Anda di halaman 1dari 19

Letter doi:10.

1038/nature22977

Whole-body imaging of lymphovascular niches


identifies pre-metastatic roles of midkine
DavidOlmeda1, DanielaCerezo-Wallis1, EricaRiveiro-Falkenbach2, PaulaC.Pennacchi1, MartaContreras-Alcalde1, NuriaIbarz3,
MetehanCifdaloz1, XavierCatena1, TonantzinG.Calvo1, EstelaCan1, DirenaAlonso-Curbelo1, JavierSuarez1, LisaOsterloh1,
OsvaldoGraa4, FranciscaMulero5, DiegoMegas6, MartaCaamero7, JorgeL.Martnez-Torrecuadrada8, ChandraniMondal9,
JulieDiMartino9, DavidLora10, InsMartinez-Corral11, J.JavierBravo-Cordero9, JavierMuoz3, SusanaPuig12,
PabloOrtiz-Romero13, JosL.Rodriguez-Peralto2, SagrarioOrtega11 & MaraS.Soengas1

Cutaneous melanoma is a type of cancer with an inherent potential Crosses to Tyr::CreERT2;BrafV600E or Tyr::CreERT2;BrafV600E;Ptenlox/lox
for lymph node colonization, which is generally preceded by immune-competent mouse strains19 were performed to monitor benign
neolymphangiogenesis13. However, sentinel lymph node removal nevi or malignant melanomas. These genetically engineered mouse
does not necessarily extend the overall survival of patients with models (GEMM) were maintained in black-coat or albino backgrounds
melanoma4,5. Moreover, lymphatic vessels collapse and become to characterize pigmented or amelanotic lesions (Fig. 1a).
dysfunctional as melanomas progress6,7. Therefore, it is unclear A panel of nine melanoma cell lines with different levels of VEGFC,
whether (and how) lymphangiogenesis contributes to visceral a main known driver of lymphangiogenesis3, were used to generate
metastasis. Soluble and vesicle-associated proteins secreted by subcutaneous xenografts in Vegfr3Luc nu/nu mice (Extended Data
tumours and/or their stroma have been proposed to condition Fig. 1a, b). This set of cell lines also recapitulated the most frequent
pre-metastatic sites in patients with melanoma814. Still, the identities melanoma-associated alterations in BRAF, NRAS, PTEN and p53
and prognostic value of lymphangiogenic mediators remain (Supplementary Table 1). Before implantation, cells were transduced
unclear2,14. Moreover, our understanding of lymphangiogenesis with mCherry for dual fluorescencebioluminescence imaging of
(in melanomas and other tumour types) is limited by the paucity tumour growth and subsequent emission of VEGFR3-associated
of mouse models for live imaging of distal pre-metastatic niches15. luciferase (V3-Luc), respectively (see examples in Fig. 1b). Histological
Injectable lymphatic tracers have been developed7, but their limited staining for VEGFR3 in Lyve1-positive lymphatic endothelial cells
diffusion precludes whole-body imaging at visceral sites16. Vascular (LEC) confirmed neo-lymphangiogenesis (see Fig. 1c for examples in
endothelial growth factor receptor 3 (VEGFR3) is an attractive the skin, and Extended Data Figs 1ce, for lymph nodes, spleen and
lymphoreporter17 because its expression is strongly downregulated lungs, respectively).
in normal adult lymphatic endothelial cells, but is activated in We identified four major patterns of tumour-driven neolym-
pathological situations such as inflammation and cancer17,18. phangiogenesis: with no V3-Luc at any site (pattern I); restricted to
Here, we exploit this inducibility of VEGFR3 to engineer mouse the primary tumour (II); occurring locally and distally (III); and,
melanoma models for whole-body imaging of metastasis generated unexpectedly, without or with highly delayed tumoural V3-Luc but with
by human cells, clinical biopsies or endogenously deregulated potent systemic emission (IV) (Fig. 1d, e, Extended Data Fig. 2ac and
oncogenic pathways. This strategy revealed early induction of distal Supplementary Information). Only patterns III and IV (those including
pre-metastatic niches uncoupled from lymphangiogenesis at primary distal V3-Luc) were found to substantially increase lymphatic vessel
lesions. Analyses of the melanoma secretome and validation in density at lymph nodes and to result in efficient nodal metastasis
clinical specimens showed that the heparin-binding factor midkine (Extended Data Fig. 2cf). These patterns did not correlate with
is a systemic inducer of neo-lymphangiogenesis that defines patient BRAF, NRAS, p53 or PTEN status (not shown). Moreover, tumoural
prognosis. This role of midkine was linked to a paracrine activation VEGFC was neither sufficient nor essential for the activation of d istal
of the mTOR pathway in lymphatic endothelial cells. These data V3-Luc and nodal metastases (Fig. 1a and Extended Data Fig. 2,
support the use of VEGFR3 reporter mice as a MetAlert discovery where VEGFC-positive cell lines are labelled in green). Patient-derived
platform for drivers and inhibitors of metastasis. xenografts from human skin metastases further confirmed early
To generate animal models for whole-body imaging of neolym- and systemic induction of V3-Luc uncoupled from prior tumoural
phangiogenesis in melanoma progression, we used Flt4 tm1.1Sgo lymphangiogenesis (Extended Data Fig. 3a, b). Systemic V3-Luc was
(Vegfr3EGFPLUC) mice17, a knock-in system in which an EGFPluciferase also obvious in GEMM melanomas (but not in nevi), in both p igmented
fusion cassette is coupled to the transcription of Vegfr3 (Fig. 1a). These and amelanotic backgrounds (Extended Data Fig. 3ce).
VEGFR3 reporter mice (herein referred to as Vegfr3Luc for simplicity) Tumour burden estimated by quantitative RTPCR at different
were crossed with immune-deficient Foxn1nu (nu/nu) mice and used as anatomical locations and time points after tumour cell implantation
hosts for human melanoma cell lines and patient-derived xenografts. revealed significant systemic activation of V3-Luc prior to tumour cell

1
Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 2Department of Pathology, Medical School, Universidad
Complutense, Instituto i+12, Hospital Universitario 12 de Octubre, Madrid 28041, Spain. 3Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO),
Madrid 28029, Spain. 4Bioinformatics Unit, Structural Biology and Biocomputing Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 5Molecular Imaging Unit,
Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 6Confocal Microscopy Unit, Biotechnology Programme, Spanish National Cancer Research
Centre (CNIO), Madrid 28029, Spain. 7Histopathology Unit, Biotechnology Programme,Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 8Crystallography and Protein
Engineering Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 9Department of Medicine, Division of Hematology and Oncology, The
Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA. 10CIBERESP, Instituto i+12, Hospital Universitario 12 de Octubre, Madrid 28041, Spain. 11Transgenic
Mice Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain. 12Melanoma Unit, Dermatology Department, Hospital Clnic de Barcelona, Institut
dInvestigacions Biomdiques dAugust Pi i Sunyer, 08036 Barcelona, Spain. 13Department of Dermatology, Medical School, Universidad Complutense, Instituto i+12, Hospital Universitario 12
deOctubre, Madrid 28041, Spain. Present addresses: Memorial Sloan Kettering Cancer Centre, New York, USA (D.A.-C.); Roche Pharma Research and Early Development, Roche Innovation
Center Munich, 82377 Penzberg, Germany (M.C.); Department of Immunology, Genetics and Pathology and Rudbeck Laboratory, University of Uppsala, 75185 Uppsala, Sweden (I.M.-C.).

6 7 6 | N A T U R E | V O L 5 4 6 | 2 9 J une 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

a b c Figure 1 | Vegfr3Luc reporter mice for whole-


Stop
0 14 24 30
Basal Tumour-induced body analysis of benign and malignant
Vegfr3(Flt4)
melanocytic lesions. a, Vegfr3Luc mouse models

Bright field
29 30 3 UTR Vegfr3
used in this study, with the five strains generated
IRES EGFP-Luc 3 UTR
indicated with numbers in circles. b, Xenografts
Vegfr3Luc 6 of mCherrySK-Mel-147 cells, imaged as

(tumour cells)
Tyr::CreERT2; BrafV600E
Lyve1 indicated. c, Confocal immunomicroscopy of

mCherry
NU( Ico)-
Foxn1nu VEGFR3 (red) and Lyve1 (green) in normal
Tyr::CreERT2; Tyr::CreERT2;
BrafV600E BrafV600E Ptenlox/lox skin or in xenografts of SK-Mel-147 cells.
1 2 White coat 4 1
d, Four main patterns (IIV) of V3-Luc emission
3 Black coat 5
10 Vegfr3/Lyve1/ identified by whole-body bioluminescence
DAPI of mice bearing xenografts of the indicated
Luciferase
(V3-Luc)
Human cells, Nevi GEMM
melanoma cell lines. Numbers represent
50 m days after implantation and dotted lines
tissue biopsies melanomas
2.5
show tumour area. e, V3-Luc emission at the
d V3-Luc e
10 2,000 10 LN Spleen Liver Lung
indicated locations and tumour sizes. Average
Tumour
0 30 55 tumour volumes are indicated on the x-axis and
1,500
visualized with a dotted red line for simplicity.
WM164

I 5 1,000 5
LN, lymph nodes. Data are mean s.d.
500 (6 mice per condition, 24 lymph nodes per
0 0 0 cell line). One-way ANOVA was used with
0 46 64 **** 2,000
10
Dunnetts correction for multiple comparisons.
10
** 1,500 Fluorescence: ps1cm2 sr1 109;
WM35

II
bioluminescence: ps1cm2 per sr 106. See
* 1,000
5
5
Source Data for quantification of V3-Luc in e.
500
Here and in all corresponding figure panels
0 0 0
15
*P 0.05, **P 0.01, ***P 0.001.
0 12 31 15 2,000
**** **** ** **** ***
SK-Mel-147

*** 1,500 10 **** * ** **


10
III ** 1,000 **** *** ** *
5
5 500

0 0 0
2,000 ** **
p s1 cm2 sr 1 106
p s1 cm2 sr1 106

(mm3)

0 45 65 10 **** ***
10
1,500
**** **
451LU

Tumour vol

IV 1,000 **
5 5
500

0 0 0
0
0
1, 0
1, 0
0

0
0
1, 0
0
0

0
0
1, 0
1, 0
0

0
0
1, 0
1, 0
0

0
0
1, 0
1, 0
0

2.5 10
25
50
00
50

25
50
00
50

25
50
00
50

25
50
00
50

25
50
00
50
1,

Av. tumour vol. (mm3) Av. tumour vol. (mm3)

colonization (see black dots in Fig. 2a, b for quantitative analyses in a


LN Lung
b
LN met. Lung met. Skin met.
V3-Luc (p s1 cm2 sr1 106)

lymph nodes and lungs; P< 0.01 and P<0.001, respectively). These
V3-Luc (p s1 cm2 sr1 106)

15 *** 8
*** 20
**** ***
data support the idea that V3-Luc positive sites represent pre-metastatic 7.0 ***
Cell
10 *** *
***
**** number ****
niches. V3-Luc imaging was then analysed before and after surgical *** 10 *** 0 **** *** *
removal of xenografts of human melanoma cells (SK-Mel-147; Fig. 2c) **** ** >10
5
10 4
>100
or GEMM melanomas (Extended Data Fig. 4). In both cases, excision 3.5 5 >1,000
of primary lesions led to a marked reduction in V3-Luc. Metastatic >10,000
0
relapses were found at later time points (involving skin, lymph node 1.0
0 1 2 3 4
1
0 1 2 3 4
0
1 2 3 4
0
1 2 3 4 1 2 3 4
and lung metastases) and were invariably preceded by V3-Luc emission Time (weeks) 1 At 2 Before 3 After 4 At
injection surgery surgery Met
(Fig. 2d). Therefore, early systemic luciferase emission in Vegfr3Luc mice c
reflects long-distance signals fuelled by the primary lesions. 0 20 30 34 38 40 45 51 58 64
Proteomic analyses were then performed to identify tumour-secreted
Visible light

drivers of distal lymphangiogenesis and metastasis. Exosome cargo was


defined using label-free liquid chromatographymass spectrometry
(Fig. 3a) in representative examples of the four V3-Luc emission
Relapse
Surgery

patterns described above. Candidate drivers were filtered for s tatistical 10

significance, selective expression in cell lines with systemic V3-Luc


V3-Luc

emission, no previous lymphangiogenic roles and no links to melanoma


(see Methods). The top scoring candidate was the heparin-binding
factor midkine (MDK; marked in blue in the volcano plots of Fig. 3b;
2.5
see additional secreted proteins in Supplementary Table 2). We found it
particularly encouraging that MDK is secreted by other cancer types2023 Figure 2 | Vegfr3Luc mice reveal pre-metastatic niches. a, V3-Luc
and has been proposed to mediate metastasis by its mitogenic, pro- emission by xenografts of mCherrySK-Mel-147 cells in sentinel lymph
nodes and lungs. Coloured dots correspond to tumour cell burden defined
inflammatory and angiogenic functions20,2325, but has no connection
by RTPCR. b, Quantification of V3-Luc emission by SK-Mel-147
to lymphovascular expansion. mCherry cells before and after surgical resection of the cutaneous lesions.
Immunohistochemical analyses confirmed that MDK was expressed Lymph nodes (n=9), lung metastases (met.) (n=7) and skin metastases
in highly metastatic melanoma xenografts (Fig. 3c and Extended (n= 5); t-test. c, Representative whole-body imaging of experiments in b.
Data Fig. 5a). Importantly, although we had found MDK as an See Source Data for quantification of V3-Luc in a, b. Numbers on mice
exosome cargo, enzyme-linked immunosorbent assay (ELISA) and represent time in days after tumour cell implantation.

2 9 J une 2 0 1 7 | V O L 5 4 6 | N A T U R E | 6 7 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

a b

SK-Mel-147
SK-Mel-28
12 139 37 180 44 188

WM164

451LU
222 554 391 582 387

WM35
432
4 4 4

log10 P
2 MDK 2 2
30
MDK MDK
0 0 0
10 5 0 5 > 10 10 5 0 5 > 10 10 5 0 5 > 10
20
log2 (451LU vs WM164) log2 (SK-Mel-28 vs WM164) log2 (SK-Mel-147 vs WM164)

c WM164 WM35 451LU SK-Mel-28 SK-Mel-147

50 m

d shMDK e f g h
* **** **** ***
shC sh1 sh5
20
** **** **** 10
***

(p s1 cm2 sr1 106)

(p s1 cm2 sr1 106)


15 6

Mice with lung mets


12
8

Metastatic LN
15
V3-Luc skin

V3-Luc LN
10 4
6
10
4
5 2
5 2

0 0 0 0
sh1 sh5 sh1 sh5 sh1 sh5 sh1 sh5
3 shC shC shC shC
shMDK shMDK shMDK shMDK

i j k l m Lyve1 MDK Merge


V3-Luc (p s1 cm2 sr1 107)

Empty MDK **** **** **


8
Mice with lung mets

*
1.5
6
Metastatic LN

12
1.0 4
4

0.5
2 20 m

0 0 0 *
Merge
M ty

M ty

M ty
DK

DK

DK
p

p
Em

Em

Em

3 50 m

Figure 3 | Proteomic analyses identify MDK as new pro- g, Metastatic lymph nodes (mCherry-positive) of mice analysed in e. Data
lymphangiogenic and pro-metastatic factor. a, Label-free quantitative correspond to mean s.d. (n6 mice per condition). One-way ANOVA
(LFQ) expression of exosome cargo identified by LCMS/MS in the with Dunnetts correction for multiple comparisons. h, Lung metastases
indicated cell lines. b, Volcano plots showing exosomal proteins (grey (blue) after tumour removal. Fishers exact test, n= 10. i, V3-Luc emission
dots) differentially expressed with respect to the non-lymphangiogenic by MDK overexpression in the otherwise negative WM164 cells. Scale,
WM164 cells. Proteins upregulated or downregulated versus the poorly ps1cm2 sr1 106. j, Relative nodal V3-Luc emission at the indicated
metastatic WM35 cells are depicted in red or green, respectively. The conditions. The number of metastatic lymph nodes per mouse is shown
hyperbolic black curve separates statistically significantly regulated in k; t-test. i, Lung metastases (mCherry positive; red) in MDK gain-of-
proteins as defined by two-sample Students t-test, false discovery function studies (overexpressing MDK in WM164 cells). Fishers exact
rate (FDR)=0.05; artificial within groups variance (S0)= 0.8. test, n= 7. m, Immunomicrographs showing MDK (green) accumulated
c, Immunological detection of MDK (pink) in tumour xenografts. at areas of neo-lymphangiogenesis and at sites of lymphatic sprouting
df, V3-Luc emission by SK-Mel-147mCherry cells transduced with in lymph nodes (arrows). Asterisks mark stromal cells. See Source Data
control or MDK shRNA1 or shRNA5 (d), with quantification for for quantification of V3-Luc in eh, jl) and Supplementary Fig. 1 for
cutaneous lesions in e and for sentinel and brachial lymph nodes in f. uncropped blots.

immunoblotting after exosome depletion showed 4060-fold higher with marked inhibition of bioluminescence and metastasis to lymph
expression of MDK in the soluble fraction of aggressive melanoma cells nodes (Fig. 3dg) and visceral organs (Fig. 3h). In turn, gain of func-
(Extended Data Fig. 5b). Therefore we proceeded with the analysis of tion (restoring MDK expression; Extended Data Fig. 6a) was sufficient
soluble MDK in the secretome of melanoma cells. to convert the non-lymphangiogenic WM164 cell line into a potent
Loss- and gain-of-function studies were performed to manipulate inducer of systemic V3-Luc (Fig. 3i, j), favoring metastatsis to lymph
the endogenous levels of MDK. For loss-of-function tests, we used nodes (Fig. 3k) and distal sites (shown for lungs in Fig. 3l and Extended
five short hairpin RNAs (shRNAs). Of those, quantitative PCR with Data Fig. 6b), without affecting VEGFC or VEGFD mRNA, or the
reverse transcription (qRTPCR) and ELISA identified shMDK(1) and growth and vascularization of the cutaneous lesions (Extended Data
shMDK(5) as the most potent blockers of MDK expression and secre- Fig. 6c, d and data not shown).
tion (Extended Data Fig. 5c, d). None of these shRNAs affected intra- In further support of a pro-lymphangiogenic role for MDK, ectopic
cellular levels of VEGFC or VEGFD mRNA (Extended Data Fig. 5e). expression of this protein in LEC induced Vegfr3 mRNA (see qRTPCR
Moreover, MDK depletion did not significantly affect the growth or analyses in Extended Data Fig. 6e). Moreover, conditioned medium
blood vessel density of melanoma xenografts (Extended Data Fig. 5f, g). from MDK-expressing melanoma cells enhanced LEC p roliferation,
This is in contrast to the mitogenic20,24 and angiogenic roles of MDK whereas medium from cells with loss of MDK function had an
in other settings26,27. Instead, in melanoma xenografts, MDK shRNAs inhibitory effect (Extended Data Fig. 6f, g). In addition, confocal
decreased lymphatic vessel density (LVD) and reduced tumoural V3-Luc immunofluorescence in MDK gain-of-function experiments demon-
(Extended Data Fig. 5h, i and Fig. 3e, respectively). Nevertheless, the strated that MDK accumulated at sites of nodal neolymphangiogenesis
most obvious effect of MDK downregulation was found systemically, in vivo (see Fig. 3m for colocalization with Lyve1-LEC in lymph nodes,

6 7 8 | N A T U R E | V O L 5 4 6 | 2 9 J une 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

a b c d + CM (no MDK) + CM (MDK)

CM LoF M1

CM LoF M5
CM GoF

12 h
24 h

12 h
24 h
GoF

10
15
30
1h

10
15
30
1h
GoF (ES)
0.2
WM164 vs mTOR 0.1 p-RPS6
CM WM164-MDK NES = 4.26
q < 0.001
LoF 0
Total RPS6
SK-Mel-147 vs
SK-Mel-147

5
M
RNA-seq

F-
oF

oF
e

Lo
G

G
0

LoF M1 (ES)
IPA

M
C

C
0.1 MDK + + + + +
GSEA NES = 3.73
Vehicle + + + + +
Oncogenic 0.2 q < 0.001
Rapamycin +
signatures
Torin 1 +
Cancer
hallmarks VEGFR3
0

LoF M5 (ES)
Functional analyses >3
0.1 P-RPS6
LEC (monolayers)

Log2FC
NES = 3.49
0.2 q < 0.001 RPS6
LEC (sprouting)
Histological validation -Actin
<3

Nevi Stage III Skin metastasis LN metastasis Visceral metastasis


f

g **** h 100

Disease-free survival (%)


****
****
**** MDK LOW
100 60

75 High
Cases (%)

Low
20 MDK HIGH
50 Medium

25
Negative
0 **
0 4 8 12
0 Years
Number at risk
i

et

et

et
ev

I-I

LOW 33 23 20 18 15 15 13 13 13 9 7 4 3 0
N

e
ag

in

LN

al
er
Sk
St

HIGH 24 11 8 7 7 6 6 4 2 2 1 0 0 0
sc
Vi

Figure 4 | MDK activates the mTOR pathway and is an indicator of poor by immunoblotting of the impact of conditioned medium (CM-GoF or
prognosis in melanoma. a, Identification of downstream effectors CM-LoF) on VEGFR3 and total and phosphorylated RPS6 in human
of MDK in LEC by RNA-seq and the corresponding validation analyses. LEC. When indicated, LEC were incubated with the mTOR inhibitors
CM, conditioned medium; GoF, gain of function; LoF, loss of function. rapamycin (1M) or torin 1 (10nM). f, g, MDK immunohistochemistry
b, Heat map of deregulated genes (fold change >1.5), with mTOR pathway- (pink) in paraffin sections of the indicated human biopsies (f), with data
associated factors in red. c, Enrichment score (ES) plots for mTOR-related quantified in g. h, KaplanMeier survival curves of patients with
pathways. d, Time-course analyses of RPS6 phosphorylation in LEC stage IIIV melanoma, classified on the basis of low and high MDK
incubated with conditioned medium from control or MDK gain-of- expression in the corresponding sentinel lymph nodes. Fishers exact
function cells. Experiments were performed in triplicate. e, Visualization test. n= 20.

including tip cells of sprouting lymphatic vessels, and Extended Data The mTOR pathway was intriguing because, in addition to its long-
Fig. 6h). MDK was also enriched in lymphatic vessels in visceral organs known contribution to tumour cell growth and motility, it has been
such as the lungs or liver before tumour cell colonization (Extended found to modulate lymphangiogenesis28,29, although with no previous
Data Fig. 6i and data not shown). link to MDK. The conditioned medium of MDK-expressing melanoma
Conceptually, increased lymphatic vessel density would not be cells promoted the phosphorylation of the mTOR target RPS6 in LEC
expected to promote metastasis per se, unless it were coupled to effec- (Fig. 4d). MDK-induced phosphorylation of RPS6 was accompanied
tive intra- or extravasation of tumour cells. Time-lapse videomicros- by induction of VEGFR3 expression (Fig. 4e) and LEC sprouting
copy revealed that incubation of LEC with MDK increased the adhesion (Extended Data Fig. 9a). The dependency on MDK and mTOR of these
and transmigration of WM164 and SK-Mel-147 cells (Supplementary LEC-associated effects was confirmed by using conditioned medium
Videos 1 and 2; Extended Data Fig. 7ac). The ability of MDK shRNA from MDK-depleted cells and the mTOR inhibitors torin 1 and rapa-
to inhibit the intravasation of melanoma cells through LEC was mycin (Fig. 4e and Extended Data Fig. 9b, c). Moreover, histological
confirmed histologically (Extended Data Fig. 7d, e). Moreover, the analyses of xenografts in Vegfr3Luc mice showed activation of the mTOR
pro-metastatic ability of MDK was further supported by intravital pathway (phosphorylation of RPS6) in vivo at sites of MDK-induced
imaging of melanoma cells invading the subcapsular sinus at sentinel neolymphangiogenesis (Extended Data Fig. 9d). Together, these results
lymph nodes (Extended Data Fig. 8; see Methods). identify a functional role of MDK in melanoma as a mechanistic link
RNA sequencing (RNA-seq) was performed in LECs incubated with between two key pro-tumorigenic processes: neolymphangiogenesis
conditioned medium from gain- and loss-of-function studies to iden- and mTOR signalling.
tify downstream effectors of MDK (Fig. 4a). Genes that were regulated Analysis of a retrospective series of human benign nevi and melano-
in opposing directions by gain or loss of MDK function are shown mas confirmed tumour-associated expression of MDK (Fig. 4f, g and
in heat maps in Fig. 4b. Ingenuity pathway analyses (IPA) and gene Extended Data Fig. 10a, b). Moreover, an independent series of sentinel
set enrichment analyses (GSEA) identified multiple cancer hallmarks lymph nodes from 62 patients with stage IIIII melanoma showed that
and oncogenic signatures as putative downstream effectors of MDK patients with high nodal MDK expression (Extended Data Fig. 10c)
(Supplementary Tables 3, 4). These included the mTOR pathway as one had significantly worse disease-free survival (DFS) than patients with
of the most enriched MDK-associated signalling cascades (Fig. 4b, c). low nodal MDK expression (P= 0.0034; Fig. 4h), even when they had

2 9 J une 2 0 1 7 | V O L 5 4 6 | N A T U R E | 6 7 9
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

tumour-negative sentinel lymph nodes (P= 0.0243; Extended Data 20. Muramatsu, T. & Kadomatsu, K. Midkine: an emerging target of drug
Fig. 10d). Multivariate analyses confirmed that the correlation between development for treatment of multiple diseases. Br. J. Pharmacol. 171,
811813 (2014).
high MDK expression and reduced DFS was independent of the age 21. Yao, J., Li, W. Y. & Gao, S. G. The advances of Midkine with peripheral invasion
and gender of the patient and, importantly, of the thickness (Breslow in pancreatic cancer. Am. J. Cancer Res. 5, 29122917 (2015).
depth) of the primary lesion (Extended Data Fig. 10e). 22. Jones, D. R. Measuring midkine: the utility of midkine as a biomarker in cancer
and other diseases. Br. J. Pharmacol. 171, 29252939 (2014).
The immunocompetent and immunodeficient VEGFR3- 23. Kishida, S. & Kadomatsu, K. Involvement of midkine in neuroblastoma
lymphoreporter mouse models of melanoma generated in this study tumourigenesis. Br. J. Pharmacol. 171, 896904 (2014).
have uncovered distinct patterns of metastatic progression that can 24. Kadomatsu, K., Kishida, S. & Tsubota, S. The heparin-binding growth factor
midkine: the biological activities and candidate receptors. J. Biochem. 153,
bypass the requirement for intra- or peri-tumoural lymphangiogenesis. 511521 (2013).
Live imaging coupled with loss- and gain-of-function studies revealed 25. Jono, H. & Ando, Y. Midkine: a novel prognostic biomarker for cancer. Cancers
that MDK influences LEC via paracrine effects of the mTOR p athway (Basel) 2, 624641 (2010).
26. Weckbach, L. T. et al. Midkine acts as proangiogenic cytokine in hypoxia-
on VEGFR3 expression (Extended Data Fig. 10f). Moreover, the induced angiogenesis. Am. J. Physiol. Heart Circ. Physiol. 303, H429H438
physiological relevance of MDK was validated by multivariate analysis (2012).
of disease-free survival of patients with melanoma. We anticipate that 27. Choudhuri, R., Zhang, H. T., Donnini, S., Ziche, M. & Bicknell, R. An angiogenic
the genome-wide proteomic RNA-seq data presented here, showing role for the neurokines midkine and pleiotrophin in tumorigenesis. Cancer Res.
57, 18141819 (1997).
modulation of pro-inflammatory molecules in melanoma cells and 28. Luo, Y. et al. Rapamycin inhibits lymphatic endothelial cell tube formation by
LEC, may open new areas of investigation. For example, the accumu- downregulating vascular endothelial growth factor receptor 3 protein
lation of MDK at the lymph node subcapsular sinus (Extended Data expression. Neoplasia 14, 228237 (2012).
29. Patel, V. et al. Decreased lymphangiogenesis and lymph node metastasis
Figs 8, 10c) may affect the function of this structure as a physical and by mTOR inhibition in head and neck cancer. Cancer Res. 71, 71037112
immunological barrier to tumour cell dissemination30. In this context, it (2011).
will be interesting to determine whether MDK (and mTOR) cooperate 30. Pucci, F. et al. SCS macrophages suppress melanoma by restricting
tumor-derived vesicle-B cell interactions. Science 352, 242246
with other factors that are enriched in the secretome of metastatic (2016).
melanoma cells. As the Vegfr3Luc reporters in this study are not limited
to melanoma, they may represent a versatile metastasis-alert platform Supplementary Information is available in the online version of the paper.
for gene discovery and drug testing in a broad spectrum of malignant
Acknowledgements We thank colleagues at the CNIO Melanoma Group,
diseases. particularly A. Tenaglia for technical assistance; J. A. Esteban for critical reading
of this manuscript; the i+12 Biobank of the Hospital 12 de Octubre for help
Online Content Methods, along with any additional Extended Data display items and
with histological analyses and for patient biopsies; L. Larue and M. McMahon
Source Data, are available in the online version of the paper; references unique to
for Tyr:CreERT2 and inducible BrafCA mouse strains, respectively; and the
these sections appear only in the online paper.
Animal Facility Unit and the Histopathology Core of CNIO for assistance.
M.S.S. is funded by grants from the Spanish Ministry of Economy and
received 10 March 2016; accepted 25 May 2017.
Innovation (project SAF2014-56868-R), the Asociacin Espaola Contra el
Cncer (AECC), the Worldwide Cancer Research, an Established Investigator
1. Zheng, W., Aspelund, A. & Alitalo, K. Lymphangiogenic factors, mechanisms, Award from the Melanoma Research Alliance (MRA), and a LOral Paris
and applications. J. Clin. Invest. 124, 878887 (2014). USA-MRA Team Science Award for Woman in Scientific Research. The CNIO
2. Stacker, S. A. et al. Lymphangiogenesis and lymphatic vessel remodelling in Proteomics Unit belongs to ProteoRed, PRB2-ISCIII, supported by grant
cancer. Nat. Rev. Cancer 14, 159172 (2014). PT13/0001. N.I. and J.M. are funded by SAF2013-45504-R (MINECO). J.M. is
3. Karaman, S. & Detmar, M. Mechanisms of lymphatic metastasis. J. Clin. Invest. also supported by Ramon y Cajal Programme (MINECO) RYC-2012-10651.
124, 922928 (2014). J.L.R.-P and P.O.-R are funded by grants FIS 2014/1737, 11/02568 and FIS
4. van Akkooi, A. C. J. & Eggermont, A. M. M. Melanoma: MSLT-1-SNB is a 2014/01784, 11/1759, respectively, from the Spanish Ministry of Health.
biomarker, not a therapeutic intervention. Nat. Rev. Clin. Oncol. 11, 248249 F.M. is funded by the AMIT Project/CDTI/CENIT Programme (MICINN), S.O. by
(2014). SAF2013-44866-R (MINECO), and J.J.B.-C. by an NCI K22CA196750 grant and
5. Morton, D. L. et al. Final trial report of sentinel-node biopsy versus nodal the TCI Young Scientist Cancer Research Award JJR Fund (P30 CA196521).
observation in melanoma. N. Engl. J. Med. 370, 599609 (2014). J.D.M. is the recipient of a postdoctoral fellowship from the ARC Foundation and
6. Padera, T. P. et al. Lymphatic metastasis in the absence of functional E.R.-F. from Fundacin Cientfica de la Asociacin Espaola Contra el Cncer.
intratumor lymphatics. Science 296, 18831886 (2002). D.C.-W. is the recipient of a predoctoral fellowship from Fundacin La Caixa,
7. Proulx, S. T. et al. Use of a PEG-conjugated bright near-infrared dye for and M.C.-A. and X.C. are recipients of the Immutrain Marie Skodowska-Curie
functional imaging of rerouting of tumor lymphatic drainage after sentinel ITN Grant.
lymph node metastasis. Biomaterials 34, 51285137 (2013).
8. Kaplan, R. N. et al. VEGFR1-positive haematopoietic bone marrow progenitors Author Contributions M.S.S. and D.O. conceived and designed all
initiate the pre-metastatic niche. Nature 438, 820827 (2005). expression studies, mechanistic analyses and prognostic evaluations.
9. Demirsoy, S., Martin, S., Maes, H. & Agostinis, P. Adapt, Recycle, and Move on: S.O. conceived, designed and developed the Vegfr3Luc (Flt4tm1.1Sgo) and
Proteostasis and Trafficking Mechanisms in Melanoma. Front. Oncol. 6, 240 Vegfr3Luc nu/nu mouse models (Strain 1), contributed to experimental design,
(2016). discussed data and revised the manuscript. I.M.-C. generated the Vegfr3Luc
10. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells immunocomptetent allele and the Vegfr3Luc nu/nu Strain 1, and contributed
toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883891 to their initial characterization. D.O. performed all spatio-temporal studies
(2012). of neolymphangiogenesis, as well as loss- and gain-of-function studies of
11. Hoshino, A. et al. Tumour exosome integrins determine organotropic MDK in vivo. D.C.-W. contributed to mechanistic and histological analyses
metastasis. Nature 527, 329335 (2015). of lymphangiogenesis, helped with in vivo experiments and revised the
12. Coelho, P., Almeida, J., Prudncio, C., Fernandes, R. & Soares, R. Effect of manuscript. E.R.-F., P.O.-R. and J.L.R.-P. performed the analysis of patient
adipocyte secretome in melanoma progression and vasculogenic mimicry. prognosis (with bioinformatic aid from D.L. and S.P.). P.C.P., M.C.-A., and
J. Cell. Biochem. 117, 16971706 (2016). X.C. contributed to functional analyses of lymphangiogenesis. N.I. and J.M.
13. Obenauf, A. C. et al. Therapy-induced tumour secretomes promote resistance performed LCMS/MS. F.M. helped with whole-body image analyses, D.M. with
and tumour progression. Nature 520, 368372 (2015). time-lapse confocal microscopy and M.Ca. with histological evaluations. O.G.
14. Liu, Y. & Cao, X. Characteristics and significance of the pre-metastatic niche. and M.Ci. performed RNA-seq. T.G.C. was in charge of animal breeding and
Cancer Cell 30, 668681 (2016). genotyping. J.J.B.-C., C.M. and J.D.M. performed intravital microscopy. J.S., L.O.,
15. Eklund, L., Bry, M. & Alitalo, K. Mouse models for studying angiogenesis and J.L.M.-T., E.C., T.G.C. and D.A.-C. provided technical assistance. The manuscript
lymphangiogenesis in cancer. Mol. Oncol. 7, 259282 (2013). was written by M.S.S. and D.O., revised by S.O. and D.A.-C., and approved by all
16. Sevick-Muraca, E. M., Kwon, S. & Rasmussen, J. C. Emerging lymphatic authors. M.S.S. supervised the project.
imaging technologies for mouse and man. J. Clin. Invest. 124, 905914 (2014).
17. Martnez-Corral, I. et al. In vivo imaging of lymphatic vessels in development, Author Information Reprints and permissions information is available
wound healing, inflammation, and tumor metastasis. Proc. Natl Acad. Sci. USA at www.nature.com/reprints. The authors declare no competing financial
109, 62236228 (2012). interests. Readers are welcome to comment on the online version of the
18. Petrova, T. V. et al. VEGFR-3 expression is restricted to blood and lymphatic paper. Publishers note: Springer Nature remains neutral with regard
vessels in solid tumors. Cancer Cell 13, 554556 (2008). to jurisdictional claims in published maps and institutional affiliations.
19. Dankort, D. et al. Braf(V600E) cooperates with Pten loss to induce metastatic Correspondence and requests for materials should be addressed to
melanoma. Nat. Genet. 41, 544552 (2009). M.S.S. (msoengas@cnio.es).

6 8 0 | N A T U R E | V O L 5 4 6 | 2 9 J une 2 0 1 7
2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Methods was minor distal luciferase emission and no significant nodal metastatic potential
Mouse breeding and induction of nevi and melanomas in Vegfr3Luc GEMM. The (second row in Fig. 1d, e; Extended Data Fig. 2c, d). Pattern III was represented by
Vegfr3Luc nu/nu immunodeficient mice and the four Tyr:CreERT2 immunocom- metastatic cell lines (WM902B, UACC62, SK-Mel-103 and SK-Mel-147) with local
petent strains (Vegfr3Luc; BrafV600E and Vegfr3Luc; BrafV600E; PTENlox/lox) generated and distal activation of Vegfr3Luc (third row in Fig. 1c, d; Extended Data Fig. 2a, b)
in albino- and black-coated mice are summarized in Fig. 1a. Strains used in this and nodal metastases (Extended Data Fig. 2d), but with no correlation to VEGFC
study for the crosses are as follows: Vegfr3Luc (Flt4tm1.1Sgo), initially engineered mRNA expression (Extended Data Fig. 1c). Pattern IV corresponded to cell lines
in CD-1;129Sv;C57BL6/J-albino background17; nu/nu mice (Crl:NU(Ico)Foxn1nu); (WM115, 451LU and SK-Mel-28) that did not express significant VEGFC and
Tyr::CreERT2/1Lru (ref. 31); BrafCA (Braftm1Mmcm)19 and Ptentm2Mak (ref. 32). Albino induced minimal or highly delayed tumoural bioluminescence, but still promoted
and black-coated animals were bred independently. Mice were genotyped as marked V3-Luc emission at lymph nodes and subsequently at spleen, liver and lung
previously described19,31,32. Nevi and melanomas in the Tyr::CreERT2 strains (bottom row in Fig. 1d, e; Extended Data Fig. 2df).
were induced in 14-week-old female mice by topical treatment with 5l of 5mM Immunocompetent Vegfr3Luc-GEMM nevi and melanoma were imaged
4-hydroxy-tamoxifen as previously described19. Animals were maintained in following similar techniques and procedures as for Vegfr3Luc nu/nu mice, with
an inbred background and genotyped as described by the manufacturers. All animals killed when tumour areas covered a maximum 700mm2 of the mice.
experiments with mice were performed in accordance with protocols approved by Sample sizes were chosen to ensure statistical significance. No randomization or
the Institutional Ethics Committee of the CNIO and the Instituto de Salud Carlos blind analyses were performed. All experiments with mice were performed in
III (see below for maximum size of the tumour lesions). No statistical methods accordance with protocols approved by the Institutional Ethics Committee of the
were used to predetermine sample size. The experiments were not randomized and CNIO and the Instituto de Salud Carlos III36.
the investigators were not blinded to allocation during experiments and outcome Protein immunoblotting and histological analyses of gene expression in mouse
assessment. tumours. For western blotting, primary and secondary antibodies were diluted
Cell culture. Human primary melanocytes were isolated from neonatal foreskins33 in 5% milk TBS-T incubated overnight at 4C or for 1h at room temperature.
and maintained in medium 254 supplemented with melanocyte growth factors Primary antibodies used were: MDK (sc-1398, Santa Cruz), VEGFR3 (Millipore),
(HMG-1) containing 10ng ml-1 phorbol 12-myristate 13-acetate (Invitrogen). RPS6 (5G10, Cell Signaling), P-RPS6 (Ser240/244; #2215, Cell Signaling) and Actin
Melanoma cells34 were cultured in DMEM (Invitrogen) supplemented with 10% (Sigma-Aldrich, A5441). HRP-conjugated secondary antibodies used were anti-
FBS, 100g ml-1 penicillin and 100g ml-1 streptomycin. See Supplementary mouse and anti-rabbit (GE Healthcare) or anti-goat (Jackson Immunoresearch).
Table 1 for a list of melanoma cells used and their genetic backgrounds. Human For analyses of tissue architecture and expression of lymphangiogenic markers,
lymph node endothelial cells (HLEC) Innoprot (Derio) were cultivated in Clonetics tissues were fixed in formalin and embedded in paraffin. Sections were prepared
EGM-2 MV BulletKit (Lonza) following the manufacturers instructions. HLEC for haematoxylin and eosin (H&E) staining. For immunostaining, 3m paraffin
were used within six passages in culture. All the melanoma cell lines were authen- sections were deparaffinized and placed in PBS. Endogenous peroxidase a ctivity
ticated using GenePrint 10 Loci Service and all the cell lines were routinely tested was quenched with 3% hydrogen peroxide. Sections were incubated with blocking
for mycoplasma contamination. solution (DAKO) to reduce non-specific staining. Incubation with the indicated
Gene silencing and overexpression by lentiviral transduction. MDK silencing antibodies (see below) was then followed by staining with the appropriate bioti-
was performed by lentiviral-driven expression of shRNAs, with pLKO-constructs nylated secondary antibodies (DAKO). Tissue sections were then incubated
purchased from Sigma: MDK-sh1 (Clone ID NM_002391.3-505s21c1); MDK-sh2 with Envision-Peroxidase Rabbit Ready-to-Use (DAKO) using diaminoben-
(238s21c1); MDK-sh3 (337s21c1); MDK-sh4 (421s21c1); MDK-sh5 (621s21c1). zidine (DAB) as a substrate for the peroxidase reaction. Sections were finally
Non-target shRNA (CAACAAGATGAAGAGCACCAA) was used as control. counterstained with haematoxylin (Anatech) and mounted with permanent
Infected cells were selected by incubation with puromycin (1g ml-1) and MDK mounting medium. For MDK and VEGFC histological analyses, the Ultravision
downregulation was determined by protein immunoblotting, ELISA or qRTPCR ONE Detection System (RTU, Thermo Scientific) was used according to the man-
(see below). For the overexpression of human MDK, ORF lentiviral expression ufacturers instructions. For immunofluorescence-based analyses, tissue s ections
vector pReceiver-Lv105-A0792 (MDK) and the corresponding empty vector were deparaffinized, incubated overnight with primary antibodies at 4C in a
were purchased from Genecopoeia. Viruses were produced in 293FT cells and humidified chamber and then rinsed and incubated with fluorescent secondary
infections were performed as previously described35. When indicated, cells were antibodies for 1h at room temperature. Nuclei were counterstained with Prolong
stably infected with mCherry pLV-puro lentiviral vectors as described35. Gold+DAPI (Invitrogen, concentration 5g ml-1) 20min before imaging.
Non-invasive imaging of tumour growth and neolymphangiogenesis in vivo by Primary antibodies used were: hVEGFR3 (MAB3757, Millipore), mVegfr3 (AF743,
bioluminescence and fluorescence optical imaging. To analyse xenografts from R&D Systems), mLyve-1 (ab14917, Millipore), hLYVE-1 (ab36993, Abcam),
human melanoma cells, subcutaneous implantations were performed with mCherry- CD-31(ab28364, Abcam), MDK (sc-1398, Santa Cruz), VEGFC (NB110-61022,
labelled cells (15 106 cells) into the flanks of 14-week old Vegfr3Luc nu/nu NB110-61022), RPS6 (5G10, Cell Signaling), P-RPS6 (Ser240/244; #2215, Cell
mice. Patient-derived xenographs (PDX) were generated from freshly isolated Signaling) CD169 (clone 3D6.112; AbD Serotec), MCSF antibody (ab183316,
biopsies of skin metastases. Tumour growth was monitored every 2d by caliper Abcam).
measurement of the two orthogonal large and small external diameters (a, b), and Assessment of metastatic tumour cell burden in Vegfr3Luc mice. mCherry-
volume was estimated using the formula V= a b2 0.52. Non-invasive imaging labelled SK-Mel-147 melanoma cells (1 106) were implanted subcutaneously
of mCherry fluorescence or V3-Luc bioluminiscence was performed using an into 14-week old female Vegfr3Luc nu/nu immunodeficient mice. Animals were
IVIS-SPECTRUM imaging system (PerkinElmer). Mice were anaesthetized with randomized into four groups for weekly analyses and collection. At the desired
isoflurane and injected intraperitoneally with 150mg kg-1 luciferin (PerkinElmer). times, V3-Luc emission was measured as described above. Sentinel lymph nodes
Sequential images were obtained after luciferin injection and the maximum light and lungs were removed and samples were preserved in RNAlater (ThermoFisher,
emission was determined for each mouse as previously described17. Photons Cat. AM7020). RNA and DNA purification from tissue samples was performed
emitted from specific regions were quantified using Living Image software (Caliper using AllPrep DNA/RNA Mini Kit (Qiagen, Cat. 80204). cDNA preparations
Life Sciences). In vivo dual fluorescence/bioluminescence imaging was performed were performed using the High-Capacity cDNA Reverse Transcription Kit
at different time points after cell implantation, until tumours reached an average (ThermoFisher, Cat. 4368814). Quantification of melanoma cells was performed
final size of 1,500mm3 or an average diameter of 1.5cm (set to follow ethical pro- by qRTPCR, using primers for mCherry and 18S as described in the qRTPCR
cedures for humane handling of animals36). Post hoc analyses were then performed section. Standard curves for the estimation of cell number were performed using
in representative cell lines for in-depth quantification of V3-Luc emission at the defined numbers of SK-Mel-147mCherry cells.
tumour, inguinal and brachial lymph nodes and in visceral organs (spleen, liver and qRTPCR. RNA purification from melanoma tissue samples and qRTPCR
lung), corresponding to animals with defined tumour sizes (i.e. 0, 250, 500, 1,000 were performed using the following primers: hVEGFC-F: TGCCAG
and 1,500mm3), to control for the varied growth rates of the different cell lines. CAACACTACCACAG; hVEGFC-R: GTGATTATTCCACATGTAATTGGTG;
The in vivo patterns of V3-Luc emission identified from the human melanoma hVEGFD-F: CAAGAAAGCGGCTTCAGGTA; hVEGFD-R: GCAACGA
cell lines tested as xenografts in Vegfr3Luc nu/nu mice were as follows: pattern I TCTTCGTCAAACA; hVEGFR3-F: CAAGAAAGCGGCTTCAGGTA; hVEGF
(for example, WM164) corresponded to the expected behaviour of xenografts R3-R: GCAGAGAAGAAAATGCTGACG; MDK-F: CCTGCAACTGGA
expressing low levels of VEGFC: namely, no signs of V3-Luc emission at any AGAAGGAG; MDK-R: CTGGCACTGAGCATTGTAGC; mCherry-F:
anatomical site (Fig. 1d, e, top panels), no altered lymphatic density (Extended CCCGCCGACATCCCCGACTA; mCherry-R: GGGTCACGGTCACCACGCC;
Data Fig. 2c) and infrequent metastatic colonization of sentinel lymph nodes, as 18S-F: TTGGAGGGCAAGTCTGGTG; 18S-R: CCGCTCCCAAGATCCAACTA.
determined by fluorescence-based imaging (Extended Data Fig. 2d). Pattern II was HLEC proliferation assay. To define the impact of MDK-overexpressing
exemplified by WM35, a VEGFC-expressing cell line (Extended Data Fig. 1c, d). melanoma cells on the growth of HLEC, conditioned medium was collected from
In this case, potent induction of V3-Luc was detected at the tumour site, but there WM164 cells infected with control (empty vector) or MDK-expressing lentiviruses.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Melanoma cells were cultured in DMEM supplemented with 10% FBS until they Trypsin (Promega; 1:100 sample concentration, overnight at 37C). The resulting
reach 80% confluence. Medium was then changed to EGM-2 MV for 24h. This peptides were desalted using micro-columns filled with Poros Oligo R3 beads
conditioned medium was collected, centrifuged for 5min at 1,500r.p.m. and (Life Technologies). Samples were dried and dissolved in 30l 0.1% formic acid
filtered through a 0.45-m pore membrane before use. Conditioned medium (FA). Peptides were separated by reversed-phase chromatography using a nanoLC
was then added to HLEC 24h after plating (6 well plates, 2 105 cells per well) Ultra system (Eksigent), directly coupled with a LTQ-Orbitrap Velos instrument
for a 5-day analysis of proliferation rate using the Promegas CellTiter MTS cell (Thermo) via nanoESI (ProxeonBiosystem). Peptides were loaded onto a Reprosil-
proliferation assay kit. Conversely, analysis of growth of HLEC was performed on Pur C18 column (3m, 400 0.075mm; Dr. Maisch), with a trapping column
cells pre-conditioned with medium from SK-Mel-147 expressing shC or shMDK(1) (Prot Trap Column 0.310mm, ReproSil C18-AQ, 5m), for 10min with a flow
and shMDK(5) to determine the inhibitory effects of MDK depletion. rate of 2.5l per min loading buffer (0.1% FA). Elution was performed with a
HLEC sprouting assay. HLECs were cultured in EGM-2 MV medium (Lonza). 120-min linear gradient (buffer A: 2% ACN, 0.1% FA; buffer B: 100% ACN, 0.1% FA)
Hanging drop cultures were generated 24h after incubation with Cell Tracker at 300nl per min. Peptides were directly electrosprayed into the mass spectrometer
(Molecular Probes, Life Technologies), using 1,000 cells per drop (25l). This using a PicoTip emitter (360/20 OD/IDm tip, ID 10m, New Objective) at 1.4kV
was performed in EBM-2 medium containing 20% Methocel. Sphere formation spray voltage with a heated capillary temperature of 325C and S-Lens of 60%.
was checked by microscopy 24h thereafter. Spheres were then collected in Mass spectra were acquired in a data-dependent manner, with an automatic switch
PBS supplemented with 10% of FBS, centrifuged for 3min at 200r.p.m., and between MS and MS/MS scans using a top 10 method. MS spectra were acquired
resuspended in the desired EGM-2 MV conditioned medium obtained as described with a resolution of 60,000 (FWHM) at 400 m/z in the Orbitrap, scanning a mass
before. When indicated, spheres were treated with 1M rapamycin or 10nM torin 1 range between 350 and 1500 m/z. Peptide fragmentation was performed using
and embedded in type I collagen. Pictures were collected 24h thereafter. The collision-induced dissociation (CID), set at 35%. Label-free quantification was
number and length of sprouts were quantified using ImageJ software. performed in MaxQuant (1.5.1.2) using default settings and further statistical
Melanoma cell attachment and transmigration through HLEC. HLEC were analysis of the quantitative data was conducted with Perseus (1.5.1.6). The mass
seeded in a 24-well glass bottom plate and then incubated at 37C with 5% CO2. spectrometry proteomics data have been deposited to the ProteomeXchange
At confluence, medium was refreshed in the absence or presence of 500g ml-1 Consortium via the PRIDE partner repository with the dataset identifier
recombinant human MDK (Peprotech) for a 16-h incubation. 105 mCherry labelled PXD002703. The proteomic analysis resulted in the identification and quantifi-
SK-Mel-147 or WM164 cells were then seeded on top of the pre-conditioned HLEC cation of a total of 1,705 exosomal proteins (Fig. 3a). A statistical filter (cut-off of
cells for immediate time-lapse imaging (fluorescence videomicroscopy using a FDR 0.05 and a threshold S0 of 0.8) was applied to identify factors enriched with
wide-field microscope coupled with a CCD camera). For SK-Mel-147 cells, images respect to the non-metastatic and non-lymphangiogenic WM164. A second filter
were captured at 2.33-min intervals for a total of 200min, and for WM164 every was used to eliminate factors shared with cell line WM35 (with lymphangiogenesis
3.33min (total of 965.7min). Videos were assembled using ImageJ software, which restricted mostly to cutaneous lesions). This approach identified 139, 180 and
was also used to quantify melanoma cell surface (mCherry-positive areas) at the 188 candidates for long range-acting metastatic factors in 451LU, SK-Mel-28 and
indicated acquisition times. Results are represented as the mean and s.e.m. of the SK-Mel-147 cells, respectively, as summarized in Fig. 3b (Supplementary Table 2).
fold-induction of the mCherry-covered area per frame, analysed in four different The top 20% of these proteins were then scored on the basis of three criteria:
wells (n>200 cells per time point and condition). t-test analyses were performed (i) being common to all cell lines that activate distal lymphangiogenesis,
on time-matched observations to define statistical differences in the spreading (ii) having described functions associated with metastasis but not previously linked
capacity of melanoma cells on control- vs MDK-conditioned HLEC. to lymphangiogenesis, and (iii) being understudied in melanoma. The highest
Transmigration analyses were performed on 8-m pore Boyden chambers ranked protein in this analysis was MDK (Fig. 3b).
containing confluent HLEC monolayer (preconditioned for 24h with empty RNA-seq and bioinformatic analyses. RNA-seq was performed by the CNIO
medium or with medium containing 500g ml-1 recombinant human MDK). Genomics Unit. One microgram of total RNA from the samples was used. The
105 mCherry-labelled SK-Mel-147 or WM164 cells were seeded on top of these pre- average sample RNA Integrity Number was 9.75 (Agilent 2100 Bioanalyzer).
treated HLEC cells using DMEM without growth factors. As a chemo-attractant, The poly(A)+ fraction was purified and randomly fragmented, converted to
complete DMEM was added to the bottom of the plates. Cells were incubated double-stranded cDNA and processed through subsequent enzymatic treatments
in these conditions for 16h and subsequently fixed in 4% paraformaldehyde. of end-repair, dA-tailing, and ligation to adapters as in Illuminas TruSeq Stranded
Chambers were then scanned using a Leica SP5-multiphoton confocal microscope mRNA Sample Preparation Part # 15031047 Rev. D kit (this kit incorporates dUTP
(Leica Microsystems). The number of melanoma-mCherry cells in the upper part during 2nd strand cDNA synthesis, which implies that only the cDNA strand
(non-migrating) and lower part (migrating) of the chamber was quantified using generated during 1st strand synthesis is eventually sequenced). The adapter-
Imaris image analysis software (Bitplane). t-tests were performed on matched ligated library was completed by PCR with Illumina PE primers (eight cycles). The
observations using Graphpad Prism software. resulting purified cDNA library was applied to an Illumina flow cell for cluster
Exosome purification. Exosomes were prepared essentially as described10. Briefly, generation and sequenced on an Illumina HiSeq 2500 following the m anufacturers
cells were cultured in medium supplemented with 10% exosome-depleted FBS protocols. Fifty-base-pair single-end sequenced reads were analysed with the
(Hyclone). FBS was depleted of bovine exosomes by ultracentrifugation at 100,000g nextpresso pipeline (http://bioinfo.cnio.es/nextpresso/), as follows: sequencing
overnight. Supernatant fractions collected from 24-h cell cultures were pelleted by quality was checked with FastQC v0.11.0 (http://www.bioinformatics.babraham.
centrifugation at 500g for 10min. The supernatant was centrifuged at 20,000g for ac.uk/projects/fastqc/). Reads were aligned to the human genome (GRCh37/
20min. Exosomes were then collected by centrifugation at 100,000g for 70min. The hg19) with TopHat-2.0.1038 using Bowtie 1.0.039 and Samtools 0.1.1940, allowing
exosome pellet was resuspended in 13ml PBS and collected by ultracentrifugation 2 mismatches and 20 multihits. Transcript quantification and differential expres-
at 100,000g for 70min. The exosome-producing cells were trypsinized, counted sion were calculated with Cufflinks 2.2.138, using the human GRCh37/hg19
and stored for proteomic analysis. transcript annotations from https://ccb.jhu.edu/software/tophat/igenomes.
ELISA-based analysis of MDK secretion. For the ELISA estimation of soluble or shtml. GSEAPreranked41 was used to perform gene set enrichment analysis of
exosome-contained MDK, cells were incubated for 24h in DMEM with 10% FBS the described gene signatures on a pre-ranked gene list, setting 1,000 gene set
before collection. The conditioned medium was then divided in two fractions, permutations. Only those gene sets with significant enrichment levels (FDR
one for elimination of cell debris by centrifugation at 13,000r.p.m. for 10min, and q<0.25) were finally considered.
another to deplete exosomes by ultracentrifugation at 100,000g for 14h. MDK Intravital multiphoton microscopy. SK-Mel-147 cells labelled with GFP and
expression in these soluble and exosome-cleared conditions was analysed by ELISA transduced with control shRNA (shC) or MDK shRNA(5) were injected subcu-
(Human MIDKINE ELISA Development Kit 900-K190; Prepotech). Readings were taneously (1 106 cells) into 6-week-old female nude mice. Intravital imaging
obtained for three independent biological replicates. To define MDK concentration of the inguinal lymph node adjacent to the tumour was performed 3 weeks after
in exosomes, these were lysed with ELISA extraction buffer 75mM Tris tumour cell injection. To label the inguinal lymph node, 20l of 155kDa TRITC-
pH 7.0; 150mM NaCl; 1mM EGTA; 1mM EDTA and 1% Triton 100; 0.5% sodium dextran was injected intradermally at the lateral tail base on the tumour side. Mice
deoxycholate and protease and phosphatase inhibitors. were anaesthetized with 5% isofluorane and the lymph node was exposed by skin
LCMS/MS analysis of exosome cargo. For exosome profiling, samples were flap surgery as previously described42. Imaging was performed using an Olympus
digested using the standard filter aided sample preparation (FASP) protocol37. FV1000 multiphoton microscope with a 251.05 NA water immersion objective
Briefly, samples were resuspended in UT buffer (8M urea in 100mM Tris-HCl, as previously described43 and adapted for the lymph node. Specifically, we focused
pH 8.01). Proteins were then reduced with 10mM DTT and alkylated using on the subcapsular sinus, the outer area of lymph nodes that serves as a main
50mM IAA for 20min in the dark. Proteins were digested with Lys-C (Wako) barrier to tumour cell entry30,44. Briefly, a Coherent Chameleon Vision II laser
for 6h (1:50). Finally, samples were diluted in 50mM ammonium bicarbonate was tuned to 880nm for excitation of GFP and TRITC-dextran. Collagen was
to reduce the urea concentration to 1M, and were subsequently digested with visualized by second harmonic generation. 3D images were reconstructed with

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Imaris (Bitplane). 4D images were acquired at a 5-m step-size ranging from 100 following settings and filters in Perseus software: 250 randomizations, FDR= 0.05
to 150m in depth from the lymph node surface with a 2-min time interval for and fudge factor S0=0.8. For simplicity, only significant P values are shown in all
30min. Individual Z-slices are provided at different time points in Extended Data experiments.
Fig. 8c (three d ifferent planes containing the cell volume, starting at 20m depth Data availability. The mass spectrometry proteomics data have been deposited
from lymph node capsule with 5m step size) showing no Z-drift. Orthogonal in the ProteomeXchange Consortium via the PRIDE partner repository with the
ZY views are also provided to visualize the 3D location of the cell in relation to dataset identifier PXD002703 (http://proteomecentral.proteomexchange.org/cgi/
the lymphatic vessel over time. Images were analysed in ImageJ. GetDataset?ID=PXD002703). RNA-seq data have been deposited in NCBIs Gene
As shown in Extended Data Fig. 8a (upper panels), MDK-expressing Expression Omnibus with accession number GSE94549. Source Data are provided
SK-Mel-147 cells were found in clusters invading the subcapsular sinus at sites for Figs 1e, 2a, b and 3el and Extended Data Figs 1d, 2df, 3be, 4a, 5a, ci, 6be,
of a large expansion of the vasculature (see dextran-labelled areas). This was in 7a, c, e and 9a, b. All other data that support the findings of this study are available
contrast to individual or small cell clusters of MDK-deficient SK-Mel-147 cells from the authors upon reasonable request.
found blocked at the border of the subcapsular sinus on the collagen-enriched
area (Extended Data Fig. 8a, bottom panels). Snap shots in Extended Data
31. Yajima, I. et al. Spatiotemporal gene control by the Cre-ERT2 system in
Fig. 8b, c further illustrate the impact of MDK deficiency on the ability of melanocytes. Genesis 44, 3443 (2006).
melanoma cells to move towards the vasculature. Therefore, these data provide 32. Marino, S. et al. PTEN is essential for cell migration but not for fate
additional mechanistic explanation as to why modulating the endogenous levels of determination and tumourigenesis in the cerebellum. Development 129,
MDK in melanoma cell lines affects not only their pro-lymphangiogenic capability, 35133522 (2002).
but also their metastatic potential. 33. Denoyelle, C. et al. Anti-oncogenic role of the endoplasmic reticulum
differentially activated by mutations in the MAPK pathway. Nat. Cell Biol. 8,
MDK expression and metastatic risk in human clinical biopsies. MDK levels
10531063 (2006).
were estimated by two independent pathologists. Lymph node and paraffin- 34. Soengas, M. S. et al. Inactivation of the apoptosis effector Apaf-1 in malignant
embedded blocks were collected from a retrospective series of 62 patients with melanoma. Nature 409, 207211 (2001).
melanoma (2185 years old, with a median age of 65; stage IIIII at diagnosis). 35. Alonso-Curbelo, D. et al. RAB7 counteracts PI3K-driven macropinocytosis
Gender distribution was 66.1% female and 33.9% male. Immunohistochemistry activated at early stages of melanoma development. Oncotarget 6,
1184811862 (2015).
was performed in 3-m sections of primary lesions or sentinel lymph nodes as
36. Workman, P. et al. Guidelines for the welfare and use of animals in cancer
indicated and MDK-specific staining was scored as negative, low or high taking research. Br. J. Cancer 102, 15551577 (2010).
into account the intensity of its expression. Survival curves were estimated with the 37. Wisniewski, J. R., Zougman, A., Nagaraj, N. & Mann, M. Universal sample
KaplanMeier product-limit method and compared using log-rank test. Hazard preparation method for proteome analysis. Nat. Methods 6, 359362 (2009).
ratios were calculated using Cox regression and adjusted with univariate and 38. Trapnell, C. et al. Differential gene and transcript expression analysis of
multivariate models for Breslow depth, age or gender. DFS was defined as the RNA-seq experiments with TopHat and Cufflinks. Nat. Protocols 7, 562578
(2012). 10.1038/nprot.2012.016
time interval between diagnosis and the development of the earliest metastasis 39. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-
detected at any anatomical site. DFS was considered censored for patients who efficient alignment of short DNA sequences to the human genome. Genome
did not p resent metastases at the last follow-up. Human tumour biopsies were Biol. 10, R25 (2009).
obtained from the i+12 Biobank (RD09/0076/00118) of the Hospital 12 Octubre, 40. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics
the biobank from the Hospital Clnic de Barcelona, and the Spanish National 25, 20782079 (2009).
41. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based
Biobank Network, with the corresponding informed consent and ethical protocols
approach for interpreting genome-wide expression profiles. Proc. Natl Acad.
approved by their Clinical Investigation Ethical Committees. Sci. USA 102, 1554515550 (2005).
Statistical analyses. Cell proliferation and tumour growth curves were analysed 42. Das, S. et al. Tumor cell entry into the lymph node is controlled by CCL1
by one-way and two-way ANOVA with Dunnetts multiple comparison test (mixed chemokine expressed by lymph node lymphatic sinuses. J. Exp. Med. 210,
model) considering matching among the measures at different time points. P values 15091528 (2013).
are indicated in each figure for statistically significant comparisons (P< 0.05). For 43. Patsialou, A. et al. Intravital multiphoton imaging reveals multicellular
streaming as a crucial component of in vivo cell migration in human breast
the analysis of proteomic data, LFQ values were log2 transformed and missing tumors. Intravital 2, e25294 (2013).
data were imputed based on the observed normal distributions (width value 0.3 44. Willard-Mack, C. L. Normal structure, function, and histology of lymph nodes.
and down-shift 2). Then, a two-sample Students test was performed using the Toxicol. Pathol. 34, 409424 (2006).

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 1 | Non-invasive imaging of c, Histological staining of Lyve1 (brown) in lymph nodes with low or
neolymphangiogenesis in Vegfr3Luc reporter mice. a, qRTPCR analysis high V3-Luc emission. d, Costaining of VEGFR3 (green) and Lyve1
of VEGFC mRNA levels in the indicated melanoma cell lines. Graphs show LEC (brown) in the spleens of mice identified as low and high V3-Luc
means.d. of three biological replicates. Data were normalized to mRNA emitters in vivo. e, Lung metastases in mice as in Fig. 1c imaged by
levels in the poorly metastatic WM164 cell line. b, Immunostaining immunofluorescence for the detection of VEGFR3 (green) and melanoma
for VEGFC (pink) in xenografts of the indicated melanoma cell lines. cells (red).

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Extended Data Figure 2 | Assessment of metastatic potential of Lyve1 in histological sections of sentinel lymph nodes from mice with
melanoma cell lines in Vegfr3Luc mice. a, b, Tumour-associated xenografts generated as in a and killed when primary tumours reached
bioluminiscence driven by subcutaneous implantation of the indicated 1,500mm3. d, Tumour-positive lymph nodes (sentinel, brachial and
cell lines (labelled with mCherry) in Vegfr3Luc nu/nu mice, with images axillar) in mice bearing xenografts of the indicated cell lines. e, Relative
centred on the tumour (a) or showing the lateral flank of the mice to V3-Luc signal in subcutaneous xenografts generated by the indicated cell
visualize sentinel-inguinal and brachial lymph nodes, as well as signal lines. f, Average signal in sentinel and brachial lymph nodes. Data are
from spleen, liver and lung (b). Four main patterns (IIV) of V3-Luc means.e. (minimum 6 mice per condition, 24 lymph nodes per cell line).
emission are shown. Red lines mark the tumour area as detected by Cell lines labelled in green in af express detectable VEGFC mRNA as
mCherry emission (data not shown). Detection of the lymphatic marker summarized in Fig. 1a.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 3 | Whole-body analysis of in Tyr::CreERT2-inducible BrafV600E and BrafV600E; Ptenlox/lox mice,
neolymphangiogenesis induced in immunodeficient and respectively (black coat background), imaged 4 weeks after topical
immunocompetent GEMM. a, Imaging of luciferase induction by a administration of 4-OH-tamoxifen. Arrows mark systemic activation of
PDX isolated from a skin melanoma metastasis and implanted in luciferase. Bottom, quantification of luciferase signal in tumour, sentinel
Vegfr3Luc nu/nu mice. Mice were imaged at different time points to and brachial lymph nodes, spleen, liver and lung. Units: ps1cm2 per
show V3-Luc induced at distal lymph nodes before detection of V3-Luc sr 106. Data correspond to means.d. (4 mice for nevi and 10 mice
at the subcutaneous lesion. Dashed red lines mark tumour area. for melanoma). Statistical analysis: one-way ANOVA with Dunnetts test
b, Quantification of the luciferase signal in tumour, lymph node (sentinel for multiple comparisons. d, Luciferase emission induced by cutaneous
and brachial), spleen, liver and lung, at the defined average tumour sizes melanomas generated in immunocompetent Vegfr3Luc; BrafV600E; Ptenlox/lox
indicated in the x-axis (visualized with a dotted red line for simplicity). (albino) mice, visualized at the indicated times after topical administration
Specific tumour volumes are indicated in the right y-axis. Bioluminescence of 4-OH-tamoxifen. e, Quantification of the luciferase signal in the
data correspond to means.d. (4 mice per condition, 16 lymph nodes indicated organs, as defined in Methods. The average tumour size at each
per PDX). Statistical analysis: t-test. c, Comparative imaging of V3-Luc point is indicated by a red dotted line and in the x-axis. Data correspond to
emission induced by benign nevi and cutaneous melanomas generated means.d. (four mice per condition). Statistical analysis: t-test.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Extended Data Figure 4 | Analysis of metastatic potential in Vegfr3Luc indicated times before and after surgical removal of primary cutaneous
mice. a, Pearson correlation analyses of the luciferase signal versus cell melanoma. Note the reduction in tumour-driven V3-Luc signal,
burden, corresponding to data presented in Fig. 2a. Shown are lymph particularly at visceral sites, and the reactivation at later time points
nodes or lungs with luciferase signal over the background. b, V3-Luc (black arrows), marking metastatic relapse. Fur was removed to facilitate
emission in immunocompetent BrafV600E;Ptenlox/lox (albino) mice at the imaging.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 5 | MDK loss of function impairs melanoma biological replicates. d, Effect of the indicated shRNAs on MDK secretion,
induced lymphangiogenesis and metastasis. a, Detection of MDK analysed by ELISA. e, qRTPCR analysis of VEGFC (left) and VEGFD
protein expression by immunohistochemistry in xenografts of the (right) mRNA expression in SK-Mel-147 cells expressing control or MDK
indicated cell lines. b, Secreted MDK in the indicated cell lines. Upper, shRNA (1) or (5). Data correspond to means.d. of three biological
immunoblots of conditioned medium containing (+) or depleted of replicates. f, Lack of significant effect of MDK shRNA (shown for shRNA
exosomes (). Lower, ELISA-based quantification of MDK in conditioned (1) and (5)) on the growth of SK-Mel-147 cells as subcutaneous xenografts
medium of the indicated cell lines containing or depleted of exosomes in Vegfr3Luc nu/nu mice. Data correspond to means.d. for six mice per
(black and grey bars, respectively). MDK levels in the purified exosome condition. g, Blood vessel density (BLV) defined by immunohistochemical
fraction (white bars) are indicted as a reference. Data were normalized staining for CD31 in xenografts of SK-Mel-147 cells expressing control or
to levels in the complete soluble fraction (black), and correspond to MDK shRNA (1) or (5). h, i, Peri-tumoural or intratumoural lymphatic
means.d. of three biological replicates. c, Depletion of MDK mRNA vessel density (LVD) determined by histological staining for Lyve1 in
detected by qPCR of SK-Mel-147 cells infected with lentiviruses coding for lesions as in f. Data correspond to means.d. for three tumours per
control or MDK shRNA (1) or (5). Data correspond to means.d. of three experimental condition.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Extended Data Figure 6 | See next page for caption.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 6 | MDK accumulates at sites of cells expressing MDK (and their parental controls). e, Increased VEGFR3
neolymphangiogenesis. a, Relative expression of secreted MDK in mRNA in MDK-expressing HLEC, determined by qRTPCR. Data in ae
WM164 cells infected with control lentiviruses (empty) or lentiviruses correspond to means.d. of three biological replicates. f, Proliferative
expressing MDK cDNA (right), analysed in parallel with respect to basal capacity of HLEC incubated with conditioned medium from control- or
levels in primary melanocytes and the indicated cell lines (left). Data MDK-overexpressing WM164 cells (left, red). Statistical analysis: t-test.
correspond to biological triplicates measured by ELISA, using purified g, Depletion of MDK in SK-Mel-147 cells (blue). Data are represented as
recombinant protein as a reference for quantification. b, Representative the relative means.d. of three biological replicates. Statistical analysis:
images of lung metastases identified by mCherry fluorescence. c, qRT one-way ANOVA. h, Dual immunofluorescence analysis to visualize MDK
PCR analysis of VEGFC (left) and VEGFD (right) mRNA showing minimal (green) and lymphatic vessels (Lyve1, red) in V3-Luc-positive lymph
changes in control compared with MDK-overexpressing WM164 cells. nodes of mice implanted with WM164 cells expressing MDK. i, The
d,Assessment of peri-tumoural lymphatic vessel density (Lyve1 staining) equivalent analysis in lungs.
in consecutive histological sections of xenografts generated by WM164

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Extended Data Figure 7 | MDK enhances the ability of melanoma cells retained (up, pseudocoloured in red) or transmigrating (down, green)
cells to interact with and migrate through HLEC. a, Attachment and through the HLEC layer defined by confocal videomicroscopy as described
spreading of mCherry-labelled WM164 or SK-MEL-147 melanoma cell in Methods. c, Percentage of migrating WM-164 or SK-MEL-147 cells
lines on a confluent monolayer of HLEC preincubated for 16h with in conditions as in b. Statistical analysis: t-test. d, Histological sections
500gml1 recombinant MDK. Statistical analysis: t-test. See of xenografts generated by SK-Mel-147 cells expressing control or
Supplementary Videos 1 and 2 for live imaging of the spreading MDK shRNA and stained for Lyve-1 (brown) to visualize tumour
capacity of WM164 and SK-Mel-147 melanoma cells, respectively, cell intravasation. e, Quantification of lymphatic vessels colonized by
in control versus MDK-treated HLEC. b, Migration of SK-Mel-147 melanoma cells in d per 25-mm2 field analysed. Data correspond to
cells through a confluent layer of HLEC in the absence or presence of means.d. for three biological replicates (with a minimum of six fields
recombinant MDK (500gml1). Pictures show representative images of analysed per tumour).

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 8 | Intravital multiphoton imaging of lymph micrometastases. Scale bar, 50m. b, Still images showing the differential
node metastases from melanoma cells expressing or lacking MDK. intralesional motility of the indicated SK-Mel-147 cell populations from
a, Maximum projection, top views or side views of lymph nodes from lesions in a. The yellow dotted lines are shown to visualize the border of
representative mice three weeks after implantation of SK-Mel-147 a dextran-labelled vessel in the context of a highly motile shC-melanoma
cells labelled with GFP and transduced with control shRNA (shC, top) cell (solid white outline). Vertical dotted white lines are included as
or MKD shRNA (shMDK, bottom). Tumour cells are visualized by reference to track cell movements. Scale bar, 10m. c, Z-slices at different
green fluorescence, and collagen fibres by second harmonic generation time points from the time images in b. Bottom panels correspond to
(blue). Vessels at the subcapsular sinus (SCS) are stained with 155-kD orthogonal zy views at the indicated time points to show the 3D location
dextran (magenta). Note the horizontal and vertical growth of shC cells of the cell (circled in b and marked with an arrow in c) with respect to the
(metastasis expressing MDK), and the disorganization of the surrounding lymphatic vasculature. Scale bar, 10m.
SCS. By contrast, shMDK cells were identified as single cells or rare

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Letter RESEARCH

Extended Data Figure 9 | Modulation of lymphangiogenic features by c, Micrographs to visualize LEC-sprouting assays in a, b. Yellow dots
MDK via the mTOR pathway. a, Sprouting ability of collagen-embedded mark the ends of sprouts. d, Immunofluorescence micrographs showing
LEC (stained with CellTracker green for fluorescence imaging). Data the expression of MDK (pink), lymphatic vessels (LYVE1, green) and
correspond to the length of sprouts generated when LEC colonies were p-RPS6 (red) in lymph nodes in an MDK gain-of-function setting, where
incubated with conditioned medium (CM) from WM164 or WM164- Vegfr3Luc mice were implanted subcutaneously with WM164 cells and their
MDK cells. b, Length of sprouts of collagen-embedded LEC colonies in the isogenic derivatives WM164-MDK. Images were collected 2 weeks after
absence or presence of mTOR inhibitors. Statistical analysis: t-test; n= 10. implantation. White boxes mark magnified areas shown to the right.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
RESEARCH Letter

Extended Data Figure 10 | MDK expression in human melanoma areas (bottom). d, KaplanMeier survival curves of melanoma patients
specimens. a, MDK expression shown by immunostaining (brown) as in Fig. 4h, considering only stage III (sentinel lymph node-negative)
in paraffin-embedded sections of representative examples of human patients classified on the basis of low and high MDK expression in the
benign nevus, stage III melanoma specimens and skin and lymph node corresponding sentinel lymph nodes. The number of patients at risk is
metasases. b, MDK levels in the indicated melanocytic lesions scored shown for each time point. e, Multivariate analysis of the prognostic value
as negative, low and high (see Methods). Statistical analysis, 2 t-test; of MDK expression on DFS, showing that it is independent of age, gender
n=20 per category. c, Micrographs of low- and high-MDK expressing and Breslow depth. f, Proposed mode of action of MDK in the induction of
sentinel lymph nodes, focusing on subcapsular sinus (top) and medullary lymphovascular niches and metastasis in melanoma.

2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

Anda mungkin juga menyukai