ArticlePDF Available

Mafba is a downstream transcriptional effector of Vegfc signaling essential for embryonic lymphangiogenesis in zebrafish

Authors:

Abstract and Figures

The lymphatic vasculature plays roles in tissue fluid balance, immune cell trafficking, fatty acid absorption, cancer metastasis, and cardiovascular disease. Lymphatic vessels form by lymphangiogenesis, the sprouting of new lymphatics from pre-existing vessels, in both development and disease contexts. The apical signaling pathway in lymphangiogenesis is the VEGFC/VEGFR3 pathway, yet how signaling controls cellular transcriptional output remains unknown. We used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for lymphatic vessel development. We found that mafba is required for the migration of lymphatic precursors after their initial sprouting from the posterior cardinal vein. mafba expression is enriched in sprouts emerging from veins, and we show that mafba functions cell-autonomously during lymphatic vessel development. Mechanistically, Vegfc signaling increases mafba expression to control downstream transcription, and this regulatory relationship is dependent on the activity of SoxF transcription factors, which are essential for mafba expression in venous endothelium. Here we identify an indispensable Vegfc-SoxF-Mafba pathway in lymphatic development. © 2015 Koltowska et al.; Published by Cold Spring Harbor Laboratory Press.
The uq 4bh mutant fails to form a lymphatic vasculature. (A) Vascular nuclei [Tg(fli1a: nlsEGFP); green] and veins and lymphatic vessels [Tg(lyve1:DsRed); white] in the trunk of sibling (top panels) and mutant (bottom panels) embryos at 5 dpf. (TD) Thoracic duct; (DLLV) dorsal longitudinal lymphatic vessel; (ISLV) intersegmental lymphatic vessel. Arrows indicate lymphatics, and asterisks indicate their absence. Bars, 50 μm. (B) Gross morphology of sibling (top) and mutant (bottom) embryos at 5 dpf. (C) Quantification of total LEC number using the overlay of transgenic marker expression from A. Mean ± SEM; scored siblings, n = 15; uq 4bh , n = 18; ttest. ( * * * * ) P < 0.0001. (D) The facial lymphatic network in sibling (top) and mutant (bottom) embryos at 5 dpf. (LFL) Lateral facial lymphatic; (OLV) otolithic lymphatic; (LAA) branchial arch lymphatics; (MFL) medial facial lymphatics. Bars, 50 μm. (E) Quantification of facial lymphatic cell numbers in individual embryos using the overlay of transgenic marker expression from A. Mean ± SEM; scored siblings, n = 6; uq 4bh , n = 8; t-test. ( * ) P < 0.05. (F ) PLs in the horizontal myoseptum in sibling (top) and mutant (bottom) embryos. Bars, 30 μm. (G) Quantification of PL numbers in individual embryos using the overlay of transgenic marker expression from A. Mean ± SEM; scored siblings, n = 10; uq 4bh , n = 12; t-test. (ns) No significant difference. (H) prox1a-expressing LECs as they emerge from the PCV at 36 h post-fertilization (hpf) in sibling (top) and mutant (bottom) embryos labeled by Tg(prox1a:Kalt4-4xUAS:uncTagRFP);Tg (10xUAS:Venus). (Green) α-GFP. Bars, 30 μm. (I) Quantification of prox1a-expressing LECs in individual embryos at 36 hpf. Mean ± SEM; scored siblings, n = 16; uq 4bh , n = 15; t-test. (ns) No significant difference.
… 
A cell-autonomous function for mafba in lymphatic vascular development. (A) Transplantation of Tg(fli1a:EGFP)-labeled (green) and dextran blue-labeled (blue) donor cells into a Tg(lyve1b:DsRed) (red) host derived from a mafba uq4bh in-cross. (A ′ ,A ′′ ) Wild-type ECs were observed to contribute to arteries (AECs), veins (VECs), and lymphatics (LECs) (white arrows) in sibling and mutant recipients. The boxed areas indicate high-power regions (shown at right). (B) Quantification of endothelial contributions in A ′ and A ′′. Percentage contributions of a cell to AECs, VECs, and LECs for all vascular grafted embryos (n = 11 wild type into sibling embryos with vascular grafts; n = 13 wild type into mutant embryos with vascular grafts). (C ) Transplantation of Tg(lyve1b:DsRed)-labeled (red), Tg (kdrl:EGFP)-labeled (green), and dextran bluelabeled (blue) donor cells derived from a mafba uq4bh in-cross into an unlabeled wild-type host. Genotyping confirmed donor identity. (C ′ ) Sibling ECs contributed to AECs, VECs, and LECs (arrows). (C ′′ ) Mutant ECs contributed AECs (n = 12/12 grafts) and VECs (n = 3/12 grafts) but not LECs. The boxed areas indicate highpower images (shown at right). (D) Quantification of endothelial contributions in C ′ and C ′′. Percentage contributions to AECs, VECs, and LECs for all vascular grafts. One mutant graft was observed with lymphatic vascular morphology but expressed Tg(lyve1b:DsRed) and Tg (kdrl:EGFP), indicative of a differentiation defect (see Supplemental Fig. 3B). Bars, 30 μm.
… 
Content may be subject to copyright.
mafba is a downstream transcriptional
effector of Vegfc signaling essential
for embryonic lymphangiogenesis
in zebrafish
Katarzyna Koltowska,
1
Scott Paterson,
1
Neil I. Bower,
1
Gregory J. Baillie,
1
Anne K. Lagendijk,
1
Jonathan W. Astin,
2
Huijun Chen,
1
Mathias Francois,
1
Philip S. Crosier,
2
Ryan J. Taft,
1
Cas Simons,
1
Kelly A. Smith,
1
and Benjamin M. Hogan
1
1
Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia,
Brisbane, Queensland 4072, Australia;
2
Department of Molecular Medicine and Pathology, School of MedicalSciences, University
of Auckland, Auckland 1023, New Zealand
The lymphatic vasculature plays roles in tissue fluid balance, immune cell trafficking, fatty acid absorption, cancer
metastasis, and cardiovascular disease. Lymphatic vessels form by lymphangiogenesis, the sprouting of new lym-
phatics from pre-existing vessels, in both development and disease contexts. The apical signaling pathway in lym-
phangiogenesis is the VEGFC/VEGFR3 pathway, yet how signaling controls cellular transcriptional output remains
unknown. We used a forward genetic screen in zebrafish to identify the transcription factor mafba as essential for
lymphatic vessel development. We found that mafba is required for the migration of lymphatic precursors after their
initial sprouting from the posterior cardinal vein. mafba expression is enriched in sprouts emerging from veins, and
we show that mafba functions cell-autonomously during lymphatic vessel development. Mechanistically, Vegfc
signaling increases mafba expression to control downstream transcription, and this regulatory relationship is de-
pendent on the activity of SoxF transcription factors, which are essential for mafba expression in venous endothe-
lium. Here we identify an indispensable VegfcSoxFMafba pathway in lymphatic development.
[Keywords: lymphatic; vascular; Mafb; Vegfc; Sox18; Sox7; zebrafish]
Supplemental material is available for this article.
Received April 2, 2015; revised version accepted July 1, 2015.
Lymphangiogenesis is the formation of new lymphatic
vessels from pre-existing vessels. Lymphangiogenesis
plays integral roles in cancer metastasis, lymphedema,
and cardiovascular disease (Lim et al. 2013; Martel et al.
2013; Stacker et al. 2014). Common molecular pathways
control lymphangiogenesis in development and disease,
and much of our current understanding has come from
the study of embryogenesis. In the embryo, the lymphatic
vasculature derives chiefly from pre-existing veins
through a process involving cellular transdifferentiation,
migration, and proliferation and vessel morphogenesis
(Oliver and Srinivasan 2010; Koltowska et al. 2013).
Lymphangiogenesis is dependent on VEGFR3 signaling
in all known contexts, including during development.
Knockout mice for Vegfr3 are embryonic-lethal due to car-
diovascular failure (Dumont et al. 1998), and heterozy-
gous mutation of Vegfr3 in the Chy mouse model leads
to lymphedema and lymphatic vascular defects (Karkkai-
nen et al. 2001). Furthermore, the transgenic overexpres-
sion of a soluble inhibitory (ligand trap) form of VEGFR3
disrupts tissue lymphangiogenesis (Makinen et al. 2001).
Mouse mutants for Vegfc fail to form the earliest lymphat-
ic sprouts from embryonic veins, and, indicative of the
instructive role for VEGFC, overexpression of this ligand
promotes ectopic tissue lymphangiogenesis (Jeltsch et
al. 1997; Karkkainen et al. 2004; Hagerling et al. 2013).
In humans, the VEGFC/VEGFR3 pathway also controls
lymphatic vessel development, and patients with muta-
tions in either VEGFR3 or VEGFC develop primary
lymphedema in familial Milroys disease or Milroys-
like lymphedema, respectively (Irrthum et al. 2000; Kark-
kainen et al. 2000; Gordon et al. 2013). Finally, several co-
receptors and modulators of VEGFC/VEGFR3 signaling
play crucial roles in lymphangiogenesis (for review, see
Corresponding author: b.hogan@imb.uq.edu.au
Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.263210.
115.
© 2015 Koltowska et al. This article is distributed exclusively by Cold
Spring Harbor Laboratory Press for the first six months after the full-issue
publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml).
After six months, it is available under a Creative Commons License (At-
tribution-NonCommercial 4.0 International), as described at http://
creativecommons.org/licenses/by-nc/4.0/.
1618 GENES &DEVELOPMENT 29:16181630 Published by Cold Spring Harbor Laboratory Press; ISSN 0890-9369/15; www.genesdev.org
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
Schulte-Merker et al. 2011; Koltowska et al. 2013; Zheng
et al. 2014), underscoring how central the pathway is in
lymphatic development.
Characterization of the zebrafish lymphatic vasculature
has demonstrated the highly conserved function of the
VEGFC/VEGFR3 signaling pathway in vertebrates (Kuch-
ler et al. 2006; Yaniv et al. 2006). Forward genetic screens
for mutants lacking lymphatics in zebrafish discovered a
function for Ccbe1 (Hogan et al. 2009a), which was subse-
quently shown to play a conserved role in mice and hu-
mans (Alders et al. 2009; Connell et al. 2010; Bos et al.
2011). CCBE1 regulates the processing and activation of
immature VEGFC to its mature, functional form neces-
sary for lymphangiogenesis (Jeltsch et al. 2014; Le Guen
et al. 2014). In addition, zebrafish mutants have been de-
scribed in vegfc and vegfr3 themselves, and we now
know that zebrafish Vegfc signaling controls all secondary
angiogenesis, including the formation of lymphatic pre-
cursors (PLs) from the posterior cardinal vein (PCV) (Ho-
gan et al. 2009a,b; Villefranc et al. 2013; Le Guen et al.
2014). The coordinated activity of both Vegfc and Vegfd
further controls lymphangiogenesis of the facial lymphat-
ic network (Okuda et al. 2012; Astin et al. 2014).
Although the apical signaling pathway that governs
lymphatic sprouting is well established, we still have a
very limited understanding of how signaling controls
downstream pathways and regulates endothelial cell (EC)
transcription. Several transcription factors are known to
control the initial specification and maintenance of lym-
phatic EC (LEC) fate in mice, including PROX1, COUPT-
FII, and SOX18 (Wigle and Oliver 1999; Francois et al.
2008; Srinivasan et al. 2010; Srinivasan and Oliver 2011).
Other transcription factors controlling later differentia-
tion and specialization of lymphatic vessels include
FOXC2 and GATA2 (Petrova et al. 2004; Norrmen et al.
2009; Kazenwadel et al. 2012; Lim et al. 2012). Interesting-
ly, emerging data have suggested that both the mainte-
nance of PROX1 and the induction of SOX18 activity can
be driven by VEGF signaling mechanisms (Deng et al.
2013; Duong et al. 2014; Srinivasan et al. 2014). While
this integration of cell-extrinsic signaling and cell-intrin-
sic transcriptional information appears to play an impor-
tant role in lymphatic development, the spatiotemporal
control of signaling and the complexity of EC transcrip-
tional mechanisms remain far from fully understood.
The initial genetic screens performed for zebrafish lym-
phatic development used a pan-endothelial marker [Tg
(fli1a:EGFP)
y1
] and scored the presence of the thoracic
duct as a proxy for systemic lymphangiogenesis. We
took advantage of the recently described Tg(-5.2lyve1b:
dsRed) strain, which labels the comprehensive network
of developing lymphatic vessels and embryonic veins
(Okuda et al. 2012), to perform a phenotypically sensitized
forward genetic screen and integrated whole-genome se-
quence mapping for rapid gene discovery. Here, we report
the first mutant characterized from this screen, a mafba
mutant. We found that mafba is an essential Vegfc-regu-
lated transcription factor controlling lymphangiogenesis
and identified a role for SoxF transcription factors in in-
ducing mafba expression.
Results
uq
4bh
mutants fail to form lymphatic vasculature
We designed and performed an ENU mutagenesis screen
in zebrafish. Briefly, we mutagenized the Tg(-5.2lyve1b:
dsRed) strain and performed a classical F3 embryonic
screen in this background. We sequenced the genomes
of the transgenic mutagenized founders and our in-house
WIK strain, which we used to generate mapping crosses
with a predefined genomic variation. In total, we used
838 mutagenized F1 genomes (419 F2 families) with, on
average, four F2 in-crosses scored per family (or 573 ge-
nomes screened). We identified 34 mutants that gave lym-
phatic deficiency/venous sprouting defects at either of
two time points: 3 d post-fertilization (dpf) and 5 dpf.
The uq
4bh
mutant showed a highly selective reduction,
up to a complete loss, of lymphatic vessels in the embry-
onic trunk and face but formed a grossly normal blood vas-
culature (Fig. 1A; Supplemental Fig. 1B). Overall, the body
plan was normal except for an otic vesicle defect and ede-
ma by 7 dpf (Figs. 1B, 2D; Supplemental Fig. 1A). Quanti-
fication of the number of LECs in the trunk and face using
a nuclear marker of ECs coupled with Tg(-5.2lyve1b:
dsRed) demonstrated a significant reduction in mutants
(Fig. 1CE). In the developing facial lymphatics, reduc-
tions were observed in medial facial lymphatics and bran-
chial arch lymphatics (Fig. 1D,E; Supplemental Fig. 1C).
The number of LECs in the otolithic lymphatic vessel
was increased but was presumed to reflect altered, local
tissue patterning. Earlier in development, examination
of parachordal PL cell number showed no difference in
mutants (Fig. 1F,G). We analyzed the expression of prox1a
in the Tg(prox1a:Kalt4-4xUAS:uncTagRFP) transgenic
line that has been previously reported (Dunworth et al.
2014; van Impel et al. 2014) crossed onto a Tg(10xUAS:Ve-
nus) reporter strain. Interestingly,we saw no change in
prox1a expression in sprouts leaving the PCV, suggesting
normal establishment of cell identity in mutants (Fig. 1H,
I). Overall, these observations indicate that the uq
4bh
mu-
tants initiate lymphatic fate and sprouting secondary an-
giogenesis, but ongoing formation of a comprehensive
lymphatic network fails.
uq
4bh
is a mafba mutant
We used a whole-genome sequence-based approach to
map the genomic location of the affected gene (Supple-
mental Material; data not shown). We identified a region
of homozygosity on chromosome 23, and, within this
linked region, a candidate mutation was identified in the
mafba gene (Fig. 2A; Supplemental Fig. 2A). A C/T non-
sense mutation was identified (encoding Q155) and pre-
dicted to truncate Mafba prior to the critical basic region
leucine zipper (BRLZ) domain and thus is a predicted
loss-of-function allele (Fig. 2B,C). The zebrafish neural
segmentation mutant valentino (Moens et al. 1996) is a
mafba mutant and displays the same otic vesicle pheno-
type as uq
4bh
(Fig. 2D), suggesting a causative mafba mu-
tation. MAFB is a bZIP transcription factor that can act
as an activator or repressor. MAFB homologs regulate
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1619
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
cellular differentiation in various developmental con-
texts, controlling posterior hindbrain/otic fate decisions
(Moens et al. 1996; Moens and Prince 2002), podocyte
development (Sadl et al. 2002; Moriguchi et al. 2006), pan-
creatic β-cell differentiation (Artner et al. 2007), and he-
matopoietic lineage decisions (Sieweke et al. 1996, 1997;
Kelly et al. 2000; Bakri et al. 2005). However, MafB has
no previously described role in either lymphangiogenesis
or vascular development.
To confirm that the mutation in mafba causes the
lymphatic vascular developmental phenotype, we used
CRISPR genome editing to generate germline mosaic
zebrafish transmitting mutations in mafba (Supplemental
Fig. 2B). Mosaic founders were then crossed to carriers
for the uq
4bh
mutation, and the progeny were analyzed
for phenotypes. Compound heterozygous embryos (con-
firmed by genotyping) displayed the described valentino
otic phenotype, and this was coincident with a loss of
lymphatic vascular development (Fig. 2D,E). This comple-
mentation test confirmed the mutant as mafba
uq4bh
(Fig.
2E; Supplemental Fig. 2C,D).
mafba is expressed in the PCV and enriched
in secondary sprouts
To determine the cell type in which mafba is active, we
examined gene expression by in situ hybridization (ISH).
mafba expression was observed in neurons, rhombo-
meres, and the pancreas (data not shown) as well as the
PCV. We observed weak PCV expression at 24 h post-fer-
tilization (hpf), which was increased by 30 and 36 hpf, pre-
ceding and concomitant with secondary angiogenesis (Fig.
3A; Supplemental Fig. 3A). At 48 hpf, when secondary
sprouts have formed, we observed enriched expression of
mafba dorsally in ECs sprouting or sprouted from the
PCV (Fig. 3A). As ISH is insensitive at later developmental
stages, we examined mafba expression in FACS (fluores-
cent-activated cell sorting)-sorted populations of arterial
ECs (AECs), venous ECs (VECs), and LECs that we isolat-
ed from 60 hpf, 3 dpf, and 5 dpf embryos (Fig. 3B,C; Coxam
et al. 2014, 2015). Indicative of lineage-restricted expres-
sion in the vasculature, we found that mafba was highly
enriched in VECs compared with AECs at early time
Figure 1. The uq
4bh
mutant fails to form a lymphat-
ic vasculature. (A) Vascular nuclei [Tg(fli1a:
nlsEGFP); green] and veins and lymphatic vessels
[Tg(lyve1:DsRed); white] in the trunk of sibling (top
panels) and mutant (bottom panels) embryos at
5 dpf. (TD) Thoracic duct; (DLLV) dorsal longitudinal
lymphatic vessel; (ISLV) intersegmental lymphatic
vessel. Arrows indicate lymphatics, and asterisks in-
dicate their absence. Bars, 50 μm. (B) Gross morphol-
ogy of sibling (top) and mutant (bottom) embryos at
5 dpf. (C) Quantification of total LEC number using
the overlay of transgenic marker expression from A.
Mean ± SEM; scored siblings, n= 15; uq
4bh
,n= 18; t-
test. (∗∗∗∗)P< 0.0001. (D) The facial lymphatic net-
work in sibling (top) and mutant (bottom) embryos
at 5 dpf. (LFL) Lateral facial lymphatic; (OLV) otolithic
lymphatic; (LAA) branchial arch lymphatics; (MFL)
medial facial lymphatics. Bars, 50 μm. (E) Quantifica-
tion of facial lymphatic cell numbers in individual
embryos using the overlay of transgenic marker ex-
pression from A. Mean ± SEM; scored siblings, n= 6;
uq
4bh
,n= 8; t-test. ()P< 0.05. (F) PLs in the horizon-
tal myoseptum in sibling (top) and mutant (bottom)
embryos. Bars, 30 μm. (G) Quantification of PL num-
bers in individual embryos using the overlay of trans-
genic marker expression from A. Mean ± SEM; scored
siblings, n= 10; uq
4bh
,n= 12; t-test. (ns) No sig-
nificant difference. (H)prox1a-expressing LECs as
they emerge from the PCV at 36 h post-fertilization
(hpf) in sibling (top) and mutant (bottom) embryos
labeled by Tg(prox1a:Kalt4-4xUAS:uncTagRFP);Tg
(10xUAS:Venus). (Green) α-GFP. Bars, 30 μm. (I)
Quantification of prox1a-expressing LECs in individ-
ual embryos at 36 hpf. Mean ± SEM; scored siblings,
n= 16; uq
4bh
,n= 15; t-test. (ns) No significant
difference.
Koltowska et al.
1620 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
points (comparable with known VEC markers) and was
enriched in LECs compared with VECs by 5 dpf (compara-
ble with known venous and lymphatic markers lyve1b
and prox1a). We examined expression of other Maf family
genes and the duplicate Mafb homolog mafbb, which
showed detectable EC expression, suggesting that it could
play a compensatory role (Supplemental Fig. 4B,C); how-
ever, given the phenotype of mafba mutants, any compen-
sation must be partial.
mafba acts cell-autonomously during zebrafish
lymphangiogenesis
mafba expression is suggestive of a cell-autonomous func-
tion; hence, we performed cellular transplantation exper-
iments to formally test autonomy. We transplanted wild-
type Tg(fli1a:EGFP) cells into Tg(-5.2lyve1b:DsRed) re-
cipients from a mafba heterozygous in-cross and scored
for the presence of grafted (EGFP-positive) cells in arteries
(AECs), veins (VECs), and lymphatics (LECs) at 5 dpf. We
found that wild-type cells could as readily contribute
AECs, VECs, and LECs in mutant embryos (n= 13 vascu-
lar grafts) as they could in sibling controls (n= 11 vascular
grafts) (Fig. 4A,B). We traced all grafted cells with dextran
and noted the positions of non-ECs, which were in vari-
able locations and not consistent in rescued mutant em-
bryos. Together, these analyses suggest that mafba is
sufficient in ECs to direct lymphangiogenesis in mutant
embryos.
Reciprocally, we transplanted mutant cells from Tg
(kdrl:EGFP);Tg(-5.2lyve1b:DsRed) double-transgenic em-
bryos into unlabeled wild-type hosts. In this experimental
setting, AECs will express only EGFP, VECs will express
EGFP plus dsRED, and LECs will express only dsRED in
successful vascular grafts. Transplanted mutant cells
contributed to AECs (n= 12/12 vascular grafts) and VECs
(n= 3/12 vascular grafts) but not Tg(lyve1:DsRed)-ex-
pressing LECs (Fig. 4C,D). Interestingly, in one trans-
planted embryo (n= 1/12), mutant cells formed a section
of vasculature that appeared to be lymphatic based on
Figure 2. Positional cloning of mafba
uq4bh
using whole-genome sequence-based ho-
mozygosity mapping and mutation detec-
tion. (A) Schematic plot of genomic
homozygosity across all 25 chromosomes
(top), chromosome 23 (middle), and the re-
gion of linkage (bottom) (see the Materials
and Methods for details). The mafba gene
is located centrally within the region of
highest homozygosity. (B) Sequence chro-
matograms confirming the nonsense allele
in the mafba gene. (Top) Wild type (n=6
reads). (Middle) Heterozygous (n= 18
reads). (Bottom) Mutant (n= 19 reads). (C)
Schematic of the location of the Q155al-
lele predicted to truncate the critical
BRLZ domain of Mafba. (D) Otic vesicle
morphology of sibling (left) and mutant
(right) embryos at 3 dpf. Bars, 80 μm. (E)
Confirmation of causative mafba
uq4bh
al-
lele by complementation test with a
mafba
5 bp del
transmitting founder. Vascu-
lature visualized by Tg(kdrl:EGFP) (green)
and Tg(-5.2lyve1b:DsRed) (red) in the sib-
ling (top) and transheterozygous maf-
ba
uq4bh
;mafba
5 bp del
(bottom). Siblings, n
= 10; mafba
uq4bh
; mafba
5bpdel
,n= 10.
(Right) Red channel. Bars, 30 μm. See also
Supplemental Figure 2.
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1621
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
morphology, but this vessel expressed Tg(kdrl:EGFP),
which is normally restricted to blood vessels. Hence,
this grafted vessel was considered to have a differentia-
tion defect (Supplemental Fig. 3B). Taken together, re-
ciprocal cellular mosaic experiments demonstrate that
mafba acts autonomously in ECs during lymphatic vessel
development.
A set of mafba-dependent endothelial genes
are responsive to Vegfc signaling
We next isolated Tg(kdrl:EGFP)-expressing ECs from sib-
ling and mafba
uq4bh
mutant embryos at 48 hpf by embryo
dissociation and FACS for EGFP (Supplemental Fig. 4A).
We performed RNA sequencing (RNA-seq) in triplicate
and generated a concordant data set identifying 23
down-regulated and 68 up-regulated genes (Fig. 5A; Sup-
plemental Table 1). These dysregulated genes were gener-
ally lowly expressed, probably indicative of low numbers
of cells derived from secondary sprouts (which express
mafba) in the larger pool of Tg(kdrl:EGFP)-expressing
ECs. To confirm that these genes were dysregulated, we
used quantitative PCR (qPCR) for 15 down-regulated
and 47 up-regulated genes and confirmed that the majori-
ty was misexpressed, as indicated in the RNA-seq data
(Fig. 5B).
To understand how selective to secondary angiogenesis
these genes are, we sorted ECs using FACS from 30-hpf
MO-vegfc knockdown embryos (prior to venous sprout-
ing) and 48-hpf Tg(prox1a:Kalt4-4xUAS:uncTagRFP);Tg
(10xUAS:Vegfc) vegfc-inducedembryos, which over-
express Vegfc in all prox1a-expressing tissues and hence
show ectopic venous angiogenesis (Helker et al. 2013;
Le Guen et al. 2014; K Koltowska, AK Lagendijk, C
Pichol-Thievend, JC Fischer, M Francois, EA Ober, AS
Yap, and BM Hogan, in prep.). We then performed qPCR
for 15 genes that were down-regulated in mafba mu-
tants. Of these 15 genes, 13 were down-regulated in
MO-vegfc ECs, and eight were up-regulated in Vegfc-in-
duced ECs (Fig. 5C). This observation suggested that, in
ECs, mafba-dependent genes are also dependent on nor-
mal Vegfc signaling.
mafba expression is up-regulated by Vegfc
We investigated mafba expression in MO-vegfc and
MO-vegfr3 embryos at 30 hpf and saw no change in mafba
expression (Supplemental Fig. 4D). At 48 hpf, when mafba
normally becomes enriched in the dorsal PCV, this en-
richment was lost in MO-vegfc and MO-vegfr3 embryos,
and overall EC mafba levels (normalized) were reduced
by qPCR (Fig. 5D,E). Venous sprouts fail to form in MO-
vegfc and MO-vegfr3 embryos, and so the loss of ex-
pression may be a consequence of failed morphogenesis.
However, the observation is consistent with selective
enrichment in cells responding to Vegfc signaling, so we
next examined mafba expression in vegfc-induced embry-
os. We observed vastly increased mafba expression in the
vasculature of vegfc-induced embryos by ISH and con-
firmed this increase by qPCR (normalized) using FACS-
sorted ECs from vegfc-induced embryos (Fig. 5F,G). We
did not find any evidence for regulation of vegfc,vegfr3,
or other Vegf family downstream from mafba (Supple-
mental Fig. 4EG).
mafba regulates LEC migration from the horizontal
myoseptum but is dispensable for vegfc-induced
proliferation
To determine the earliest cellular defect in trunk lym-
phangiogenesis in mafba mutants, we time-lapse-imaged
PLs in the horizontal myoseptum from 48 hpf onward
in Tg(-5.2lyve1:DsRed);Tg(kdrl:EGFP) double-transgenic
embryos. While wild-type PLs actively migrated out of
the myoseptum and elongated along their arterial sub-
strates (Bussmann et al. 2010; Cha et al. 2012), mutant
PLs commonly failed to migrate from the myoseptum
(Fig. 6A; Supplemental Movies 1, 2). Some PLs dis-
played distinctly broad and rounded morphology during
Figure 3. mafba is expressed in endothelium
during lymphangiogenesis. (A) Expression of
mafba in the zebrafish trunk at 24 hpf and 36
hpf (left) and 48 hpf (right). (Bottom right pan-
els) Higher-magnification images of secondary
sprouts (PCV; arrowheads). (Boxed area) High-
magnification region; (bracket) dorsal aorta
(DA). Bars, 100 μm. (B) Quantitative PCR
(qPCR) on FACS-sorted AEC versus VEC popu-
lations at 60 hpf; mafba expression is VEC-en-
riched, comparable with vegfr3 and in contrast
to AEC marker flt1. Mean ± SEM. Expression is
relative to kdrl.(C) qPCR on FACS-sorted LEC
and VEC populations from 3 dpf and 5 dpf; the
mafba expression profile is similar to LEC
markers prox1a and lyve1b. Mean ± SEM. Ex-
pression is relative to rpl13.
Koltowska et al.
1622 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
migration, which was not observed in wild-type cells (Fig.
6A). Interestingly, we did not find any evidence for dysre-
gulation of chemokine signaling components that control
this migratory event (Cha et al. 2012) or ephrinb2a/ephB4,
which have been implicated in EC migration (Supplemen-
tal Fig. 5AC). We next examined mafba mutants in the
vegfc-induced transgenic overexpression background and
found that vegfc overexpression induced the proliferation
of VECs at the same level in mafba mutants as in wild-
type transgenic embryos (Fig. 6B,C). Together, these data
show that mafba controls the ongoing migration of PLs af-
ter initial secondary angiogenesis but is not required for
vegfc transgene-induced venous proliferation.
Vegfc up-regulates mafba expression in a SoxF
transcription factor-dependent manner
We and others have previously shown that VEGF signal-
ing can control the activity of SOX18 in human ECs,
mice, and zebrafish (Deng et al. 2013; Duong et al.
2014). To determine whether SoxF family transcription
factors play a role in mafba induction, we examined
mafba expression in sox7/sox18 double morpholino
(dMO)-injected embryos. We found a strong reduction to
complete absence of mafba expression in the PCV at 30
and 48 hpf (Fig. 7A), an unexpected observation because
sox7/sox18 dMO embryos display expanded expression
of classical VEC markers (Herpers et al. 2008; Pendeville
et al. 2008). Given that SOXF transcription factors can re-
spond to VEGFs and that we observed that mafba expres-
sion is also Vegfc-responsive, we next examined whether
the up-regulation of mafba by Vegfc is dependent on
SoxF transcription factors. We found that knockdown of
sox7 alone did not have an impact on the induction of
mafba by Vegfc (Fig. 7B,C). sox18 knockdown strongly
reduced the intensity of induction, and the dMO knock-
down further reduced the induction of mafba expres-
sion (Fig. 7B,C). Supporting this regulatory relationship
in vitro, transfection of human umbilical vein ECs with
SOX18 but not SOX7 induced MAFB expression (Supple-
mental Fig. 6A,B). Furthermore, expression of sox18 and
sox7 was normal in mafba
uq4bh
mutants, suggesting that
these genes are upstream of but not downstream from
mafba (Supplemental Fig. 6D,E).
Given that the transgenic overexpression of Vegfc in-
duces proliferation of VECs, we asked whether the reduc-
tion of mafba VEC expression in vegfc-induced/MO-soxF-
injected embryos was concurrent with a reduction in VEC
proliferation. Importantly, Vegfc overexpression in sox7/
sox18 double-knockdown embryos still induced robust
VEC proliferation (Fig. 7D). This observation is strikingly
in line with the fact that mafba also played no role in
vegfc-induced VEC proliferation in this transgenic back-
ground. Finally, we examined sox7 and sox18 expression
by qPCR on embryonic ECs FACS-sorted [using Tg(kdrl:
EGFP)] from MO-vegfc and vegfc-induced embryos at
48 hpf. We found that both were mildly reduced in loss-
of-function scenarios and increased in gain-of-function
Figure 4. A cell-autonomous function for
mafba in lymphatic vascular development. (A)
Transplantation of Tg(fli1a:EGFP)-labeled (green)
and dextran blue-labeled (blue) donor cells into a
Tg(lyve1b:DsRed) (red) host derived from a maf-
ba
uq4bh
in-cross. (A,A′′) Wild-type ECs were ob-
served to contribute to arteries (AECs), veins
(VECs), and lymphatics (LECs) (white arrows)
in sibling and mutant recipients. The boxed ar-
eas indicate high-power regions (shown at right).
(B) Quantification of endothelial contributions
in Aand A′′. Percentage contributions of a cell
to AECs, VECs, and LECs for all vascular grafted
embryos (n= 11 wild type into sibling embryos
with vascular grafts; n= 13 wild type into mu-
tant embryos with vascular grafts). (C) Trans-
plantation of Tg(lyve1b:DsRed)-labeled (red), Tg
(kdrl:EGFP)-labeled (green), and dextran blue-
labeled (blue) donor cells derived from a maf-
ba
uq4bh
in-cross into an unlabeled wild-type
host. Genotyping confirmed donor identity. (C)
Sibling ECs contributed to AECs, VECs, and
LECs (arrows). (C′′) Mutant ECs contributed
AECs (n= 12/12 grafts) and VECs (n= 3/12 grafts)
but not LECs. The boxed areas indicate high-
power images (shown at right). (D) Quantifica-
tion of endothelial contributions in Cand C′′.
Percentage contributions to AECs, VECs, and
LECs for all vascular grafts. One mutant graft
was observed with lymphatic vascular morphol-
ogy but expressed Tg(lyve1b:DsRed) and Tg
(kdrl:EGFP), indicative of a differentiation defect (see Supplemental Fig. 3B). Bars, 30 μm.
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1623
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
scenarios, where normalized expression of endothelial
egfp was not changed (Fig. 7E; Supplemental Fig. 6C).
Discussion
Taken together, the observations above demonstrate that
the transcription factor Mafba is essential for embryon-
ic lymphangiogenesis during zebrafish development. In
mice, MAFB has established roles in directing hindbrain
segmentation and podocyte, pancreatic β-cell, and hema-
topoietic lineage differentiation but no known function
in vascular lineages (Sieweke et al. 1996, 1997; Moens
et al. 1998; Kelly et al. 2000; Sadl et al. 2002; Bakri et al.
2005; Moriguchi et al. 2006; Artner et al. 2007). Our find-
ings of a cell-autonomous role in lymphatic vessel devel-
opment, given the previously described functions in cell
fate and differentiation, suggest a likely role in endothelial
lineage decisions or differentiation. A function in ongoing
LEC differentiation could explain the migration defect ob-
served if critical machinery was not switched on in PLs
after they initially sprout from the PCV. To fully under-
stand how Mafba elicits such a specific phenotype in
LECs, it is clear that the characterization of downstream
genes and pathways is now needed.
Vegfc is the major driver of developmental lymphangio-
genesis in vertebrates (Karkkainen et al. 2004). In zebra-
fish, Vegfc induces VEC proliferation in the PCV (Helker
et al. 2013; Le Guen et al. 2014) and the sprouting of ve-
nous precursors and PLs from the PCV during secondary
angiogenesis (Hogan et al. 2009a; Villefranc et al. 2013).
Our observations of mafba expression and the expression
of a subset of mafba-dependent genes in Vegfc loss-of-
function and gain-of-function embryos indicate that
mafba is responsive to Vegfc signaling during develop-
ment. We showed previously that mutations in genes
within the Vegfc pathway completely block the pheno-
types caused by overexpression of Vegfc (Le Guen et al.
2014), yet here we observed that loss of mafba has no im-
pact on transgene-induced proliferation while being cru-
cial for lymphatic development. This suggests that Vegfc
signaling has multiple downstream outcomes in ECs
Figure 5. mafba-dependent endothelial
genes are Vegfc-dependent, and mafba is
up-regulated by Vegfc signaling. (A) Sche-
matic summary of RNA-seq from FACS-
sorted sibling and mafba
uq4bh
mutants at
48 hpf. n= 23 down-regulated genes; n= 68
up-regulated genes. P< 0.01 using EdgeR
analysis. (B) qPCR validation of RNA-seq.
RNA-seq relative changes and qPCR fold
changes are displayed side by side for a sub-
set of up-regulated and down-regulated
genes. Mean ± SEM. The boxed area con-
tains down-regulated genes. (C) Fifteen
down-regulated genes analyzed by qPCR
in FACS-sorted ECs from 48-hpf vegfc-in-
duced and 30-hpf MO-vegfc embryos. Re-
ductions in expression were observed in
both mafba
uq4bh
and MO-vegfc embryonic
endothelium. Mean ± SEM. (D) Expression
of mafba in control uninjected (n= 30/33),
MO-vegfc-injected (n= 18/18), and MO-
vegfr3-injected (n= 33/33) embryos at 48
hpf. Enriched dorsal PCV expression (ar-
rows) is absent in MO-vegfc and MO-vegfr3
embryos (brackets). Bars, 100 μm. (E) qPCR
validation of mafba reduction in FACS-
sorted ECs from MO-Vegfc embryos. Ex-
pression is given relative to the geometric
average of kdrl,cdh5, and lyve1b expres-
sion. Mean ± SEM. (F) Expression of mafba
in control and vegfc-induced [Tg(prox1a:
Kalt4);Tg(10xUAS:Vegfc)] embryos. mafba
expression is induced throughout the PCV
in vegfc-induced embryos. n= 16/16. ISH
staining was for shorter periods than in E
to highlight increased expression. Bars,
100 μm. (G) qPCR validation of mafba in-
duction in FACS-sorted ECs from Tg
(prox1a:Kalt4);Tg(10xUAS:Vegfc) embryos. Expression is given relative to the geometric average of kdrl,cdh5, and lyve1b expression.
Mean ± SEM.
Koltowska et al.
1624 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
and that different cellular responses are controlled by dif-
ferent effectors. In line with this observation, the cellular
defect in mafba
uq4bh
mutants occurs later than in vegfc,
vegfr3, or ccbe1 mutants (Hogan et al. 2009a; Villefranc
et al. 2013; Le Guen et al. 2014), with mafba controlling
LEC migration from the horizontal myoseptum rather
than sprouting of LEC precursors from the PCV. It will
be intriguing to discover whether MAFB proteins play spe-
cialized roles regulating a discrete subset of functional
genes rather than broad roles in LEC identity/transcrip-
tion and determine how this compares with other tran-
scription factors, such as PROX1, which is maintained
by VEGFC signaling in mice (Srinivasan et al. 2014).
We further investigated potential mechanisms by
which Vegfc might regulate the levels of mafba in zebra-
fish. In mice, MafB can act together with Ets1, and there
is evidence that Vegfc can modulate Ets-mediated tran-
scription (Sieweke et al. 1996; Yoshimatsu et al. 2011);
however, we examined morpholino knockdown models
and gene expression levels and found no evidence that
Mafba acts coordinately with Ets factors in lymphatic de-
velopment (Supplemental Fig. 6FJ). In mice, the tran-
scription factor Sox18 controls lymphangiogenesis in a
partially redundant manner with other SOXF transcrip-
tion factors in different mouse strains (Francois et al.
2008; Hosking et al. 2009). The transcription factors
Sox18 and Sox7 function redundantly during zebrafish
blood vascular development, with dMO-injected embryos
displaying early arterialvenous defects that are recapitu-
lated in genetic mutants and do not allow for controlled
analysis of later lymphangiogenesis (Cermenati et al.
2008; Herpers et al. 2008; Pendeville et al. 2008; Herm-
kens et al. 2015). Interestingly, recent work has shown
that SOXF transcription factors can be up-regulated and
nuclear-localized in response to VEGF/VEGFCERK sig-
naling to control downstream gene expression and vascu-
lar lineage decisions (Deng et al. 2013; Duong et al. 2014).
We investigated whether SOXF transcription factors
could link Vegfc signaling to mafba expression. We found
that Sox18 and Sox7 are necessary for normal mafba ex-
pression and further showed that they indeed mediate
the observed induction of mafba by transgenic overex-
pression of Vegfc. Taken together, these observations led
us to a working model of how this pathway functions to
control lymphatic development (Fig. 7F).
The question of how cells acquire LEC identity and
differentiate has led to the characterization of a number
of transcriptional regulators of LEC fate and differentia-
tion (Wigle and Oliver 1999; Francois et al. 2008; Sriniva-
san et al. 2010; Srinivasan and Oliver 2011). How these
transcription factors combine and integrate as a function-
al regulatory network and how they are controlled by
extrinsic signals to modulate precise spatiotemporally
controlled gene expression remain to be elucidated. The
Figure 6. mafba controls LEC migration and is dispensable for Vegfc transgene-induced proliferation. (A) Representative images from
time lapse of LEC migration from sibling (n= 3) and mafba
uq4bh
mutant (n= 3) embryos. Visualized in Tg(lyve1:DsRed) (white) embryos
from 48 hpf. See Supplemental Movies 1 and 2. (Arrows) LECs. Bars, 20 μm. (B)vegfc-induced embryos display increased VEC numbers
(bottom left panel) compared with control embryos (top left panel). (Right panels) Increased VEC number is still observed in vegfc-induced
mafba
uq4bh
mutant embryos. Tg(fli1a:nlsEGFP) (white) labels endothelial nuclei. Bars, 30 μm. (C) Quantification of VEC number in sib-
ling (n= 4), sibling vegfc-induced (n= 5), mafba
uq4bh
(n= 6), and mafba
uq4bh
vegfc-induced (n= 6). Mean ± SEM; t-test. (ns) No significant
difference.
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1625
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
addition of Mafb as a crucial regulator of lymphangiogen-
esis provides a new direction and increases the complexity
of LEC gene regulation. Importantly, many transcription-
al regulators of LEC development play central roles in
lymphatic vascular diseases (Fang et al. 2000; Finegold
et al. 2001; Irrthum et al. 2003; Ostergaard et al. 2011).
In addition to gaining a deeper mechanistic understanding
of LEC fate acquisition and differentiation, it will be inter-
esting to determine whether MAFB transcription factors
contribute to vascular pathologies in the future.
Materials and methods
Zebrafish
Animal work followed the guidelines of the animal ethics com-
mittee at the University of Queensland. The forward genetic
screen was based on previous studies, with mutagenesis as de-
scribed previously (de Bruijn et al. 2009). The genomic sequenc-
ing pipeline was based on previous studies (Leshchiner et al.
2012) and will be described in full elsewhere. Published zebrafish
lines were Tg(fli1a:nEGFP)
y7
(Lawson and Weinstein 2002), Tg
(-5.2lyve1b:DsRed)
nz101
(Okuda et al. 2012), TgBAC(prox1a:
KalTA4-4xUAS-ADV.E1b:TagRFP)
nim5
(Dunworth et al. 2014;
van Impel et al. 2014), Tg(flt1:YFP)
hu4624
(Hogan et al. 2009a),
and Tg(kdrl:EGFP)
s84 3
(Jin et al. 2005).
Transgenesis, morpholinos, genotyping, and genome editing
10xUAS:vegfc plasmid DNA was generated using the full-length
zebrafish vegfc cDNA cloned into the Gateway pME vector
(pDON-221) using Gateway technology (Hartley et al. 2000). To
generate the Tg(10xUAS:vegfc)
uq2bh
strain, 20 ng/μL plasmid
DNA and 25 ng/μLtol2 transposase mRNA were injected in
one-cell stage embryos, and F1 founders were identified by
Figure 7. mafba is downstream from Vegfc and SoxF transcription factors. (A) Expression of mafba at 30 hpf in control uninjected (n=38/
41) and MO-sox18/sox7 dMO-injected (n= 24/25) embryos (left) and at 48 hpf in control uninjected (n= 30/33) and MO-sox18/sox7 dMO-
injected (n= 18/18) embryos (right). Arrows indicate PCV, and an asteriskindicates reduced expression. Bars, 100 μm. (B)mafba expression
at 48 hpf in control and vegfc-induced embryos as well as vegfc-induced embryos injected with MO-sox7,MO-sox18, and MO-sox18/sox7
dMO. Arrows indicate PCV expression, and asterisks indicate reduced/absent expression. Bars, 100 μm. (C) Quantification of mafba ex-
pression in embryos from E, with embryos scored as high, medium, and low mafba expressors. Control, 88%, n= 37/42, high; MO-sox7,
89%, n= 39/44, high; MO-sox18, 71%, n= 25/35, medium; dMO, 71%, n= 34/48, low. (D) Quantification of VEC number in vegfc-induced
control and MO-sox18/sox7 dMO-injected embryos. Mean ± SEM. Control, n= 12; control vegfc-induced, n= 34; dMO, n= 11; dMO vegfc-
induced, n= 25; t-test. (ns) No significant difference. (E) qPCR for sox18 expression in FACS-sorted zebrafish ECs from MO-vegfc,vegfc-
induced, and control embryos. egfp served as a control for EC expression levels. Expression is relative to the geometric average of kdrl,
cdh5, and lyve1b expression. Mean ± SEM. (F) Working model of Mafbafunction in lymphangiogenesis: Vegfc up-regulates mafba expres-
sion in secondary sprouts and in a Sox18-dependent (redundancy with Sox7) manner. Mafba is essential for PL migration. Vegfc controls
venous proliferation independently of Mafba or Sox18/7.
Koltowska et al.
1626 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
PCR. mafba full-length cDNA was cloned into the pCS2
+
vector
and used as a template for riboprobe synthesis. MO-vegfr3,MO-
vegfc,MO-sox7, and MO-sox18 were described previously
(Herpers et al. 2008; Le Guen et al. 2014). Two morpholino
against ets1 (Pham et al. 2007) were used as described previously,
and two morpholinos against ets2 were designed and injected
(5 ng per embryo). CRISPR genome editing for mafba was
performed as described in Gagnon et al. (2014) to generate the
mafba
uq5bh
(5-base-pair [bp] del) allele. All primer and morpholino
sequence details are provided in the Supplemental Material.
Whole-mount ISH
Whole-mount ISH was performed as described previously (Karto-
pawiro et al. 2014) using the mafba probe (see above) generated
from plasmid linearized using ClaI. Probes for vegfc (Ober et al.
2004), vegfr3 (Hogan et al. 2009b), cxcr4a and cxcl12b (Coxam
et al. 2014), efnb2a (Durbin et al. 1998), ephb4a (Cooke et al.
1997), and sox18 and sox7 (Herpers et al. 2008) were used as pre-
viously described.
FACS and gene expression analysis
Isolation of zebrafish embryonic ECs, RNA extraction, cDNA
preparation, and qPCR were performed as described previously
(Coxam et al. 2014; Kartopawiro et al. 2014). For isolating cells
from sibling and mafba
uq4bh
mutants, embryos were divided
into the phenotypic categories based on the otic phenotype at
48 hpf. For isolating cells from vegfc-induced and siblings, embry-
os were divided into phenotypic categories based on vascular phe-
notype at 48 hpf. For MO-vegfc and uninjected control, cells were
isolated at 30 hpf and 48 hpf. Primer sequences are in the Supple-
mental Material. RNA-seq was performed using the Illumina
NextSeq500 and generated average 76-bp reads, which were
then mapped to the reference genome (danRer7/Zv9). Full details
on library preparation, sequencing, and analysis are in the Supple-
mental Material.
Immunohistochemistry
Immunohistochemistry for anti-GFP was performed according to
the following protocol. Embryos were fixed in 4% PFA (parafor-
maldehyde) overnight and washed five times with PBST (0.1%
Tween in PBS [phosphate buffered saline]). Embryos were blocked
in PBDT (PBS with 1% BSA, 1% DMSO, 0.1% Triton-100) with
10% horse serum for 3 h, anti-GFP (chicken polyclonal to GFP,
1:200; ab13970) was added, and embryos were incubated over-
night. Embryos were washed five times for 30 min in PBDT and
then incubated in PBDT and 10% horse serum with secondary an-
tibody (goat anti-chicken IgG, 1:400; Alexa 488; Invitrogen,
A11039) and DAPI (1:1000; Sigma Aldrich) overnight. Embryos
were washed five times for 30 min in PBST and imaged.
Transplantation
Transplantation was performed essentially as described previ-
ously (Hogan et al. 2009a) with the following changes. Donor em-
bryos were injected with dextran cascade blue (10,000 MW,
Invitrogen) at 5 ng/nL (1 nL per embryo). Cells from wild-type
donor embryos [Tg(fli1a:EGFP)] were transplanted into host
embryos derived from mafba heterozygous in-crosses [Tg
(-5.2lyve1b:DsRed)]. For reciprocal transplants, mutant cells [Tg
(kdrl:EGFP);Tg(-5.2lyve1b:DsRed)] were transplanted into unla-
beled wild-type hosts, and donors were genotyped. Embryos
with successfully transplanted ECs were cultured until 5 dpf.
Imaging and quantification
Live and fixed embryos were mounted laterallyand imaged using
a Zeiss LSM 710 FCS confocal microscope. All images were pro-
cessed using either ImarisX64 7.70 and/or ImageJ 1.47 (National
Institutes of Health) software. The number of LEC nuclei [ex-
pressing Tg(fli1a:nEGFP)] coexpressing Tg(-5.2lyve1b:DsRed)
across five somites through a Z-stack was manually counted
using ImageJ 1.47 (National Institutes of Health) software
(Fig. 1A,C,DG; Supplemental Fig. 1C) The number of Prox1-
positive LECs expressing Tg(prox1aBAC:KalTA4-4xUAS-E1b:
uncTagRFP)
nim5
;Tg(10xUAS:Venus) detected by α-GFP in green
and costained with DAPI (to label nuclei) across five somites
was manually counted using ImageJ 1.47 (National Institutes
of Health) software (Fig. 1H,I). For the quantification of EC num-
bers shown in Figure 5, the spot tool in ImarisX64 7.70 software
was used. For each sample, the Tg(fli1a:nEGFP) nuclei were se-
lected based on fluorescence intensity, and the total number of
GFP-positive nuclei (across five somites) in a full Z-stack was
calculated.
Acknowledgments
Imaging was performed in the Australian Cancer Research Foun-
dations Dynamic Imaging Facility at the Institute for Molecular
Bioscience. Sequencing was performed by the Institute for Molec-
ular Bioscience Sequencing Facility. K.K. was supported by a
Lymphatic Education and Research Network Post-doctoral Fel-
lowship, A.K.L. was supported by a University of Queensland
Post-doctoral Fellowship, B.M.H. was supported in part by an
Australian Research Council Future Fellowship (FT100100165)
and in part by a National Health and Medical Research Coun-
cil/National Heart Foundation Career Development Fellowship
(1083811), M.F. was supported by a National Health and Medical
Research Council Career Development Fellowship (1011242),
and K.A.S. was supported by an Australian Research Council Fu-
ture Fellowship (FT110100496). J.W.A. and P.S.C. receive funding
from The Ministry of Business, Innovation, and Employment; the
Health Research Council of New Zealand; and the Auckland
Medical Research Council. This research was supported by Na-
tional Health and Medical Research Council grant 1050138 and
the Cariplo Foundation.
References
Alders M, Hogan BM, Gjini E, Salehi F, Al-Gazali L, Hennekam
EA, Holmberg EE, Mannens MM, Mulder MF, Offerhaus GJ,
et al. 2009. Mutations in CCBE1 cause generalized lymph ves-
sel dysplasia in humans. Nat Genet 41: 12721274.
Artner I, Blanchi B, Raum JC, Guo M, Kaneko T, Cordes S, Sie-
weke M, Stein R. 2007. MafB is required for islet βcell matu-
ration. Proc Natl Acad Sci 104: 38533858.
Astin JW, Haggerty MJ, Okuda KS, Le Guen L, Misa JP, Tromp A,
Hogan BM, Crosier KE, Crosier PS. 2014. Vegfd can compen-
sate for loss of Vegfc in zebrafish facial lymphatic sprouting.
Development 141: 26802690.
Bakri Y, Sarrazin S, Mayer UP, Tillmanns S, Nerlov C, Boned A,
Sieweke MH. 2005. Balance of MafB and PU.1 specifies al-
ternative macrophage or dendritic cell fate. Blood 105:
27072716.
Bos FL, Caunt M, Peterson-Maduro J, Planas-Paz L, Kowalski J,
Karpanen T, van Impel A, Tong R, Ernst JA, Korving J, et al.
2011. CCBE1 is essential for mammalian lymphatic vascular
development and enhances the lymphangiogenic effect of
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1627
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
vascular endothelial growth factor-C in vivo. Circ Res 109:
486491.
Bussmann J, Bos FL, Urasaki A, Kawakami K, Duckers HJ,
Schulte-Merker S. 2010. Arteries provide essential guidance
cues for lymphatic endothelial cells in the zebrafish trunk.
Development 137: 26532657.
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: 26572666.
Cha YR, Fujita M, Butler M, Isogai S, Kochhan E, Siekmann AF,
Weinstein BM. 2012. Chemokine signaling directs trunk lym-
phatic network formation along the preexisting blood vascula-
ture. Dev Cell 22: 824836.
Connell F, Kalidas K, Ostergaard P, Brice G, Homfray T, Roberts
L, Bunyan DJ, Mitton S, Mansour S, Mortimer P, et al. 2010.
Linkage and sequence analysis indicate that CCBE1 is mutat-
ed in recessively inherited generalised lymphatic dysplasia.
Hum Genet 127: 231241.
Cooke JE, Xu QL, Wilson SW, Holder N. 1997. Characterisation of
five novel zebrafish Eph-related receptor tyrosine kinases sug-
gests roles in patterning the neural plate. Dev Genes Evol 206:
515531.
Coxam B, Sabine A, Bower NI, Smith KA, Pichol-Thievend C,
Skoczylas R, Astin JW, Frampton E, Jaquet M, Crosier PS,
et al. 2014. Pkd1 regulates lymphatic vascular morphogenesis
during development. Cell Rep 7: 623633.
Coxam B, Neyt C, Grassini DR, Le Guen L, Smith KA, Schulte-
Merker S, Hogan BM. 2015. Carbamoyl-phosphate synthetase
2, aspartate transcarbamylase, and dihydroorotase (cad) regu-
lates Notch signaling and vascular development in zebrafish.
Dev Dyn 244: 19.
de Bruijn E, Cuppen E, Feitsma H. 2009. Highly efficient ENU
mutagenesis in zebrafish. Methods Mol Biol 546: 312.
Deng Y, Atri D, Eichmann A, Simons M. 2013. Endothelial ERK
signaling controls lymphatic fate specification. J Clin Invest
123: 12021215.
Dumont DJ, Jussila L, Taipale J, Lymboussaki A, Mustonen T,
Pajusola K, Breitman M, Alitalo K. 1998. Cardiovascular fail-
ure in mouse embryos deficient in VEGF receptor-3. Science
282: 946949.
Dunworth WP, Cardona-Costa J, Bozkulak EC, KimJD, Meadows
S, Fischer JC, Wang Y, Cleaver O, Qyang Y, Ober EA, et al.
2014. Bone morphogenetic protein 2 signaling negatively
modulates lymphatic development in vertebrate embryos.
Circ Res 114: 5666.
Duong T, Koltowska K, Pichol-Thievend C, Le Guen L, Fontaine
F, Smith KA, Truong V, Skoczylas R, Stacker SA, Achen MG,
et al. 2014. VEGFD regulates blood vascular development by
modulating SOX18 activity. Blood 123: 11021112.
Durbin L, Brennan C, Shiomi K, Cooke J, Barrios A, Shanmugalin-
gam S, Guthrie B, Lindberg R, Holder N. 1998. Eph signaling is
required for segmentation and differentiation of the somites.
Genes Dev 12: 30963109.
Fang J, Dagenais SL, Erickson RP, Arlt MF, Glynn MW, Gorski JL,
Seaver LH, Glover TW. 2000. Mutations in FOXC2(MFH-1), a
forkhead family transcription factor, are responsible for the
hereditary lymphedema-distichiasis syndrome. Am J Hum
Genet 67: 13821388.
Finegold DN, Kimak MA, Lawrence EC, Levinson KL, Cherniske
EM, Pober BR, Dunlap JW, FerrellRE. 2001. Truncating muta-
tions in FOXC2 cause multiple lymphedema syndromes.
Hum Mol Genet 10: 11851189.
Francois 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 vas-
culature in mice. Nature 456: 643647.
Gagnon JA, Valen E, Thyme SB, Huang P, Ahkmetova L, Pauli A,
Montague TG, Zimmerman S, Richter C, Schier AF. 2014. Ef-
ficient mutagenesis by Cas9 protein-mediated oligonucleo-
tide insertion and large-scale assessment of single-guide
RNAs. PLoS One 9: e98186.
Gordon K, Schulte D, Brice G, Simpson MA, Roukens MG, van
Impel A, Connell F, Kalidas K, Jeffery S, Mortimer PS, et al.
2013. Mutation in vascular endothelial growth factor-C, a li-
gand for vascular endothelial growth factor receptor-3, is asso-
ciated with autosomal dominant milroy-like primary
lymphedema. Circ Res 112: 956960.
Hagerling R, Pollmann C, Andreas M, Schmidt C, Nurmi H, Ad-
ams RH, Alitalo K, Andresen V, Schulte-Merker S, Kiefer F.
2013. A novel multistep mechanism for initial lymphangio-
genesis in mouse embryos based on ultramicroscopy. EMBO
J32: 629644.
Hartley JL, Temple GF, Brasch MA. 2000. DNA cloning using in
vitro site-specific recombination. Genome Res 10: 17881795.
Helker CS, Schuermann A, Karpanen T, Zeuschner D, Belting
HG, Affolter M, Schulte-Merker S, Herzog W. 2013. The
zebrafish common cardinal veins develop by a novel mecha-
nism: lumen ensheathment. Development 140: 27762786.
Hermkens DM, van Impel A, Urasaki A, Bussmann J, Duckers HJ,
Schulte-Merker S. 2015. Sox7 controls arterial specification in
conjunction with hey2 and efnb2 function. Development 142:
16951704.
Herpers R, van de Kamp E, Duckers HJ, Schulte-Merker S. 2008.
Redundant roles for sox7 and sox18 in arteriovenous specifica-
tion in zebrafish. Circ Res 102: 1215.
Hogan BM, Bos FL, Bussmann J, Witte M, Chi NC, Duckers HJ,
Schulte-Merker S. 2009a. Ccbe1 is required for embryonic
lymphangiogenesis and venous sprouting. Nat Genet 41:
396398.
Hogan BM, Herpers R, Witte M, Helotera H, Alitalo K, Duckers
HJ, Schulte-Merker S. 2009b. Vegfc/Flt4 signalling is sup-
pressed by Dll4 in developing zebrafish intersegmental arter-
ies. Development 136: 40014009.
Hosking B, Francois M, Wilhelm D, Orsenigo F, Caprini A, Svin-
gen T, Tutt D, Davidson T, Browne C, Dejana E, et al. 2009.
Sox7 and Sox17 are strain-specific modifiers of the lymphan-
giogenic defects caused by Sox18 dysfunction in mice. Devel-
opment 136: 23852391.
Irrthum A, Karkkainen MJ, Devriendt K, Alitalo K, Vikkula M.
2000. Congenital hereditary lymphedema caused by a muta-
tion that inactivates VEGFR3 tyrosine kinase. Am J Hum Ge-
net 67: 295301.
Irrthum A, Devriendt K, Chitayat D, Matthijs G, Glade C, Steij-
len PM, Fryns JP, Van Steensel MA, Vikkula M. 2003. Muta-
tions in the transcription factor gene SOX18 underlie
recessive and dominant forms of hypotrichosis-lymphede-
ma-telangiectasia. Am J Hum Genet 72: 14701478.
Jeltsch M, Kaipainen A, Joukov V, Meng X, Lakso M, Rauvala H,
Swartz M, Fukumura D, Jain RK, Alitalo K. 1997. Hyperplasia
of lymphatic vessels in VEGF-C transgenic mice. Science 276:
14231425.
Jeltsch M, Jha SK, Tvorogov D, Anisimov A, Leppanen VM, Hol-
opainen T, Kivela R, Ortega S, Karpanen T, Alitalo K. 2014.
CCBE1 enhances lymphangiogenesis via ADAMTS3-mediat-
ed VEGF-C activation. Circulation 129: 19621971.
Jin SW, Beis D, Mitchell T, Chen JN, Stainier DY. 2005. Cellular
and molecular analyses of vascular tube and lumen formation
in zebrafish. Development 132: 51995209.
Koltowska et al.
1628 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
Karkkainen MJ, Ferrell RE, Lawrence EC, Kimak MA, Levinson
KL, McTigue MA, Alitalo K, Finegold DN. 2000. Missense
mutations interfere with VEGFR-3 signalling in primary lym-
phoedema. Nat Genet 25: 153159.
Karkkainen MJ, Saaristo A, Jussila L, Karila KA, Lawrence EC,
Pajusola K, Bueler H, Eichmann A, Kauppinen R, Kettunen
MI, et al. 2001. A model for gene therapy of human hereditary
lymphedema. Proc Natl Acad Sci 98: 1267712682.
Karkkainen MJ, Haiko P, Sainio K, Partanen J, Taipale J, Petrova
TV, Jeltsch M, Jackson DG, 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: 7480.
Kartopawiro J, Bower NI, Karnezis T, Kazenwadel J, Betterman
KL, Lesieur E, Koltowska K, Astin J, Crosier P, Vermeren S,
et al. 2014. Arap3 is dysregulated in a mouse model of hypotri-
chosis-lymphedema-telangiectasia and regulates lymphatic
vascular development. Hum Mol Genet 23: 12861297.
Kazenwadel J, Secker GA, Liu YJ, Rosenfeld JA, Wildin RS, Cuel-
lar-Rodriguez J, Hsu AP, Dyack S, Fernandez CV, Chong CE,
et al. 2012. Loss-of-function germline GATA2 mutations in
patients with MDS/AML or MonoMAC syndrome and prima-
ry lymphedema reveal a key role for GATA2 in the lymphatic
vasculature. Blood 119: 12831291.
Kelly LM, Englmeier U, Lafon I, Sieweke MH, Graf T. 2000. MafB
is an inducer of monocytic differentiation. EMBO J 19:
19871997.
Koltowska K, Betterman KL, Harvey NL, Hogan BM. 2013. Get-
ting out and about: the emergence and morphogenesis of the
vertebrate lymphatic vasculature. Development 140:
18571870.
Kuchler AM, Gjini E, Peterson-Maduro J, Cancilla B, Wolburg H,
Schulte-Merker S. 2006. Development of the zebrafish lym-
phatic system requires VEGFC signaling. Curr Biol 16:
12441248.
Lawson ND, Weinstein BM. 2002. In vivo imaging of embryonic
vascular development using transgenic zebrafish. Dev Biol
248: 307318.
Le Guen L, Karpanen T, Schulte D, Harris NC, Koltowska K,
Roukens G, Bower NI, van Impel A, Stacker SA, Achen MG,
et al. 2014. Ccbe1 regulates Vegfc-mediated induction of
Vegfr3 signaling during embryonic lymphangiogenesis. De-
velopment 141: 12391249.
Leshchiner I, Alexa K, Kelsey P, Adzhubei I, Austin-Tse CA, Coo-
ney JD, Anderson H, King MJ, Stottmann RW, Garnaas MK,
et al. 2012. Mutation mapping and identification by whole-ge-
nome sequencing. Genome Res 22: 15411548.
Lim KC, Hosoya T, Brandt W, Ku CJ, Hosoya-Ohmura S, Camper
SA, Yamamoto M, Engel JD. 2012. Conditional Gata2 inacti-
vation results in HSC loss and lymphatic mispatterning. J
Clin Invest 122: 37053717.
Lim HY, Thiam CH, Yeo KP, Bisoendial R, Hii CS, McGrath KC,
Tan KW, Heather A, Alexander JS, Angeli V. 2013. Lymphatic
vessels are essential for the removal of cholesterol from pe-
ripheral tissues by SR-BI-mediated transport of HDL. Cell
Metab 17: 671684.
Makinen T, Jussila L, Veikkola T, Karpanen T, Kettunen MI,
Pulkkanen KJ, Kauppinen R, Jackson DG, Kubo H, Nishikawa
S, et al. 2001. Inhibition of lymphangiogenesis with resulting
lymphedema in transgenic mice expressing soluble VEGF re-
ceptor-3. Nat Med 7: 199205.
Martel C, Li W, Fulp B, Platt AM, Gautier EL, Westerterp M,Bitt-
man R, Tall AR, Chen SH, Thomas MJ, et al. 2013. Lymphatic
vasculature mediates macrophage reverse cholesterol trans-
port in mice. J Clin Invest 123: 15711579.
Moens CB, Prince VE. 2002. Constructing the hindbrain: insights
from the zebrafish. Dev Dyn 224: 117.
Moens CB, Yan YL, Appel B, Force AG, Kimmel CB. 1996. valen-
tino: a zebrafish gene required for normal hindbrain segmenta-
tion. Development 122: 39813990.
Moens CB, Cordes SP, Giorgianni MW, Barsh GS, Kimmel
CB. 1998. Equivalence in the genetic control of hindbrain
segmentation in fish and mouse. Development 125: 381
391.
Moriguchi T, Hamada M, Morito N, Terunuma T, Hasegawa K,
Zhang C, Yokomizo T, Esaki R, Kuroda E, Yoh K, et al.
2006. MafB is essential for renal development and F4/80 ex-
pression in macrophages. Mol Cell Biol 26: 57155727.
Norrmen C, Ivanov KI, Cheng J, Zangger N, Delorenzi M, Jaquet
M, Miura N, Puolakkainen P, Horsley V, Hu J, et al. 2009.
FOXC2 controls formation and maturation of lymphatic col-
lecting vessels through cooperation with NFATc1. J Cell
Biol 185: 439457.
Ober EA, Olofsson B, Makinen T, Jin SW, Shoji W, Koh GY, Ali-
talo K, Stainier DY. 2004. Vegfc is required for vascular devel-
opment and endoderm morphogenesis in zebrafish. EMBO
Rep 5: 7884.
Okuda KS, Astin JW, Misa JP, Flores MV, Crosier KE, Crosier PS.
2012. lyve1 expression reveals novel lymphatic vessels and
new mechanisms for lymphatic vessel development in zebra-
fish. Development 139: 23812391.
Oliver G, Srinivasan RS. 2010. Endothelial cell plasticity: how to
become and remain a lymphatic endothelial cell. Develop-
ment 137: 363372.
Ostergaard P, Simpson MA, Connell FC, Steward CG, Brice G,
Woollard WJ, Dafou D, Kilo T, Smithson S, Lunt P, et al.
2011. Mutations in GATA2 cause primary lymphedema asso-
ciated with a predisposition to acute myeloid leukemia
(Emberger syndrome). Nat Genet 43: 929931.
Pendeville H, Winandy M, Manfroid I, Nivelles O, Motte P, Pas-
que V, Peers B, Struman I, Martial JA, Voz ML. 2008. Zebrafish
Sox7 and Sox18 function together to control arterial-venous
identity. Dev Biol 317: 405416.
Petrova TV, Karpanen T, Norrmen C, Mellor R, Tamakoshi T,
Finegold D, Ferrell R, Kerjaschki D, Mortimer P, Yla-Hert-
tuala S, et al. 2004. Defective valves and abnormal mural
cell recruitment underlie lymphatic vascular failure in lymph-
edema distichiasis. Nat Med 10: 974981.
Pham VN, Lawson ND, Mugford JW, Dye L, Castranova D, Lo B,
Weinstein BM. 2007. Combinatorial function of ETS tran-
scription factors in the developing vasculature. Dev Biol
303: 772783.
Sadl V, Jin F, Yu J, Cui S, Holmyard D, Quaggin S, Barsh G, Cordes
S. 2002. The mouse Kreisler (Krml1/MafB) segmentation gene
is required for differentiation of glomerular visceral epithelial
cells. Dev Biol 249: 1629.
Schulte-Merker S, Sabine A, Petrova TV. 2011. Lymphatic vascu-
lar morphogenesis in development, physiology, and disease. J
Cell Biol 193: 607618.
Sieweke MH, Tekotte H, Frampton J, Graf T. 1996. MafB is an in-
teraction partner and repressor of Ets-1 that inhibits erythroid
differentiation. Cell 85: 4960.
Sieweke MH, Tekotte H, Frampton J, Graf T. 1997. MafB repress-
es erythroid genes and differentiation through direct interac-
tion with c-Ets-1. Leukemia 11(Suppl 3):486488.
Srinivasan RS, Oliver G. 2011. Prox1 dosage controls the number
of lymphatic endothelial cell progenitors and the formation of
the lymphovenous valves. Genes Dev 25: 21872197.
Srinivasan RS, Geng X, Yang Y, Wang Y, Mukatira S, Studer M,
Porto MP, Lagutin O, Oliver G. 2010. The nuclear hormone
Mafb in lymphangiogenesis
GENES &DEVELOPMENT 1629
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
receptor Coup-TFII is required for the initiation and early
maintenance of Prox1 expression in lymphatic endothelial
cells. Genes Dev 24: 696707.
Srinivasan RS, Escobedo N, Yang Y, Interiano A, Dillard ME, Fin-
kelstein D, Mukatira S, Gil HJ, Nurmi H, Alitalo K, et al. 2014.
The Prox1Vegfr3 feedback loop maintains the identity and
the number of lymphatic endothelial cell progenitors. Genes
Dev 28: 21752187.
Stacker SA, Williams SP, Karnezis T, Shayan R, Fox SB, Achen
MG. 2014. Lymphangiogenesis and lymphatic vessel remodel-
ling in cancer. Nat Rev Cancer 14: 159172.
van Impel A, Zhao Z, Dorien MA, Hermkens M, Roukens G, Fi-
scher JC, Peterson-Maduro J, Duckers H, Ober EA, Ingham
PW, et al. 2014. Divergence of zebrafish and mouse lymphatic
cell fate specification pathways. Development 141: 1228
1238.
Villefranc JA, Nicoli S, Bentley K, Jeltsch M, Zarkada G, Moore
JC, Gerhardt H, Alitalo K, Lawson ND. 2013. A truncation al-
lele in vascular endothelial growth factor c reveals distinct
modes of signaling during lymphatic and vascular develop-
ment. Development 140: 14971506.
Wigle JT, Oliver G. 1999. Prox1 function is required for the devel-
opment of the murine lymphatic system. Cell 98: 769778.
Yaniv K, Isogai S, Castranova D, Dye L, Hitomi J, Weinstein BM.
2006. Live imaging of lymphatic development in the zebra-
fish. Nat Med 12: 711716.
Yoshimatsu Y, Yamazaki T, Mihira H, Itoh T, Suehiro J, Yuki K,
Harada K, MorikawaM, Iwata C, Minami T, et al. 2011. Etsfam-
ily members induce lymphangiogenesis through physical and
functional interaction with Prox1. J Cell Sci 124: 27532762.
Zheng W, Aspelund A, Alitalo K. 2014. Lymphangiogenic factors,
mechanisms, and applications. J Clin Invest 124: 878887.
Koltowska et al.
1630 GENES &DEVELOPMENT
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
10.1101/gad.263210.115Access the most recent version at doi:
2015 29: 1618-1630 Genes Dev.
Katarzyna Koltowska, Scott Paterson, Neil I. Bower, et al.
essential for embryonic lymphangiogenesis in zebrafish
is a downstream transcriptional effector of Vegfc signalingmafba
Material
Supplemental
http://genesdev.cshlp.org/content/suppl/2015/08/07/29.15.1618.DC1.html
References
http://genesdev.cshlp.org/content/29/15/1618.full.html#ref-list-1
This article cites 77 articles, 43 of which can be accessed free at:
License
Commons
Creative
.http://creativecommons.org/licenses/by-nc/4.0/at
Creative Commons License (Attribution-NonCommercial 4.0 International), as described
). After six months, it is available under ahttp://genesdev.cshlp.org/site/misc/terms.xhtml
six months after the full-issue publication date (see
This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first
Service
Email Alerting click here.right corner of the article or
Receive free email alerts when new articles cite this article - sign up in the box at the top
http://genesdev.cshlp.org/subscriptions
go to: Genes & Development To subscribe to
© 2015 Koltowska et al.; Published by Cold Spring Harbor Laboratory Press
Cold Spring Harbor Laboratory Press on January 10, 2016 - Published by genesdev.cshlp.orgDownloaded from
... Heterozygous hah mutants were out-crossed to WIK WT to establish a mapping cross. To identify the genetic cause of the hah phenotype, we used a genome sequencing mapping approach, as previously published [22,23]. Briefly, 45 homozygous hah vcc43 mutant embryos and 45 siblings at 4 dpf were pooled and homogenized, and genomic DNA was extracted using the Qiagen DNeasy ® Blood & Tissue Kit (Qiagen, Hilden, Germany). ...
... Reads were then mapped to the Danio rerio reference genome (danRer7/Zv9). Regions of high parent strain homozygosity were identified [24], and candidate variants were detected [22,23]. ...
... This mutant was named heart and head (hah vcc43 ). Genome sequencing and linkage analysis [22][23][24] of hah vcc43−/− embryos identified a single 18 Mb linkage peak on chromosome 17 ( Figure 1A). This interval is syntenic to a region on human chromosome 14q12 and contained several single nucleotide sequence variants ( Figure 1B). ...
Article
Full-text available
Dilated cardiomyopathy (DCM) is a common heart muscle disorder that frequently leads to heart failure, arrhythmias, and death. While DCM is often heritable, disease-causing mutations are identified in only ~30% of cases. In a forward genetic mutagenesis screen, we identified a novel zebrafish mutant, heart and head (hahvcc43), characterized by early-onset cardiomyopathy and craniofacial defects. Linkage analysis and next-generation sequencing identified a nonsense variant in the highly conserved scfd1 gene, also known as sly1, that encodes sec1 family domain-containing 1. Sec1/Munc18 proteins, such as Scfd1, are involved in membrane fusion regulating endoplasmic reticulum (ER)/Golgi transport. CRISPR/Cas9-engineered scfd1vcc44 null mutants showed severe cardiac and craniofacial defects and embryonic lethality that recapitulated the phenotype of hahvcc43 mutants. Electron micrographs of scfd1-depleted cardiomyocytes showed reduced myofibril width and sarcomere density, as well as reticular network disorganization and fragmentation of Golgi stacks. Furthermore, quantitative PCR analysis showed upregulation of ER stress response and apoptosis markers. Both heterozygous hahvcc43 mutants and scfd1vcc44 mutants survived to adulthood, showing chamber dilation and reduced ventricular contraction. Collectively, our data implicate scfd1 loss-of-function as the genetic defect at the hahvcc43 locus and provide new insights into the role of scfd1 in cardiac development and function.
... Importantly, a number of these TFs are already known to play important roles in lymphatics (e.g. NFAT Kulkarni et al, 2009;Norrmen et al, 2009, MAFB Dieterich et al, 2015Koltowska et al, 2015b, TBX1 Chen et al, 2010, and TCF Nicenboim et al, 2015Cha et al, 2018), supporting the prediction that members of these TF families will play important functional roles in LEC development. TF motifs enriched in DAPs that were more open in VECs were associated with blood vascular and venous identity as would be expected (e.g. ...
... To determine the functional relevance of the above observations, we took advantage of a notch1b uq53bh mutant strain that we had identified in an earlier forward genetic screen (Koltowska et al, 2015b). This mutant phenocopied earlier Notch1 morphant phenotypes (Geudens et al, 2010), mapped to the notch1b locus on Chromosome 5 by whole genome sequencing analysis and sequencing confirmed a nonsense mutation at amino acid position 128 (Fig 8D, Appendix Fig S2I) predicted to be a loss of function mutation. ...
... Published mutant lines used were prox1a i278 (van Impel et al, 2014) and prox1b sa35 (Tao et al, 2011;Koltowska et al, 2015a). The uq53bh mutant was isolated in a previously described genetic screen (Koltowska et al, 2015b). The genetic mapping was performed as previously described (Koltowska et al, 2015b) and identified a single region of linkage on Chromosome 5 containing the notch1b locus (data not shown). ...
Article
During development, the lymphatic vasculature forms as a second network derived chiefly from blood vessels. The transdifferentiation of embryonic venous endothelial cells (VECs) into lymphatic endothelial cells (LECs) is a key step in this process. Specification, differentiation and maintenance of LEC fate are all driven by the transcription factor Prox1, yet the downstream mechanisms remain to be elucidated. We here present a single-cell transcriptomic atlas of lymphangiogenesis in zebrafish, revealing new markers and hallmarks of LEC differentiation over four developmental stages. We further profile single-cell transcriptomic and chromatin accessibility changes in zygotic prox1a mutants that are undergoing a LEC-VEC fate shift. Using maternal and zygotic prox1a/prox1b mutants, we determine the earliest transcriptomic changes directed by Prox1 during LEC specification. This work altogether reveals new downstream targets and regulatory regions of the genome controlled by Prox1 and presents evidence that Prox1 specifies LEC fate primarily by limiting blood vascular and haematopoietic fate. This extensive single-cell resource provides new mechanistic insights into the enigmatic role of Prox1 and the control of LEC differentiation in development.
... Studies of mutants isolated from forward genetic screens or associated with human diseases led to the establishment of the Vegfc/Flt4 axis as the central pathway for lymphangiogenesis [26][27][28][29]. Accordingly, the currently growing list of trunk lymphangiogenesis regulators almost exclusively relates to molecules involved in Vegfc/ Flt4 signaling, some of them acting directly upstream such as CCBE1 [30][31][32][33] or the transcription factor HHEX [9] or downstream like the transcription factors Mafba [34] and Yap1 [35]. Acting through multiple intracellular events, including the activation of the common effector of Vegf receptors Erk [7], Vegfc signaling controls several aspects of lymphangiogenesis including LEC differentiation through Prox1 expression, proliferation, and migration after cell cycle arrest [36]. ...
... After their simultaneous budding from the PCV, venous sprouts rapidly diverge and develop into two differently fated structures: the intermediate pool of midline PLs that later give rise to the trunk lymphatic system and the ISVs that establish the venous intersegmental network. Expression of Prox1a in some ECs from the PCV correlates with their lymphatic fate [15,34]. Yet, recent data suggest that the arterial or venous fate of secondary sprouts could rather be defined upstream of secondary sprouting, at least partially by a Notch-driven heterogeneity preexisting in the primary ISVs [21]. ...
Article
Full-text available
Background Lymphangiogenesis, the formation of lymphatic vessels, is tightly linked to the development of the venous vasculature, both at the cellular and molecular levels. Here, we identify a novel role for Sorbs1, the founding member of the SoHo family of cytoskeleton adaptor proteins, in vascular and lymphatic development in the zebrafish. Results We show that Sorbs1 is required for secondary sprouting and emergence of several vascular structures specifically derived from the axial vein. Most notably, formation of the precursor parachordal lymphatic structures is affected in sorbs1 mutant embryos, severely impacting the establishment of the trunk lymphatic vessel network. Interestingly, we show that Sorbs1 interacts with the BMP pathway and could function outside of Vegfc signaling. Mechanistically, Sorbs1 controls FAK/Src signaling and subsequently impacts on the cytoskeleton processes regulated by Rac1 and RhoA GTPases. Inactivation of Sorbs1 altered cell-extracellular matrix (ECM) contacts rearrangement and cytoskeleton dynamics, leading to specific defects in endothelial cell migratory and adhesive properties. Conclusions Overall, using in vitro and in vivo assays, we identify Sorbs1 as an important regulator of venous and lymphatic angiogenesis independently of the Vegfc signaling axis. These results provide a better understanding of the complexity found within context-specific vascular and lymphatic development.
... LEC initial migration from the cardinal vein is dependent on the production of vascular endothelial growth factor C (Vegfc) and the activation of its primary receptor fms-related tyrosine kinase 4 (FLT4), also known as VEGFR3 (10,11). Activation of canonical or non-canonical VEGFR3-signaling, and enzymes controlling the proteolytic activation of VEGFC play an essential role in LEC initial migration as well as development and maintenance of lymphatic vessels (10,(12)(13)(14)(15). In addition, directed migration of LECs is controlled by the CXCL12/CXCR4 chemokine axis (16)(17)(18). ...
... Besides, decreased Vegfc expression in mesenchymal cells did not implicate a reduction at the transcriptional level of genes involved in the VEGFR3-signaling axis in E13.5 LECs from Osr1 GCE/GCE embryos. Indeed, Flt4, Hhex and other downstream targets of the signaling pathway such as Maf, or Egr1 (15,32,43,44) were upregulated in LECs from E13.5 Osr1 GCE/GCE embryos ( figure 6C). Since BECs are also a source of Vegfc, we analyzed Vegfc expression in E13.5 Osr1 GCE/GCE embryos via RT-qPCR analysis of FACS isolated BECs and did not observe a change in Vegfc expression ( figure S5C). ...
Preprint
Full-text available
The lymphatic system is formed during embryonic development by the commitment of specialized lymphatic endothelial cells (LECs) and their subsequent assembly in primary lymphatic vessels. While lymphatic cells are in continuous contact with mesenchymal cells during development and in adult tissues, the role of mesenchymal cells in lymphatic vasculature development remains poorly characterized. Here, we show that a subpopulation of mesenchymal cells expressing the transcription factor Osr1 are in close association with migrating LECs and established lymphatic vessels in mice. Lineage tracing experiments revealed that Osr1+ cells precede LEC arrival during lymphatic vasculature assembly in the back of the embryo. Using Osr1-deficient embryos and functional in vitro assays, we show that Osr1 acts in a non-cell autonomous manner controlling proliferation and early migration of LECs to peripheral tissues. Thereby, mesenchymal Osr1+ cells control in a bimodal manner the production of extracellular matrix scaffold components and signal ligands critical for lymphatic vessels formation.
... Upstream of Prox1, two transcription factors, SOX18 (SRY -Sex Determining Region Y- Box 18) and COUP-TFII (Chicken Ovalbumin Upstream Promoter Transcription Factor II -also known as NR2F2-), are expressed in the anterior cardinal vein during initial lymphatic fate specification and directly promote Prox1 expression by binding to its promoter 24,25 . More recently, the musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB) transcription factor has also been shown to propagate Prox1, Sox18, and Coup-TFII expression 26,27 (Fig. 1). ...
Article
Full-text available
Lymphangiogenesis refers to the generation of new lymphatic vessels from pre-existing ones. During development and particular adult states, lymphatic endothelial cells (LEC) undergo reprogramming of their transcriptomic and signaling networks to support the high demands imposed by cell proliferation and migration. Although there has been substantial progress in identifying growth factors and signaling pathways controlling lymphangiogenesis in the last decades, insights into the role of metabolism in lymphatic cell functions are just emerging. Despite numerous similarities between the main metabolic pathways existing in LECs, blood ECs (BEC) and other cell types, accumulating evidence has revealed that LECs acquire a unique metabolic signature during lymphangiogenesis, and their metabolic engine is intertwined with molecular regulatory networks, resulting in a tightly regulated and interconnected process. Considering the implication of lymphatic dysfunction in cancer and lymphedema, alongside other pathologies, recent findings hold promising opportunities to develop novel therapeutic approaches. In this review, we provide an overview of the status of knowledge in the molecular and metabolic network regulating the lymphatic vasculature in health and disease.
... APLNR signaling is required for the generation of cells that give rise to HSCs [34], and there is a prominent role of the IGF2/IGF2R system in promoting endothelial progenitor cells homing [35]. The MAFB gene encodes a transcription factor that controls endothelial sprouting in vitro and in vivo [36] and plays an important role in the embryonic development of the lymphatic vascular system [62,63]. IGFBP2 is known as a developmentally regulated gene that is highly expressed in embryonic and fetal tissues and markedly decreases after birth [64]. ...
Article
Full-text available
Human pluripotent stem cells (hPSCs) can be used as a renewable source of endothelial cells for treating cardiovascular disease and other ischemic conditions. Here, we present the derivation and characterization of a panel of distinct clonal embryonic endothelial progenitor cells (eEPCs) lines that were differentiated from human embryonic stem cells (hESCs). The hESC line, ESI-017, was first partially differentiated to produce candidate cultures from which eEPCs were cloned. Endothelial cell identity was assessed by transcriptomic analysis, cell surface marker expression, immunocytochemical marker analysis, and functional analysis of cells and exosomes using vascular network forming assays. The transcriptome of the eEPC lines was compared to various adult endothelial lines as well as various non-endothelial cells including both adult and embryonic origins. This resulted in a variety of distinct cell lines with functional properties of endothelial cells and strong transcriptomic similarity to adult endothelial primary cell lines. The eEPC lines, however, were distinguished from adult endothelium by their novel pattern of embryonic gene expression. We demonstrated eEPC line scalability of up to 80 population doublings (pd) and stable long-term expansion of over 50 pd with stable angiogenic properties at late passage. Taken together, these data support the finding that hESC-derived clonal eEPC lines are a potential source of scalable therapeutic cells and cell products for treating cardiovascular disease. These eEPC lines offer a highly promising resource for the development of further preclinical studies aimed at therapeutic interventions.
... APLNR signaling is required for the generation of cells that give rise to HSCs [33] and there is a prominent role of the IGF2/IGF2R system in promoting EPC homing [34]. The MAFB gene encodes a transcription factor that controls endothelial sprouting in vitro and in vivo [35] and plays an important role in the embryonic development of the lymphatic vascular system [61,62]. IGFBP2 is known as a developmentally regulated gene that is highly expressed in embryonic and fetal tissues and markedly decreases after birth [63]. ...
Preprint
Human pluripotent stem cells (hPSCs) can be used as a renewable source of endothelial cells for treating cardiovascular disease and other ischemic conditions. Here, we present the derivation and characterization of a panel of distinct clonal embryonic endothelial progenitor cell (eEPC) lines that were differentiated from human embryonic stem cells (hESCs). The hESC line, ESI-017, was first partially differentiated to produce candidate cultures from which eEPC were cloned. Endothelial cell identity was assessed by transcriptomic analysis, cell surface marker expression, immunocytochemical marker analysis, and functional analysis using a vascular network forming assay. The transcriptome of the eEPC lines was compared to various adult endothelial lines as well as various non-endothelial cells including both adult and embryonic origins. This resulted in a variety of distinct cell lines with functional properties of endothelial cells and strong transcriptomic similarity to adult endothelial primary cell lines. The eEPC lines, however, were distinguished from adult endothelium by a novel pattern of embryonic gene expression. We demonstrated scalability of up to 80 population doublings and stable with long-term expansion over 50 passages and stable angiogenic properties at late passage in the EPC line. Taken together, these data support the finding that hESC-derived clonal eEPC lines are useful as a source of scalable therapeutic cells and cell products for treating cardiovascular disease. These eEPC lines offer a highly promising resource for preclinical studies and therapeutic interventions.
... MAF bZIP transcription factor B (Mafb) has been identified as a downstream transcriptional effector of the VEGF-C/VEGFR3 axis, regulating the transcription factors PROX1, SOX18, COUP-TFII, and the kruppel-like transcription factor 4 in LECs (Dieterich et al., 2015;Koltowska et al., 2015). Mafb regulates morphogenesis of a subset of lymphatic beds, including the skin and diaphragm in mice (Dieterich et al., 2020;Rondon-Galeano et al., 2020). ...
Article
Full-text available
During development, lymphatic endothelial cell (LEC) progenitors differentiate from venous endothelial cells only in limited regions of the body. Thus, LEC migration and subsequent tube formation are essential processes for the development of tubular lymphatic vascular network throughout the body. In this review, we discuss chemotactic factors, LEC-extracellular matrix interactions and planar cell polarity regulating LEC migration and formation of tubular lymphatic vessels. Insights into molecular mechanisms underlying these processes will help in understanding not only physiological lymphatic vascular development but lymphangiogenesis associated with pathological conditions such as tumors and inflammation.
... MAFB, a member of the large MAF TF subfamily, has been implicated in the regulation of both lymphangiogenesis and sprouting angiogenesis. 261,262 Analysis of the actively translated transcriptome at different stages of postnatal retinal angiogenesis combined with promoter analysis identified MAFB as a key regulator of postnatal angiogenesis. 261 Mafb is enriched at the angiogenic front in postnatal retinas, while induced EC-specific deletion results in defective angiogenic expansion. ...
Article
Full-text available
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Article
During embryonic development, lymphatic endothelial cell (LEC) precursors are distinguished from blood endothelial cells by the expression of Prospero-related homeobox 1 (Prox1), which is essential for lymphatic vasculature formation in mouse and zebrafish. Prox1 expression initiation precedes LEC sprouting and migration, serving as the marker of specified LECs. Despite its crucial role in lymphatic development, Prox1 upstream regulation in LECs remains to be uncovered. SOX18 and COUP-TFII are thought to regulate Prox1 in mice by binding its promoter region. However, the specific regulation of Prox1 expression in LECs remains to be studied in detail. Here, we used evolutionary conservation and chromatin accessibility to identify enhancers located in the proximity of zebrafish prox1a active in developing LECs. We confirmed the functional role of the identified sequences through CRISPR/Cas9 mutagenesis of a lymphatic valve enhancer. The deletion of this region results in impaired valve morphology and function. Overall, our results reveal an intricate control of prox1a expression through a collection of enhancers. Ray-finned fish-specific distal enhancers drive pan-lymphatic expression, whereas vertebrate-conserved proximal enhancers refine expression in functionally distinct subsets of lymphatic endothelium.
Article
Full-text available
SoxF family members have been linked to arterio-venous specification events and human pathological conditions, but in contrast to Sox17 and Sox18, a detailed in vivo analysis of a Sox7 mutant model is still lacking. In this study we generated zebrafish sox7 mutants to understand the role of Sox7 during vascular development. By in vivo imaging of transgenic zebrafish lines we show that sox7 mutants display a short circulatory loop around the heart as a result of aberrant connections between the lateral dorsal aorta (LDA) and either the venous primary head sinus (PHS) or the common cardinal vein (CCV). In situ hybridization and live observations in flt4:mCitrine transgenic embryos revealed increased expression levels of flt4 in arterial endothelial cells at the exact location of the aberrant vascular connections in sox7 mutants. An identical circulatory short loop could also be observed in newly generated mutants for hey2 and efnb2. By genetically modulating levels of sox7, hey2 and efnb2 we demonstrate a genetic interaction of sox7 with hey2 and efnb2. The specific spatially confined effect of loss of Sox7 function can be rescued by overexpressing the Notch intracellular domain (NICD) in arterial cells of sox7 mutants, placing Sox7 upstream of Notch in this aspect of arterial development. Hence, sox7 levels are crucial in arterial specification in conjunction with hey2 and efnb2 function, with mutants in all three genes displaying shunt formation and an arterial block. © 2015. Published by The Company of Biologists Ltd.
Article
Full-text available
The mammalian lymphatic vasculature is important for returning fluids from the extracellular tissue milieu back to the blood circulation. We showed previously that Prox1 dosage is important for the development of the mammalian lymphatic vasculature. The lack of Prox1 activity results in the complete absence of lymphatic endothelial cells (LECs). In Prox1 heterozygous embryos, the number of LECs is reduced because of a decrease in the progenitor pool in the cardinal vein. This reduction is caused by some progenitor cells being unable to maintain Prox1 expression. In this study, we identified Vegfr3, the cognate receptor of the lymphangiogenic growth factor Vegfc, as a dosage-dependent, direct in vivo target of Prox1. Using various mouse models, we also determined that Vegfr3 regulates Prox1 by establishing a feedback loop necessary to maintain the identity of LEC progenitors and that Vegfc-mediated activation of Vegfr3 signaling is necessary to maintain Prox1 expression in LEC progenitors. We propose that this feedback loop is the main sensing mechanism controlling the number of LEC progenitors and, as a consequence, the number of budding LECs that will form the embryonic lymphatic vasculature.
Article
Full-text available
Lymphangiogenesis is a dynamic process that involves the sprouting of lymphatic endothelial cells (LECs) from veins to form lymphatic vessels. Vegfr3 signalling, through its ligand Vegfc and the extracellular protein Ccbe1, is essential for the sprouting of LECs to form the trunk lymphatic network. In this study we determined whether Vegfr3, Vegfc and Ccbe1 are also required for development of the facial and intestinal lymphatic networks in the zebrafish embryo. Whereas Vegfr3 and Ccbe1 are required for the development of all lymphatic vessels, Vegfc is dispensable for facial lymphatic sprouting but not for the complete development of the facial lymphatic network. We show that zebrafish vegfd is expressed in the head, genetically interacts with ccbe1 and can rescue the lymphatic defects observed following the loss of vegfc. Finally, whereas knockdown of vegfd has no phenotype, double knockdown of both vegfc and vegfd is required to prevent facial lymphatic sprouting, suggesting that Vegfc is not essential for all lymphatic sprouting and that Vegfd can compensate for loss of Vegfc during lymphatic development in the zebrafish head.
Article
Full-text available
The CRISPR/Cas9 system has been implemented in a variety of model organisms to mediate site-directed mutagenesis. A wide range of mutation rates has been reported, but at a limited number of genomic target sites. To uncover the rules that govern effective Cas9-mediated mutagenesis in zebrafish, we targeted over a hundred genomic loci for mutagenesis using a streamlined and cloning-free method. We generated mutations in 85% of target genes with mutation rates varying across several orders of magnitude, and identified sequence composition rules that influence mutagenesis. We increased rates of mutagenesis by implementing several novel approaches. The activities of poor or unsuccessful single-guide RNAs (sgRNAs) initiating with a 5' adenine were improved by rescuing 5' end homogeneity of the sgRNA. In some cases, direct injection of Cas9 protein/sgRNA complex further increased mutagenic activity. We also observed that low diversity of mutant alleles led to repeated failure to obtain frame-shift mutations. This limitation was overcome by knock-in of a stop codon cassette that ensured coding frame truncation. Our improved methods and detailed protocols make Cas9-mediated mutagenesis an attractive approach for labs of all sizes.
Article
Full-text available
Lymphatic vessels arise during development through sprouting of precursor cells from veins, which is regulated by known signaling and transcriptional mechanisms. The ongoing elaboration of vessels to form a network is less well understood. This involves cell polarization, coordinated migration, adhesion, mixing, regression, and shape rearrangements. We identified a zebrafish mutant, lymphatic and cardiac defects 1 (lyc1), with reduced lymphatic vessel development. A mutation in polycystic kidney disease 1a was responsible for the phenotype. PKD1 is the most frequently mutated gene in autosomal dominant polycystic kidney disease (ADPKD). Initial lymphatic precursor sprouting is normal in lyc1 mutants, but ongoing migration fails. Loss of Pkd1 in mice has no effect on precursor sprouting but leads to failed morphogenesis of the subcutaneous lymphatic network. Individual lymphatic endothelial cells display defective polarity, elongation, and adherens junctions. This work identifies a highly selective and unexpected role for Pkd1 in lymphatic vessel morphogenesis during development.
Article
Full-text available
Lymphangiogenesis, the growth of lymphatic vessels, is essential in embryonic development. In adults, it is involved in many pathological processes such as lymphedema, inflammatory diseases, and tumor metastasis. Advances during the past decade have dramatically increased the knowledge of the mechanisms of lymphangiogenesis, including the roles of transcription factors, lymphangiogenic growth factors and their receptors, and intercellular and intracellular signaling cascades. Strategies based on these mechanisms are being tested in the treatment of various human diseases such as cancer, lymphedema, and tissue allograft rejection. This Review summarizes the recent progress on lymphangiogenic mechanisms and their applications in disease treatment.
Article
Full-text available
The generation of new lymphatic vessels through lymphangiogenesis and the remodelling of existing lymphatics are thought to be important steps in cancer metastasis. The past decade has been exciting in terms of research into the molecular and cellular biology of lymphatic vessels in cancer, and it has been shown that the molecular control of tumour lymphangiogenesis has similarities to that of tumour angiogenesis. Nevertheless, there are significant mechanistic differences between these biological processes. We are now developing a greater understanding of the specific roles of distinct lymphatic vessel subtypes in cancer, and this provides opportunities to improve diagnostic and therapeutic approaches that aim to restrict the progression of cancer.
Article
As a result of numerous genome sequencing projects, large numbers of candidate open reading frames are being identified, many of which have no known function. Analysis of these genes typically involves the transfer of DNA segments into a variety of vector backgrounds for protein expression and functional analysis. We describe a method called recombinational cloning that uses in vitro site-specific recombination to accomplish the directional cloning of PCR products and the subsequent automatic subcloning of the DNA segment into new vector backbones at high efficiency. Numerous DNA segments can be transferred in parallel into many different vector backgrounds, providing an approach to high-throughput, in-depth functional analysis of genes and rapid optimization of protein expression. The resulting subclones maintain orientation and reading frame register, allowing amino- and carboxy-terminal translation fusions to be generated. In this paper, we outline the concepts of this approach and provide several examples that highlight some of its potential.
Article
Background: The interplay between Notch and Vegf signaling regulates angiogenesis in the embryo. Notch signaling limits the responsiveness of endothelial cells to Vegf to control sprouting. Despite the importance of this regulatory relationship, much remains to be understood about extrinsic factors that modulate the pathway. Results: During a forward genetic screen for novel regulators of lymphangiogenesis, we isolated a mutant with reduced lymphatic vessel development. This mutant also exhibited hyperbranching arteries, reminiscent of Notch pathway mutants. Positional cloning identified a missense mutation in the carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (cad) gene. Cad is essential for UDP biosynthesis, which is necessary for protein glycosylation and de novo biosynthesis of pyrimidine-based nucleotides. Using a transgenic reporter of Notch activity, we demonstrate that Notch signaling is significantly reduced in cad(hu10125) mutants. In this context, genetic epistasis showed that increased endothelial cell responsiveness to Vegfc/Vegfr3 signaling drives excessive artery branching. Conclusions: These findings suggest important posttranslational modifications requiring Cad as an unappreciated mechanism that regulates Notch/Vegf signaling during angiogenesis.