In mammals, the homeodomain transcription factor Prox1 acts as the central regulator of lymphatic cell fate. Its restricted expression in a subset of cardinal vein cells leads to a switch towards lymphatic specification and hence represents a prerequisite for the initiation of lymphangiogenesis. Murine Prox1-null embryos lack lymphatic structures, and sustained expression of Prox1 is indispensable for the maintenance of lymphatic cell fate even at adult stages, highlighting the unique importance of this gene for the lymphatic lineage. Whether this pre-eminent role of Prox1 within the lymphatic vasculature is conserved in other vertebrate classes has remained unresolved, mainly owing to the lack of availability of loss-of-function mutants. Here, we re-examine the role of Prox1a in zebrafish lymphangiogenesis. First, using a transgenic reporter line, we show that prox1a is initially expressed in different endothelial compartments, becoming restricted to lymphatic endothelial cells only at later stages. Second, using targeted mutagenesis, we show that Prox1a is dispensable for lymphatic specification and subsequent lymphangiogenesis in zebrafish. In line with this result, we found that the functionally related transcription factors Coup-TFII and Sox18 are also dispensable for lymphangiogenesis. Together, these findings suggest that lymphatic commitment in zebrafish and mice is controlled in fundamentally different ways.

Lymphatic vessels play pivotal roles in tissue fluid homeostasis, immune cell trafficking and the uptake of dietary fats in the small intestine. A failure in lymphatic vessel development (lymphangiogenesis) or in lymphatic function is causative for several inherited or acquired pathological conditions that lead to tissue swelling by accumulation of extravasated fluids (Tammela and Alitalo, 2010; Schulte-Merker et al., 2011).

In mice, combined activity of the transcription factors Coup-TFII (Nr2f2 - Mouse Genome Informatics) (Srinivasan et al., 2010) and Sox18 (François et al., 2008) leads to the polarized expression of Prox1 in a subset of endothelial cells (ECs) within the cardinal vein at embryonic day (E) 9.5. Shortly thereafter, Prox1-positive ECs leave the cardinal vein in a dorsal direction, mediated by Vegfc- and Flt4-driven processes of polarized sprouting and migration, resulting in the formation of the first lymphatic structures in the embryo (Karkkainen et al., 2004; Hägerling et al., 2013). Prox1 expression in lymphatic precursor cells is essential for the initiation of a lymphatic gene expression program, and Prox1 knockout mice lack all lymphatic structures (Wigle et al., 2002). Forced expression of Prox1 is sufficient to confer lymphatic identity to blood ECs, demonstrating the pivotal role of the gene for lymphatic specification (Hong et al., 2002; Petrova et al., 2002). Continuous expression of Prox1 in lymphatic ECs is also indispensable for the maintenance of lymphatic cell fate during later stages of development (Johnson et al., 2008), thus firmly establishing murine Prox1 as the central determining factor of lymphatic identity.

In the zebrafish trunk, the process of vasculogenesis establishes an initial primitive circulatory loop consisting of the dorsal aorta (DA) and the posterior cardinal vein (PCV). Then, in a wave of angiogenic sprouting from the DA (primary or arterial sprouting), a set of ∼30 arterial intersegmental vessels (ISVs) is formed on each side of the embryo. Shortly thereafter, at about 36 hours post-fertilization (hpf), another group of ∼30 sprouts emerges from each side of the PCV. These venous (or secondary) sprouts also migrate dorsally, and about half of them make a stable connection to pre-existing arterial ISVs, thereby remodeling them into intersegmental veins. Venous sprouts that fail to connect to arteries migrate further dorsally towards the midline of the embryo where they populate the region of the horizontal myoseptum as parachordal lymphangioblasts (PLs) (Hogan et al., 2009a), which constitute a pool of lymphatic precursors in the embryonic trunk. These PLs will subsequently migrate away from the horizontal myoseptum [at 2.5 days post-fertilization (dpf)] using arterial ISVs as migration routes to populate the different regions of the trunk, eventually giving rise to the thoracic duct (TD; situated between the DA and PCV), a number of intersegmental lymphatic vessels (ISLVs) in close proximity to arterial ISVs, and the dorsal longitudinal lymphatic vessel (DLLV) (Bussmann et al., 2010).

Previous work has indicated a strong conservation of the genes controlling lymphangiogenesis between zebrafish and mammals. In all organisms examined, mutations in the transmembrane receptor Flt4, its secreted ligand Vegfc, or the more recently discovered gene ccbe1 lead to a block of lymphangiogenesis already at the level of sprouting from the venous endothelium (Schulte-Merker et al., 2011; Koltowska et al., 2013). Although several publications have suggested that Prox1 function in lymphatic specification might be conserved both in amphibians (Ny et al., 2005) and fish, the evidence in the case of the latter has remained open to interpretation, complicated by the existence of duplicated prox1 genes in zebrafish (Del Giacco et al., 2010; Tao et al., 2011). Although expression of prox1a within lymphatic structures has been reported (Yaniv et al., 2006), it remains unclear whether this expression consistently marks all lymphatic structures during different stages of lymphangiogenesis. No mutant allele of prox1a has previously been described and its morpholino-mediated knockdown results in severely malformed embryos, making a conclusive assessment of its requirement for lymphatic development impossible (Küchler et al., 2006). Thus, although mutations in prox1b do not interfere with normal lymphatic development (Tao et al., 2011), the possibility remains that prox1a could indeed be required during lymphatic specification in fish.

Using a novel transgenic reporter line, we show here that prox1a exhibits a dynamic expression pattern in different endothelial compartments during early vascular development. In contrast to the situation in mice, we found that expression of this reporter gene only becomes a specific and reliable marker for lymphatic ECs at later stages of lymphangiogenesis, arguing against a lymphatic specification function during the onset of venous sprouting. In line with this, using a novel targeted allele of prox1a in combination with the previously described prox1b mutation, we show that lymphangiogenesis can proceed in the complete absence of Prox1 in zebrafish. In addition, we show that the functionally related transcription factors Nr2f2 (referred to as Coup-TFII in the following) and Sox18 are also dispensable during lymphangiogenesis. These results indicate that the Sox18/Coup-TFII/Prox1 lymphatic specification code is not conserved in fish, suggesting an alternative mode of lymphatic commitment in this vertebrate class.

To obtain a more precise and sensitive readout for lymphatic structures during development, we created a bacterial artificial chromosome (BAC) reporter line for flt4 [Tg(flt4BAC:mCitrine), flt4:mCit hereafter]. Similar to previous observations in mice (Kaipainen et al., 1995), we found that flt4 is initially expressed in all blood ECs but becomes progressively restricted to venous and lymphatic endothelial cells (LECs) after 26 hpf (supplementary material Fig. S1A,B) and 36 hpf (supplementary material Fig. S1C-F), respectively. In contrast to Flt4 expression in mice (Hägerling et al., 2013), flt4:mCit expression is not lost in venous structures, making it a lymphatic-enriched, but not lymphatic-specific reporter at 5 dpf. In pan-endothelial transgenic lines such as fli1a:eGFP, the TD is the only lymphatic structure that is readily distinguishable from blood vessels. By comparison, the flt4 reporter line also reliably highlights more delicate lymphatic structures such as ISLVs or the DLLV in the trunk (supplementary material Fig. S1G,H) as well as the facial lymphatic system (Fig. 1A,B). Combined with the arterial-specific flt1enh:tdTom reporter (Bussmann et al., 2010), all lymphatic, venous and arterial structures in the embryo can be easily distinguished (see below).

Fig. 1.

prox1a expression marks different endothelial compartments during vascular development. In all pictures (except for E), flt4:mCit expression is shown in green and prox1a:KalTA4,UAS:tagRFP expression is highlighted in red. (A) In full z-projections, prox1a expression at 5 dpf is apparent in various tissues, including liver (white arrowhead), lens (arrow) and myotome (blue arrowhead). (A′) Same z-projection displaying only the flt4:mCit expression in venous and lymphatic ECs. (B) Partial z-projection of the same embryo (comprising only optical sections without myotome signal) reveals additional expression in the spinal cord (blue arrowhead) and in the lymphatic vasculature of the head (arrow) and the trunk (white arrowhead). Images in A and B are composed of several overlapping z-projections because the embryo was too large to fit in a single field of view. (C,D) Higher magnification of trunk lymphatics at 5 dpf exhibiting combinatorial prox1a (red) and flt4 (green) reporter expression restricted to the TD (arrows) and ISLVs (arrowheads). (E) Depth color-coded z-projection (projecting each slice in a different color according to its position within the stack; see color bar) of boxed region in B (only prox1a channel) reveals the presence of two separate prox1a-positive vessels above the swim bladder (white arrows point to lymphatics on the right, arrowheads to lymphatics on the left body side), which merge near the sixth ISVs with the TD in the trunk region (yellow arrow). (F) Dorsal view of the prox1a-positive bilateral anterior TD (arrowheads), which connects to the trunk TD at the indicated location (yellow arrow). The image has been assembled from a set of partial z-projections (see dotted lines) owing to interference of other prox1a-positive structures. (G,H) Full (G) and partial (H) z-projections of the area (compare with box in A) where the anterior TD (arrowhead in G) and the facial lymphatic network (arrow in G) fuse and drain into the common cardinal vein (arrow in H) in a 5 dpf embryo. ACV, anterior cardinal vein; CCV, common cardinal vein; JLV, jugular lymphatic vessel; PCV, posterior cardinal vein; TD, thoracic duct. (I,J) At 32 hpf, prox1a-positive cells (red, arrows) are located within the PCV (green). Note that in this lateral view, prox1a-positive cells are located in both the dorsal and ventral aspect of the PCV. (K) A sprouting venous EC (arrow) at 38 hpf expressing both flt4 and prox1a transgenes. (L,O) At 48 hpf, only a proportion of PLs are prox1a:KalTA4-positive (white arrows) whereas the majority shows no signs of reporter expression (yellow arrows). In addition, venous ISVs positive for the prox1a reporter are evident (white arrowheads). (L) Overlay of a full z-projection of the flt4:mCit signal (green, to outline the complete vasculature) and identical partial z-projections of the prox1a (red, see also O) and flt4 reporter (blue, to highlight the part of the vasculature that is included in the partial z-projection of the prox1a signal). Note that prox1a-positive ECs will appear white in the overlay. (M,N) The prox1a reporter line also labels individual arterial sprouts (arrowheads) at 27 hpf. (P,Q) Single z-plane of the trunk region in a 25 hpf embryo showing DA cells positive for prox1a reporter expression (arrowheads). Note ventral domain expression of the DA. (R) Intersegmental vein showing both prox1a and flt4 reporter expression at 48 hpf. (S,T) At 3.5 dpf, all LECs display expression of the prox1a reporter but expression in other endothelial domains has disappeared. (U) Expression of prox1a in the head region at 3.5 dpf, highlighting the facial lymphatic network (arrow) including the drainage point with the common cardinal vein (arrowhead).

Fig. 1.

prox1a expression marks different endothelial compartments during vascular development. In all pictures (except for E), flt4:mCit expression is shown in green and prox1a:KalTA4,UAS:tagRFP expression is highlighted in red. (A) In full z-projections, prox1a expression at 5 dpf is apparent in various tissues, including liver (white arrowhead), lens (arrow) and myotome (blue arrowhead). (A′) Same z-projection displaying only the flt4:mCit expression in venous and lymphatic ECs. (B) Partial z-projection of the same embryo (comprising only optical sections without myotome signal) reveals additional expression in the spinal cord (blue arrowhead) and in the lymphatic vasculature of the head (arrow) and the trunk (white arrowhead). Images in A and B are composed of several overlapping z-projections because the embryo was too large to fit in a single field of view. (C,D) Higher magnification of trunk lymphatics at 5 dpf exhibiting combinatorial prox1a (red) and flt4 (green) reporter expression restricted to the TD (arrows) and ISLVs (arrowheads). (E) Depth color-coded z-projection (projecting each slice in a different color according to its position within the stack; see color bar) of boxed region in B (only prox1a channel) reveals the presence of two separate prox1a-positive vessels above the swim bladder (white arrows point to lymphatics on the right, arrowheads to lymphatics on the left body side), which merge near the sixth ISVs with the TD in the trunk region (yellow arrow). (F) Dorsal view of the prox1a-positive bilateral anterior TD (arrowheads), which connects to the trunk TD at the indicated location (yellow arrow). The image has been assembled from a set of partial z-projections (see dotted lines) owing to interference of other prox1a-positive structures. (G,H) Full (G) and partial (H) z-projections of the area (compare with box in A) where the anterior TD (arrowhead in G) and the facial lymphatic network (arrow in G) fuse and drain into the common cardinal vein (arrow in H) in a 5 dpf embryo. ACV, anterior cardinal vein; CCV, common cardinal vein; JLV, jugular lymphatic vessel; PCV, posterior cardinal vein; TD, thoracic duct. (I,J) At 32 hpf, prox1a-positive cells (red, arrows) are located within the PCV (green). Note that in this lateral view, prox1a-positive cells are located in both the dorsal and ventral aspect of the PCV. (K) A sprouting venous EC (arrow) at 38 hpf expressing both flt4 and prox1a transgenes. (L,O) At 48 hpf, only a proportion of PLs are prox1a:KalTA4-positive (white arrows) whereas the majority shows no signs of reporter expression (yellow arrows). In addition, venous ISVs positive for the prox1a reporter are evident (white arrowheads). (L) Overlay of a full z-projection of the flt4:mCit signal (green, to outline the complete vasculature) and identical partial z-projections of the prox1a (red, see also O) and flt4 reporter (blue, to highlight the part of the vasculature that is included in the partial z-projection of the prox1a signal). Note that prox1a-positive ECs will appear white in the overlay. (M,N) The prox1a reporter line also labels individual arterial sprouts (arrowheads) at 27 hpf. (P,Q) Single z-plane of the trunk region in a 25 hpf embryo showing DA cells positive for prox1a reporter expression (arrowheads). Note ventral domain expression of the DA. (R) Intersegmental vein showing both prox1a and flt4 reporter expression at 48 hpf. (S,T) At 3.5 dpf, all LECs display expression of the prox1a reporter but expression in other endothelial domains has disappeared. (U) Expression of prox1a in the head region at 3.5 dpf, highlighting the facial lymphatic network (arrow) including the drainage point with the common cardinal vein (arrowhead).

prox1a is expressed within different endothelial cell populations during development

To assess whether prox1a might represent the functional Prox1 ortholog in zebrafish and would therefore be expressed specifically in LECs, we generated different prox1a BAC reporter lines that were analyzed in combination with the aforementioned flt4:mCit line. As simple fluorophore expression under the control of the prox1aBAC promoter resulted in extremely weak signals (not shown), we took advantage of the enhancing effect of an optimized Gal4 variant [KalTA4 (Distel et al., 2009)] in a prox1aBAC:KalTA4, UAS:tagRFP expression cassette. Consistent with previously published data (Glasgow and Tomarev, 1998; Thisse and Thisse, 2005; Pistocchi et al., 2009), the transgene marked a wide range of tissues comprising the lens, retina, liver, neuromasts and myotome (Fig. 1A). To identify expression domains masked by the myotomal expression in full z-projections, partial z-projections of the trunk were analyzed, revealing expression in all lymphatic structures within the head and trunk, where the signal colocalized with flt4:mCit expression at 5 dpf (Fig. 1B-D). In contrast to previously published lymphatic markers such as stabilin1:YFP or lyve1:DsRed2 (Hogan et al., 2009a; Okuda et al., 2012) or the flt4:mCit line, at 5 dpf the prox1a reporter exhibits no additional expression domains in other endothelial compartments, such as the PCV, and hence reflects the first truly LEC-specific marker at this stage of development in zebrafish. Taking advantage of this feature, we found that the anterior part of the TD, which connects the head lymphatics with the TD in the trunk, comprises a bilateral structure above the developing swim bladder. The TD splits up near the sixth intersegmental vessel pair and runs in close proximity to the correspondingly bilateral PCVs (Fig. 1E,F; supplementary material Fig. S2A), finally connecting to the facial lymphatic network (Okuda et al., 2012) and ultimately the common cardinal veins (Fig. 1G,H).

Next, we investigated whether zebrafish prox1a expression would, like its murine counterpart, mark LECs at earlier stages, possibly even within the venous endothelium of the PCV at a time point prior to venous (secondary) sprouting. At 32 hpf, shortly before the emergence of the first sprouts from the PCV, prox1a could indeed be detected in a subset of venous cells (Fig. 1I,J) and subsequently in sprouting venous ECs (Fig. 1K). To determine whether prox1a might exclusively mark secondary sprouts that will give rise to lymphangioblasts, expression was analyzed at 44 hpf, a stage when PL cells have extended to the horizontal myoseptum and venous destined sprouts have already established a connection to the respective ISV. We found prox1a reporter expression only in a proportion of PLs (Fig. 1L; 17/78 PLs), indicating that not all lymphatic precursor cells are positive for prox1a at this stage. Equally important, the transgene also transiently marked individual venous ISVs (in 12/21 imaged regions) (Fig. 1L,O,R), suggesting no clear correlation between prox1a expression and the fate of secondary sprouts.

Earlier during development, prox1a reporter activity could also be detected in ECs of developing arterial ISVs (Fig. 1M,N) and in particular of the DA (Fig. 1P,Q), as well as in undefined cells within the axial vessels before the onset of circulation (supplementary material Fig. S2B-E). In addition, transgene expression highlighted a subpopulation of ECs in the caudal vein region already at 26 hpf, an expression domain that could be detected up to day 3 (supplementary material Fig. S2D-G). Probably as a consequence of this early and relatively wide expression within the caudal vein, prox1a-positive venous ISVs and PL sprouts were more frequently detected in this area at 48 hpf (supplementary material Fig. S2G).

Because prox1a is initially expressed within different endothelial subpopulations, we wanted to establish the earliest stage when prox1a expression would reliably and exclusively mark the lymphatic part of the vasculature. We found that, at least at 3.5 dpf, when LECs leave the horizontal myoseptum region and form the different lymphatic structures within the trunk, all LECs were highlighted by expression of the reporter, which was also the case in the head region (Fig. 1S-U). However, because the strong myotomal signal did not allow a conclusive assessment of prox1a expression in lymphangioblasts at the level of the horizontal myoseptum, it cannot be excluded that the gene was already expressed in all LECs while still aligning with the horizontal myoseptum.

Taken together, expression of prox1a becomes a reliable and very useful maker for LECs only at later stages of lymphangiogenesis. Therefore, the expression data presented here would be in line with a prox1a function in specifying PLs at the level of secondary sprouts but not at the level of the vein, as is the case in mice.

Lymphatic development is not blocked in prox1a mutant embryos

To assess the role of Prox1a in lymphangiogenesis, we generated a loss-of-function allele for prox1a using zinc-finger nucleases (supplementary material Fig. S3A). The prox1ai278 allele harbors a 10 bp deletion in the first coding exon, resulting in a frame-shift after amino acid 149, and a premature stop codon after additional four amino acids (Fig. 2A). Homozygous mutants are devoid of full-length Prox1a protein as assessed by immunostaining of slow muscle fibers at 30 hpf (Fig. 2B,C). The overall appearance of homozygous prox1ai278 mutants was normal until between days 4 and 5, at which stage mutant larvae started to develop severe edema around the gut and the eye (Fig. 2D,E). At 5.5-6 dpf, most mutants had impaired blood circulation and showed signs of tissue necrosis. Importantly, timing as well as severity of edema was different from ccbe1, vegfc and flt4 mutant scenarios: although completely lacking lymphatic structures, mutants of each of those genes only develop mild edema starting from 5-6 dpf. The later onset of edema in those lymphatic mutants presumably results from the lymphatic system beginning to perform its drainage function at early day 5 (Karpanen and Schulte-Merker, 2011); thus, atypical tissue fluid accumulations can only arise from this time point. It follows that the edema observed in prox1a mutants does not indicate impaired lymphatic drainage function.

Fig. 2.

Lymphangiogenesis in prox1a mutant embryos. (A) Schematic of the homeodomain (HD) containing Prox1a protein, indicating the predicted effect of the 10 bp deletion in the prox1ai278 allele, leading to a frame-shift (red amino acids) and a truncated protein after 153 amino acids. (B,C) Prox1a immunostaining of slow muscle fibers in sibling (B) and homozygous mutant prox1ai278 (C) embryos demonstrates a complete loss of wild-type Prox1a protein (green) at 30 hpf (slow myosin heavy chain-1 is shown in red). (D,E) Brightfield pictures of 5 dpf sibling (D) and homozygous prox1ai278 mutant (E) embryos. Note the strong edema formation around the eye and gut area (arrowheads), which can be even more pronounced in other prox1a mutants at this stage. (F,G) In both heterozygous siblings (F) and homozygous prox1ai278 mutants (G), PLs appear at the level of the horizontal myoseptum at 2 dpf (arrows). (H) Average PL numbers per embryo are mildly reduced in prox1a mutants at 2 dpf (Student’s t-test, *P=0.025). Error bars indicate s.d. of wild-type (green), heterozygous (orange) and mutant (red) groups in embryos from a prox1a+/- incross. (I-K) flt4:mCit; flt1enh:tdTom double transgenic embryos highlighting arterial ISVs in red and venous and lymphatic structures in green. Compared with heterozygous siblings (I), most homozygous prox1ai278 mutants do not display TD defects at 5 dpf (J), whereas others display a mild reduction (K) in some areas of the trunk (arrows point at TD; asterisks mark the lack of TD). Note the overall unaffected ratio of venous and arterial ISVs in mutants (J,K). (L) Average number of segments positive for TD cells, scored in the first ten segments above the yolk extension at 5 dpf. Error bars indicate the s.d. for the respective genotypic class from a prox1a+/- incross. ***P=2.3E-08 (Student’s t-test, comparison of wild-type and mutant population). (M,N) The average percentage of intersegmental veins (M) and arteries (N) does not differ between genotypic classes in an incross of prox1ai278 carriers. (O) In prox1ai278 mutants, the average number of ISVs is not altered. Error bars represent s.d. n.s., not statistically significant.

Fig. 2.

Lymphangiogenesis in prox1a mutant embryos. (A) Schematic of the homeodomain (HD) containing Prox1a protein, indicating the predicted effect of the 10 bp deletion in the prox1ai278 allele, leading to a frame-shift (red amino acids) and a truncated protein after 153 amino acids. (B,C) Prox1a immunostaining of slow muscle fibers in sibling (B) and homozygous mutant prox1ai278 (C) embryos demonstrates a complete loss of wild-type Prox1a protein (green) at 30 hpf (slow myosin heavy chain-1 is shown in red). (D,E) Brightfield pictures of 5 dpf sibling (D) and homozygous prox1ai278 mutant (E) embryos. Note the strong edema formation around the eye and gut area (arrowheads), which can be even more pronounced in other prox1a mutants at this stage. (F,G) In both heterozygous siblings (F) and homozygous prox1ai278 mutants (G), PLs appear at the level of the horizontal myoseptum at 2 dpf (arrows). (H) Average PL numbers per embryo are mildly reduced in prox1a mutants at 2 dpf (Student’s t-test, *P=0.025). Error bars indicate s.d. of wild-type (green), heterozygous (orange) and mutant (red) groups in embryos from a prox1a+/- incross. (I-K) flt4:mCit; flt1enh:tdTom double transgenic embryos highlighting arterial ISVs in red and venous and lymphatic structures in green. Compared with heterozygous siblings (I), most homozygous prox1ai278 mutants do not display TD defects at 5 dpf (J), whereas others display a mild reduction (K) in some areas of the trunk (arrows point at TD; asterisks mark the lack of TD). Note the overall unaffected ratio of venous and arterial ISVs in mutants (J,K). (L) Average number of segments positive for TD cells, scored in the first ten segments above the yolk extension at 5 dpf. Error bars indicate the s.d. for the respective genotypic class from a prox1a+/- incross. ***P=2.3E-08 (Student’s t-test, comparison of wild-type and mutant population). (M,N) The average percentage of intersegmental veins (M) and arteries (N) does not differ between genotypic classes in an incross of prox1ai278 carriers. (O) In prox1ai278 mutants, the average number of ISVs is not altered. Error bars represent s.d. n.s., not statistically significant.

In Prox1-null mutant mice, the failure of LEC progenitors to leave the cardinal vein results in the absence of lymphatic structures (Yang et al., 2012) and even heterozygous embryos die shortly after birth with dysfunctional lymphatics and a lack of lymphovenous valves (Harvey et al., 2005; Srinivasan and Oliver, 2011). If prox1a was equally important in specifying lymphatic cell fate in zebrafish, homozygous mutants should be deficient in lymphatic structures. However, when analyzing the appearance of PLs at the horizontal myoseptum at 2 dpf, we found that prox1ai278 mutants show only a marginal reduction in the number of PLs (Fig. 2F-H). As it is possible that this initially mild defect results in more dramatic effects at later stages, we evaluated whether the formation of the TD was impaired. At 5 dpf, no defects in heterozygous embryos and only minor defects in a proportion of homozygous mutants were evident (Fig. 2I-K). On average, homozygous mutants lacked the TD only in 1.4 of the first ten somites, a mild phenotype that could also reflect a secondary defect caused by the early edema formation and overall impaired appearance of mutants at 5 dpf (Fig. 2L). Hence, expression of prox1a in the venous and lymphatic endothelium as revealed by the reporter gene described above is not essential for the specification of lymphatic structures. To confirm that the loss of prox1a did not affect secondary sprouts that are committed to a venous fate, the ratio of arteries and veins as well as the total number of ISVs were quantified. Overall, no defects on the blood vasculature could be observed and the arterio-venous ratio was also unaffected in mutants (Fig. 2I-K,M-O). We also found that expression of the prox1a reporter line was present in homozygous prox1ai278 mutants, indicating that, in contrast to the situation in mice (Srinivasan et al., 2010), Prox1a protein is not required for maintaining its own lymphatic expression (supplementary material Fig. S4A-D). Furthermore, considering that the prox1a reporter represents a specific lymphatic marker at 5 dpf, the unaltered expression of both flt4:mCit and prox1a:KalTA4,UAS:tagRFP in the TD of homozygous prox1a mutants argues for correct lymphatic specification of LECs even in the absence of functional Prox1a protein. Given the specific zygotic expression pattern of prox1a in different subsets of ECs, our finding that Prox1a protein was undetectable in slow muscle fibers of prox1ai278 mutants prior to secondary sprouting as well as the fact that maternally expressed prox1a transcripts have not been identified (Harvey et al., 2013), it seems unlikely that the lack of zygotic phenotype can be attributed to maternal rescue. We therefore conclude that prox1a function is not essential for any aspect of lympho-venous sprouting and that the gene is dispensable for lymphatic cell fate determination in zebrafish.

Double mutants for prox1a and prox1b only show minor lymphatic defects

Although neither of the two prox1 orthologs appears to be essential for zebrafish lymphatic development (Tao et al., 2011) (this study), it remains possible that they function redundantly. We therefore generated double mutants for prox1ai278 and the previously characterized loss-of-function allele prox1bSA0035 (Tao et al., 2011). In all allelic combinations, including double mutant embryos, PLs were observed at the horizontal myoseptum (Fig. 3A-C), indicating that even the absence of all Prox1 function fails to block the appearance of lymphatic-fated secondary sprouts. Subsequent analysis of double mutants for the formation of TD at 5 dpf demonstrated that a moderate but significant reduction in the length of the TD within the first ten segments was evident (Fig. 3D-H); whether this is a specific effect of prox1 deficiency, or whether this phenotype represents a secondary consequence of more general defects within the embryo (see Fig. 2E) is difficult to assess. The presence of PL cells, facial lymphatics (supplementary material Fig. S5A-D) and TD tissue in double mutants, however, clearly shows that lymphatic commitment is not governed by zygotic expression of the Prox1a and Prox1b transcription factors. It follows that lymphatic specification in zebrafish must be achieved in a different way, possibly through the broadened activity of another functionally related transcription factor.

Fig. 3.

Lymphatic specification is not blocked in prox1ai278;prox1bSA0035 double mutants. (A-C) In all genotype combinations of a prox1ai278;prox1bSA0035 double heterozygous incross, including double heterozygotes (A), homozygous prox1b mutants (B) and homozygous double mutants (C), PLs are apparent at the horizontal myoseptum at 2 dpf (arrows). (D-F) As in double heterozygous embryos (D), the loss of both copies of prox1b (E) does not lead to lymphatic defects on the level of TD formation. In prox1ai278;prox1bSA0035 double homozygous embryos (F), mild TD defects are occasionally visible (arrows indicate the TD and asterisks highlight segments without TD). (G) Average number of TD-positive segments within the first ten segments above the yolk extension for the indicated genotypic classes of a prox1a+/-;prox1b+/- incross. The moderate reduction in TD length is statistically significant in the prox1a single mutants (Student’s t-test, *P=0.01) and the prox1a;prox1b double mutant embryos (**P=0.002) when compared with wild-type siblings. Error bars represent s.d. (H) Overview about the average TD length (TD) in ten scored segments, the corresponding s.d. and the number of scored embryos (n) in a prox1a+/-;prox1b+/- incross. n.s., not statistically significant.

Fig. 3.

Lymphatic specification is not blocked in prox1ai278;prox1bSA0035 double mutants. (A-C) In all genotype combinations of a prox1ai278;prox1bSA0035 double heterozygous incross, including double heterozygotes (A), homozygous prox1b mutants (B) and homozygous double mutants (C), PLs are apparent at the horizontal myoseptum at 2 dpf (arrows). (D-F) As in double heterozygous embryos (D), the loss of both copies of prox1b (E) does not lead to lymphatic defects on the level of TD formation. In prox1ai278;prox1bSA0035 double homozygous embryos (F), mild TD defects are occasionally visible (arrows indicate the TD and asterisks highlight segments without TD). (G) Average number of TD-positive segments within the first ten segments above the yolk extension for the indicated genotypic classes of a prox1a+/-;prox1b+/- incross. The moderate reduction in TD length is statistically significant in the prox1a single mutants (Student’s t-test, *P=0.01) and the prox1a;prox1b double mutant embryos (**P=0.002) when compared with wild-type siblings. Error bars represent s.d. (H) Overview about the average TD length (TD) in ten scored segments, the corresponding s.d. and the number of scored embryos (n) in a prox1a+/-;prox1b+/- incross. n.s., not statistically significant.

coup-TFII mutants develop a wild-type lymphatic vasculature and do not show arterio-venous defects

The principal factor that promotes venous cell identity in many vertebrate embryos is the transcription factor COUP-TFII (NR2F2), which suppresses expression of arterial genes in venous ECs (You et al., 2005). As lymphatics are venous-derived, loss of endothelial Coup-TFII expression in mice not only causes venous specification defects but also results in a lack of LECs (Srinivasan et al., 2007). Coup-TFII has been reported to be required for the initiation of Prox1 expression within the cardinal vein by direct binding to its promoter region (Srinivasan et al., 2010) and subsequently also for the maintenance of Prox1 expression in future LECs during early lymphangiogenesis. The latter function is thought to be dependent on a direct physical interaction between Coup-TFII and Prox1, and heterodimers have been reported to act as co-regulators of several lymphatic lineage specific genes such as FLT4 in cultured LECs (Lee et al., 2009). More recently, vascular defects including a reduction in PL number and aberrant TD formation has been reported to result from morpholino knockdown of zebrafish coup-TFII (Aranguren et al., 2011). We therefore wondered whether lymphatic lineage specification in fish might be entirely regulated by coup-TFII instead of prox1 genes. To explore this possibility, we employed TALEN constructs targeting the first exon of coup-TFII and generated a 1 bp insertion allele nr2f2hu10330 (coup-TFIIhu10330 hereafter). Sequencing of cDNA from coup-TFIIhu10330 mutant embryos did not reveal alternative transcripts lacking the 1 bp insertion (data not shown). This suggests that the allele represents a loss-of-function situation because the insertion results in a frame-shift and premature stop codon 5′ to both the nuclear receptor-DNA-binding domain and the ligand-binding domain (Fig. 4A).

Fig. 4.

Specification of the lymphatic lineage is independent of coup-TFII and sox18. (A) Schematic of the Coup-TFII protein indicating the position of the first affected amino acid in the 1 bp insertion allele nr2f2hu10330 (coup-TFIIhu10330 hereafter) which leads to a premature stop codon after an additional four amino acids N-terminal to the nuclear receptor-DNA binding and ligand-binding domain. (B,C) Brightfield images of wild-type (B) and homozygous mutant coup-TFIIhu10330 (C) embryos, with no signs of edema or morphological abnormalities at 6 dpf. (D,E) flt4:mCit;flt1enh:tdTom-positive embryos from a coup-TFIIhu10330 incross. Compared with heterozygous siblings (D), coup-TFII mutants (E) show only marginal TD defects (asterisk) in a small proportion of embryos at 5 dpf. Note the overall normal vascular morphology with proper PCV, DA and ISVs present in homozygous mutants. Arrowheads indicate the presence of TD. (F) The average length of the TD in the first ten segments above the yolk extension is not significantly affected in coup-TFIIhu10330 mutants. (G) The proportion of venous ISVs is unaltered in homozygous coup-TFIIhu10330 mutants at 5 dpf. (H) Schematic overview of the Sox18 protein, indicating the first affected amino acid in the 1-bp insertion allele sox18hu10320, preceding the HMG-box. The resulting frame-shift leads to a premature stop codon after additional 50 amino acids. (I,J) Bright-field images of heterozygous (I) and homozygous (J) mutant sox18hu10320 embryos at 5 dpf. Note the overall wild-type appearance of mutant embryos (J). (K) The average length of the TD in wild-type (green), heterozygous (yellow) and homozygous mutant (red) sox18hu10320 embryos does not differ significantly. (L,M) sox18hu10320 heterozygous (L) and homozygous (M) mutant embryos expressing fli1a:eGFP in all ECs. In a sox18 loss-of-function situation, embryos do not show defects in the formation of the TD (arrowheads). (N) The average number of PLs present in sox18hu10320 mutants is not significantly affected at 54 hpf. Error bars represent s.d. n.s., not statistically significant.

Fig. 4.

Specification of the lymphatic lineage is independent of coup-TFII and sox18. (A) Schematic of the Coup-TFII protein indicating the position of the first affected amino acid in the 1 bp insertion allele nr2f2hu10330 (coup-TFIIhu10330 hereafter) which leads to a premature stop codon after an additional four amino acids N-terminal to the nuclear receptor-DNA binding and ligand-binding domain. (B,C) Brightfield images of wild-type (B) and homozygous mutant coup-TFIIhu10330 (C) embryos, with no signs of edema or morphological abnormalities at 6 dpf. (D,E) flt4:mCit;flt1enh:tdTom-positive embryos from a coup-TFIIhu10330 incross. Compared with heterozygous siblings (D), coup-TFII mutants (E) show only marginal TD defects (asterisk) in a small proportion of embryos at 5 dpf. Note the overall normal vascular morphology with proper PCV, DA and ISVs present in homozygous mutants. Arrowheads indicate the presence of TD. (F) The average length of the TD in the first ten segments above the yolk extension is not significantly affected in coup-TFIIhu10330 mutants. (G) The proportion of venous ISVs is unaltered in homozygous coup-TFIIhu10330 mutants at 5 dpf. (H) Schematic overview of the Sox18 protein, indicating the first affected amino acid in the 1-bp insertion allele sox18hu10320, preceding the HMG-box. The resulting frame-shift leads to a premature stop codon after additional 50 amino acids. (I,J) Bright-field images of heterozygous (I) and homozygous (J) mutant sox18hu10320 embryos at 5 dpf. Note the overall wild-type appearance of mutant embryos (J). (K) The average length of the TD in wild-type (green), heterozygous (yellow) and homozygous mutant (red) sox18hu10320 embryos does not differ significantly. (L,M) sox18hu10320 heterozygous (L) and homozygous (M) mutant embryos expressing fli1a:eGFP in all ECs. In a sox18 loss-of-function situation, embryos do not show defects in the formation of the TD (arrowheads). (N) The average number of PLs present in sox18hu10320 mutants is not significantly affected at 54 hpf. Error bars represent s.d. n.s., not statistically significant.

Homozygous coup-TFIIhu10330 mutants were viable beyond 6 dpf (Fig. 4B,C) and developed a normal blood vasculature without any sign of arterio-venous identity defects in the main trunk vessels. In addition, when scoring TD formation at 5 dpf, we observed only negligible defects and the majority of mutants were indistinguishable from siblings (Fig. 4D-F). As the ratio of intersegmental arteries and veins was also unaffected in coup-TFII mutant embryos (Fig. 4G), these results demonstrate that zygotically expressed coup-TFII is not essential for venous specification and lymphatic development.

Loss of sox18 does not interfere with lymphatic development

Another transcription factor that has been implicated in lymphatic development in mice and humans is Sox18 (Irrthum et al., 2003). Mice lacking functional Sox18 develop edema in certain genetic backgrounds (François et al., 2008) because of a failure in LEC development. Sox18 is expressed in a subset of cardinal vein cells that subsequently initiate Prox1 expression. In vitro experiments further showed that Sox18 can activate Prox1 expression by direct binding to its promoter region, placing Sox18 upstream of Prox1 during lymphatic specification (François et al., 2008). More recent morpholino knockdown studies as well as overexpression experiments employing a dominant-negative mouse Sox18 variant in zebrafish suggested a specific requirement for sox18 during sprouting of lymphatic-fated secondary sprouts from the PCV. In contrast to mouse, however, zebrafish sox18 is expressed in both the DA and PCV in a non-polarized fashion during venous sprouting (Cermenati et al., 2013), which is counterintuitive in the context of lymphatic specification.

We generated a mutant with a 1 bp insertion 5′ to the HMG-box encoding sequence (Fig. 4H). This allele is predicted to encode a truncated protein that contains neither the essential HMG nor the trans-activation domains (reviewed by Downes and Koopman, 2001), and as no alternative transcripts could be detected in mutants (data not shown) we consider it to represent a loss-of-function allele. Loss of Sox18 function did not lead to any obvious differences in the formation of arterial and venous ISVs, which is in line with previous morpholino knockdown data (Cermenati et al., 2008; Herpers et al., 2008; Pendeville et al., 2008; Cermenati et al., 2013). At 5 dpf, the overall appearance of homozygous sox18hu10320 mutants was indistinguishable from siblings (Fig. 4I,J) and when scored for the presence of the TD, no lymphatic or blood vascular phenotypes were detectable (Fig. 4K-M). Accordingly, the quantification of PLs at the horizontal myoseptum at 48 hpf did not reveal any significant differences (Fig. 4N), demonstrating that Sox18 is dispensable for venous sprouting and lymphatic cell fate determination.

Overexpression of prox1a in the venous endothelium does not affect endothelial cell behavior

It has been reported that overexpression of Prox1 in blood ECs in vitro is sufficient to shift their gene expression profile towards an LEC phenotype (Hong et al., 2002; Petrova et al., 2002). Similar results were also obtained from mouse models, where the forced expression of Prox1 in blood ECs led to an LEC-like gene expression pattern, severe edema formation, and embryonic lethality at E13.5 (Kim et al., 2010). Interestingly, the ability of Prox1 to transform blood into lymphatic ECs seems to be restricted to the venous endothelium in vivo (Kim et al., 2013), indicating that the additional expression of only one transcription factor is sufficient to differentiate a venous EC into an LEC. Although we showed here that mutations in the zebrafish orthologs of Prox1 and two other central genes that govern and maintain lymphatic commitment in mammals did not block lymphatic specification, we wondered whether prox1a expression might nevertheless be sufficient to force blood ECs into a lymphatic cell fate. Therefore, we aimed to mis-express prox1a in blood ECs and injected an UAS:prox1a construct into a flt4:Gal4FF driver-line (Fig. 5A) which drives expression from a 3.8 kb flt4 promoter fragment from Medaka (Deguchi et al., 2012). Upon transient injection of the UAS-construct, overexpression of prox1a transcripts was verified by whole-mount in situ hybridization (ISH). As expected, embryos injected with the UAS-construct expressed prox1a in a mosaic pattern in venous and arterial ECs at considerably higher levels than the endogenous endothelial expression in uninjected siblings at 32 hpf (Fig. 5B-E). We checked at 38 hpf (data not shown) and 48 hpf whether this prox1a mis-expression would have an influence on the behavior of positive ECs (marked by the co-expression of mRFP) during venous sprouting. However, neither the overall number of secondary sprouts nor the positioning of RFP-positive cells within the vasculature indicated a differential cell behavior upon prox1a overexpression (Fig. 5F,G,J,K). Furthermore, and in contrast to the mouse experiments, continuous mis-expression of prox1a did also not result in any vascular or lymphatic defects at later stages of lymphangiogenesis. In fact, at 5 dpf no differences in vascular morphology or in the expression pattern of the flt4:Gal4FF;UAS:GFP marker could be appreciated and cells expressing the construct ended up in all endothelial compartments (Fig. 5H,I,L,M; data not shown). Together, these results indicate that zebrafish prox1a is not only dispensable for lymphatic specification, but is also unable to commit ECs to a lymphatic fate.

Fig. 5.

Forced expression of prox1a does not commit venous ECs to a lymphatic phenotype. (A) Schematic of the prox1a overexpression construct and the flt4:Gal4FF line, which drives Gal4FF expression under the control of a Medaka 3.8 kb flt4 promoter fragment. (B-E) Whole-mount in situ hybridization against prox1a in uninjected flt4:Gal4FF siblings (B,C) and embryos injected with the prox1a mis-expression construct at 32 hpf (D,E). Note the domain of forced prox1a expression within the axial vessels in injected embryos (arrows in E), which does not reflect endogenous prox1a expression (C). Arrowheads in B,D highlight endogenous prox1a expression in the lateral line primordium; arrowhead in C points at a signal in the corpuscles of Stannius. (F-M) flt4:Gal4FF;UAS:eGFP wild-type siblings (F-I) and embryos injected with UAS:prox1a_IRES_mTurq-NLS-UAS:mRFP plasmid (J-M). At 2 dpf, forced expression of prox1a in ECs (marked by mRFP expression in J,K) does not lead to the emergence of ectopic secondary sprouts (F,G). At 5 dpf, UAS:prox1a-UAS:mRFP positive ECs are still evident in arterial and venous ECs without any signs of lymphatic or venous defects (L), suggesting that prox1a expression is not capable of influencing the cell fate and behavior of arterial and venous ECs (red in L,M). Arrows in F,J point at PLs. In H,L, arrows highlight the TD and ISLVs and white arrowheads in J-M mark exemplary ECs harboring the prox1a mis-expression construct. Note that the flt4:Gal4FF reporter line shows occasional expression in myotome cells as well as in a subset of neurons in the spinal cord. UIC, uninjected control.

Fig. 5.

Forced expression of prox1a does not commit venous ECs to a lymphatic phenotype. (A) Schematic of the prox1a overexpression construct and the flt4:Gal4FF line, which drives Gal4FF expression under the control of a Medaka 3.8 kb flt4 promoter fragment. (B-E) Whole-mount in situ hybridization against prox1a in uninjected flt4:Gal4FF siblings (B,C) and embryos injected with the prox1a mis-expression construct at 32 hpf (D,E). Note the domain of forced prox1a expression within the axial vessels in injected embryos (arrows in E), which does not reflect endogenous prox1a expression (C). Arrowheads in B,D highlight endogenous prox1a expression in the lateral line primordium; arrowhead in C points at a signal in the corpuscles of Stannius. (F-M) flt4:Gal4FF;UAS:eGFP wild-type siblings (F-I) and embryos injected with UAS:prox1a_IRES_mTurq-NLS-UAS:mRFP plasmid (J-M). At 2 dpf, forced expression of prox1a in ECs (marked by mRFP expression in J,K) does not lead to the emergence of ectopic secondary sprouts (F,G). At 5 dpf, UAS:prox1a-UAS:mRFP positive ECs are still evident in arterial and venous ECs without any signs of lymphatic or venous defects (L), suggesting that prox1a expression is not capable of influencing the cell fate and behavior of arterial and venous ECs (red in L,M). Arrows in F,J point at PLs. In H,L, arrows highlight the TD and ISLVs and white arrowheads in J-M mark exemplary ECs harboring the prox1a mis-expression construct. Note that the flt4:Gal4FF reporter line shows occasional expression in myotome cells as well as in a subset of neurons in the spinal cord. UIC, uninjected control.

Specification of the lymphatic lineage in mice is intimately linked to the restricted expression of the transcription factor Prox1 in a subpopulation of cardinal vein cells, and Prox1 function is essential for future LECs to migrate out of the cardinal vein (Yang et al., 2012). In the absence of mutants for Prox1 orthologs in other vertebrate species, a stringent assessment of conserved Prox1 function during lymphangiogenesis has been hampered. Nevertheless, expression studies in, for example, Xenopus (Ny et al., 2005) and zebrafish (Yaniv et al., 2006) suggest that there is evolutionary conservation.

Previous morpholino-knockdown strategies for zebrafish prox1a did not provide a fully conclusive picture and we had previously pointed out that different morpholinos directed against prox1a result in developmental anomalies, which we interpreted as being unspecific (Küchler et al., 2006). Hence, we generated a prox1a loss-of-function model to address conclusively the role of prox1-like genes for lymphangiogenesis in fish. Our results demonstrate that prox1a expression is not essential and also not sufficient for specification of LECs. Even the combined loss of prox1a and prox1b does not inhibit lymphatic development.

In combination with our finding that coup-TFII and sox18 are also not essential for lymphangiogenesis to occur, this suggests that lymphatic specification must be achieved in a different way and that the Coup-TFII/Sox18/Prox1 function has been evolving only in higher vertebrates or has been eliminated in the zebrafish/teleost lineage. How can one explain the strict requirement for Prox1 in mice, while in fish the Coup-TFII/Sox18/Prox1 code seems dispensable?

In mice, angiogenesis and lymphangiogenesis are two temporally separated processes. Initially, starting at E8.0, a network of intersegmental arteries and veins is formed by angiogenesis (Fig. 6A1) (Walls et al., 2008). In a second step, a subpopulation of cardinal vein cells is specified to the lymphatic lineage by Prox1 expression at E9.5 (Fig. 6A2), making only those cells responsive to the VegfC signal. These lymphatic precursors become motile then and leave the epithelial layer of the vein to form the first lymphatic structures (Fig. 6A3,4).

Fig. 6.

Chronology of angiogenic and lymphangiogenic events in mice and zebrafish. (A) In mice, angiogenic sprouting from the DA and cardinal vein starts at E8.0 and leads to the formation of intersegmental arteries and veins (A1). Subsequently, a subpopulation of venous ECs is specified to the lymphatic lineage by the restricted expression of Prox1 (green cells; A2). These lymphatic precursor cells then start to leave the venous epithelium in a dorsal direction (A3), where they will eventually form transient lymphatic structures comprising the primordial TD and the dorsal peripheral longitudinal lymphatic vessel (Hägerling et al., 2013). (B) In contrast to the situation in mice, zebrafish angiogenic cell behavior is initially restricted to arterial ECs, which form ISVs in the trunk (B1). In a second step, venous angiogenesis and lymphangiogenesis take place simultaneously and both processes depend on the same genes (vegfc, flt4, ccbe1). All emerging sprouts from the PCV initially migrate dorsally but shortly thereafter they display two different cell behaviors, reflecting the first visible signs of differential cell fate among the sprouts: some of them will connect to intersegmental arteries (venous fated cells) and others proceed towards the horizontal myoseptum (lymphatic fated cells), often even passing an ISV on their route (B2, dotted arrows). As a result, both a set of intersegmental veins and a pool of parachordal lymphangioblast at the horizontal myoseptum are formed (B3). The latter will subsequently migrate away from this region, to form the initial lymphatic structures in the trunk (B4).

Fig. 6.

Chronology of angiogenic and lymphangiogenic events in mice and zebrafish. (A) In mice, angiogenic sprouting from the DA and cardinal vein starts at E8.0 and leads to the formation of intersegmental arteries and veins (A1). Subsequently, a subpopulation of venous ECs is specified to the lymphatic lineage by the restricted expression of Prox1 (green cells; A2). These lymphatic precursor cells then start to leave the venous epithelium in a dorsal direction (A3), where they will eventually form transient lymphatic structures comprising the primordial TD and the dorsal peripheral longitudinal lymphatic vessel (Hägerling et al., 2013). (B) In contrast to the situation in mice, zebrafish angiogenic cell behavior is initially restricted to arterial ECs, which form ISVs in the trunk (B1). In a second step, venous angiogenesis and lymphangiogenesis take place simultaneously and both processes depend on the same genes (vegfc, flt4, ccbe1). All emerging sprouts from the PCV initially migrate dorsally but shortly thereafter they display two different cell behaviors, reflecting the first visible signs of differential cell fate among the sprouts: some of them will connect to intersegmental arteries (venous fated cells) and others proceed towards the horizontal myoseptum (lymphatic fated cells), often even passing an ISV on their route (B2, dotted arrows). As a result, both a set of intersegmental veins and a pool of parachordal lymphangioblast at the horizontal myoseptum are formed (B3). The latter will subsequently migrate away from this region, to form the initial lymphatic structures in the trunk (B4).

In fish, however, the timing of events is different: angiogenesis begins with a first wave of VegfA- and Vegfr2-governed sprouting from the DA, giving rise to a set of intersegmental arteries (Fig. 6B1). Subsequently, a second set of sprouts arises exclusively from the PCV (Fig. 6B2). The emergence of these venous sprouts that eventually will give rise to both intersegmental veins and lymphatic precursors, depends on the Vegfc/Flt4 pathway and on Ccbe1, indicating that in zebrafish the mechanisms that drive the initial migration of both subpopulations is identical. This might also explain why genes that at some point specifically mark LECs in mice (such as Flt4 or Lyve1) are expressed within the whole venous endothelium in zebrafish. It is only slightly later, when secondary sprouts reach the level of the arterial ISVs that both populations display a differential behavior, suggesting that only from this point in time are lymphatic and venous fates separating (Fig. 6B3,4). It is therefore plausible that it is not an intrinsic mechanism but rather external cues, possibly provided by the ISVs themselves, that control which sprout will connect to an ISV and which will migrate further to the horizontal myoseptum. In fact, the only known signaling pathway so far that has been shown to affect the cell fate decision made by secondary sprouts is Dll4/Notch. Knockdown of dll4 leads to a dramatically enhanced number of venous ISVs and only a small number of PL cells, suggesting that the fate of secondary sprouts is strongly shifted towards venous identity (Geudens et al., 2010). Whether this requirement for Notch signaling is artery intrinsic (dll4 and notch1b are both expressed in arterial ECs) or reflects a signaling interaction between ISV and secondary sprouts requires further analysis, but in either case arterial ISVs directly or indirectly influence the fate decisions made by venous sprouts.

In summary, the scheme depicted in Fig. 6 offers an explanation as to why the Coup-TFII/Sox18/Prox1 signaling axis is required in mice, but dispensable in zebrafish: in mice (and probably other vertebrate classes) Prox1 function specifies LEC fate within a subset of venous ECs within the cardinal vein. In zebrafish (and probably all teleosts), this specification step is not required within the endothelial epithelium of the cardinal vein. Our findings therefore reveal an astonishing difference in the earliest step of lymphangiogenesis between vertebrates. In the absence of a full understanding of the exact point in time when LECs become specified in zebrafish, it remains difficult to appreciate fully the degree of similarity between mice and other vertebrates at the level of genetic control and cellular behavior. However, other steps of lymphangiogenesis, such as the migration of future LECs away from the major vein, are conserved in vertebrates, as evidenced by the conserved functions of vegfc (Karkkainen et al., 2004; Ny et al., 2008; Villefranc et al., 2013), flt4 (Jeltsch et al., 1997; Ny et al., 2008; Hogan et al., 2009b) and ccbe1 (Hogan et al., 2009a; Bos et al., 2011).

Zebrafish husbandry

Strains were maintained under standard husbandry conditions. Animal experiments have been performed according to the rules of the Animal Experimentation Committee (DEC) of the KNAW and the A*STAR Biological Resource Centre Institutional Animal Care and Use Committee (IACUC Project #110638). The following published transgenic lines have been employed in this study: Tg(flt1enh:tdTomato) (Bussmann et al., 2010), Tg(fli1a:eGFP)y1 (Lawson and Weinstein, 2002), Tg(kdrl:HRAS-mCherry)s916 (Hogan et al., 2009a).

Transgenesis

The Tg(flt4BAC:mCitrine)hu7135 line was generated from BAC DKEY-58G10 and the Tg(prox1aBAC:KalTA4-4xUAS-E1b:uncTagRFP)nim5 from BAC DKEY-5J3 following standard BAC recombineering procedures (Bussmann and Schulte-Merker, 2011). For the generation of the flt4 promoter construct Tg(flt4:Gal4FF)hu9236, a previously reported 3.8-kb promoter fragment (Deguchi et al., 2012) was amplified from Medaka genomic DNA (supplementary material Table S1) and cloned into the miniTol2 vector 5′ of the Gal4FF coding sequence. In case of the UAS:prox1a mis-expression construct, the prox1a cDNA was placed behind a 5xUAS cassette in the pT2A vector followed by an IRES sequence and a mTurquoise-NLS cassette including PolyA sequences. Because mTurq-NLS expression behind the IRES turned out to be very low, an additional 5xUAS:mRFP cassette was inserted into the plasmid to identify Gal4FF-positive cells harboring the prox1a overexpression construct after injection. BAC DNA (100 pg/embryo) or plasmids (25 pg/embryo) were co-injected with Tol2 transposase mRNA (25 pg/embryo) into one-cell-stage embryos and the progeny was screened for germline transmission.

Genome editing by zinc-finger nuclease and transcription activator-like effector nucleases (TALENs)

For the generation of prox1a mutants, plasmids encoding zinc-finger nucleases targeting the locus were obtained from ToolGen (Korea). The zinc-finger target sites in the first exon of prox1a were: 5′-TGAGATGGAGAG-3′ and 5′-GGTCATGGAGGG-3′ (supplementary material Fig. S3). TALEN-mediated genome editing for the generation of coup-TFII and sox18 mutants was performed as described (Cermak et al., 2011; Bedell et al., 2012). The TALEN binding sites in sox18 exon1 were: TAL1, 5′-TGCCTGGGTCTGGAAC-3′; TAL2, 5′-TGGCCTCCGCTGCTGTT-3′. For coup-TFII, the TALEN recognition sites were TAL1, 5′-TCCGACCCCTCAGACACCCGT-3′ and TAL2, 5′-AACAATAACAACACACAGTCA-3′.

Genotyping

The prox1ai278, sox18hu10320 as well as the coup-TFII allele nr2f2hu10330 were genotyped by KASPAR using the primers indicated in supplementary material Table S1. KASPAR genotyping of the prox1bSA0035 allele was performed as described (Tao et al., 2011).

Immunohistochemistry and in situ hybridization (ISH)

Antibody staining using the following antibodies was performed as described (Elworthy et al., 2008): mAb F59 [anti-slow myosin heavy chain-1, Developmental Studies Hybridoma Bank (DSHB); 1:100], rabbit anti-Prox1 (AB5475, Chemicon, USA; 1:5000), goat anti-rabbit Alexa488 and goat anti-mouse Alexa546 (Invitrogen; 1:1000). ISH was carried out as described previously (Schulte-Merker, 2002).

Microscopy

Confocal imaging was performed on living embryos embedded laterally (unless otherwise stated) in 0.5-1% low melting point agarose (Invitrogen) on Leica SPE and SP8 microscopes. Brightfield images were taken on an Olympus SZX16 Stereomicroscope. Images were processed using Adobe Photoshop CS5.1 and Fiji (http://fiji.sc/Fiji). Stitching of composite pictures was performed using Leica LAS AF software or Adobe Illustrator CS5.1.

We thank M. Witte and Audrey Iyu for technical assistance; D. Schulte, S. van de Pavert and F. Kiefer for critical comments on the manuscript; and A. de Graaff and the Hubrecht Imaging Centre for supporting the imaging.

Author contributions

A.v.I. generated the flt4 transgenic lines, analyzed the prox1a/b mutants and the prox1a reporter line and wrote the manuscript. Z.Z., D.M.A.H. and M.G.R. generated and analyzed new mutant alleles, in part using TALEN constructs generated by J.P.-M. J.C.F. generated the prox1a reporter line. H.D., E.A.O., P.W.I. and S.S.-M. conceived and supervised the study and edited the manuscript.

Funding

This work was supported by a Marie Curie Intra-European Fellowship award (to A.v.I.); a VENI grant from the Netherlands Organisation for Scientific Research (NWO) [863.11.022 to M.G.R.]; the Agency for Science Technology and Research (A*STAR) (Z.Z. and P.W.I.); and a Medical Research Council grant [U117581329 to J.F. and E.A.O.]. Deposited in PMC for release after 6 months.

Aranguren
X. L.
,
Beerens
M.
,
Vandevelde
W.
,
Dewerchin
M.
,
Carmeliet
P.
,
Luttun
A.
(
2011
).
Transcription factor COUP-TFII is indispensable for venous and lymphatic development in zebrafish and Xenopus laevis
.
Biochem. Biophys. Res. Commun.
410
,
121
126
.
Bedell
V. M.
,
Wang
Y.
,
Campbell
J. M.
,
Poshusta
T. L.
,
Starker
C. G.
,
Krug
R. G.
II
,
Tan
W.
,
Penheiter
S. G.
,
Ma
A. C.
,
Leung
A. Y.
, et al. 
. (
2012
).
In vivo genome editing using a high-efficiency TALEN system
.
Nature
491
,
114
118
.
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 essential for mammalian lymphatic vascular development and enhances the lymphangiogenic effect of vascular endothelial growth factor-C in vivo
.
Circ. Res.
109
,
486
491
.
Bussmann
J.
,
Schulte-Merker
S.
(
2011
).
Rapid BAC selection for tol2-mediated transgenesis in zebrafish
.
Development
138
,
4327
4332
.
Bussmann
J.
,
Bos
F. L.
,
Urasaki
A.
,
Kawakami
K.
,
Duckers
H. J.
,
Schulte-Merker
S.
(
2010
).
Arteries provide essential guidance cues for lymphatic endothelial cells in the zebrafish trunk
.
Development
137
,
2653
2657
.
Cermak
T.
,
Doyle
E. L.
,
Christian
M.
,
Wang
L.
,
Zhang
Y.
,
Schmidt
C.
,
Baller
J. A.
,
Somia
N. V.
,
Bogdanove
A. J.
,
Voytas
D. F.
(
2011
).
Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting
.
Nucleic Acids Res.
39
,
e82
.
Cermenati
S.
,
Moleri
S.
,
Cimbro
S.
,
Corti
P.
,
Del Giacco
L.
,
Amodeo
R.
,
Dejana
E.
,
Koopman
P.
,
Cotelli
F.
,
Beltrame
M.
(
2008
).
Sox18 and Sox7 play redundant roles in vascular development
.
Blood
111
,
2657
2666
.
Cermenati
S.
,
Moleri
S.
,
Neyt
C.
,
Bresciani
E.
,
Carra
S.
,
Grassini
D. R.
,
Omini
A.
,
Goi
M.
,
Cotelli
F.
,
François
M.
, et al. 
. (
2013
).
Sox18 genetically interacts with VegfC to regulate lymphangiogenesis in zebrafish
.
Arterioscler. Thromb. Vasc. Biol.
33
,
1238
1247
.
Deguchi
T.
,
Fujimori
K. E.
,
Kawasaki
T.
,
Maruyama
K.
,
Yuba
S.
(
2012
).
In vivo visualization of the lymphatic vessels in pFLT4-EGFP transgenic medaka
.
Genesis
50
,
625
634
.
Del Giacco
L.
,
Pistocchi
A.
,
Ghilardi
A.
(
2010
).
prox1b Activity is essential in zebrafish lymphangiogenesis
.
PLoS ONE
5
,
e13170
.
Distel
M.
,
Wullimann
M. F.
,
Köster
R. W.
(
2009
).
Optimized Gal4 genetics for permanent gene expression mapping in zebrafish
.
Proc. Natl. Acad. Sci. USA
106
,
13365
13370
.
Downes
M.
,
Koopman
P.
(
2001
).
SOX18 and the transcriptional regulation of blood vessel development
.
Trends Cardiovasc. Med.
11
,
318
324
.
Elworthy
S.
,
Hargrave
M.
,
Knight
R.
,
Mebus
K.
,
Ingham
P. W.
(
2008
).
Expression of multiple slow myosin heavy chain genes reveals a diversity of zebrafish slow twitch muscle fibres with differing requirements for Hedgehog and Prdm1 activity
.
Development
135
,
2115
2126
.
François
M.
,
Caprini
A.
,
Hosking
B.
,
Orsenigo
F.
,
Wilhelm
D.
,
Browne
C.
,
Paavonen
K.
,
Karnezis
T.
,
Shayan
R.
,
Downes
M.
, et al. 
. (
2008
).
Sox18 induces development of the lymphatic vasculature in mice
.
Nature
456
,
643
647
.
Geudens
I.
,
Herpers
R.
,
Hermans
K.
,
Segura
I.
,
Ruiz de Almodovar
C.
,
Bussmann
J.
,
De Smet
F.
,
Vandevelde
W.
,
Hogan
B. M.
,
Siekmann
A.
, et al. 
. (
2010
).
Role of delta-like-4/Notch in the formation and wiring of the lymphatic network in zebrafish
.
Arterioscler. Thromb. Vasc. Biol.
30
,
1695
1702
.
Glasgow
E.
,
Tomarev
S. I.
(
1998
).
Restricted expression of the homeobox gene prox 1 in developing zebrafish
.
Mech. Dev.
76
,
175
178
.
Hägerling
R.
,
Pollmann
C.
,
Andreas
M.
,
Schmidt
C.
,
Nurmi
H.
,
Adams
R. H.
,
Alitalo
K.
,
Andresen
V.
,
Schulte-Merker
S.
,
Kiefer
F.
(
2013
).
A novel multistep mechanism for initial lymphangiogenesis in mouse embryos based on ultramicroscopy
.
EMBO J.
32
,
629
644
.
Harvey
N. L.
,
Srinivasan
R. S.
,
Dillard
M. E.
,
Johnson
N. C.
,
Witte
M. H.
,
Boyd
K.
,
Sleeman
M. W.
,
Oliver
G.
(
2005
).
Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity
.
Nat. Genet.
37
,
1072
1081
.
Harvey
S. A.
,
Sealy
I.
,
Kettleborough
R.
,
Fenyes
F.
,
White
R.
,
Stemple
D.
,
Smith
J. C.
(
2013
).
Identification of the zebrafish maternal and paternal transcriptomes
.
Development
140
,
2703
2710
.
Herpers
R.
,
van de Kamp
E.
,
Duckers
H. J.
,
Schulte-Merker
S.
(
2008
).
Redundant roles for sox7 and sox18 in arteriovenous specification in zebrafish
.
Circ. Res.
102
,
12
15
.
Hogan
B. M.
,
Bos
F. L.
,
Bussmann
J.
,
Witte
M.
,
Chi
N. C.
,
Duckers
H. J.
,
Schulte-Merker
S.
(
2009a
).
Ccbe1 is required for embryonic lymphangiogenesis and venous sprouting
.
Nat. Genet.
41
,
396
398
.
Hogan
B. M.
,
Herpers
R.
,
Witte
M.
,
Heloterä
H.
,
Alitalo
K.
,
Duckers
H. J.
,
Schulte-Merker
S.
(
2009b
).
Vegfc/Flt4 signalling is suppressed by Dll4 in developing zebrafish intersegmental arteries
.
Development
136
,
4001
4009
.
Hong
Y. K.
,
Harvey
N.
,
Noh
Y. H.
,
Schacht
V.
,
Hirakawa
S.
,
Detmar
M.
,
Oliver
G.
(
2002
).
Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate
.
Dev. Dyn.
225
,
351
357
.
Irrthum
A.
,
Devriendt
K.
,
Chitayat
D.
,
Matthijs
G.
,
Glade
C.
,
Steijlen
P. M.
,
Fryns
J. P.
,
Van Steensel
M. A.
,
Vikkula
M.
(
2003
).
Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia
.
Am. J. Hum. Genet.
72
,
1470
1478
.
Jeltsch
M.
,
Kaipainen
A.
,
Joukov
V.
,
Meng
X.
,
Lakso
M.
,
Rauvala
H.
,
Swartz
M.
,
Fukumura
D.
,
Jain
R. K.
,
Alitalo
K.
(
1997
).
Hyperplasia of lymphatic vessels in VEGF-C transgenic mice
.
Science
276
,
1423
1425
.
Johnson
N. C.
,
Dillard
M. E.
,
Baluk
P.
,
McDonald
D. M.
,
Harvey
N. L.
,
Frase
S. L.
,
Oliver
G.
(
2008
).
Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity
.
Genes Dev.
22
,
3282
3291
.
Kaipainen
A.
,
Korhonen
J.
,
Mustonen
T.
,
van Hinsbergh
V. W.
,
Fang
G. H.
,
Dumont
D.
,
Breitman
M.
,
Alitalo
K.
(
1995
).
Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development
.
Proc. Natl. Acad. Sci. USA
92
,
3566
3570
.
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
).
Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins
.
Nat. Immunol.
5
,
74
80
.
Karpanen
T.
,
Schulte-Merker
S.
(
2011
).
Zebrafish provides a novel model for lymphatic vascular research
.
Methods Cell Biol.
105
,
223
238
.
Kim
H.
,
Nguyen
V. P.
,
Petrova
T. V.
,
Cruz
M.
,
Alitalo
K.
,
Dumont
D. J.
(
2010
).
Embryonic vascular endothelial cells are malleable to reprogramming via Prox1 to a lymphatic gene signature
.
BMC Dev. Biol.
10
,
72
.
Kim
H.
,
Cruz
M.
,
Bourdeau
A.
,
Dumont
D. J.
(
2013
).
Cell-cell interactions influence vascular reprogramming by Prox1 during embryonic development
.
PLoS ONE
8
,
e52197
.
Koltowska
K.
,
Betterman
K. L.
,
Harvey
N. L.
,
Hogan
B. M.
(
2013
).
Getting out and about: the emergence and morphogenesis of the vertebrate lymphatic vasculature
.
Development
140
,
1857
1870
.
Küchler
A. M.
,
Gjini
E.
,
Peterson-Maduro
J.
,
Cancilla
B.
,
Wolburg
H.
,
Schulte-Merker
S.
(
2006
).
Development of the zebrafish lymphatic system requires VEGFC signaling
.
Curr. Biol.
16
,
1244
1248
.
Lawson
N. D.
,
Weinstein
B. M.
(
2002
).
In vivo imaging of embryonic vascular development using transgenic zebrafish
.
Dev. Biol.
248
,
307
318
.
Lee
S.
,
Kang
J.
,
Yoo
J.
,
Ganesan
S. K.
,
Cook
S. C.
,
Aguilar
B.
,
Ramu
S.
,
Lee
J.
,
Hong
Y. K.
(
2009
).
Prox1 physically and functionally interacts with COUP-TFII to specify lymphatic endothelial cell fate
.
Blood
113
,
1856
1859
.
Ny
A.
,
Koch
M.
,
Schneider
M.
,
Neven
E.
,
Tong
R. T.
,
Maity
S.
,
Fischer
C.
,
Plaisance
S.
,
Lambrechts
D.
,
Héligon
C.
, et al. 
. (
2005
).
A genetic Xenopus laevis tadpole model to study lymphangiogenesis
.
Nat. Med.
11
,
998
1004
.
Ny
A.
,
Koch
M.
,
Vandevelde
W.
,
Schneider
M.
,
Fischer
C.
,
Diez-Juan
A.
,
Neven
E.
,
Geudens
I.
,
Maity
S.
,
Moons
L.
, et al. 
. (
2008
).
Role of VEGF-D and VEGFR-3 in developmental lymphangiogenesis, a chemicogenetic study in Xenopus tadpoles
.
Blood
112
,
1740
1749
.
Okuda
K. S.
,
Astin
J. W.
,
Misa
J. P.
,
Flores
M. V.
,
Crosier
K. E.
,
Crosier
P. S.
(
2012
).
lyve1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish
.
Development
139
,
2381
2391
.
Pendeville
H.
,
Winandy
M.
,
Manfroid
I.
,
Nivelles
O.
,
Motte
P.
,
Pasque
V.
,
Peers
B.
,
Struman
I.
,
Martial
J. A.
,
Voz
M. L.
(
2008
).
Zebrafish Sox7 and Sox18 function together to control arterial-venous identity
.
Dev. Biol.
317
,
405
416
.
Petrova
T. V.
,
Mäkinen
T.
,
Mäkelä
T. P.
,
Saarela
J.
,
Virtanen
I.
,
Ferrell
R. E.
,
Finegold
D. N.
,
Kerjaschki
D.
,
Ylä-Herttuala
S.
,
Alitalo
K.
(
2002
).
Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor
.
EMBO J.
21
,
4593
4599
.
Pistocchi
A.
,
Feijóo
C. G.
,
Cabrera
P.
,
Villablanca
E. J.
,
Allende
M. L.
,
Cotelli
F.
(
2009
).
The zebrafish prospero homolog prox1 is required for mechanosensory hair cell differentiation and functionality in the lateral line
.
BMC Dev. Biol.
9
,
58
.
Schulte-Merker
S.
(
2002
).
Looking at embryos
. In
Zebrafish: A Practical Approach
, Vol.
261
(ed.
Nusslein-Volhard
C.
), pp.
39
58
.
New York, NY
:
Oxford University Press
.
Schulte-Merker
S.
,
Sabine
A.
,
Petrova
T. V.
(
2011
).
Lymphatic vascular morphogenesis in development, physiology, and disease
.
J. Cell Biol.
193
,
607
618
.
Srinivasan
R. S.
,
Oliver
G.
(
2011
).
Prox1 dosage controls the number of lymphatic endothelial cell progenitors and the formation of the lymphovenous valves
.
Genes Dev.
25
,
2187
2197
.
Srinivasan
R. S.
,
Dillard
M. E.
,
Lagutin
O. V.
,
Lin
F. J.
,
Tsai
S.
,
Tsai
M. J.
,
Samokhvalov
I. M.
,
Oliver
G.
(
2007
).
Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature
.
Genes Dev.
21
,
2422
2432
.
Srinivasan
R. S.
,
Geng
X.
,
Yang
Y.
,
Wang
Y.
,
Mukatira
S.
,
Studer
M.
,
Porto
M. P.
,
Lagutin
O.
,
Oliver
G.
(
2010
).
The nuclear hormone receptor Coup-TFII is required for the initiation and early maintenance of Prox1 expression in lymphatic endothelial cells
.
Genes Dev.
24
,
696
707
.
Tammela
T.
,
Alitalo
K.
(
2010
).
Lymphangiogenesis: Molecular mechanisms and future promise
.
Cell
140
,
460
476
.
Tao
S.
,
Witte
M.
,
Bryson-Richardson
R. J.
,
Currie
P. D.
,
Hogan
B. M.
,
Schulte-Merker
S.
(
2011
).
Zebrafish prox1b mutants develop a lymphatic vasculature, and prox1b does not specifically mark lymphatic endothelial cells
.
PLoS ONE
6
,
e28934
.
Thisse
C.
,
Thisse
B.
(
2005
).
High Throughput Expression Analysis of ZF-Models Consortium Clones. ZFIN Direct Data Submission
(http://zfin.org)
Villefranc
J. A.
,
Nicoli
S.
,
Bentley
K.
,
Jeltsch
M.
,
Zarkada
G.
,
Moore
J. C.
,
Gerhardt
H.
,
Alitalo
K.
,
Lawson
N. D.
(
2013
).
A truncation allele in vascular endothelial growth factor c reveals distinct modes of signaling during lymphatic and vascular development
.
Development
140
,
1497
1506
.
Walls
J. R.
,
Coultas
L.
,
Rossant
J.
,
Henkelman
R. M.
(
2008
).
Three-dimensional analysis of vascular development in the mouse embryo
.
PLoS ONE
3
,
e2853
.
Wigle
J. T.
,
Harvey
N.
,
Detmar
M.
,
Lagutina
I.
,
Grosveld
G.
,
Gunn
M. D.
,
Jackson
D. G.
,
Oliver
G.
(
2002
).
An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype
.
EMBO J.
21
,
1505
1513
.
Yang
Y.
,
García-Verdugo
J. M.
,
Soriano-Navarro
M.
,
Srinivasan
R. S.
,
Scallan
J. P.
,
Singh
M. K.
,
Epstein
J. A.
,
Oliver
G.
(
2012
).
Lymphatic endothelial progenitors bud from the cardinal vein and intersomitic vessels in mammalian embryos
.
Blood
120
,
2340
2348
.
Yaniv
K.
,
Isogai
S.
,
Castranova
D.
,
Dye
L.
,
Hitomi
J.
,
Weinstein
B. M.
(
2006
).
Live imaging of lymphatic development in the zebrafish
.
Nat. Med.
12
,
711
716
.
You
L. R.
,
Lin
F. J.
,
Lee
C. T.
,
DeMayo
F. J.
,
Tsai
M. J.
,
Tsai
S. Y.
(
2005
).
Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity
.
Nature
435
,
98
104
.

Competing interests

The authors declare no competing financial interests.

Supplementary information