp70lpromoter. We used the human form because the proteolytic processingsites of zebrafish Vegfc are not experimentally validated whereas thehuman form has been examined in detail and processing sites are known (Joukov et al., 1997). We injected DNA forhsp70l:∆N∆CVEGFC.t2a.mCherry and generated mosaic embryos.After a 1-hour heatshock at 26 hpf, embryos injected withhsp70l:∆N∆CVEGFC.t2a.mCherry displayed ISV hyperbranching (at54 hpf, Fig. 6A) as observed previously upon vegfc overexpression(Fig. 3C; Fig. 5A) and expressed mosaic mCherry. Co-injection of thehsp70l:∆N∆CVEGFC.t2a.mCherry with MO-ccbe1 generated arobust, quantifiable rescue of PL formation in these embryoscompared with MO-ccbe1 controls (Fig. 6A,B). Co-injectinghsp70l:∆N∆CVEGFC.t2a.mCherry with MO-vegfr3 did not rescue PLformation at the horizontal myoseptum by 54 hpf (Fig. 6A,B). Finally,we found that ISV hyperbranching was present in ccbe1 and vegfr3morphants injected with hsp70l:∆N∆CVEGFC.t2a.mCherry(Fig. 6C), which is consistent with the ability of processed VEGFC toactivate VEGFR2 (KDR – Human Gene Nomenclature Database)signaling. The ability of a processed form of VEGFC to rescue theMO-ccbe1 loss-of-function phenotype demonstrates that a deficit inVegfc maturation is responsible for venous angiogenesis andlymphangiogenesis defects in the absence of Ccbe1.

DISCUSSIONOur results show that ccbe1 genetically interacts with vegfc andvegfr3 and that ccbe1 mutation or depletion suppresses theformation of excess and abnormal aISV and vISV sprouts driven byectopic Vegfc/Vegfr3 signaling. These data highlight the necessityfor functional Ccbe1 for the propagation of (ectopic) Vegfc-drivensignals in the developing embryo. Furthermore, we show that inccbe1 and vegfr3 morphants, venous Erk signaling and Vegfr3activation at Tyr1071/1076 are reduced. Our finding of a block inVegfc/Vegfr3 signaling is consistent with phenotypes observed inhumans (Alders et al., 2009) and mice (Bos et al., 2011). Previousstudies have used proximity ligation assays as a readout for Vegfr3signaling (Bos et al., 2011), and did not observe the reduction ofVegfr3 signaling in the absence of Ccbe1 that we see here. However,the previous study was unable to examine the entirety of signalingin cells at equivalent developmental stages during the induction ofsprouting of lymphatics from the vein. We overcame previouslimitations by utilizing the immunofluorescence visualization ofactivated (phosphorylated) Erk in the PCV. We found that during theinduction of secondary sprouting Erk activation is dorsally enrichedand segmentally patterned in the PCV in a Vegfr3-dependentmanner. This assay hence provides improved sensitivity to detect

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Dev

elop

men

t

Page 9: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

Vegfc-Vegfr3 induced signaling during secondary angiogenesis invivo compared with previous approaches. Using this approach andalso cross-reacting antibodies to phospho-Vegfr3, we found a crucialrequirement for Ccbe1 for in vivo Vegfc-Vegfr3-Erk signaling.

Given this function of Ccbe1, its non-autonomous role in zebrafish(Hogan et al., 2009a) and its extracellular localization (Bos et al.,2011), we asked if Ccbe1 can directly modulate the levels of bioactiveVegfc. We did this in cell-based models, and found that upon CCBE1overexpression, the amount of mature bioactive VEGFC is increasedin the culture medium. CCBE1 can induce higher levels of VEGFCin trans and hence performs this function outside of the cell in this invitro context. Fully processed VEGFC was increased more than thepartially processed and full-length forms in the medium, indicatingthat CCBE1 regulates processing. However, CCBE1 can increaselevels of the partially processed and full-length forms to some degree,also suggesting enhanced release from the cell surface or matrix.Confirming that this capability has relevance in vivo, we observed arescue of the ccbe1 loss-of-function phenotype when we reintroduceda secreted and fully processed form of VEGFC in zebrafish. Thisrescue, combined with the observation that full-length Vegfc failed torescue the phenotype, demonstrates that Ccbe1 regulates Vegfcmaturation and bioavailability. Ccbe1 does not appear to be a proteaseand the precise molecular mechanisms involved, including the rolesof additional proteins in the CCBE1/VEGFC/VEGFR3 pathway,require further analysis.

One pertinent question is: why would the developing embryoneed an additional factor to regulate Vegfc-driven activation ofVegfr3? It is crucial to note that vegfc in zebrafish is transcribed inthe hypochord and dorsal aorta in the midline of the embryonictrunk. Despite this, secondary sprouts from the vein migrate past thistranscriptional source of ligand on the dorsoventral and mediolateralaxes of the embryo to the horizontal myoseptum and give rise toPLs. This suggests that spatial presentation of active Vegfc proteinduring secondary sprouting must be regulated independently of thetranscriptional source of vegfc. ccbe1 transcript expression precedes

and predicts the migration route of lymphatic precursors in the trunk(Hogan et al., 2009a) suggesting that Ccbe1 serves to impart thenecessary post-translational activation of Vegfc (see proposed modelin Fig. 7) in a spatially and temporally regulated manner. Hence, amigrating lymphatic endothelial cell could be directed through theembryonic environment by a route pre-determined by the earlier,local concentration of Ccbe1. To test this idea directly is inherentlydifficult, but the hypothesis is supported by overexpression regimesof ccbe1 and vegfc in an ectopic location, the FP of the neural tube:here we found that Vegfc-driven sprouting of endothelial cells wasenhanced in the presence of ectopic Ccbe1.

9

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Fig. 6. Ectopic expression of matureVEGFC rescues secondary sprouting inccbe1 morphants. (A) Confocalprojections of Tg(fli1a:EGFP) at 54 hpf.Knock down of ccbe1 or vegfr3 leads to aloss of PLs at the horizontal myoseptum(arrowheads and asterisks). Ectopicexpression of the mature form of VEGFCstrongly rescues PL formation in ccbe1morphants but not in vegfr3 morphants.Arrows indicate hyperbranched ISVs.(B) Quantification of PL formation at 54hpf. In wild type, 98% (n=54/55) ofembryos develop PLs, whereas in MO-ccbe1-injected embryos <4% (n=2/52) do.PL development is rescued to 74%(n=29/39) in ccbe1 morphants transientlyoverexpressing ΔNΔCVEGFC (P<0.0001).This rescue was never observed in vegfr3morphants with all embryos devoid of PLs(n=23/23). (C) Quantification of ISVhypersprouting at 54 hpf. ISVhypersprouting was observed in wild-typeembryos (93%; n=40/43), with mildreductions in ccbe1 morphants (79%;n=31/39) and vegfr3 morphants (65%;n=15/23) after ΔNΔCVEGFCoverexpression.

Fig. 7. Ccbe1 activates Vegfc to induce Vegfr3 signaling. Proposedmodel for coordination of angiogenesis by Ccbe1, Vegfc and Vegfr3 in thedeveloping embryo. Vegfc is produced in a largely inactive full-length formthat is processed and released from the cell surface/ECM in a Ccbe1-dependent manner to generate the mature, highly active form. Downstream,arteries respond in a manner dampened by Dll4-dependent suppression ofVegfr3 signaling (Hogan et al., 2009b), whereas Vegfr3 signaling in veinsinduces secondary angiogenesis, which produces both intersegmental veinsand lymphatic vascular precursor cells. D

evel

opm

ent

Page 10: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

10

CCBE1 mutations have been shown to be responsible forgeneralized lymph vessel dysplasia in humans (Alders et al., 2009;Connell et al., 2010). Our finding that Ccbe1 is a crucial modulatorof the Vegfc/Vegfr3 pathway during embryo development, suggeststhat a deficiency of VEGFR3 signaling may be responsible for thelymphatic aspects of Hennekam Syndrome. Other aspects of thissyndrome that are distinct from Milroy’s disease might be due toother, yet to be identified, functions of CCBE1.

MATERIALS AND METHODSZebrafish strains and transgenesisAnimal work followed guidelines of the animal ethics committees at theUniversity of Queensland, and the Royal Netherlands Academy of Arts andSciences (DEC). Zebrafish transgenic lines Tg(fli1a:EGFP), Tg(−6.5kdr-l:mCherry), Tg(−0.8flt1:tdTomato) and Tg(hsp70l:Gal4)1.5kca4 lines weredescribed previously (Scheer et al., 2001; Lawson and Weinstein, 2002;Hogan et al., 2009a; Bussmann et al., 2010). N-ethyl-N-nitrosourea (ENU)mutagenesis was performed as previously described (Wienholds et al., 2002).

The Tg(4xUAS:vegfc) line was generated using a 4xUAS promoterupstream of the full length zebrafish vegfc cDNA (supplementary materialFig. S3A) cloned using the Gateway system (Hartley et al., 2000; Walhoutet al., 2000). The Tg(flt4:YFP) reporter line was generated from BACDKEY-58G10 as previously described (Bussmann and Schulte-Merker,2011) and is characterized in detail elsewhere (van Impel et al., 2014). TheTg(s1173:Gal4) line was kindly provided by Ethan Scott (School ofBiomedical Sciences, University of Queensland). sonic hedgehog (shh)promoter and floorplate enhancer (Ertzer et al., 2007) was used to driveCcbe1 IRES tagRFP or Vegfc IRES mturquoise (supplementary materialFig. S3C), cloned into the MiniTol2 vector (Balciunas et al., 2006); plasmids(25 ng/μl) were injected with tol2 transposase mRNA (25 ng/μl) into 1- to2-cell-stage embryos. All genotyping details and primers are given insupplementary material Tables S1 and S3. KASPar (KBioscience) was usedfor the indicated vegfc alleles.

DNA, RNA and morpholino injectionsConstructs for transient DNA injection of theTg(hsp70l:∆∆VEGFC.t2a.mCherry) transgene were generated by PCRamplifying the N- and C-terminally truncated form of human VEGFC,containing the endogenous secretion peptide (Joukov et al., 1997) followed byGateway recombineering. Primer sequences are given in supplementarymaterial Table S2. DNA was injected at a concentration of 90 ng/μl at single-cell stage. Morpholinos used are shown in supplementary material Table S4.The vegfc cDNA used was described previously (Hogan et al., 2009a).

Analysis of ERK and Vegfr3 signaling in zebrafish embryosPhospho-ERK was detected by immunofluorescence, using the previouslydescribed protocol (Inoue and Wittbrodt, 2011) but modified as follows:embryos were blocked and incubated with p-ERK primary antibody (1/250;Cell Signaling, #4370) in TBS containing 0.1% Triton X-100, 1% bovineserum albumin, 10% goat serum and then with horseradish peroxidase-conjugated anti-goat secondary antibody (1/1000; Cell Signaling, #7074) in2% blocking solution (Roche) dissolved in 100 mM maleic acid with 150mM NaCl (pH 7.4). Phospho-ERK was subsequently detected using theTSA reagent (Perkin Elmer) as per manufacturer’s guidelines. As a controlfor antibody specificity, embryos were incubated in 100 μM PD98059 (CellSignaling) from 24 hpf. Embryos were vibratome sectioned (100 μm) (LeicaVT100S) and imaged as described below.

Zebrafish phospho-Vegfr3 was detected following immunoprecipitation(IP) with anti phospho-VEGFR3 (Cell Applications, CB5793) using thesame antibody. One hundred and fifty embryos were lysed overnight at 4°Cin modified RIPA buffer containing 1 mM EDTA, 4% protease inhibitor(Sigma), 1% phenylmethylsulfonyl fluoride (PMSF; Sigma) and 1% Haltphosphatase inhibitor cocktail (Thermo Fisher Scientific). IP was carried outovernight at 4°C with the phospho-Vegfr3 antibody (1/1000), followed by2 hours at 4°C with protein A agarose beads (1/10) (Thermo FisherScientific). Standard western blotting approaches were used to detect Vegfr3

with both anti-phospho-VEGFR3 (1/1000; Cell Applications, CB5793) andanti (non-phosphorylated) VEGFR3 (1/1000; Cell Applications, CB5792).Mouse anti-myosin light chain 1 and 3f (1/100; Developmental StudiesHybridoma Bank, F310) was used to detect a loading control band.Quantification relative to IgG light chain (phospho-Vegfr3 only) or Myosinused ImageJ across three biological replicates. Only phospho-Vegfr3 wasassessed and not total Vegfr3, owing to a limitation of the cross-reactingantibodies in only detecting bands post-IP for phospho-Vegfr3.

VEGFC processing/secretion in vitroFor analysis of VEGFC processing and secretion two approaches were used.Briefly, using 293EBNA-1 cells stably expressing a full-length form ofhuman VEGFC (Karnezis et al., 2012) were transiently transfected[Lipofectamine 2000 (Invitrogen)] with a CCBE1 expression vector after amedium change to a serum-free chemically defined medium (Pro293a-CDM, Lonza). Both supernatant and cell lysates were subsequently collectedfor western blotting. 293EBNA-1 cells were maintained in Dulbecco’sModified Eagle Medium, supplemented with 10% fetal bovine serum,50 mM L-glutamine, 50 μg/ml penicillin, 50 μg/ml streptomycin and100 μg/ml hygromycin B (Roche Diagnostics). Cells were lysed in 0.1%SDS, 50 mM Tris, 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate,pH 8.0, 1 mM PMSF, 10 μg/ml aprotinin and 10 μg/ml leupeptin. Lysateswere incubated at 4°C for 20 minutes with gentle agitation and centrifugedat 13,000 g for 10 minutes. Western blotting was performed with antibodiestargeting VEGFC (R&D Systems, BAF752) or CCBE1 (Abcam, ab101967).

For cell mixing experiments, separate populations of HEK293T cells weretransfected with constructs expressing CCBE1, VEGFC or both. Cells werepassaged and mixed as indicated after 24 hours. From mixed cultures,conditioned supernatants were collected and cells were lysed in RIPA/SDSbuffer after another 48 hours. Supernatant and cell lysates were isolated andwestern blotting analysis performed using VEGFC (VEGFC isoform 103antibody, antibodies-online) and CCBE1 (HPA041374, Sigma) antibodies.

qPCR detection of Vegfc and Ccbe1 expressionFor expression analysis in the cultured cells, RNA was isolated usingRNeasy Mini Kit (Qiagen), and cDNA synthesized using High CapacitycDNA Reverse Transcription Kit (Applied Biosystems). For expressionanalysis, Taqman Gene Expression Assays were employed [VEGFC(Hs01099203_m1), GAPDH (Mm99999915_g1)] using Taqman FastUniversal PCR Master Mix (2×) (Applied Biosystems) as permanufacturer’s instructions and analyzed on a 7300 RT-PCR machine(Applied Biosystems). For zebrafish qPCR, RNA was extracted at 24 hpffrom 20 zebrafish embryos. cDNA was synthesized using the Superscript IIIKit (Invitrogen) and reactions used SYBR Green kit (Applied Biosystems)analyzed on a 7500 RT-PCR machine (Applied Biosystems). Data werenormalized using the geometric average of ef1α (eef1a1l1), rpl13 and rps29,which were found to be the most stable reference genes using GeNorm withdata displayed as arbitrary units (A.U.).

ImagingConfocal imaging was performed on live embryos mounted laterally usinga Zeiss 510 or a Leica SPE confocal microscope at the indicated stages.Sections were imaged using a Zeiss 710 FCS confocal microscope.

Statistical analysis and mutation effect predictionFor genetic interaction data, χ2 tests were performed using GraphPad(http://graphpad.com/quickcalcs/chisquared1.cfm). PolyPhen-2 was used toevaluate mutation pathogenicity (http://genetics.bwh.harvard.edu/pph2/).

AcknowledgementsWe thank C. Neyt, G. van de Hoek, N. Chrispijn and M. Witte for technicalassistance; Dagmar Wilhelm for advice; Nathan Lawson for providing an initial P-Erk immunofluorescence protocol; and Ethan Scott for providing the Tg(s1173:Gal4)line. Imaging was performed in the Australian Cancer Research Foundation’sDynamic Imaging Facility at IMB and at the Hubrecht Imaging Center (HIC).

Competing interestsThe authors declare no competing financial interests.

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Dev

elop

men

t

Page 11: Ccbe1 regulates Vegfc-mediated induction of Vegfr3 ... › ... › 12 › dev.100495.full.pdfterminus of the protein. The protein localizes to secretory vesicles (Alders et al., 2009),

Author contributionsL.L.G. designed and performed experiments, analyzed data and co-wrote themanuscript; T.K., D.S., N.C.H., K.K., G.R., N.I.B. and A.v.I. designed andperformed experiments, analyzed data and edited the manuscript; S.A.S. andM.G.A. designed experiments, analyzed data and edited the manuscript; S.S.-M.and B.M.H. designed experiments, analyzed data and co-wrote the manuscript.

FundingThis work was supported by an Australian Research Council Future Fellowship[FT100100165 to B.M.H.]; a European Molecular Biology Organization Long-TermFellowship [LTRF 52-2007 to T.K.]; Veni grants from The Netherlands Organisationfor Scientific Research (NWO) [916.10.132 to T.K.; 863.11.022 to G.R.]; MarieCurie Intra-European Fellowships (to D.S. and A.v.I.); National Health and MedicalResearch Council of Australia (NHMRC) project grants [631657 andAPP1050138]; a Program Grant [487900 to M.G.A. and S.A.S.] and ResearchFellowships (to M.G.A. and S.A.S.) from the National Health and MedicalResearch Council; and KNAW (S.S.-M.).

Supplementary materialSupplementary material available online athttp://dev.biologists.org/lookup/suppl/doi:10.1242/dev.100495/-/DC1

ReferencesAdzhubei, I. A., Schmidt, S., Peshkin, L., Ramensky, V. E., Gerasimova, A., Bork,

P., Kondrashov, A. S. and Sunyaev, S. R. (2010). A method and server forpredicting damaging missense mutations. Nat. Methods 7, 248-249.

Alders, M., Hogan, B. M., Gjini, E., Salehi, F., Al-Gazali, L., Hennekam, E. A.,Holmberg, E. E., Mannens, M. M. A. M., Mulder, M. F., Offerhaus, G. J. A. et al.(2009). Mutations in CCBE1 cause generalized lymph vessel dysplasia in humans.Nat. Genet. 41, 1272-1274.

Bahram, F. and Claesson-Welsh, L. (2010). VEGF-mediated signal transduction inlymphatic endothelial cells. Pathophysiology 17, 253-261.

Balciunas, D., Wangensteen, K. J., Wilber, A., Bell, J., Geurts, A., Sivasubbu, S.,Wang, X., Hackett, P. B., Largaespada, D. A., McIvor, R. S. et al. (2006).Harnessing a high cargo-capacity transposon for genetic applications in vertebrates.PLoS Genet. 2, e169.

Bos, F. L., Caunt, M., Peterson-Maduro, J., Planas-Paz, L., Kowalski, J., Karpanen,T., van Impel, A., Tong, R., Ernst, J. A., Korving, J. et al. (2011). CCBE1 is essentialfor mammalian lymphatic vascular development and enhances the lymphangiogeniceffect of vascular endothelial growth factor-C in vivo. Circ. Res. 109, 486-491.

Bussmann, J. and Schulte-Merker, S. (2011). Rapid BAC selection for tol2-mediatedtransgenesis in zebrafish. Development 138, 4327-4332.

Bussmann, J., Bos, F. L., Urasaki, A., Kawakami, K., Duckers, H. J. and Schulte-Merker, S. (2010). Arteries provide essential guidance cues for lymphatic endothelialcells in the zebrafish trunk. Development 137, 2653-2657.

Cha, Y. R., Fujita, M., Butler, M., Isogai, S., Kochhan, E., Siekmann, A. F. andWeinstein, B. M. (2012). Chemokine signaling directs trunk lymphatic networkformation along the preexisting blood vasculature. Dev. Cell 22, 824-836.

Connell, F., Kalidas, K., Ostergaard, P., Brice, G., Homfray, T., Roberts, L.,Bunyan, D. J., Mitton, S., Mansour, S., Mortimer, P. et al.; LymphoedemaConsortium (2010). Linkage and sequence analysis indicate that CCBE1 is mutatedin recessively inherited generalised lymphatic dysplasia. Hum. Genet. 127, 231-241.

Dixelius, J., Makinen, T., Wirzenius, M., Karkkainen, M. J., Wernstedt, C., Alitalo,K. and Claesson-Welsh, L. (2003). Ligand-induced vascular endothelial growthfactor receptor-3 (VEGFR-3) heterodimerization with VEGFR-2 in primary lymphaticendothelial cells regulates tyrosine phosphorylation sites. J. Biol. Chem. 278, 40973-40979.

Dumont, D. J., Jussila, L., Taipale, J., Lymboussaki, A., Mustonen, T., Pajusola,K., Breitman, M. and Alitalo, K. (1998). Cardiovascular failure in mouse embryosdeficient in VEGF receptor-3. Science 282, 946-949.

Ertzer, R., Müller, F., Hadzhiev, Y., Rathnam, S., Fischer, N., Rastegar, S. andSträhle, U. (2007). Cooperation of sonic hedgehog enhancers in midline expression.Dev. Biol. 301, 578-589.

Gordon, K., Schulte, D., Brice, G., Simpson, M. A., Roukens, M. G., van Impel, A.,Connell, F., Kalidas, K., Jeffery, S., Mortimer, P. S. et al. (2013). Mutation invascular endothelial growth factor-C, a ligand for vascular endothelial growth factorreceptor-3, is associated with autosomal dominant milroy-like primary lymphedema.Circ. Res. 112, 956-960.

Hägerling, R., Pollmann, C., Andreas, M., Schmidt, C., Nurmi, H., Adams, R. H.,Alitalo, K., Andresen, V., Schulte-Merker, S. and Kiefer, F. (2013). A novelmultistep mechanism for initial lymphangiogenesis in mouse embryos based onultramicroscopy. EMBO J. 32, 629-644.

Hartley, J. L., Temple, G. F. and Brasch, M. A. (2000). DNA cloning using in vitro site-specific recombination. Genome Res. 10, 1788-1795.

Hogan, B. M., Bos, F. L., Bussmann, J., Witte, M., Chi, N. C., Duckers, H. J. andSchulte-Merker, S. (2009a). Ccbe1 is required for embryonic lymphangiogenesisand venous sprouting. Nat. Genet. 41, 396-398.

Hogan, B. M., Herpers, R., Witte, M., Heloterä, H., Alitalo, K., Duckers, H. J. andSchulte-Merker, S. (2009b). Vegfc/Flt4 signalling is suppressed by Dll4 indeveloping zebrafish intersegmental arteries. Development 136, 4001-4009.

Inoue, D. and Wittbrodt, J. (2011). One for all – a highly efficient and versatile methodfor fluorescent immunostaining in fish embryos. PLoS ONE 6:e19713.

Irrthum, A., Devriendt, K., Chitayat, D., Matthijs, G., Glade, C., Steijlen, P. M.,Fryns, J. P., Van Steensel, M. A. and Vikkula, M. (2003). Mutations in thetranscription factor gene SOX18 underlie recessive and dominant forms ofhypotrichosis-lymphedema-telangiectasia. Am. J. Hum. Genet. 72, 1470-1478.

Isogai, S., Lawson, N. D., Torrealday, S., Horiguchi, M. and Weinstein, B. M.(2003). Angiogenic network formation in the developing vertebrate trunk.Development 130, 5281-5290.

Isogai, S., Hitomi, J., Yaniv, K. and Weinstein, B. M. (2009). Zebrafish as a newanimal model to study lymphangiogenesis. Anat. Sci. Int. 84, 102-111.

Jeltsch, M., Kaipainen, A., Joukov, V., Meng, X., Lakso, M., Rauvala, H., Swartz,M., Fukumura, D., Jain, R. K. and Alitalo, K. (1997). Hyperplasia of lymphaticvessels in VEGF-C transgenic mice. Science 276, 1423-1425.

Joukov, V., Sorsa, T., Kumar, V., Jeltsch, M., Claesson-Welsh, L., Cao, Y., Saksela,O., Kalkkinen, N. and Alitalo, K. (1997). Proteolytic processing regulates receptorspecificity and activity of VEGF-C. EMBO J. 16, 3898-3911.

Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V. W., Fang, G. H.,Dumont, D., Breitman, M. and Alitalo, K. (1995). Expression of the fms-liketyrosine kinase 4 gene becomes restricted to lymphatic endothelium duringdevelopment. Proc. Natl. Acad. Sci. USA 92, 3566-3570.

Karkkainen, M. J., Ferrell, R. E., Lawrence, E. C., Kimak, M. A., Levinson, K. L.,McTigue, M. A., Alitalo, K. and Finegold, D. N. (2000). Missense mutationsinterfere with VEGFR-3 signalling in primary lymphoedema. Nat. Genet. 25, 153-159.

Karkkainen, M. J., Haiko, P., Sainio, K., Partanen, J., Taipale, J., Petrova, T. V.,Jeltsch, M., Jackson, D. G., Talikka, M., Rauvala, H. et al. (2004). Vascularendothelial growth factor C is required for sprouting of the first lymphatic vesselsfrom embryonic veins. Nat. Immunol. 5, 74-80.

Karnezis, T., Shayan, R., Caesar, C., Roufail, S., Harris, N. C., Ardipradja, K.,Zhang, Y. F., Williams, S. P., Farnsworth, R. H., Chai, M. G. et al. (2012). VEGF-Dpromotes tumor metastasis by regulating prostaglandins produced by the collectinglymphatic endothelium. Cancer Cell 21, 181-195.

Koltowska, K., Betterman, K. L., Harvey, N. L. and Hogan, B. M. (2013). Getting outand about: the emergence and morphogenesis of the vertebrate lymphaticvasculature. Development 140, 1857-1870.

Küchler, A. M., Gjini, E., Peterson-Maduro, J., Cancilla, B., Wolburg, H. andSchulte-Merker, S. (2006). Development of the zebrafish lymphatic system requiresVEGFC signaling. Curr. Biol. 16, 1244-1248.

Lawson, N. D. and Weinstein, B. M. (2002). In vivo imaging of embryonic vasculardevelopment using transgenic zebrafish. Dev. Biol. 248, 307-318.

Leslie, J. D., Ariza-McNaughton, L., Bermange, A. L., McAdow, R., Johnson, S. L.and Lewis, J. (2007). Endothelial signalling by the Notch ligand Delta-like 4 restrictsangiogenesis. Development 134, 839-844.

McColl, B. K., Baldwin, M. E., Roufail, S., Freeman, C., Moritz, R. L., Simpson, R.J., Alitalo, K., Stacker, S. A. and Achen, M. G. (2003). Plasmin activates thelymphangiogenic growth factors VEGF-C and VEGF-D. J. Exp. Med. 198, 863-868.

Nicoli, S., Knyphausen, C. P., Zhu, L. J., Lakshmanan, A. and Lawson, N. D.(2012). miR-221 is required for endothelial tip cell behaviors during vasculardevelopment. Dev. Cell 22, 418-429.

Okuda, K. S., Astin, J. W., Misa, J. P., Flores, M. V., Crosier, K. E. and Crosier, P. S.(2012). lyve1 expression reveals novel lymphatic vessels and new mechanisms forlymphatic vessel development in zebrafish. Development 139, 2381-2391.

Salameh, A., Galvagni, F., Bardelli, M., Bussolino, F. and Oliviero, S. (2005). Directrecruitment of CRK and GRB2 to VEGFR-3 induces proliferation, migration, andsurvival of endothelial cells through the activation of ERK, AKT, and JNK pathways.Blood 106, 3423-3431.

Scheer, N., Groth, A., Hans, S. and Campos-Ortega, J. A. (2001). An instructivefunction for Notch in promoting gliogenesis in the zebrafish retina. Development 128,1099-1107.

Siegfried, G., Basak, A., Cromlish, J. A., Benjannet, S., Marcinkiewicz, J.,Chrétien, M., Seidah, N. G. and Khatib, A. M. (2003). The secretory proproteinconvertases furin, PC5, and PC7 activate VEGF-C to induce tumorigenesis. J. Clin.Invest. 111, 1723-1732.

Siekmann, A. F. and Lawson, N. D. (2007). Notch signalling limits angiogenic cellbehaviour in developing zebrafish arteries. Nature 445, 781-784.

van Impel, A. and Schulte-Merker, S. (2014). A fisheye view on lymphangiogenesis.Adv. Anat. Embryol. Cell Biol. 214, 153-165.

van Impel, A., Zhao, Z., Hermkens, D. M. A., Roukens, M. G., Fischer, J. C.,Peterson-Maduro, J., Duckers, H., Ober, E. A., Ingham, P. W. and Schulte-Merker, S. (2014). Divergence of zebrafish and mouse lymphatic cell fatespecification pathways. Development 141, (in press).

Veikkola, T., Jussila, L., Makinen, T., Karpanen, T., Jeltsch, M., Petrova, T. V.,Kubo, H., Thurston, G., McDonald, D. M., Achen, M. G. et al. (2001). Signallingvia vascular endothelial growth factor receptor-3 is sufficient for lymphangiogenesisin transgenic mice. EMBO J. 20, 1223-1231.

Villefranc, J. A., Nicoli, S., Bentley, K., Jeltsch, M., Zarkada, G., Moore, J. C.,Gerhardt, H., Alitalo, K. and Lawson, N. D. (2013). A truncation allele in vascularendothelial growth factor c reveals distinct modes of signaling during lymphatic andvascular development. Development 140, 1497-1506.

Walhout, A. J., Temple, G. F., Brasch, M. A., Hartley, J. L., Lorson, M. A., van denHeuvel, S. and Vidal, M. (2000). GATEWAY recombinational cloning: application tothe cloning of large numbers of open reading frames or ORFeomes. MethodsEnzymol. 328, 575-592, IN7.

Wienholds, E., Schulte-Merker, S., Walderich, B. and Plasterk, R. H. (2002). Target-selected inactivation of the zebrafish rag1 gene. Science 297, 99-102.

Yaniv, K., Isogai, S., Castranova, D., Dye, L., Hitomi, J. and Weinstein, B. M. (2006).Live imaging of lymphatic development in the zebrafish. Nat. Med. 12, 711-716.

11

RESEARCH ARTICLE Development (2014) doi:10.1242/dev.100495

Dev

elop

men

t