ArticlePDF Available

Mandriota SJ, Jussila L, Jeltsch M, Compagni A, Baetens D, Prevo R, Banerji S, Huarte J, Montesano R, Jackson DG, Orci L, Alitalo K, Christofori G, Pepper MSVascular endothelial growth factor-C mediated lymphangiogenesis promotes tumour metastasis. EMBO 20: 672-682

Authors:
  • Fondation des Grangettes

Abstract and Figures

Metastasis is a frequent and lethal complication of cancer. Vascular endothelial growth factor-C (VEGF-C) is a recently described lymphangiogenic factor. Increased expression of VEGF-C in primary tumours correlates with dissemination of tumour cells to regional lymph nodes. However, a direct role for VEGF-C in tumour lymphangiogenesis and subsequent metastasis has yet to be demonstrated. Here we report the establishment of transgenic mice in which VEGF-C expression, driven by the rat insulin promoter (Rip), is targeted to beta-cells of the endocrine pancreas. In contrast to wild-type mice, which lack peri-insular lymphatics, RipVEGF-C transgenics develop an extensive network of lymphatics around the islets of Langerhans. These mice were crossed with Rip1Tag2 mice, which develop pancreatic beta-cell tumours that are neither lymphangiogenic nor metastatic. Double-transgenic mice formed tumours surrounded by well developed lymphatics, which frequently contained tumour cell masses of beta-cell origin. These mice frequently developed pancreatic lymph node metastases. Our findings demonstrate that VEGF-C-induced lymphangiogenesis mediates tumour cell dissemination and the formation of lymph node metastases.
Content may be subject to copyright.
Stefano J.Mandriota
1
, Lotta Jussila
2
,
Michael Jeltsch
2
, Amelia Compagni
3
,
Danielle Baetens, Remko Prevo
4
,
Suneale Banerji
4
, Joachim Huarte,
Roberto Montesano, David G.Jackson
4
,
Lelio Orci, Kari Alitalo
2
, Gerhard Christofori
3
and Michael S.Pepper
5
Department of Morphology, University Medical Centre, 1 rue Michel
Servet, 1211 Geneva 4, Switzerland,
2
Molecular/Cancer Biology
Laboratory and Ludwig Institute for Cancer Research, Haartman
Institute, University of Helsinki, Finland,
3
Institute of Molecular
Pathology, Vienna, Austria and
4
MRC Human Immunology Unit,
Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
1
Present address: The Wellcome Trust Centre for Human Genetics,
Roosevelt Drive, Oxford, UK
5
Corresponding author
e-mail: michael.pepper@medecine.unige.ch
Metastasis is a frequent and lethal complication
of cancer. Vascular endothelial growth factor-C
(VEGF-C) is a recently described lymphangiogenic
factor. Increased expression of VEGF-C in primary
tumours correlates with dissemination of tumour cells
to regional lymph nodes. However, a direct role for
VEGF-C in tumour lymphangiogenesis and subse-
quent metastasis has yet to be demonstrated. Here we
report the establishment of transgenic mice in which
VEGF-C expression, driven by the rat insulin pro-
moter (Rip), is targeted to b-cells of the endocrine
pancreas. In contrast to wild-type mice, which lack
peri-insular lymphatics, RipVEGF-C transgenics
develop an extensive network of lymphatics around
the islets of Langerhans. These mice were crossed
with Rip1Tag2 mice, which develop pancreatic b-cell
tumours that are neither lymphangiogenic nor meta-
static. Double-transgenic mice formed tumours
surrounded by well developed lymphatics, which
frequently contained tumour cell masses of b-cell
origin. These mice frequently developed pancreatic
lymph node metastases. Our ®ndings demonstrate
that VEGF-C-induced lymphangiogenesis mediates
tumour cell dissemination and the formation of lymph
node metastases.
Keywords: islet of Langerhans/lymphangiogenesis/
tumour metastasis/VEGF-C
Introduction
The metastatic spread of tumour cells is responsible for the
majority of cancer deaths. Tumour cell dissemination is
mediated by a number of mechanisms, including local
tissue invasion, lymphatic spread, haematogenous spread,
or direct seeding of body cavities or surfaces. Clinical and
pathological observations have long suggested that, for
many tumours, the most common pathway of initial
dissemination is via lymphatics, with patterns of spread
via afferent vessels following routes of natural drainage
(reviewed by Fidler, 1997; Cotran et al., 1999; Sleeman,
2000). However, the lymphatic system has traditionally
been overshadowed by the greater emphasis placed on the
blood vascular system. This has been due in part to the
absence of suitable markers that distinguish lymphatic
from blood vascular endothelium, and to the lack of
identi®cation of lymphatic-speci®c growth factors.
In recent years, these limitations have been relieved by
the discovery of a small number of potential lymphatic-
speci®c markers (reviewed by Jackson, 2001). These
include: LYVE-1, a lymphatic endothelial receptor for
the extracellular matrix/lymphatic ¯uid mucopolysac-
charide hyaluronan (Banerji et al., 1999); Prox-1, a
homeobox gene product involved in regulating early
lymphatic development (Wigle and Oliver, 1999); podo-
planin, a glomerular podocyte membrane mucoprotein
(Breiteneder-Geleff et al., 1999); and the vascular
endothelial growth factor receptor-3 (VEGFR-3), a
transmembrane tyrosine kinase receptor for the lymphatic
endothelial growth factors vascular endothelial growth
factor-C (VEGF-C) and VEGF-D (reviewed in Veikkola
et al., 2000). Targeted inactivation of the VEGFR-3 gene
has revealed that it also plays an important role in the
development of the early blood vascular system, prior to
the emergence of lymphatic vessels (Dumont et al., 1998).
VEGF-C, the ®rst ligand to be discovered for VEGFR-3
(Joukov et al., 1996; Lee et al., 1996), is a member of the
VEGF family of polypeptide growth factors, which
comprises VEGF-A, -B, -C, -D and orf virus VEGFs (or
VEGF-E) (reviewed in Eriksson and Alitalo, 1999;
Ferrara, 1999). VEGF-C is produced in a pre-pro-peptide
form that is proteolytically processed to a mature
homodimer of ~40 kDa (Joukov et al., 1997). Proteolytic
processing increases the af®nity of VEGF-C for VEGFR-3
some 400-fold, and also enables it to bind to and activate
VEGFR-2 (Joukov et al., 1997). Based on its expression
pro®le and its binding to VEGFR-3, VEGF-C has been
implicated in the development of the lymphatic system
(Kukk et al., 1996; Lymboussaki et al., 1999). In addition,
transgenic overexpression of VEGF-C using the keratin 14
promoter induces lymphatic vessel enlargement/dilatation
in the skin (Jeltsch et al., 1997), and recombinant VEGF-C
induces lymphangiogenesis in the chick chorioallantoic
membrane (Oh et al., 1997). The capacity of VEGF-C to
bind to and activate VEGFR-2 may partially explain why
it also stimulates angiogenesis under certain experimental
conditions (Cao et al., 1998; Witzenbichler et al., 1998).
In contrast to VEGF-A, whose crucial role in tumour
angiogenesis is well established (Ferrara, 1999), very little
is known about the function of VEGF-C in tumour
Vascular endothelial growth factor-C-mediated
lymphangiogenesis promotes tumour metastasis
The EMBO Journal Vol. 20 No. 4 pp. 672±682, 2001
672 ã European Molecular Biology Organization
formation and/or progression. Nonetheless, a correlation
between VEGF-C expression, tumour lymphangiogenesis
and the formation of metastases in regional lymph nodes
has recently been described. Thus, levels of VEGF-C in
primary tumours are signi®cantly correlated with lymph
node metastases in a variety of cancers, including thyroid,
prostate, gastric, colorectal and lung (Bunone et al., 1999;
Tsurusaki et al., 1999; Yonemura et al., 1999; Akagi et al.,
2000; Niki et al., 2000; Ohta et al., 2000). One study has
described a strong correlation between lymphatic vessel
density and VEGF-C expression (Ohta et al., 1999).
However, in this study, no correlation was observed
between lymphatic vessel density and lymph node
metastases. Despite the continuing accumulation of
correlative clinical data, a functional role for VEGF-C in
tumour lymphangiogenesis and/or lymphatic enlargement,
and its role in tumour cell dissemination, have yet to be
demonstrated directly.
In order to test the hypothesis that VEGF-C-induced
lymphangiogenesis can promote tumour metastasis, we
generated transgenic mouse lines in which VEGF-C
expression, driven by the rat insulin promoter, is targeted
to b-cells of the islets of Langerhans, and tested their
capacity to form metastases by crossing them with the
Rip1Tag2 transgenic line, a transgenic mouse model of
non-metastatic b-cell carcinogenesis (Hanahan, 1985).
Results
A full-length human VEGF-C cDNA was cloned between
the rat insulin II gene promoter (Rip) and the SV40 small
T antigen intron and polyadenylation signal (Figure 1A).
Two founder (F0) mice (designated nos 23 and 24) were
identi®ed, which were capable of germline transmission.
RipVEGF-C transgenic mice were viable, similar in size to
wild-type littermates, normoglycaemic and fertile. Mice
derived from F0 no. 23 had a single copy of the
transgene, whereas those derived from F0 no. 24 had
four to six copies integrated in a head-to-tail array
(Figure 1B and data not shown). Pancreas-speci®c
expression of the VEGF-C transgene was con®rmed by
reverse transcription±polymerase chain reaction (RT±
PCR) screening of several organs, using oligonucleotide
primers designed to amplify VEGF-C cDNA of human but
not of mouse origin (Figure 1C and data not shown). In
contrast to wild-type littermates (Figure 2A), immuno-
histochemical analysis of the pancreata of RipVEGF-C
transgenic mice revealed strong VEGF-C staining in the
majority of islet cells (Figure 2B), as would be expected
from the relative abundance of insulin-expressing b-cells
in islets (~80%).
Striking morphological differences were observed
between transgenic and wild-type pancreata. Clearly
demarcated spaces, lined by a single layer of ¯attened
cells, which frequently contained lymphocytes rather than
red blood cells, were observed around the majority of
transgenic islets (Figure 3C and E). These morphological
features were observed in all RipVEGF-C mice killed
between E15.5 and 18 months. These structures, which
were never observed around islets of wild-type mice
(Figure 3A), were identi®ed as lymphatic vessels on the
basis of three separate criteria. First, they were lined by
endothelial cells that were immunoreactive for VEGFR-3
(Figure 2C). Weak VEGFR-3 immunostaining was
also observed within some islets of both wild-type and
transgenic mice (Figure 2C and data not shown).
Secondly, as revealed by transmission electron micro-
scopy (TEM), the basement membrane of these vessels
was either absent or discontinuous, and the endothelial
cells were devoid of the characteristic fenestrations found
in contiguous capillary blood vesselsÐboth hallmarks of
lymphatic endothelium (Figure 4A and B). Thirdly, as
revealed by immunohistochemistry and immuno-EM, they
stained with an antibody to the novel lymphatic endothe-
lial marker LYVE-1 (Banerji et al., 1999; Prevo et al.,
2001) on both their luminal and abluminal surfaces
(Figure 3D, F and 4C), similar to lymphatic vessels in
normal wild-type mouse tissues (Prevo et al., 2001).
Fig. 1. Molecular characterization of RipVEGF-C transgenic mice.
(A) The transgene was constructed by cloning the complete human
VEGF-C cDNA (nucleotides 1±1997; DDBJ/EMBL/GenBank
accession No. X94216) between the ~695 bp BamHI±XbaI fragment
of Rip (Hanahan, 1985) and the SV40 small T antigen intron and
polyadenylation signal. The L-shaped arrow indicates insulin gene
transcription initiation. E, EcoRI restriction sites. (B) Ten micrograms
of genomic DNA from two RipVEGF-C transgenic mice from families
23 and 24 or from a wild-type littermate (wt) (the latter spiked with the
RipVEGF-C transcriptional unit as indicated in picograms) were
digested with EcoRI and analysed by Southern blotting using the SV40
moiety of the transgene as a probe. Single or double asterisks indicate
the 3¢ end of the insertion site in family 23 and 24, respectively.
Markers on the left indicate kilobases. (C) Reverse transcription
products from oligo dT-primed total RNAs from pancreata of
RipVEGF-C transgenic mice from families 23 and 24 or from wild-
type mice (wt) were analysed by PCR using hVEGF-C speci®c primers
or acidic ribosomal phosphoprotein P0 (P0) primers. Where indicated,
RT was omitted, or PCR mix alone (mix) was analysed. Markers on the
left indicate base pairs.
VEGF-C-mediated lymphangiogenesis and metastasis
673
A quantitative analysis of lymphatic vessel density in
RipVEGF-C mice, based on LYVE-1 immunoreactivity,
revealed that 98% of islets were in intimate association
with LYVE-1-positive lymphatics (Figures 3D, F and 5;
Table I), which contrasts with the ®nding that only 4% of
islets had closely apposed lymphatics in wild-type
littermates (Figures 3B and 5; Table I). In some
RipVEGF-C mice, peri-insular lymphatics were particu-
larly well developed, and extended between the lobules of
exocrine acini (Figure 3F). Previous studies in the rodent
pancreas have revealed the absence of an association
between lymphatics and islets of Langerhans (Bertelli
et al., 1993; Navas et al., 1995; Ji and Kato, 1997;
reviewed by O'Morchoe, 1997), which is consistent with
our LYVE-1 and VEGFR-3 immunohistochemical data.
From these data we conclude that RipVEGF-C mice form
lymphatics de novo (lymphangiogenesis). In contrast,
mice expressing VEGF-C in the skin under the control of
the keratin 14 promoter display an increase both in the size
and number of lymphatic vessels (Jeltsch et al., 1997;
T.Veikkola, unpublished data).
Vascular density and the percentage islet volume
occupied by blood vessels were determined by immuno-
histochemistry with the vascular endothelial-speci®c anti-
body MECA-32 (Hallmann et al., 1995). In contrast to
LYVE-1, which stains lymphatic vessels (Banerji et al.,
1999), MECA-32 only stains blood vessels (Figure 6A and
B). No differences were observed between wild-type and
RipVEGF-C transgenic islets (Table I). Similar results
were obtained using the pan-endothelial marker CD31
(data not shown), indicating that forced expression of
VEGF-C did not promote vascular angiogenesis.
No alterations were observed in the normal proportion
or spatial distribution of a, b, d or PP cells in the islets of
RipVEGF-C mice when compared with wild-type litter-
mates, as assessed by immunohistochemical analysis for
glucagon, insulin, somatostatin and pancreatic polypep-
tide, respectively (data not shown).
We thus conclude that increased expression of VEGF-C
in the b-cells of RipVEGF-C transgenic mice speci®cally
promotes lymphangiogenesis, which is observed around
but not within the islets of Langerhans, and which does not
affect islet architecture or cellular composition.
To assess the role of VEGF-C-induced lymphangio-
genesis in tumour metastasis, we crossed RipVEGF-C
mice with Rip1Tag2 mice, a well characterized transgenic
model of non-metastatic b-cell carcinogenesis (Hanahan,
1985; Perl et al., 1998, 1999) that displays morphological
features typical of human pancreatic b-cell tumours (Holm
et al., 1988). Of note, b-cell tumours that develop in
Rip1Tag2 mice are capable of local invasion, but do not
induce extensive lymphangiogenesis (Figure 7A and E).
When RipVEGF-C mice were crossed with Rip1Tag2
mice, the resulting double-transgenic animals displayed a
modest but signi®cant increase in tumour incidence
(de®ned as the number of tumours per pancreas having a
size >0.5 mm) compared with single Rip1Tag2 transgenic
animals, yet neither tumour volume (expressed in cubic
millimetres) nor the ratio of benign adenoma to invasive
carcinoma was altered (Table II). RipVEGF-C 3
Rip1Tag2 double-transgenic tumours consisted almost
exclusively of insulin-producing b cells (data not shown),
similar to what has been reported for Rip1Tag2 single-
transgenic tumours (Hanahan, 1985). Detailed immuno-
histochemical analyses revealed heterogeneous VEGF-C
immunoreactivity in the majority of double-transgenic
tumour cells, whereas VEGF-C immunoreactivity was
absent from Rip1Tag2 tumours (Figure 7C and D).
Furthermore, there was an increase in the number of
VEGFR-3 immunoreactive peri-tumoural lymphatic
vessels in double-transgenic mice compared with single-
transgenic mice (data not shown). The extent of this new
lymphatic vessel formation was demonstrated dramatic-
ally by LYVE-1 immunostaining, which revealed that
insulinomas were either partly surrounded by or fully
enclosed within such vessels (Figure 7F). Indeed, quan-
titative analyses con®rmed that 62% of insulinomas/islets
in double-transgenic animals had closely apposed lym-
phatics encompassing most or all of the insulinoma/islet
circumference (Figures 5, 7B and F). In contrast, only 29%
of insulinomas/islets in the Rip1Tag2 mice had associated
lymphatics (Figures 5, 7A and E). The lower levels of
lymphangiogenesis in the latter tumours correlate with the
®nding that endogenous VEGF-C was not upregulated
during tumourigenesis in Rip1Tag2 single-transgenic mice
(Figure 7C and data not shown). In small adenomas of
double-transgenic mice, LYVE-1-positive lymphatics
surrounded the tumour tissue without penetrating it
(Figure 7F). In contrast, in large adenomas and carcin-
omas, in addition to surrounding the tumours, lymphatics
(positive for LYVE-1 and VEGFR-3) were infrequently
observed in the peripheral rim of the tumour, but never
extended into the body of the tumour itself (data not
shown).
Fig. 2. Expression of VEGF-C and VEGFR-3 in RipVEGF-C
transgenic mice. Immunohistochemistry for (A and B) VEGF-C and
(C) VEGFR-3. (A) Ten-month-old female wild-type-littermate; (B and
C) 11-month-old female RipVEGF-C mouse, family 24. (B) and (C)
are serial sections. Bar: (A and B) 50 mm; (C) 30 mm.
S.J.Mandriota et al.
674
It has recently been reported that VEGFR-3, although
absent from mature blood vessels, is induced in angiogenic
tumour blood vessel endothelium (Partanen et al., 1999;
Valtola et al., 1999). We have also observed VEGFR-3-
positive intra-tumoural vessels in our system, although
a large degree of heterogeneity was observed within
individual islet cell tumours: some regions were positive,
while other regions in the same tumour were devoid of
immunoreactivity (data not shown). Since we have never
observed LYVE-1-positive vessels within normal or
tumourigenic islets (Figures 3B, D, F, 6B, 7E and F), we
are con®dent that the VEGFR-3-positive intra-islet or
intra-tumoural staining we have seen is con®ned to vessels
of the blood vascular system. VEGFR-3-positive blood
vessels were observed within tumours in both Rip1Tag2
and RipVEGF-C 3 Rip1Tag2 double-transgenic mice
(data not shown).
Since VEGF-C can be both lymphangiogenic and
angiogenic, our next objective was to compare blood
vessel density in Rip1Tag2 mice versus RipVEGF-C 3
Rip1Tag2 double transgenics. Blood vessel density is
frequently used as a measure of tumour-induced angio-
genesis (reviewed by Weidner, 1998). Using the pan-
endothelial marker CD31 (Figure 6C and D), we found no
differences between the two genotypes (Table III).
Similarly, no differences were observed when vessel
density was determined in the peripheral or central parts of
the tumour (Table III). These CD31-positive structures are
blood vessels, since we have never observed LYVE-1-
positive vessels within tumours (see above). We thus
conclude that VEGF-C does not induce angiogenesis in
double-transgenic RipVEGF-C 3 Rip1Tag2 mice.
Further examination of pancreata from RipVEGF-C 3
Rip1Tag2 double transgenics aged between 12 and 15
weeks revealed aggregates of tumour cells within the
lumen of lymphatic vessels in every case (Figure 8 and
data not shown). These intra-lymphatic tumour cells,
which were very rarely seen in Rip1Tag2 single-transgenic
mice, were clearly derived from primary b-cell tumours, as
established by their ultrastructural characteristics and
insulin immunoreactivity (Figure 8C and data not
shown). A proportion of the cells within these aggregates
contained mitotic ®gures (Figure 8A and C), indicating
that they were actively dividing. As before, the lymphatic
nature of these vessels was con®rmed by both LYVE-1
and VEGFR-3 immunoreactivity (Figure 8B and data not
shown).
We reasoned that intravascular tumour cells, by follow-
ing routes of natural drainage, would reach the marginal
sinus of pancreatic mesenteric lymph nodes, thus forming
Fig. 3. Histological analysis of wild-type and RipVEGF-C mice. (A and C) Haematoxylin and eosin staining; (B, D and F) immunohistochemistry for
LYVE-1 (DAB brown) and insulin (B and D) (fast red); (E) semithin section stained with methylene blue. (A) Twelve-month-old male wild-type
littermate; (B) 2-month-old male wild-type littermate; (C) 12-month-old male RipVEGF-C mouse, family 24; (D) 2-month-old male RipVEGF-C
mouse, family 24; (E) 11-month-old male RipVEGF-C mouse, family 24; (F) 10-month-old male RipVEGF-C mouse, family 24. Arrows in (B)
indicate ducts; En, endocrine; Ex, exocrine. Bar: (A±D) 50 mm; (E) 25 mm; (F) 100 mm.
VEGF-C-mediated lymphangiogenesis and metastasis
675
metastases at these sites. We therefore analysed regional
lymph nodes in double-transgenic mice for the presence of
tumour cells. Tumour cells were indeed observed in
regional mesenteric lymph nodes in 37% of RipVEGF-C
3 Rip1Tag2 mice (Table II; Figure 9A). In relatively
early metastases, which contained a thin rim of tumour
cells con®ned to the periphery of affected lymph nodes,
tumour cells displayed ultrastructural features of b-cells or
were insulin immunoreactive (Figure 9B and C), thus
con®rming their metastatic nature and their derivation
from primary b-cell tumours. No PP-, somatostatin- or
glucagon-immunoreactive cells could be detected in
metastases (data not shown). At later stages of metastatic
growth, lymph node tissue was almost completely
replaced by tumour cells (data not shown). These
advanced metastases displayed morphological hetero-
geneity, including an altered nucleus/cytoplasm ratio and
nuclear atypia, both hallmarks of the cellular anaplasia that
accompanies the later phases of tumour progression (data
not shown). Consistent with these observations, insulin
immunoreactivity was only retained by a minority of
tumour cells in these metastases (data not shown). At the
time points analysed, no overt metastases were observed in
distant lymph nodes or in other organs of RipVEGF-C 3
Rip1Tag2 double-transgenic mice.
Peri-tumoural lymphatics, intra-lymphatic tumour cell
aggregates and lymph node metastases have been observed
Fig. 4. Ultrastructural analysis of RipVEGF-C transgenic mouse
pancreas. (A and B) TEM of the pancreas from a 2-month-old male
RipVEGF-C transgenic mouse from family 24. (B) is a higher
magni®cation of the area delimited in (A). Arrows in (B) indicate the
basement membrane surrounding a capillary blood vessel (Bv). Note
the absence of a basement membrane along the basal surface of the
lymphatic endothelium. Arrowheads indicate endothelial fenestrations
in the capillary blood vessel. Note the absence of fenestrations in the
lymphatic endothelium. (C) Immunoelectron microscopy analysis of
LYVE-1 distribution on lymphatic endothelium in the same animal as
shown in (A) and (B). Cross-striated collagen ®brils are on the
abluminal side of the vessel. Lv, lymphatic vessel; Ex, exocrine tissue;
En, endocrine tissue. Bar: (A) 5 mm; (B) 500 nm; (C) 250 nm.
Fig. 5. Quantitation of lymphatic vessel density and disposition
in wild-type, single- (RipVEGF-C and Rip1Tag2) and double-
(RipVEGF-C/Rip1Tag2) transgenic mice. Lymphatic vessels were
identi®ed by LYVE-1 immunoreactivity, and the per cent islet/
insulinoma perimeter surrounded by LYVE-1-positive structures was
determined. For wild type, 185 islets were analysed from ®ve mice; for
RipVEGF-C, 99 islets were analysed from ®ve mice; for Rip1Tag2,
232 islets were analysed from seven mice; for RipVEGF-C/Rip1Tag2,
322 islets were analysed from eight mice.
Table I. Blood vascular and lymphatic phenotypes in wild-type versus
RipVEGF-C mice
Wild type RipVEGF-C
MECA-32-positive 13.2 6 3.6 13.6 6 4.5
vascular pro®les/10 000 mm
2
(n = 30) (n = 30)
% islet volume occupied by 13.0 6 3.4 12.9 6 4.4
MECA-32-positive blood vessels (n = 30) (n = 30)
% islets with tightly apposed 3.6 98
LYVE-1-positive lymphatics (n = 185) (n = 99)
For MECA-32 staining, six RipVEGF-C mice and six wild-type
littermates were assessed (three mice from family 23 and three from
family 24 in each case); ®ve islets were analysed per animal. Values
are mean 6 SD. For LYVE-1 staining, a total of 185 islets were
assessed in ®ve wild-type mice, and 99 islets were assessed in ®ve
RipVEGF-C mice.
n = number of islets.
S.J.Mandriota et al.
676
in double-transgenic mice derived from the two founder
families (Nos 23 and 24). Thus, we can ®rmly exclude a
transgene integration effect as the reason for the metastatic
phenotype.
Discussion
In the present study we have generated transgenic mice to
investigate the consequences of VEGF-C overexpression
on tumour cell dissemination in a well de®ned model of
b-cell carcinogenesis. Two lines were generated, which
express VEGF-C under the control of Rip. Although in
post-natal life this promoter speci®cally directs transgene
expression to pancreatic b-cells, during embryogenesis it
is transiently active in cells of the neural tube and neural
crest (Alpert et al., 1988). In almost all (98%) islets of
Langerhans analysed in RipVEGF-C transgenic mice,
extensive lymphatic channel formation was observed
around (but not within) the islets, the anatomical units in
which the transgene is expressed. In contrast, in wild-type
littermates, islets are very rarely in direct contact with
LYVE-1-positive vessels, indicating that in RipVEGF-C
mice lymphatics have formed de novo (lymphangio-
genesis). The latter result is consistent with the lymph-
angiogenic effects of VEGF-C observed in transgenic
mice which express VEGF-C in the skin under the control
of the keratin 14 promoter (T.Veikkola, unpublished data)
and in the differentiated avian chorioallantoic membrane
(Oh et al., 1997). K14-VEGF-C mice also display an
increase in the size of lymphatic vessels (Jeltsch et al.,
1997). The parameters that dictate whether VEGF-C will
induce lymphangiogenesis or lymphatic enlargement/
dilatation are not known. However, it has been well
documented that the activity of many cytokines is context
dependent, and it will therefore be important in the future
to determine what (additional) factors are involved in
determining this selectivity.
To assess the role of VEGF-C-induced lymphangio-
genesis in tumour metastasis, we crossed RipVEGF-C
mice with Rip1Tag2 mice, a well characterized transgenic
model of b-cell carcinogenesis (Hanahan, 1985; Perl et al.,
1998, 1999). Of note, b-cell tumours that develop in
Rip1Tag2 mice are capable of local invasion but are not
metastatic. In our model, increased VEGF-C expression in
double transgenics resulted in the de novo formation of
lymphatics in intimate association with b-cell tumours.
Concomitant with lymphangiogenesis, all of these mice
exhibited tumour cell aggregates within lymphatic vessels.
This was associated with the formation of metastases in
the draining regional mesenteric lymph nodes of the
pancreata in 37% of mice. While intra-lymphatic tumour
cell masses were occasionally seen in Rip1Tag2 mice,
lymph node metastases have never been observed in these
animals.
Although the metastatic dissemination of tumour cells
to regional lymph nodes is a common feature of many
human cancers, it is not clear whether tumours utilize
existing lymphatic channels or whether tumour dissemin-
ation requires the de novo formation of lymphatics
(lymphangiogenesis) (Saaristo et al., 2000; reviewed by
Fig. 6. MECA-32 and CD31 immunohistochemistry in wild-type and transgenic islets. (A) MECA-32 and (B) LYVE-1 immunolabelling in
consecutive frozen sections of a RipVEGF-C islet. CD31 immunolabelling of adenomas from 14-week-old (C) Rip1Tag2 and (D) double-transgenic
RipVEGF-C 3 Rip1Tag2 mice. L, lymphatic vessel; A, adenoma. Bar: 100 mm.
VEGF-C-mediated lymphangiogenesis and metastasis
677
Pepper, 2001). Our ®ndings suggest that Rip1Tag2 tumour
cells are intrinsically metastatic, but are unable to realize
their metastatic potential because they lack access to
appropriate portals and routes to distant sites. When such
routes are provided, in this instance via the lymphangio-
genesis induced by ectopically expressed VEGF-C, then
metastases are observed. The ®nding that increased peri-
tumoural lymphatic density results in metastasis in only
37% of the mice that develop islet tumours suggests that
additional, rate-limiting steps must also exist.
RipVEGF-C 3 Rip1Tag2 double-transgenic tumours
were similar to Rip1Tag2 single-transgenic tumours in
terms of tumour volume, transition from adenoma to
carcinoma and the relative distribution of pancreatic
hormone immunoreactive cells. Interestingly, however,
tumour incidence was increased in RipVEGF-C 3
Rip1Tag2 mice. Although we have no explanation for
this increase, we speculate that this could be due to the
formation of local intra-pancreatic metastases, which
occur as a consequence of tumour cell dissemination
through the extensive peri-tumoural lymphatic network
induced by VEGF-C. Alternatively, trophic and/or mito-
genic signals released by lymphatic endothelial cells in
the nearby parenchyma of islets/tumours might have a
positive effect on islet/tumour cell survival and/or pro-
liferation, thus increasing the fraction of islets that form
primary tumours.
In addition to its capacity to induce lymphangiogenesis
and an increase in the size of lymphatic vessels, VEGF-C
has also been demonstrated to be angiogenic under certain
circumstances (Cao et al., 1998; Witzenbichler et al.,
1998). However, we found no evidence for increased
angiogenesis in RipVEGF-C or RipVEGF-C 3 Rip1Tag2
double-transgenic mice, as determined by blood vessel
density. Thus, there was no increase in the number of
MECA-32- or CD31-positive blood vascular pro®les
Fig. 7. Histological analysis of RipVEGF-C/Rip1Tag2 double-
transgenic mice. (A and B) Haematoxylin and eosin staining; (C and
D) anti-VEGF-C immunohistochemistry; (E and F) anti-LYVE-1
immunohistochemistry. (A, C and E) Rip1Tag2 single-transgenic mice.
(B, D and F) RipVEGF-C/Rip1Tag2 double-transgenic mice. Ad,
adenoma; Lv, lymphatic vessel; Ex, exocrine tissue; D, duct. The white
space around the adenoma in (E) is artefactual. All mice were killed at
14 weeks; double-transgenic mice are from RipVEGF-C family 23.
Bar: 50 mm.
Table II. Tumour phenotypes and metastasis in Rip1Tag2 versus
RipVEGF-C 3 Rip1Tag2 mice
Rip1Tag2 RipVEGF-C 3
Rip1Tag2
Tumour incidence
a
5.4 6 1.4 12.3 6 2.9
(per mouse) (n = 27) (n = 33)
Tumour volume
b
56.5 6 35.8 38.3 6 29.1
(per mouse) (n = 27) (n = 33)
Adenoma 36% 46%
(n = 17) (n = 27)
Carcinoma 64% 54%
(n = 17) (n = 27)
Lymph node metastases
c
0% 37%
(0/17) (10/27)
a
Tumour incidence per mouse was determined macroscopically by
counting all apparent tumours >0.5 mm in the whole pancreas.
b
Tumour volume per mouse (in mm
3
) was calculated from all
macroscopically apparent tumours in the whole pancreas, with a
diameter >0.5 mm.
c
As detected by haematoxylin and eosin staining.
Values are mean 6 SD; n = number of mice.
Table III. Vascular phenotype in Rip1Tag2 versus
RipVEGF-C 3 Rip1Tag2 mice
CD31-positive vascular pro®les/
10 000 mm
2
Rip1Tag2 RipVEGF-C 3
Rip1Tag2
Tumour periphery 6.6 6 2.8 4.8 6 2.4
(n = 30) (n = 30)
Tumour centre 3.9 6 2.5 3.7 6 2.3
(n = 30) (n = 30)
For CD31 staining, two Rip1Tag2 and three RipVEGF-C 3 Rip1Tag2
mice were assessed. Ten adenomas were analysed per genotype;
vascular pro®les were counted in three (~300 3 300 mm) randomly
selected peripheral and three central areas per adenoma. Values are
mean 6 SD; n = number of ®elds.
S.J.Mandriota et al.
678
within RipVEGF-C islets when compared with wild-type
littermate controls, nor could we detect alterations in the
percentage of islet volume occupied by these vessels
(demonstrating that vessel size was likewise unaltered).
Similarly, there was no difference in vessel density
between Rip1Tag2 versus RipVEGF-C 3 Rip1Tag2
mice, either in the centre of the tumour or in the tumour
periphery. We have recently generated Rip1Tag2 3
RipVEGF-A double-transgenic mice (manuscript in pre-
paration). Extensive phenotypic characterization of these
animals has revealed that VEGF-A has an exclusively
angiogenic effect during tumour progression. This observ-
ation points to the fact that in the setting of the pancreas,
exquisite selectivity with regard to angiogenesis and
lymphangiogenesis can be achieved with VEGF-A and
VEGF-C, respectively, despite the fact that both cytokines
can activate VEGFR-2. Although currently there is no
explanation for this selectivity, this may depend on the
extent of VEGF-C proteolytic processing or on the
repertoire of VEGFRs expressed (including heterodimers
between different VEGFRs).
VEGFR-3 is downregulated in blood vascular endo-
thelium during development, and becomes restricted
almost exclusively to lymphatic endothelium and fenes-
trated blood vascular endothelium in post-natal life
(Kaipainen et al., 1995; Kukk et al., 1996; Jussila et al.,
1998; Partanen et al., 2000). It has been shown that
VEGFR-3 is re-expressed in the blood vascular endo-
thelium of several types of tumours (Partanen et al., 1999;
Valtola et al., 1999). Although we observed a variable
degree of VEGFR-3 staining on blood vascular endo-
thelium in Rip1Tag2 tumours, this was not increased in the
tumours of double-transgenic RipVEGF-C 3 Rip1Tag2
mice.
It has long been argued that lymphatic vessels may be
lost and/or collapsed in expanding primary tumours
because of high endogenous tumour interstitial pressure
(Leu et al., 2000). However, their presence can be
Fig. 8. Intra-lymphatic tumour cell masses in RipVEGF-C/Rip1Tag2 double-transgenic mice. (A) TEM showing a tumour cell mass within an
endothelial-lined lymphatic space. (B) Anti-LYVE-1 immunohistochemistry. (C) Enlargement of the area delimited in (A) showing a mitotic ®gure
and endocrine secretory granules. Mice were killed at 14 weeks and were generated from RipVEGF-C family 23. Arrowheads in (A) indicate mitotic
®gures. Bars: (A) 20 mm; (B) 50 mm; (C) 2 mm.
VEGF-C-mediated lymphangiogenesis and metastasis
679
demonstrated in the peri-tumoural stromal component
intimately associated with several types of tumours. In our
system, LYVE-1-positive lymphatics were occasionally
observed in the periphery of large adenomas, but were
never seen within islets of Langerhans or b-cell tumours.
The reasons for this selective localization of lymphatic
vessels around but not within normal and tumorigenic
islets of Langerhans is not known.
A correlation between VEGF-C expression, tumour
lymphangiogenesis and the formation of metastases in
regional lymph nodes has recently been described (Bunone
et al., 1999; Ohta et al., 1999, 2000; Tsurusaki et al., 1999;
Yonemura et al., 1999; Akagi et al., 2000; Niki et al.,
2000). Although these ®ndings provide no information on
the mechanisms of tumour cell dissemination, they raise
the possibility that VEGF-C may increase metastasis by
increasing the number and size of lymphatic vessels, or
alternatively by altering the functional properties of
existing lymphatics. For example, it is possible that
lymphatic endothelial cells locally release paracrine
factors, which in¯uence primary tumour cell invasiveness,
possibly through altered tumour cell±extracellular matrix
adhesion. Alternatively, VEGF-C may facilitate tumour
cell intravasation by altering the functional properties of
pre-existing or newly formed lymphatic endothelium (e.g.
tumour cell±endothelial cell adhesion). Future studies in
this area will need to be conducted on cultured lymphatic
endothelial cells. Although to date all in vitro studies have
been performed on large-vessel lymphatic endothelial
cells (reviewed by Pepper, 2001), it will be important in
the future to work with primary endothelial cells from
lymphatic capillaries. These cells would be useful (i) for
the identi®cation of VEGF-C-induced phenotypic alter-
ations (e.g. molecules involved in adhesive interactions
with tumour cells) and (ii) for application of techniques of
differential gene expression to identify molecular differ-
ences between blood and lymphatic capillary endothelial
cells. The utility of these techniques in identifying gene
expression pro®les in normal versus tumour endothelial
cells has recently been demonstrated (St Croix et al.,
2000).
Tumour cell metastasis to regional lymph nodes is an
early event in metastatic tumour spread, and is frequently
used as a prognostic factor to predict disease outcome or to
determine therapeutic strategies. Our observations provide
the ®rst direct evidence for a causal role for VEGF-C-
mediated lymphangiogenesis in the dissemination of
tumour cells. They also provide a relevant animal model
in which the mechanisms of lymphangiogenesis and
lymphatic tumour metastasis can be dissected, and in
which potential inhibitors of these processes can be tested.
Materials and methods
Transgenic mice
RipVEGF-C transgenic mice were generated according to standard
procedures (Hogan et al., 1994). The transgene was constructed by
cloning the human VEGF-C cDNA between the ~695 bp BamHI±XbaI
fragment of Rip (Hanahan, 1985) and the SV40 small T antigen intron and
polyadenylation signal. Genotypes were con®rmed by Southern blotting
and PCR analysis. Ten micrograms of genomic DNA from mouse tails
Fig. 9. Lymph node metastases in RipVEGF-C/Rip1Tag2 double-transgenic mice. (A) Haematoxylin and eosin staining of a lymph node containing
a peripheral ring of tumour cells (T). (B) Immuno¯uorescent antibody staining for insulin. (C) TEM showing tumour cells (T) surrounded by
lymphocytes. The inset is an enlargement of the delimited area, and shows the presence of endocrine secretory granules in tumour cells. Mice were
killed at 14 weeks and were generated from RipVEGF-C family 23. Bar: (A) 200 mm; (B) 50 mm; (C) 10 mm; inset = 2 mm.
S.J.Mandriota et al.
680
were digested with EcoRI, run in 0.7% agarose gels and analysed by
Southern blotting using the ~450 bp SV40 moiety of the RipVEGF-C
transgene as a probe. Routine PCR screening of RipVEGF-C
heterozygotes was performed using a pair of primers speci®c for the
human VEGF-C cDNA (forward: 5¢-TCCGGACTCGACCTCTCGGAC;
reverse 5¢-CCCCACATCTATACACACCTCC), starting from standard
tail or ®nger genomic DNA preparations. PCR cycles were: 94°C, 3 min
(13); 94°C, 1 min, 60°C, 1 min, 72°C, 1 min (303); 72°C, 3 min (13).
PCR products were analysed on 6% polyacrylamide gels.
Expression of the RipVEGF-C transgene was assessed by RT±PCR
using primers speci®c for the human VEGF-C cDNA, and by
immunohistochemical analysis as described below. Total RNA puri®ed
from mouse pancreas using TRIZOL Reagent (Gibco-BRL, Life
Technologies) was reverse transcribed using oligo-dT (Boehringer) and
Superscript II (Life Technologies). RT products were subjected to PCR
analysis using a pair of primers speci®c for human VEGF-C (see above)
or a pair of primers for the acidic ribosomal phosphoprotein P0 (Pepper
and Mandriota, 1998). Where indicated, RT was omitted. PCR cycles
were: 94°C, 3 min (13); 94°C, 1 min, 60°C, 1 min, 72°C, 1 min (303 for
VEGF-C; 25±303 for P0); 72°C, 3 min (13). Equal volumes of PCR
products were analysed on 6% polyacrylamide gels.
The generation and phenotypic characterization of Rip1Tag2 mice
have been described previously (Hanahan, 1985). Glucose (5% w/v) was
added to the drinking water of all tumour-bearing mice, beginning at
10 weeks of age, to counteract hypoglycaemia, which results from
insulinoma development.
Histopathological analyses
For light microscopy, pancreata were either frozen or ®xed in Bouin's
®xative or 4% paraformaldehyde, and embedded in paraf®n. Antibodies
used for immunohistochemistry were as follows: mouse monoclonal anti-
human insulin (Storch et al., 1985); rabbit polyclonal anti-human
VEGF-C (Joukov et al., 1997); rabbit polyclonal anti-human VEGFR-3
(Pajusola et al., 1993); rat monoclonal anti-mouse VEGFR-3 (Kubo et al.,
2000); MECA-32 (Hallmann et al., 1995); rat monoclonal anti-mouse
CD31 (clone MEC13.3; PharMingen); rabbit polyclonal anti-mouse
LYVE-1 ectodomain. The latter was generated by immunizing rabbits
with the mouse LYVE-1 ectodomain fused with human IgFc followed by
pre-absorption of the antiserum on human Ig±Sepharose (Prevo et al.,
2001). MECA-32 was applied to cryostat sections. All other antibodies
were applied to paraf®n-embedded material following antigen retrieval
(microwave), with the exception of MEC13.3, for which no antigen
retrieval was used. Immunoreactivity was visualized using either
peroxidase±DAB or fast red- or ¯uorescein isothiocyanate-conjugated
streptavidin or secondary antibodies. For CD31 immunostaining,
antigen±antibody complexes were revealed by means of a rabbit anti-
rat linker antibody (Dako) followed by anti-rabbit/HRP Envision system
(Dako).
For determination of blood vessel density, the number of MECA-32- or
CD31-positive vascular pro®les per 10 000 mm
2
of islet or adenoma tissue
was determined using an in-house morphometric apparatus. For
adenomas, three randomly selected ®elds (~300 3 300 mm each) at the
periphery and in the centre of each tumour were counted. The per cent
islet volume occupied by MECA-32-positive vessels was determined
using the point counting method (Weibel, 1973). Values are mean 6 SD.
For electron microscopy, mice were either perfused with 1±2%
glutaraldehyde in 100 mM phosphate buffer pH 7.4 and the pancreas
isolated, or freshly isolated pancreatic tissue was minced and ®xed by
immersion in 2% glutaraldehyde for 2 h. After washing in 100 mM
phosphate buffer, tissue fragments were post-®xed in 2% osmium
tetroxide, dehydrated in graded ethanols and embedded in Epon 812. Thin
sections were stained with uranyl acetate and lead citrate, and viewed in a
Philips CM10 electron microscope. For immunoelectron microscopy,
islets were dissected from perfused pancreatic tissue, infused with 2.3 M
sucrose and processed for cryo-ultramicrotomy (Tokuyasu, 1980).
Ultrathin cryosections were incubated using the protein A±gold technique
(Roth et al., 1978) with LYVE-1 antibody (diluted 1/50) followed by
protein A coupled to 10 nm gold particles.
Acknowledgements
The project presented in this manuscript was conceived and started in
Helsinki, and the work is the result of an equal contribution from the
laboratories in Helsinki, Vienna and Geneva, together with a major
contribution from the Oxford group. We thank Drs Hajime Kubo and
Shin-Ishi Nishikawa for the anti-VEGFR-3 antibody, and Dr Jocelyn
Holash for advice with CD31 immunohistochemistry. We also thank
Danielle Ben Nasr, Mireille Quayzin, Marie Ebrahim Malek, Gorana
Perrelet and Petra Wilgenbus for excellent technical assistance, and
Nadine Dupont and Gerard Negro for photographic work. R.M., M.S.P.
and L.O. are supported by grants from the Swiss National Science
Foundation (Nos 31-43364.95 and 31-43366.95). A.C. and G.C.
gratefully acknowledge support by Boehringer Ingelheim and by the
Austrian Industrial Research Promotion Fund. K.A., L.J. and M.J. thank
the Finnish Academy, the Sigrid Juselius Foundation, the University of
Helsinki Hospital (TYH 8105), the State Technology Development
Centre as well as the EU Biomed programs (BMH-CT96-0669 and 98-
3380) for support. D.G.J. is supported by the UK Medical Research
Council (MRC Human Immunology Unit) and a project grant
(No. 00-311) from the Association for International Cancer Research.
References
Akagi,K., Ikeda,Y., Miyazaki,M., Abe,T., Kinoshita,J., Maehara,Y. and
Sugimachi,K. (2000) Vascular endothelial growth factor-C (VEGF-C)
expression in human colorectal cancer tissues. Br. J. Cancer, 83,
887±891.
Alpert,S., Hanahan,D. and Teitelman,G. (1988) Hybrid insulin genes
reveal a developmental lineage for pancreatic endocrine cells and
imply a relationship with neurons. Cell, 53, 295±308.
Banerji,S., Ni,J., Wang,S.X., Clasper,S., Su,J., Tammi,R., Jones,M. and
Jackson,D.G. (1999) LYVE-1, a new homologue of the CD44
glycoprotein, is a lymph-speci®c receptor for hyaluronan. J. Cell
Biol., 144, 789±801.
Bertelli,E., Regoli,M. and Comparini,L. (1993) Histotopographic and
ultrastructural study on the lymphatic network of the pancreas in the
guinea pig. Acta Anat. (Basel), 147, 233±239.
Breiteneder-Geleff,S. et al. (1999) Angiosarcomas express mixed
endothelial phenotypes of blood and lymphatic capillaries:
podoplanin as a speci®c marker for lymphatic endothelium. Am. J.
Pathol., 154, 385±394.
Bunone,G., Vigneri,P., Mariani,L., Buto,S., Collini,P., Pilotti,S.,
Pierotti,M.A. and Bongarzone,I. (1999) Expression of angiogenesis
stimulators and inhibitors in human thyroid tumors and correlation
with clinical±pathologic features. Am. J. Pathol., 155, 1967±1976.
Cao,Y., Linden,P., Farnebo,J., Cao,R., Eriksson,A., Kumar,V., Qi,J.H.,
Claesson-Welsh,L. and Alitalo,K. (1998). Vascular endothelial growth
factor C induces angiogenesis in vivo. Proc. Natl Acad. Sci. USA, 95,
14389±14394.
Cotran,R.S., Kumar,V. and Tucker,C. (1999) Neoplasia. In Robbins
Pathologic Basis of Disease, 6th edn. W.B.Saunders Co.,
Philadelphia, PA, pp. 268±271.
Dumont,D.J., Jussila,L., Taipale,J., Lymboussaki,A., Mustonen,T.,
Pajusola,K., Breitman,M. and Alitalo,K. (1998) Cardiovascular
failure in mouse embryos de®cient in VEGF receptor-3. Science,
282, 946±949.
Eriksson,U. and Alitalo,K. (1999) Structure, expression and receptor-
binding properties of novel vascular endothelial growth factors. Curr.
Top. Microbiol. Immunol., 237, 41±57.
Ferrara,N. (1999) Vascular endothelial growth factor: molecular and
biological aspects. Curr. Top. Microbiol. Immunol., 237, 1±30.
Fidler,I.J. (1997) Molecular biology of cancer: invasion and metastasis.
In DeVita,V.T., Hellman,S. and Rosenberg,S.A. (eds), Cancer:
Principles and Practice of Oncology, 5th edn. Lippincott-Raven,
Philadelphia, PA, pp. 135±152.
Hallmann,R., Mayer,D.N., Berg,E.L., Broermann,R. and Butcher,E.C.
(1995) Novel mouse endothelial cell surface marker is suppressed
during differentiation of the blood brain barrier. Dev. Dyn., 202,
325±332.
Hanahan,D. (1985) Heritable formation of pancreatic b-cell tumors in
transgenic mice expressing recombinant insulin/simian virus 40
oncogenes. Nature, 315, 115±122.
Hogan,B., Beddington,R., Costantini,F. and Lacy,E. (1994)
Manipulating the Mouse Embryo. Cold Spring Harbor Laboratory
Press, Cold Spring Harbor, NY, 497 pp.
Holm,R., Vardnell,I.M., Power,R.F., Bishop,A.E., Madsen,O.D.,
Alpert,S., Hanahan,D. and Polak,J.M. (1988) Ultrastructure and
electron immunocytochemistry of insulin-producing B-cell tumors
from transgenic mice: comparison with counterpart human tumors.
Ultrastruct. Pathol., 12, 547±559.
Jackson,D.G. (2001) New molecular markers for the study of tumour
lymphangiogenesis. Anticancer Res., in press.
VEGF-C-mediated lymphangiogenesis and metastasis
681
Jeltsch,M. et al. (1997) Hyperplasia of lymphatic vessels in VEGF-C
transgenic mice. Science, 276, 1423±1425.
Ji,R.C. and Kato,S. (1997) Demonstration of the intralobular lymphatics
in the guinea pig pancreas by an enzyme±histochemical method. J.
Anat., 191, 15±22.
Joukov,V., Pajusola,K., Kaipainen,A., Chilov,D., Lahtinen,I., Kukk,E.,
Saksela,O., Kalkkinen,N. and Alitalo,K. (1996) A novel vascular
endothelial growth factor, VEGF-C, is a ligand for the Flt4
(VEGFR-3) and KDR (VEGFR-2) receptor tyrosine kinases.
EMBO J., 15, 290±298.
Joukov,V., Sorsa,T., Kumar,V., Jeltsch,M., Claesson-Welsh,L., Cao,Y.,
Saksela,O., Kalkkinen,N. and Alitalo,K. (1997) Proteolytic processing
regulates receptor speci®city and activity of VEGF-C. EMBO J., 16,
3898±3911.
Jussila,L. et al. (1998) Lymphatic endothelium and Kaposi's sarcoma
spindle cells detected by antibodies against the vascular endothelial
growth factor receptor-3. Cancer Res., 58, 1599±1604.
Kaipainen,A., Korhonen,J., Mustonen,T., van Hinsbergh,V.W.M.,
Fang,G.-H., Dumont,D., Breitman,M. and Alitalo,K. (1995)
Expression of the fms-like tyrosine kinase 4 gene becomes restricted
to lymphatic endothelium during development. Proc. Natl Acad. Sci.
USA, 92, 3566±3570.
Kubo,H. et al. (2000) Involvement of vascular endothelial growth factor
receptor-3 in maintenance of integrity of endothelial cell lining during
tumor angiogenesis. Blood, 96, 546±553.
Kukk,E., Lymboussaki,A., Taira,S., Kaipainen,A., Jeltsch,M., Joukov,V.
and Alitalo,K. (1996) VEGF-C receptor binding and pattern of
expression with VEGFR-3 suggests a role in lymphatic vascular
development. Development, 122, 3829±3827.
Lee,J., Gray,A., Yuan,J., Luoh,S.M., Avraham,H. and Wood,W.I. (1996)
Vascular endothelial growth factor-related protein: a ligand and
speci®c activator of the tyrosine kinase receptor Flt4. Proc. Natl Acad.
Sci. USA, 93, 1988±1992.
Leu,A.J., Berk,D.A., Lymboussaki,A., Alitalo,K. and Jain R.K. (2000)
Absence of functional lymphatics within a murine sarcoma: a
molecular and functional evaluation. Cancer Res., 60, 4324±4327.
Lymboussaki,A., Olofsson,B., Eriksson,U. and Alitalo,K. (1999)
Vascular endothelial growth factor (VEGF) and VEGF-C show
overlapping binding sites in embryonic endothelia and distinct sites
in differentiated adult endothelia. Circ. Res., 85, 992±999.
Navas,V., O'Morchoe,P.J. and O'Morchoe,C.C.C. (1995) Lymphatic
system of the rat pancreas. Lymphology, 28, 4±20.
Niki,T., Iba,S., Tokunou,M., Yamada,T., Matsuno,Y. and Hirohashi,S.
(2000) Expression of vascular endothelial growth factors A, B, C and
D and their relationships to lymph node status in lung
adenocarcinoma. Clin. Cancer Res., 6, 2431±2439.
Oh,S.J., Jeltsch,M.M., Birkenhager,R., McCarthy,J.E., Weich,H.A.,
Christ,B., Alitalo,K. and Wilting,J. (1997) VEGF and VEGF-C:
speci®c induction of angiogenesis and lymphangiogenesis in the
differentiated avian chorioallantoic membrane. Dev. Biol., 188,
96±109.
Ohta,Y., Shridhar,V., Bright,R.K., Kalemkerian,G.P., Du,W.,
Carbone,M., Watanabe,Y. and Pass,H.I. (1999) VEGF and VEGF
type C play an important role in angiogenesis and lymphangiogenesis in
human malignant mesothelioma tumors. Br. J. Cancer, 81, 54±61.
Ohta,Y., Nozawa,H., Tanaka,Y., Oda,M. and Watanabe,Y. (2000)
Increased vascular endothelial growth factor and vascular
endothelial growth factor-C and decreased nm23 expression
associated with microdissemination in the lymph nodes in stage I
non-small cell lung cancer. J. Thorac. Cardiovasc. Surg., 119,
804±813.
O'Morchoe,C.C.C. (1997) Lymphatic system of the pancreas. Microsc.
Res. Tech., 37, 456±477.
Pajusola,K., Aprelikova,O., Armstrong,E., Morris,S. and Alitalo,K.
(1993) Two human FLT4 receptor tyrosine kinase isoforms with
distinct carboxy terminal tails are produced by alternative processing
of primary transcripts. Oncogene, 8, 2931±2937.
Partanen,T.A., Alitalo,K. and Miettinen,M. (1999) Lack of lymphatic
vascular speci®city of vascular endothelial growth factor receptor-3 in
185 vascular tumors. Cancer, 86, 2406±2412.
Partanen,T.A., Arola,J., Saaristo,A., Jussila,L., Ora,A., Miettinen,M.,
Stacker,S.A., Achen,M.G. and Alitalo,K. (2000) VEGF-C and VEGF-
D expression in neuroendocrine cells and their receptor, VEGFR-3, in
fenestrated blood vessels in human tissues. FASEB J., 14, 2087±2096.
Pepper,M.S. (2001) Lymphangiogenesis and tumor metastasis: myth or
reality? Clin. Cancer Res., in press.
Pepper,M.S. and Mandriota,S.J. (1998) Regulation of vascular
endothelial growth factor receptor-2 (Flk-1) expression in vascular
endothelial cells. Exp. Cell Res., 241, 414±425.
Perl,A.K., Wilgenbus,P., Dahl,U., Semb,H. and Christofori,G. (1998) A
causal role for E-cadherin in the transition from adenoma to
carcinoma. Nature, 392, 190±193.
Perl,A.K., Dahl,U., Wilgenbus,P., Cremer,H., Semb,H. and Christofori,G.
(1999) Reduced expression of neural cell adhesion molecule (N-CAM)
induces metastatic dissemination of pancreatic b tumor cells. Nature
Med., 5, 286±291.
Prevo,R., Banerji,S., Ferguson,D., Clasper,S. and Jackson,D. (2001)
Mouse LYVE-1 is an endocytic receptor for hyaluronan in lymphatic
endothelium. J. Biol. Chem., in press.
Roth,J., Bendayan,M. and Orci,L. (1978) Ultrastructural localization of
intracellular antigens by the use of protein A±gold technique. J.
Histochem. Cytochem., 26, 1074±1081.
Saaristo,A., Karpanen,T. and Alitalo,K. (2000) Mechanisms of
angiogenesis and lymphangiogenesis and their use in the inhibition
of tumor growth and metastasis. Oncogene, 19, 6122±6129.
Sleeman,J.P. (2000). The lymph node as a bridgehead in the metastatic
dissemination of tumors. Recent Results Cancer Res., 157, 55±81.
St Croix,B. et al. (2000) Genes expressed in human tumor endothelium.
Science, 289, 1197±1202.
Storch,M.J., Petersen,K.G., Licht,T. and Kerp,L. (1985) Recognition of
human insulin and proinsulin by monoclonal antibodies. Diabetes, 34,
808±811.
Tokuyasu,K.T. (1980) Immunochemistry on ultrathin frozen sections.
Histochem. J., 12, 381±403.
Tsurusaki,T., Kanda,S., Sakai,H., Kanetake,H., Saito,Y., Alitalo,K. and
Koji,T. (1999) Vascular endothelial growth factor-C expression in
human prostatic carcinoma and its relationship to lymph node
metastasis. Br. J. Cancer, 80, 309±313.
Valtola,R. et al. (1999) VEGFR-3 and its ligand VEGF-C are associated
with angiogenesis in breast cancer. Am. J. Pathol., 154, 1381±1390.
Veikkola,T., Karkkainen,M., Claesson-Welsh,L. and Alitalo,K. (2000)
Regulation of angiogenesis via vascular endothelial growth factor
receptors. Cancer Res., 60, 203±212.
Weibel,E.R. (1973) Stereological techniques for electron microscope
morphometry. In Hayat,M.A. (ed.), Principles and Techniques of
Electron Microscopy, Biological Applications. Vol. 3. Van Nostrand
Reinholdt, New York, NY, pp. 237±296.
Weidner,N. (1998) Tumoural vascularity as a prognostic factor in cancer
patients: the evidence continues to grow. J. Pathol., 184, 119±122.
Wigle,J.T. and Oliver,G. (1999) Prox1 function is required for the
development of the murine lymphatic system. Cell, 98, 769±778.
Witzenbichler,B. et al. (1998) Vascular endothelial growth factor-C
(VEGF-C/VEGF-2) promotes angiogenesis in the setting of tissue
ischemia. Am. J. Pathol., 153, 381±394.
Yonemura,Y. et al. (1999) Role of vascular endothelial growth factor C
expression in the development of lymph node metastasis in gastric
cancer. Clin. Cancer Res., 5, 1823±1829.
Received October 20, 2000; revised November 22, 2000;
accepted January 3, 2001
Note added in proof
The reader is referred to the following two papers, which, using VEGF-C-
and VEGF-D-transfected tumour cells in xenotransplantation models,
obtained ®ndings comparable to those described in the present paper:
Skobe,M., Hawighorst,T., Jackson,D.G., Prevo,R., Velasco,P.,
Riccardi,L., Alitalo,K., Claffey,K. and Detmar,M. (2001) Induction of
tumour lymphangiogenesis by VEGF-C promotes breast cancer
metastasis. Nature Med., 7, in press.
Stacker,S.A. et al. (2001) VEGF-D promotes metastatic spread of tumor
cells by the lymphatics. Nature Med., 7, in press.
S.J.Mandriota et al.
682
... Nonetheless, tumor-infiltrating lymphocytes and immuneassociated gene expression signatures have prognostic value in BC, particularly in triple-negative BC and BC with a BC cells into lymphatic vessels [36]. Inflammatory cytokines and growth factors released in the TME have been found to reduce junctional vascular endothelial-cadherin in lymphatic vessels in BC models [37,38], thus making these vessels more permissive for cellular intravasation, whereas expression of the lipoxygenase ALOX15 in BC cells has been reported to induce lymphatic defects allowing invasion of entire cell clusters [39]. Of note, in a rat BC model, lymphatic vessels predominantly contained tumor cell clusters that had a high metastatic capacity [40]. ...
... While the extent of tumor-associated lymphangiogenesis in the various subtypes of BC is still not clear, its relevance for metastases has been demonstrated in cancer models where tumor cell overexpression of the lymphangiogenic growth factors VEGF-C and -D is sufficient to increase lymphatic metastases [37,38,41]. Clinical support in several (but not all) comprehensive meta-analyses representing thousands of BC cases (Table 1) observed tumor lymphatic vascular density and the expression of VEGF-C (and -D) to correlate with poor outcome in BC. ...
Article
Full-text available
This paper is a cross fertilization of ideas about the importance of molecular aspects of breast cancer metastasis by basic scientists, a pathologist, and clinical oncologists at the Henry Ford Health symposium. We address four major topics: (i) the complex roles of lymphatic endothelial cells and the molecules that stimulate them to enhance lymph node and systemic metastasis and influence the anti-tumor immunity that might inhibit metastasis; (ii) the interaction of molecules and cells when breast cancer spreads to bone, and how bone metastases may themselves spread to internal viscera; (iii) how molecular expression and morphologic subtypes of breast cancer assist clinicians in determining which patients to treat with more or less aggressive therapies; (iv) how the outcomes of patients with oligometastases in breast cancer are different from those with multiple metastases and how that could justify the aggressive treatment of these patients with the hope of cure.
... Fundamentally cancerous cells release proangiogenic factors like VEGF (angiogenic factor) that induce tumor microenvironment network for lymphatics and blood vessels. In the next step, cells have to translocate to the mesenchymal site for further localization; here translocation is triggered with the removal of adhesive cellular components like β-catenin and E-cadherin (Mandriota et al. 2001). Removal of these adhesive proteins is crucial for cancerous cell drift from epithelial origin to mesenchymal sites. ...
Chapter
Globally, cancer ranks second for mortality rate with the mark of >ten million deaths in 2020 only. Cancer is characterized by the transmutation of normal healthy cells to abnormal cancerous cells in a multistage tumor progression. Generally, cancer is categorized as malignant (or metastatic) and benign. In metastatic embedment, lymphatics are the favorable site for carcinogenic foreign invaders. The progression of lymphatic cancer causes uncontrolled growth of all the immune cells which ensures infection of almost every organ. However, in normal individuals, lymphatics are regarded as a prominent framework for host defense with the major components of helper T-cells and B cells. Currently, available treatment modalities include painful procedures of surgical interventions, chemotherapy in combination with immunotherapy, and radiation therapy. These traditional treatments encounter various limitations which affect the socioeconomic status of a patient and their family wards. However, promising pharmaceutical nanoengineered approaches change the face of treatment and keep the pace in the patient’s recovery when compared with conventional treatment. From all the nanotechnological approaches, nanocrystal is regarded as a smart nanosized carrier as it offers advantages like targeting precision, high drug loading, devoid of organic solvents or solubilizing agents, and ease of entry within the lymphatic system via leaky tumor vasculature. Recent advancements in multifunctional nanocrystal have shown the potential of nanocrystal as a versatile delivery vehicle for lymphatic targeting with anticancer therapeutics. In this chapter, we will describe lymphatic cancer development, the need for amalgamation of nanotechnology with anticancer therapeutics, nanocrystals feasibility in lymphatic cancer targeting,
... VEGFC binds to the receptor tyrosine kinase VEGF receptor-3 (VEGFR3) with high affinity, but the processed form of VEGFC can also bind to the related VEGFA receptor VEGFR2 (Joukov et al, 1997). Mouse models overexpressing VEGFC show marked increase in lymphangiogenesis and metastasis to sentinel lymph nodes (Mandriota et al, 2001;Skobe et al, 2001), and inhibition of VEGFR3 by blocking antibodies inhibits lymphangiogenesis and restricts lymph node metastasis (He et al, 2005;Burton et al, 2008). VEGFA/VEGFR2 are also important in lymphangiogenesis, promoting sentinel node metastasis in different mouse models of skin cancer (Nagy et al, 2002;Björndahl et al, 2005;Hirakawa et al, 2005). ...
Article
Full-text available
The endothelial junction component vascular endothelial (VE)–cadherin governs junctional dynamics in the blood and lymphatic vasculature. Here, we explored how lymphatic junction stability is modulated by elevated VEGFA signaling to facilitate metastasis to sentinel lymph nodes. Zippering of VE-cadherin junctions was established in dermal initial lymphatic vessels after VEGFA injection and in tumor-proximal lymphatics in mice. Shape analysis of pan-cellular VE-cadherin fragments revealed that junctional zippering was accompanied by accumulation of small round-shaped VE-cadherin fragments in the lymphatic endothelium. In mice expressing a mutant VEGFR2 lacking the Y949 phosphosite ( Vegfr2 Y949F/Y949F ) required for activation of Src family kinases, zippering of lymphatic junctions persisted, whereas accumulation of small VE-cadherin fragments was suppressed. Moreover, tumor cell entry into initial lymphatic vessels and subsequent metastatic spread to lymph nodes was reduced in mutant mice compared with WT, after challenge with B16F10 melanoma or EO771 breast cancer. We conclude that VEGFA mediates zippering of VE-cadherin junctions in initial lymphatics. Zippering is accompanied by increased VE-cadherin fragmentation through VEGFA-induced Src kinase activation, correlating with tumor dissemination to sentinel lymph nodes.
... These breeding crosses involve other oncogenes and can alter tumor phenotype and behavior dramatically. For example, RT2 crossed with RIP mice expressing higher IGF-1R, VEGF-A, VEGF-C, VEGF-D, tenascin-C, or heparinase have been observed to increase tumor growth, angiogenesis, and/or invasion in separate studies [89][90][91][92][93][94]. Interestingly, in a model of Rag −/− adaptive-immunity-depleted mice crossed with RT2, anti-angiogenic treatment would lead to smaller tumors more capable of metastases [95]. ...
Article
Full-text available
Simple Summary Pancreatic neuroendocrine neoplasms (pNENs) are a rare and understudied cancer. Some of this low knowledge base is due to a historical lack of study models. Study models have tremendous implications for validating data for a range of cancer research topics, including treatment development. Therefore, correctly choosing a model is imperative and needs to consider a range of factors pertinent to the research question. In this review, we summarized the current field of models in pNENs. We considered factors, including complexity, accuracy, and cost, in models ranging from cell line cultures, 3D cultures, and whole organismal models, including mice and zebrafish. Improving the number and quality of models available will lead to new breakthroughs in treating pNENs and may lead to findings beneficial for other cancers. Abstract Pancreatic neuroendocrine neoplasms (pNENs) are a heterogeneous group of tumors derived from multiple neuroendocrine origin cell subtypes. Incidence rates for pNENs have steadily risen over the last decade, and outcomes continue to vary widely due to inability to properly screen. These tumors encompass a wide range of functional and non-functional subtypes, with their rarity and slow growth making therapeutic development difficult as most clinically used therapeutics are derived from retrospective analyses. Improved molecular understanding of these cancers has increased our knowledge of the tumor biology for pNENs. Despite these advances in our understanding of pNENs, there remains a dearth of models for further investigation. In this review, we will cover the current field of pNEN models, which include established cell lines, animal models such as mice and zebrafish, and three-dimensional (3D) cell models, and compare their uses in modeling various disease aspects. While no study model is a complete representation of pNEN biology, each has advantages which allow for new scientific understanding of these rare tumors. Future efforts and advancements in technology will continue to create new options in modeling these cancers.
... In the context of cancer, overexpression of VEGF-C induces the formation and remodeling of lymphatic vessels within and around primary tumors [96]. In addition, the aberrant activation of VEGFR-3 signaling by tumor-derived VEGF-C promotes metastatic spread of tumor cells via the lymphatics [96,97]. Moreover, VEGF-C secreted by primary tumors stimulates lymphangiogenesis in the draining LNs even before metastasis occurs [98], inducing a permissive "lymphovascular niche" to ensure successful colonization and long-term survival of metastatic cells at the secondary site [99]. ...
Article
Full-text available
Recent advances in RNA sequencing technologies helped uncover what was once uncharted territory in the human genome-the complex and versatile world of long noncoding RNAs (lncRNAs). Previously thought of as merely transcriptional "noise", lncRNAs have now emerged as essential regulators of gene expression networks controlling development, homeostasis and disease progression. The regulatory functions of lncRNAs are broad and diverse, and the underlying molecular mechanisms are highly variable, acting at the transcriptional, post-transcriptional, translational, and post-translational levels. In recent years, evidence has accumulated to support the important role of lncRNAs in the development and functioning of the lymphatic vasculature and associated pathological processes such as tumor-induced lymphangiogenesis and cancer metastasis. In this review, we summarize the current knowledge on the role of lncRNAs in regulating the key genes and pathways involved in lymphatic vascular development and disease. Furthermore, we discuss the potential of lncRNAs as novel therapeutic targets and outline possible strategies for the development of lncRNA-based therapeutics to treat diseases of the lymphatic system.
Article
Full-text available
The tumor microenvironment is a complex network of cells, extracellular matrix, and signaling molecules that plays a critical role in tumor progression and metastasis. Lymphatic and blood vessels are major routes for solid tumor metastasis and essential parts of tumor drainage conduits. However, recent studies have shown that lymphatic endothelial cells (LECs) and blood endothelial cells (BECs) also play multifaceted roles in the tumor microenvironment beyond their structural functions, particularly in hepatocellular carcinoma (HCC). This comprehensive review summarizes the diverse roles played by LECs and BECs in HCC, including their involvement in angiogenesis, immune modulation, lymphangiogenesis, and metastasis. By providing a detailed account of the complex interplay between LECs, BECs, and tumor cells, this review aims to shed light on future research directions regarding the immune regulatory function of LECs and potential therapeutic targets for HCC.
Article
Background & Aims The class I‐ phosphatidylinositol‐3 kinases (PI3Ks) signalling is dysregulated in almost all human cancers whereas the isoform‐specific roles remain poorly investigated. We reported that the isoform δ (PI3Kδ) regulated epithelial cell polarity and plasticity and recent developments have heightened its role in hepatocellular carcinoma (HCC) and solid tumour progression. However, its role in cholangiocarcinoma (CCA) still lacks investigation. Approach & Results Immunohistochemical analyses of CCA samples reveal a high expression of PI3Kδ in the less differentiated CCA. The RT‐qPCR and immunoblot analyses performed on CCA cells stably overexpressing PI3Kδ using lentiviral construction reveal an increase of mesenchymal and stem cell markers and the pluripotency transcription factors. CCA cells stably overexpressing PI3Kδ cultured in 3D culture display a thick layer of ECM at the basement membrane and a wide single lumen compared to control cells. Similar data are observed in vivo, in xenografted tumours established with PI3Kδ‐overexpressing CCA cells in immunodeficient mice. The expression of mesenchymal and stemness genes also increases and tumour tissue displays necrosis and fibrosis, along with a prominent angiogenesis and lymphangiogenesis, as in mice liver of AAV8‐based‐PI3Kδ overexpression. These PI3Kδ‐mediated cell morphogenesis and stroma remodelling were dependent on TGFβ/Src/Notch signalling. Whole transcriptome analysis of PI3Kδ using the cancer cell line encyclopedia allows the classification of CCA cells according to cancer progression. Conclusions Overall, our results support the critical role of PI3Kδ in the progression and aggressiveness of CCA via TGFβ/src/Notch‐dependent mechanisms and open new directions for the classification and treatment of CCA patients.
Chapter
Lymphangiogenesis is the process of formation of new lymphatic vessels from already existing lymphatic capillaries. This process is especially important in cancer metastasis as lymphangiogenesis is the major pathway of cancer cells metastasizing to the regional lymph nodes. Therefore, the mechanism underlying lymphangiogenesis in the local onco-sphere is crucial in determining the metastatic potential of a solid tumor. In this chapter, we will summarize the detailed mechanism and function of cancer lymphangiogenesis. We will also outline the various tumor-derived chemokines that drives activation of lymphangiogenesis, that ultimately leads to cancer metastasis in the regional and distal onco-sphere.
Chapter
During cancer metastasis, cancer cells spread by dissemination into nearby normal tissue mostly via two major routes (i) lymph nodes and (ii) blood vessels. Cancer cells can travel through lymphatic system or extravasate into blood vessels to other parts of the body and forming new tumors at metastatic sites. In this chapter, we will focus on how vascular remodeling in the local onco-sphere could affect lymph node metastasis. We will also discuss the importance of the crosstalk between tumoral endothelial cells (lymphatic vessels, perivascular cells, endothelial progenitor cells) and tumor cells in lymph node metastasis in the regional onco-sphere.
Article
Background: The prognosis of nodal recurrence after surgery for non-functioning pancreatic neuroendocrine tumors (NF-PanNETs) and its predictors have been poorly investigated. This study aimed to compare clinicopathologic features and survival between patients with nodal relapse and those with distant relapse and to identify predictors of nodal relapse after surgery for NF-PanNETs. Methods: All patients (n = 321) submitted to surgery for NF-PanNETs were included. Nodal recurrence was defined as the presence of one or more enlarged LNs at high-quality radiologic examinations and always confirmed by 68Ga-DOTA-PET or biopsy. Results: Altogether, 21 patients (6 %) experienced nodal (± distant) relapse, and 35 patients (11 %) had distant recurrence alone. Isolated nodal recurrence occurred for 23 % of patients with recurrence. Overall, 11 patients died of disease, one of whom (pT3N1G3) had an isolated nodal relapse. The rate of LN metastases (81 % vs 54 %; p = 0.044) and median number of positive LNs (PLN) (3 vs 0; p = 0.019) both were significantly higher for the patients with nodal (± distant) relapse than for those with distant relapse alone. Microvascular invasion (p = 0.046), T stage (p = 0.004), N stage (N1 [p = 0.049]; N2 [p = 0.001]), M stage (p < 0.001), and necrosis (p = 0.011) independently predicted nodal relapse. After distal pancreatectomy (n = 182), 13 patients experienced nodal recurrence, 9 of whom had left paraortic LNs involvement. Discussion: Lymph nodes are not rare sites of recurrence after surgery for NF-PanNETs. Lymph node involvement is a powerful determinant of nodal relapse. Nodal relapse frequently involves LNs that are not removed during standard lymphadenectomy.
Article
Full-text available
No growth factors specific for the lymphatic vascular system have yet been described. Vascular endothelial growth factor (VEGF) regulates vascular permeability and angiogenesis, but does not promote lymphangiogenesis. Overexpression of VEGF-C, a ligand of the VEGF receptors VEGFR-3 and VEGFR-2, in the skin of transgenic mice resulted in lymphatic, but not vascular, endothelial proliferation and vessel enlargement. Thus, VEGF-C induces selective hyperplasia of the lymphatic vasculature, which is involved in the draining of interstitial fluid and in immune function, inflammation, and tumor metastasis. VEGF-C may play a role in disorders involving the lymphatic system and may be of potential use in therapeutic lymphangiogenesis.
Article
The vascular endothelial growth factor family has recently been expanded by the isolation of two new VEGF-related factors, VEGF-B and VEGF-C. The physiological functions of these factors are largely unknown. Here we report the cloning and characterization of mouse VEGF-C, which is produced as a disulfide-linked dimer of 415 amino acid residue polypeptides, sharing an 85% identity with the human VEGF-C amino acid sequence. The recombinant mouse VEGF-C protein was secreted from transfected cells as VEGFR-3 (Flt4) binding polypeptides of 30–32x10(3) Mr and 22–23x10(3) Mr which preferentially stimulated the autophosphorylation of VEGFR-3 in comparison with VEGFR-2 (KDR). In in situ hybridization, mouse VEGF-C mRNA expression was detected in mesenchymal cells of postimplantation mouse embryos, particularly in the regions where the lymphatic vessels undergo sprouting from embryonic veins, such as the perimetanephric, axillary and jugular regions. In addition, the developing mesenterium, which is rich in lymphatic vessels, showed strong VEGF-C expression. VEGF-C was also highly expressed in adult mouse lung, heart and kidney, where VEGFR-3 was also prominent. The pattern of expression of VEGF-C in relation to its major receptor VEGFR-3 during the sprouting of the lymphatic endothelium in embryos suggests a paracrine mode of action and that one of the functions of VEGF-C may be in the regulation of angiogenesis of the lymphatic vasculature.
Article
Angiogenesis, the sprouting of new blood vessels from pre-existing ones, and the permeability of blood vessels are regulated by vascular endothelial growth factor (VEGF) via its two known receptors Flt1 (VEGFR-1) and KDR/Flk-1 (VEGFR-2). The Flt4 receptor tyrosine kinase is related to the VEGF receptors, but does not bind VEGF and its expression becomes restricted mainly to lymphatic endothelia during development. In this study, we have purified the Flt4 ligand, VEGF-C, and cloned its cDNA from human prostatic carcinoma cells. While VEGF-C is homologous to other members of the VEGF/platelet derived growth factor (PDGF) family, its C-terminal half contains extra cysteine-rich motifs characteristic of a protein component of silk produced by the larval salivary glands of the midge, Chironomus tentans. VEGF-C is proteolytically processed, binds Flt4, which we rename as VEGFR-3 and induces tyrosine autophosphorylation of VEGFR-3 and VEGFR-2. In addition, VEGF-C stimulated the migration of bovine capillary endothelial cells in collagen gel. VEGF-C is thus a novel regulator of endothelia, and its effects may extend beyond the lymphatic system, where Flt4 is expressed.
Article
Intralobular lymphatics in the guinea pig pancreas were demonstrated enzyme-histochemically showing the extent, distribution and fine structure by combined light and transmission electron microscopy. 5-Nase)-positive lymphatic vessels were present throughout the pancreas. Intralobular lymphatics among the acini were comparatively rare and generally independent of the blood capillaries, pancreatic ducts and acini. These lymphatics revealed the usual structural features, such as typical intercellular junctions and very tenuous vascular walls without continuous basal laminae. Fine precipitates of the cerium-based reaction product for 5-Nase activity were also detected in the interlobular connective tissue. ALPase activity, absent in the lymphatics, was positive in the blood capillaries, suggesting that it is also a useful way of demonstrating, histochemically, the blood capillaries in the guinea pig pancreas.
Article
To gain a molecular understanding of tumor angiogenesis, we compared gene expression patterns of endothelial cells derived from blood vessels of normal and malignant colorectal tissues. Of over 170 transcripts predominantly expressed in the endothelium, 79 were differentially expressed, including 46 that were specifically elevated in tumor-associated endothelium. Several of these genes encode extracellular matrix proteins, but most are of unknown function. Most of these tumor endothelial markers were expressed in a wide range of tumor types, as well as in normal vessels associated with wound healing and corpus luteum formation. These studies demonstrate that tumor and normal endothelium are distinct at the molecular level, a finding that may have significant implications for the development of anti-angiogenic therapies.