Prox-1 Promotes Invasion of Kaposiform Hemangioendotheliomas

9
Prox-1 Promotes Invasion of Kaposiform Hemangioendotheliomas Soheil S. Dadras 1,2 , Adrienne Skrzypek 1 , Lynh Nguyen 1 , Jay W. Shin 1,3 , Martin M.P. Schulz 3 , Jack Arbiser 4 , Martin C. Mihm 2 and Michael Detmar 1,3 Kaposi’s sarcoma (KS) is the most frequently occurring malignant tumor in patients infected with HIV. Recent studies have revealed that infection of vascular endothelial cells with KS-associated herpes virus in vitro results in a lymphatic reprogramming of these cells, with potent induction of the lymphatic marker genes podoplanin and vascular endothelial growth factor receptor-3, which is mediated by upregulation of the transcription factor Prox1. However, the potential effects of Prox1 expression on the biology of KS and, in particular, on the aggressive and invasive behavior of KS tumors in vivo have remained unknown. We stably expressed Prox1 cDNA in the two mouse hemangioendothelioma cell lines EOMA and Py-4-1, well-established murine models for kaposiform hemangioendothelioma. Surprisingly, we found that expression of Prox1 was sufficient to induce a more aggressive behavior of tumors growing in syngenic mice, leading to enhanced local invasion into the muscular layer and to cellular anaplasia in vivo, and increased migration rate in vitro. This enhanced malignant phenotype was associated with upregulation of several genes involved in proteolysis, cell adhesion, and migration. Together, these results indicate that Prox1 plays an important, previously unanticipated role in mediating the aggressive behavior of vascular neoplasms such as KS. Journal of Investigative Dermatology (2008) 128, 2798–2806; doi:10.1038/jid.2008.176; published online 26 June 2008 INTRODUCTION Kaposi’s sarcoma (KS) is the most frequently occurring malignant tumor in patients infected with HIV (Su et al., 1990), but also occurs in HIV-negative immunosuppressed patients. KS mainly affects the skin and forms lesions of various types, including early inflammatory and patch stage lesions, as well as tumors with a predominant population of spindle cells. Infection with Kaposi’s sarcoma-associated herpesvirus (KSHV) is essential for KS tumor formation (Chang et al., 1994). In KS lesions, KSHV-infected cells characteristically appear spindle shaped and are associated with slit-like vessels/spaces that often contain red blood cells. KS has been considered to be a neoplasm of KSHV-infected lymphatic endothelium, due to the morphological characteristics of the tumor cells and the expression of several lymphatic lineage-specific genes includ- ing vascular endothelial growth factor receptor-3 and podo- planin by KS tumor cells. However, recent studies have revealed that infection of vascular endothelial cells with KSHV in vitro results in a lymphatic reprogramming of these cells, with potent induction of the lymphatic marker genes podoplanin and vascular endothelial growth factor receptor- 3 (Hong et al., 2004; Wang et al., 2004). Importantly, we found that the transcription factor Prox1 is induced by infection of vascular endothelium with KSHV, and that knockdown of Prox1 interfered with the KSHV-mediated induction of a lymphatic phenotype (Hong et al., 2004). However, the potential effects of Prox1 expression on the biology of KS and, in particular, on the aggressive and invasive behavior of KS tumors in vivo have remained unknown. The homeobox gene Prox1 is a transcription factor related to the Drosophila gene prospero, which plays a major role in the development of the central nervous system (Lavado and Oliver, 2007), liver, and pancreas (Sosa-Pineda et al., 2000; Burke and Oliver, 2002) and in lens fiber elongation (Wigle et al., 1999). Importantly, Prox1 is a master gene controlling the normal embryonic development and differentiation of the lymphatic vasculature. Prox1-deficient mice are devoid of lymphatic vasculature, and in these animals, endothelial cells fail to acquire the lymphatic phenotype; instead, they remain as blood vascular endothelium (Wigle and Oliver, 1999). We and others have shown that ectopic expression of Prox1 in differentiated blood vascular endothelial cells leads to lymphatic endothelial reprogramming of these cells, asso- ciated with downregulation of blood vascular-specific genes ORIGINAL ARTICLE 2798 Journal of Investigative Dermatology (2008), Volume 128 & 2008 The Society for Investigative Dermatology Received 31 October 2007; accepted 15 April 2008; published online 26 June 2008 1 Department of Dermatology, Cutaneous Biology Research Center, Boston, Massachusetts, USA; 2 Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA; 3 Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETH Zurich, Switzerland and 4 Department of Dermatology, Emory University, Atlanta, Georgia, USA Correspondence: Dr Soheil S. Dadras, Pathology and Dermatology, Stanford University Medical Center, 300 Pasteur Drive, Stanford, CA 94305-5324, USA. E-mail: [email protected] Abbreviations: AFP, alpha fetoprotein; KS, Kaposi’s sarcoma; KSHV, Kaposi’s sarcoma-associated herpesvirus; QRT-PCR, quantitative real-time reverse transcription-PCR

Transcript of Prox-1 Promotes Invasion of Kaposiform Hemangioendotheliomas

Prox-1 Promotes Invasion of KaposiformHemangioendotheliomasSoheil S. Dadras1,2, Adrienne Skrzypek1, Lynh Nguyen1, Jay W. Shin1,3, Martin M.P. Schulz3, Jack Arbiser4,Martin C. Mihm2 and Michael Detmar1,3

Kaposi’s sarcoma (KS) is the most frequently occurring malignant tumor in patients infected with HIV. Recentstudies have revealed that infection of vascular endothelial cells with KS-associated herpes virus in vitro resultsin a lymphatic reprogramming of these cells, with potent induction of the lymphatic marker genes podoplaninand vascular endothelial growth factor receptor-3, which is mediated by upregulation of the transcription factorProx1. However, the potential effects of Prox1 expression on the biology of KS and, in particular, on theaggressive and invasive behavior of KS tumors in vivo have remained unknown. We stably expressed Prox1cDNA in the two mouse hemangioendothelioma cell lines EOMA and Py-4-1, well-established murine modelsfor kaposiform hemangioendothelioma. Surprisingly, we found that expression of Prox1 was sufficient toinduce a more aggressive behavior of tumors growing in syngenic mice, leading to enhanced local invasion intothe muscular layer and to cellular anaplasia in vivo, and increased migration rate in vitro. This enhancedmalignant phenotype was associated with upregulation of several genes involved in proteolysis, cell adhesion,and migration. Together, these results indicate that Prox1 plays an important, previously unanticipated role inmediating the aggressive behavior of vascular neoplasms such as KS.

Journal of Investigative Dermatology (2008) 128, 2798–2806; doi:10.1038/jid.2008.176; published online 26 June 2008

INTRODUCTIONKaposi’s sarcoma (KS) is the most frequently occurringmalignant tumor in patients infected with HIV (Su et al.,1990), but also occurs in HIV-negative immunosuppressedpatients. KS mainly affects the skin and forms lesions of varioustypes, including early inflammatory and patch stage lesions, aswell as tumors with a predominant population of spindle cells.Infection with Kaposi’s sarcoma-associated herpesvirus (KSHV)is essential for KS tumor formation (Chang et al., 1994). In KSlesions, KSHV-infected cells characteristically appear spindleshaped and are associated with slit-like vessels/spaces thatoften contain red blood cells. KS has been considered to be aneoplasm of KSHV-infected lymphatic endothelium, due to themorphological characteristics of the tumor cells and theexpression of several lymphatic lineage-specific genes includ-

ing vascular endothelial growth factor receptor-3 and podo-planin by KS tumor cells. However, recent studies haverevealed that infection of vascular endothelial cells with KSHVin vitro results in a lymphatic reprogramming of these cells,with potent induction of the lymphatic marker genespodoplanin and vascular endothelial growth factor receptor-3 (Hong et al., 2004; Wang et al., 2004). Importantly, wefound that the transcription factor Prox1 is induced byinfection of vascular endothelium with KSHV, and thatknockdown of Prox1 interfered with the KSHV-mediatedinduction of a lymphatic phenotype (Hong et al., 2004).However, the potential effects of Prox1 expression on thebiology of KS and, in particular, on the aggressive and invasivebehavior of KS tumors in vivo have remained unknown.

The homeobox gene Prox1 is a transcription factor relatedto the Drosophila gene prospero, which plays a major role inthe development of the central nervous system (Lavado andOliver, 2007), liver, and pancreas (Sosa-Pineda et al., 2000;Burke and Oliver, 2002) and in lens fiber elongation (Wigleet al., 1999). Importantly, Prox1 is a master gene controllingthe normal embryonic development and differentiation of thelymphatic vasculature. Prox1-deficient mice are devoid oflymphatic vasculature, and in these animals, endothelial cellsfail to acquire the lymphatic phenotype; instead, they remainas blood vascular endothelium (Wigle and Oliver, 1999). Weand others have shown that ectopic expression of Prox1 indifferentiated blood vascular endothelial cells leads tolymphatic endothelial reprogramming of these cells, asso-ciated with downregulation of blood vascular-specific genes

ORIGINAL ARTICLE

2798 Journal of Investigative Dermatology (2008), Volume 128 & 2008 The Society for Investigative Dermatology

Received 31 October 2007; accepted 15 April 2008; published online26 June 2008

1Department of Dermatology, Cutaneous Biology Research Center, Boston,Massachusetts, USA; 2Department of Pathology, Massachusetts GeneralHospital and Harvard Medical School, Boston, Massachusetts, USA; 3Instituteof Pharmaceutical Sciences, Swiss Federal Institute of Technology, ETHZurich, Switzerland and 4Department of Dermatology, Emory University,Atlanta, Georgia, USA

Correspondence: Dr Soheil S. Dadras, Pathology and Dermatology, StanfordUniversity Medical Center, 300 Pasteur Drive, Stanford, CA 94305-5324,USA. E-mail: [email protected]

Abbreviations: AFP, alpha fetoprotein; KS, Kaposi’s sarcoma; KSHV, Kaposi’ssarcoma-associated herpesvirus; QRT-PCR, quantitative real-time reversetranscription-PCR

and upregulation of a number of lymphatic-specific genessuch as vascular endothelial growth factor receptor-3 andpodoplanin (Hong et al., 2002; Petrova et al., 2002).

Although the role of Prox1 in embryonic organogenesisand lymphatic vasculogenesis is well established, its activityin tumorigenesis remains poorly understood. Recently, Prox1loss of function has been detected in diffuse large B-celllymphomas (Nagai et al., 2003), adenocarcinomas of thebiliary tract (Laerm et al., 2007), and hepatocellularcarcinomas (Shimoda et al., 2006), suggesting a possiblerole of Prox1 as a tumor suppressor gene. In these types ofcancers, Prox1 loss of function was mainly due to genomicdeletions and epigenetic silencing. In contrast, Prox1 isstrongly expressed by human KSs in situ. Owing to the lack ofsuitable mouse models of KS, however, the biological role ofProx1 in KS pathology has been difficult to assess.

In this study, we used two established mouse modelsof Prox1-negative kaposiform hemangioendotheliomas,hemangioendothelioma (EOMA) cells (Hoak et al., 1971),and Py-4-1 cells (Dubois-Stringfellow et al., 1994), to investi-gate the biological effects of stable Prox1 expression ontumor growth and invasion. Although nodular KS in childrencan histologically mimic kaposiform hemangioendotheliomathe latter is a distinct entity and vascular tumor predomi-nantly occurring in the retroperitoneum or soft tissues ofinfants and the skin of young adults (Zukerberg et al., 1993),associated with thrombocytopenia and hemorrhage (Sarkaret al., 1997; Enjolras et al., 2000). Surprisingly, we found thatthe expression of Prox1 was sufficient to induce a moreaggressive behavior of tumors growing in syngenic mice,leading to enhanced local invasion into the muscular layerand cellular anaplasia in vivo, and increased migrationin vitro. This enhanced malignant phenotype was associatedwith upregulation of several genes involved in proteolysis,cell adhesion, and migration. Our results indicate that Prox1plays an important, previously unanticipated role in mediat-ing the aggressive behavior of vascular neoplasms such as KS.

RESULTSStable overexpression of Prox1 in hemangioendotheliomacell lines

We first investigated the expression of Prox1 in the parentalEOMA and Py-4-1 mouse hemangioendothelioma cell linesby quantitative real-time reverse transcription-PCR (QRT-PCR) and western blotting. We found that neither EOMA norPy-4-1 cells expressed endogenous murine Prox1 at themRNA (data not shown) or protein level (Figure 1a). Next, westably expressed a human Prox1 expression vector (pcDNA/Prox1) or the empty vector alone (pcDNA) in both cell lines.Human Prox1 shares more than 98% homology with themurine protein at the amino-acid level. We isolated morethan 50 Prox1-expressing cell clones and comparedtheir human Prox1 mRNA levels to the endogenous murineProx1 expression levels of primary human lymphaticendothelial cells, which are known to express high Prox1levels (Hirakawa et al., 2003). EOMA and Py-4-1 stableclones expressed human Prox1 mRNA in a wide range (datanot shown). Western blot analysis of cell lysates confirmed

efficient Prox1 protein expression in stable Py-4-1 Prox1transfectants but not in the control clones (Figure 1a).Differential immunofluorescence stains for Prox1 and forthe pan-endothelial marker CD31 confirmed strong nuclearexpression of Prox1 protein (Figure 1c), compared with aweak background signal in the control clones (Figure 1b).Human Prox1 protein expression was also confirmed in thestable EOMA Prox1 clones (data not shown). Three indepen-dent clones of control and Prox1-transfected EOMA andPy-4-1 cells were selected for further studies.

In vivo growth and morphology of Prox1-expressinghemangioendotheliomasWe first investigated the effect of different numbers ofimplanted cells and the effect of added Matrigel matrix onthe growth and morphological characteristics of parentalEOMA and Py-4-1 tumors. We found that injection of 5�105

EOMA or Py4-1 cells per site gave rise to tumors of700–800 mm3 in volume after 16 days. Injection of highercell numbers, that is, 1� 106 or 2�106, resulted in a lethalityof up to 43%, due to tumor burden, hemorrhage, and plateletcoagulopathy, similar to a previous study (Dubois-String-fellow et al., 1994). Without the addition of Matrigel matrixto the cell suspension, large cystic tumors formed butfrequently collapsed (Figure S1a). Admixture of cell cloneswith Matrigel resulted in a rather regular, soft tumor mass(Figure S1b). Addition of Matrigel accelerated the tumorgrowth rate during the initial 4 days; however, this difference

80 kD-

PC1 PC2 PC3 PP12 PP16 PP21

Prox1Control

Control clone Prox1 clone

Prox1/CD31 Prox1/CD31

Figure 1. Overexpression of human Prox1 in stably transfected mouse

Py-4-1 cell clones. (a) Western blot analysis confirmed high amounts of Prox1

protein (80 kDa) in cell lysates obtained from Py-4-1 cells stably transfected

with pcDNA3-Prox1 vector (PP12, PP16, and PP21), as compared with

control transfectants (PC1-3). (b) Differential immunofluorescence stains for

Prox1 (green) and CD31 (red) demonstrated very weak background nuclear

signal for Prox1 and strong staining for the panvascular marker CD31 in a

control Py-4-1cell clone. (c) In contrast, a Py-4-1-Prox1 stable clone showed

strong Prox1 protein expression in the nucleus. Bar¼ 50 mm.

www.jidonline.org 2799

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

normalized after 8 days (Figure S1c) when the average tumorvolume was similar in the presence (490.6 mm3) or absence(416 mm3) of Matrigel. Lectin perfusion studies revealed thatthe luminal surfaces of intra- and peritumoral vessels werestained by fluorescent Lycopersicon esculentum (FigureS1d–f), confirming efficient tumor perfusion. The tumorsusually formed an intradermal nodule with solid and cysticarchitecture without well-formed vascular lumina, whichextended deep into the subcutis (Figure S1e). Tumor-associated arteries and veins, stained by green fluorescentlectin, were present deep within the subcutis (Figure S1f). Nosignificant necrosis was identified by routine histology.

We then injected three control (empty vector-expressing)clones and three stably Prox1-expressing EOMA and Py-4-1clones intradermally into the back skin of 129P3 and B6D2F1mice, respectively. Analysis of total RNA extracted fromstable cell clones in vitro and from tumors, after 16 days ofin vivo growth, by QRT-PCR for Prox1 revealed that alltumors maintained high levels of human Prox1 mRNAexpression (Figure 2a). The expression level of human Prox1mRNA was comparable in the mouse cell clones and mousetumors with that of cultured human lymphatic endothelialcells. Differential immunofluorescence analysis revealedCD31-positive, intradermal tumor with solid and cysticarchitecture without well-formed vascular lumina, in both

the control and Prox1-expressing tumors (data not shown),whereas Prox1 showed a strong nuclear signal only in theProx1-transfected tumors (Figure 2c)—confirming stable androbust exogenous Prox1 expression—but not in the controltumors (Figure 2b).

Prox1 induces local invasion, anaplasia, and migrationof tumor cells

Histological analysis of tumor sections revealed two distinctand reproducible morphologic alterations: (i) invasion of theskeletal muscle by Prox1-expressing hemangioendotheliomacells and (ii) severe cytologic atypia (anaplasia) of the Prox1-expressing tumor cells, independent of cell line (both EOMAand Py-4-1). Infiltration and destruction of the adjacentskeletal muscle layer was seen in 45.5% of Prox1-expressingtumors (10 of 22), usually located at the deep tumor margin,but only in 6.3% of the control tumors (1 of 16, P¼0.004)(Figure 3a–c). Given the local invasive behavior of Prox1tumors in vivo, we examined the rate of migration in vitro(Figure 3d). Monolayer wounding assays demonstrated asignificant increase in migration rates for three independentProx1-expressing Py-4-1 cell clones (PP8, PP11, PP17,P¼0.0185) as compared with clones transfected only withcontrol vector (PC1, PC2, PC12). Despite the local invasivebehavior and increased migration, necropsy of all tumor-bearing mice did not reveal any evidence of macroscopic ormicroscopic metastases in the liver, lung, spleen, or lymphnodes.

Human Prox1 mRNA

Py4-1 Cells

Py4-1 Tumor

LEC

12

9

6

3

0PC1 PC2 PC3 PP8 PP11 PP12

Control Prox1

LEC

Rel

ativ

e ge

ne e

xpre

ssio

n

DermisDermis

Control tumor Prox1 tumor

TumorTumor

CD31/Prox1 CD31/Prox1

Figure 2. Prox1 cell clones and their respective mouse tumor implants

maintained high levels of Prox1 expression. (a) QRT-PCR of total RNA

extracted from stable Prox1 clones (open bar) and from tumors (closed bar)

confirmed high levels of Prox1 mRNA expression, compared with primary

human lymphatic endothelial cells. All measurements are shown relative to

the expression levels of the human GAPDH housekeeping gene. Data shown

are the mean of RNA isolations from independent cell clones (n¼6) and

tumors (n¼ 6), all tested in triplicates±SEM. (b and c) Immunofluorescence

stains for Prox1 (green) and CD31 (red) demonstrated a strong nuclear signal

for Prox1 in Prox1-expressing tumor cells, but not in control tumor cells, and

in endothelial cells lining adjacent dermal lymphatic vessels (arrowheads).

Bar¼50 mm.

Freq

uenc

y of

mus

cle

inva

sion

(%

)

60

40

20

0

10

0

20

30

40

Control (n =16) Prox1 (n =22)

P =0.004P =0.0185

Mon

olay

er w

ound

ing

assa

y

o

pen

area

(%

)

PC1 PC2 PC12 PP8 PP11 PP17

Control Prox1

Muscle

Muscle

Prox1 tumorControl tumor

Figure 3. Prox1 expression induced tumor cell invasion in vivo and

enhanced migration in vitro. (a and b) Histology of control and

Prox1-expressing tumor implants showed invasion of Prox1 tumors.

Prox1-expressing tumor cells (arrows) but not control tumors infiltrated the

skeletal muscle layer. Bar¼ 50mm. (c) The frequency of skeletal muscle

invasion was significantly increased in the Prox1-expressing tumors (n¼ 22,

P¼ 0.004) in comparison with controls (n¼ 16). (d) Monolayer wounding

in vitro assay showed significantly increased migration rates for

Prox1-expressing Py-4-1 cell clones (PP8, PP11, PP17, P¼ 0.0185) as

compared with clones transfected with only control vector (PC1, PC2, PC12).

Data are shown as mean±SEM. Each bar represents the result of three

independent experiments.

2800 Journal of Investigative Dermatology (2008), Volume 128

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

When examined under low-power magnification, bothcontrol and Prox1 tumors showed similar histologicalfindings: solid and cystic architecture without well-formedvascular lumina, composed of sheets of tumor cells, pools oferythrocytes, and remnants of Matrigel matrix (Figure S2a–b).Surprisingly, when examined under high-power magnifica-tion, we frequently identified severe cytologic atypia(anaplasia), as evidenced by nuclear pleomorphism andhyperchromasia, in the Prox1 tumors (59.1%, 13 of 22)(Figure S2d), but only rarely in the control tumors (11.8%, 2of 17; P¼ 0.001) (Figure S2c). An enhanced mitotic activitywas not identified during routine histological examination.These findings, together with the previously suggestedpotential role of Prox1 in cell proliferation during eyedevelopment (Wigle et al., 1999), prompted us to measurethe cellular proliferation rate of Prox1-expressing cell clones.However, we detected no change in the growth rate of Prox1cell clones in vitro, as compared with the control clones(Figure S2e). Similarly, Prox1 tumors did not exhibit anaccelerated growth rate compared with the control tumors(Figure S2f), and no significant change in the average weightwas detected between Prox1 tumors (0.33±0.23 g, n¼32)and control tumors (0.40±0.18 g, n¼16; P¼0.14). Similarresults were obtained for Prox1-expressing EOMA cells (datanot shown). Overall, these results show that Prox1 expressionleads to enhanced cellular invasion, nuclear pleomorphism,and migration without enhancing the cellular proliferationrate.

Differential immunofluorescence analysis of CD31 andLYVE-1 expression demonstrated numerous peritumoralblood vessels, but only a few peritumoral lymphatic vessels(Figure 4a–b). Computer-assisted morphometric analysis, inboth the control and Prox1 tumor sections, revealed nosignificant difference in the peritumoral blood vessel density,size, or relative blood vascular area in the Prox1 tumors(n¼18), compared with the control tumors (n¼19)(Figure 4c). Similarly, no significant changes were detectedin the peritumoral lymphatic vessel density, size, or relativelymphatic vascular area (data not shown).

Prox1 induces genes involved in proteolysis and lymphaticdifferentiation

The enhanced invasiveness of Prox1-expressing hemangioen-dotheliomas prompted us to investigate the underlyingmolecular mechanisms. Comparative gene array analyses ofcultured control and Prox1 clones revealed that stableoverexpression of Prox1-induced genes that are involved inproteolysis, cell adhesion, migration, differentiation, regula-tion of gene transcription, and fatty acid metabolism (Table 1).Two of the most potently induced genes were von Willebrandfactor and alpha fetoprotein (AFP; Table S1). Prox1 alsoregulated several hyaluronan-related genes, including thelymphatic-specific hyaluronan receptor LYVE-1, which ap-pears as extracellular link domain-containing 1 (Xlkd1) (TableS1). We next used QRT-PCR of total RNA extracted fromcultured cells and from tumors to validate select genes thatwere most potently regulated by Prox1 based on the gene-array results (Table 2). We found that the expressions of von

Willebrand factor, alpha fetoprotein, and LYVE-1 werestrongly upregulated in cultured Prox1-expressing clonesPP8, PP11, and PP12, as well as in their respective tumors.Although Prox1 did not upregulate the expression of thelymphatic marker podoplanin in cultured cells, podoplaninwas significantly upregulated in the Prox1 tumors (Table 2).

Then, we performed immunofluorescence analyses ofselected Prox1-induced targets in paraffin-embedded tumorsections. We found a strong increase in LYVE-1 proteinexpression in the cytoplasm and cell membrane of Prox1-expressing tumors compared with control tumors (FigureS3a–b and Figure 4b). In particular, LYVE-1-positive tumorcells were found to infiltrate the skeletal muscle layer (FigureS3c). Immunohistochemical analysis of von Willebrandfactor, AFP, and podoplanin confirmed focal expression inthe cytoplasm of Prox1 tumor cells but not in the controltumors (data not shown).

DISCUSSIONOur results reveal that stable (chronic) overexpression of thetranscription factor Prox1 in mouse hemangioendotheliomacells induces an invasive phenotype in vivo and enhancedmigration rate in vitro, promotes expression of genes involvedin cell migration and proteolysis, and also partially repro-grams these cells to acquire the expression of lymphaticendothelium-specific genes. Previous studies have revealedthat Prox1 is a master gene controlling the embryonicdevelopment of the lymphatic vascular system in mice (Honget al., 2002). Moreover, Prox1 is also involved in cellfate decisions in other organ systems, such as the centralnervous system (Lavado and Oliver, 2007), liver, and

120

100

80

60

40

20

0Control (n =18)

Prox1 (n =19)

Control (n =18)

Prox1 (n =19)

Control (n =18)

Prox1 (n =19)

Blo

od v

esse

ls p

er m

m2

Blo

od v

esse

l siz

e (�

m2 )

600

500

400

300

200

100

0

8

6

4

2

0

Rel

ativ

e bl

ood

vasc

ular

a

rea

(%)

DermisDermis

Tumor Tumor

Control tumor Prox1 tumorCD31/LYVE-1 CD31/LYVE-1

Figure 4. Prox1 expression did not induce tumor angiogenesis or

lymphangiogenesis. (a and b) Immunofluorescence stains for LYVE-1 (green)

and CD31 (red) revealed a few dermal lymphatics (arrowheads) and

numerous tumor-associated blood vessels. Note: Prox1 tumor cells

demonstrated significantly enhanced LYVE-1 expression localized to the

cytoplasm and membrane. Bar¼ 50mm. (c) No change of tumor-associated

blood vascular density, size, or relative area was detected in the Prox1 tumors

(n¼18), compared with the control (n¼19). Data are shown as mean±SEM.

www.jidonline.org 2801

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

pancreas (Sosa-Pineda et al., 2000; Burke and Oliver, 2002),and in lens fiber elongation (Wigle et al., 1999). Importantly,recent studies have shown that transient (short-term) over-expression of Prox1 in vascular endothelial cells promotesexpression of a number of lymphatic-specific genes whiledownregulating several blood vascular genes (Hong et al.,2002; Petrova et al., 2002), and that Prox1 is involved in thelymphatic reprogramming of vascular endothelium by theKS-associated herpes virus (Hong et al., 2004). Moreover,Prox1 expression has been found in some types of tumorswhere its role has remained unclear (Nagai et al., 2003;

Shimoda et al., 2006; Laerm et al., 2007). However, thebiological effects of stable overexpression of Prox1 haveremained unknown.

Our study reveals that stable overexpression of Prox1 doesnot promote in vitro proliferation of tumor cells or in vivotumor growth. Instead, Prox1 expression leads to a moreaggressive tumor phenotype of vascular tumors, as evidencedby increased local invasion into the muscle layer and bycellular anaplasia. This local invasive behavior is corrobo-rated by the enhanced migration rate of Prox1-expressingPy-4-1 cells in vitro. These results are in agreement with

Table 1. Selected genes upregulated in Prox1-expressing cells according to biological processes

Fold change (sorted by mean)

Prox1/control microarray Prox1/control microarray

Symbol Name I II

Proteolysis

Prss23 Protease, serine, 23 2.537 25.233

Serpina3g Serine (or cysteine) peptidase inhibitor, clade A member 3G 5.864 7.317

Casp12 Caspase 12 2.077 6.862

Usp45 Ubiquitin-specific peptidase 45 2.555 3.605

Tll1 Tolloid-like 3.080 1.698

Wwp1 WW domain-containing E3 ubiquitin protein ligase 1 2.599 1.820

Lipid, fatty acid, and steroid metabolism

Sc4 mol Sterol-C4-methyl oxidase-like 3.355 12.441

Cyp1b1 Cytochrome P450, family 1, subfamily b, polypeptide 1 2.497 4.616

Plscr2 Phospholipid scramblase 2 2.998 3.164

Fabp4 Fatty acid-binding protein 4, adipocyte 1.781 3.896

Pik3c2a Phosphatidylinositol 3-kinase, C2 domain-containing, alpha 2.023 3.262

Fdft1 Farnesyl diphosphate farnesyl transferase 1 2.696 2.296

Ltc4s Leukotriene C4 synthase 1.968 2.634

Pparg Peroxisome proliferator activated receptor gamma 2.042 2.258

Impad1 Inositol monophosphatase domain containing 1 2.444 1.808

Cell adhesion

Vwf Von Willebrand factor homolog 38.429 23.760

Cdh1 Cadherin 1 19.111 2.069

Selp Selectin, platelet 3.901 7.798

Nid2 Nidogen 2 4.491 3.358

Hspg2 Perlecan (heparan sulfate proteoglycan 2) 2.109 4.806

Ctnnal1 Catenin (cadherin associated protein), alpha-like 1 4.253 2.659

Col8a1 Procollagen, type VIII, alpha 1 1.762 4.932

Pkp2 Plakophilin 2 1.630 2.824

Cell structure and motility

Gem GTP-binding protein 3.795 4.693

Ctnnal1 Catenin (cadherin-associated protein), alpha-like 1 4.253 2.659

Foxp1 Forkhead box P1 1.815 4.894

Fblim1 Filamin-binding LIM protein 1 2.208 2.445

Klhl7 Kelch-like 7 (Drosophila) 2.717 1.915

2802 Journal of Investigative Dermatology (2008), Volume 128

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

findings in Prox1-deficient mice, where the migration oflymphatic precursor cells away from the cardinal vein isstrongly inhibited (Wigle and Oliver, 1999). It remains tobe investigated whether Prox1 expression is upregulated inwound-associated lymphatic endothelial cells and mightthereby also contribute to lymphatic vessel migration intothe granulation tissue. Gene array profiling studies revealedthat the invasive tumor phenotype is associated withupregulation of genes involved in proteolysis, cell adhesion,and migration. For example, Prox1 induced the expression ofgenes encoding tissue proteases, including serine protease23. Thus, Prox1 likely enhances cellular invasion bypromoting proteolysis of extracellular matrix proteins andenhancing cellular motility and migration. The observedcellular anaplasia in Prox1 tumors may be related to theupregulated expression of genes controlling cell differentia-tion, such as AFP. AFP is normally synthesized in the liver,the intestinal tract, and the yolk sac of the fetus; however, thisfetal protein is reexpressed in hepatocellular carcinomas andin some adult tumors that exhibit an embryonic state ofdifferentiation, such as germ cell neoplasms and yolk sactumors.

Prox1 overexpression did not significantly accelerate theproliferation rate of EOMA and Py-4-1 cells in vitro or in vivo.These findings are supported by our gene array analysis,which did not detect major stimulation of the expression ofgenes linked to cell cycle progression, such as cyclins A2, B2,D3, E1, H, histone H3, histone H4, or proliferating cellnuclear antigen. However, the observed downregulation ofCdkn1b in Prox1-expressing clones is in agreement withrecent findings during embryonic lens fiber development inProx1-null mice where downregulated expression of the cellcycle inhibitors Cdkn1b and Cdkn1c was reported (Wigleet al., 1999). These differences may be explained by celltype- and differentiation state-specific differences of theProx1 response.

Importantly, the chronic expression of Prox1 in Prox1-negative hemangioendothelioma cells resulted in the acquisi-tion of a partial lymphatic phenotype by these cells, inagreement with recent studies with transient Prox1 expressionin normal human vascular endothelial cells (Hong et al.,2002; Petrova et al., 2002). The chronic versus transient Prox1expression has generated distinct biological consequences of

gene expression pattern exhibiting some interesting differ-ences. First, we did not detect a significant upregulation ofpodoplanin in vitro; however, its strong expression in Prox1tumor cells in situ was evident. Although the mechanism ofthis finding is unknown at this point, a recent study hasindicated some intrinsic in vitro versus in vivo differencesin the regulation of lymphatic lineage-specific genes(Amatschek et al., 2007), likely controlled by microenviron-mental cues. Second, LYVE-1 expression is upregulated bothin vitro and in vivo. Although our data does not providemechanistic explanation for this finding, LYVE-1 is upregu-lated at both mRNA and protein levels in Prox1-expressingcell lines and tumors, which is also corroborated by our genearray analysis. The observed cytoplasmic LYVE-1 staining inProx1-expressing tumor cells, which can also be found onlymphatic endothelium, indicates that LYVE-1 might beexpressed on intracytoplasmic structures, in addition to itswell-established localization at the membrane level.

Although kaposiform hemangioendothelioma (O’Reillyet al., 1997) and KSs are two distinct clinicopathologicalentities, we demonstrate that mouse hemangioendotheliomacells expressing chronic and ectopic human Prox1 exhibitgenotypic and phenotypic features resembling human KS.These characteristics include expression of Prox1, otherlymphatic markers (such as LYVE-1 and podoplanin), lackof well-formed vascular lumina, a connection to the systemiccirculation, and local invasive behavior. Erythrocyte-filledvascular spaces and detection of fluorescently labeled lectin,after intravenous injection, provide direct morphologicalevidence that Prox1-expressing tumors are efficiently per-fused because they are connected to the systemic circulation.This is in contrast to lymphangioma circumscriptum, alymphatic vascular lesion, disconnected from the bloodvascular system, as evidenced by the absence of erythrocytesin vascular spaces (Flanagan and Helwig, 1977). It isimportant to note that infantile hemangiomas—that areusually noninvasive and that do not show signs of cellularanaplasia—express several lymphatic markers but not Prox1(Dadras et al., 2004), whereas KSs that upregulate Prox1 via aKSHV-mediated mechanism (Hong et al., 2004) displayinvasive properties. Thus, it is tempting to speculate that theupregulation of Prox1 contributes to the malignant behaviorof KSs.

Table 2. Quantitative real-time RT-PCR analysis of Prox1-induced genes in Prox1 clones and respective tumors

Gene Control clone Prox1 clone P-value Control tumor Prox1 tumor P-value

von Willebrand factor ND 4.4±0.60 o0.0001 0.96±0.17 4.1±0.43 o0.0001

AFP ND 3.3±0.07 o0.0001 0.26±0.16 1.5±0.07 0.0046

LYVE-1 0.28±0.12 4.4±0.59 o0.0001 0.97±0.17 4.3±0.50 o0.0001

Podoplanin ND 0.38±0.27 0.2302 1.98±0.57 8.57±1.47 0.0028

Flt-4 0.29±0.12 1.8±0.25 0.0001 0.43±0.12 0.49±0.14 0.8102

Data are derived from three individual clones for control and Prox1 constructs and their respective tumors in triplicate. All measurements (fold-induction) areshown relative to the expression levels of mouse GADPH housekeeping gene. ND, not detected.

www.jidonline.org 2803

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

Unexpectedly, Prox1 overexpression resulted in theupregulation of several genes involved in fatty acid metabo-lism, such as very low density lipoprotein receptor, fatty acidbinding protein 4, and peroxisome proliferator activatedreceptor gamma, among others (Table 1; Table S1). Lympha-tic vessels in the intestine—that strongly express Prox1—playa major role in the uptake of fatty acids and of the lipid-soluble vitamins A, D, E, and K (Cueni and Detmar, 2006).Recent studies in Prox1-deficient mice have shown thatheterozygous Prox1-deficient mice are characterized byextreme obesity specifically caused by reduced Prox1expression in lymphatic endothelium (Harvey et al., 2005).Moreover, there has been a long-known association oflymphatic dysfunction with fat accumulation in tissues (Witteet al., 1997). Thus, our study also provides novel insights intothe potential mechanisms by which Prox1 might modulatelipid metabolism and that might link lymphatic dysfunctionand obesity.

In conclusion, our data show that Prox1 gain of functionresults in a more aggressive vascular tumor phenotypewith partial lymphatic differentiation, representing keyfeatures of KS. To our knowledge, this is previouslyunreported. Further studies are needed to evaluate thepotential biological role of Prox1 in different types ofnonvascular malignant tumors.

MATERIALS AND METHODSCell transfection and selection

The mouse hemangioendothelioma cell lines EOMA (Obeso et al.,

1990) and Py-4-1 (Dubois-Stringfellow et al., 1994) were maintained

in complete DMEM (high glucose) with 20% fetal bovine serum,

4 mM L-glutamine, and 1% antibiotic–antimycotic solution (Invitro-

gen, Carlsbad, CA). EOMA and Py-4-1 cells were transfected with a

pcDNA vector alone or with a pcDNA/Prox1 expression vector,

containing the full-length human Prox1 cDNA and a neomycin-

resistance cassette (kindly provided by Dr S.I. Tomarev, National Eye

Institute, Bethesda, MD), using the Superfect transfection reagent

(Qiagen, Chatsworth, CA). Transfected cells were selected in

medium containing 0.4 mg ml�1 (for EOMA) and 0.8 mg ml�1 (for

Py-4-1) G418 (Invitrogen). Stably transfected cell clones were

individually expanded and analyzed for Prox1 mRNA and protein

expression (Hong et al., 2002).

Gene array analyses

Total cellular RNA was extracted from two stably Prox1-expressing

Py-4-1 cell clones and two control-transfected Py-4-1 cell clones.

Microarray analyses were performed using the mouse 430A 2.0

Array GeneChip (Affymetrix, Santa Clara, CA; containing B22,690

genes) according to the manufacturer’s instructions. Arrays were

scanned using an Affymetrix confocal scanner and analyzed by the

Microarray Suite 5.0 software (Affymetrix) as described (Hong et al.,

2004) (Geo accession number GSE8171). Intensity values were

scaled so that the overall fluorescence intensity of each chip of the

same type was equivalent. Genes with a fold change of 1.7 or greater

in both array comparisons (Prox1-expressing clones versus control-

transfected clones) were considered as increased or decreased.

Panther software (http://www.pantherdb.org/) was used to assign

biological processes to significantly overrepresented genes in

Prox1-expressing cells, as compared with control-transfected cells

(Table S1).

RNA isolation and QRT-PCR analysis

Total cellular RNA was isolated from cultured cells and from tumors

using the TRIzol method (Invitrogen) as described (Streit et al., 1999).

After treatment with RQ1 RNAse-free DNase (Promega, Madison,

WI), dual-labeled probe-based real-time RT-PCR reactions were

performed as described (Hong et al., 2004), using an ABI Prism 7000

Sequence Detection System and TaqMan EZ RT-PCR Core Reagent

(Applied Biosystems, Foster City, CA). The gene-specific probes were

labeled with 6-FAM and TAMRA, and then multiplexed with

GAPDH primers, labeled with JOE and TAMRA, as an internal

control (Table S2), as described (Hong et al., 2002). For each sample,

0.2 mg of total RNA were used and all reactions were performed in

triplicates.

Western blot analyses

Cells grown to 80% confluency in 10-cm plates were scraped in

lysis buffer (50 mmol l�1 HEPES, 150 mmol l�1 NaCl, 1% Triton

X-100, 30 mmol l�1 Na-pyrophosphate, 50 mmol l�1 Na-fluoride,

1 mmol l�1 Na-orthovanadate, pH 7.4) containing protease inhibi-

tors (0.02 mol l�1 phenylmethylsulfonyl fluoride, 50 mg ml�1 leupep-

tin, and 50 mg ml�1 aprotinin) (Sigma, St Louis, MO). The cell lysates

were collected and centrifuged at 14,000 r.p.m. for 10 minutes. After

measuring the protein concentration using the BCA protein assay

(Pierce, Rockford, IL), the supernatants were denatured in Laemmli

sample buffer (Bio-Rad, Hercules, CA) using b-mercaptoethanol at

95 1C for 5 minutes, and 30 mg of the total protein was subjected to

SDS-PAGE in duplicates. One protein gel was stained with

Coomassie blue for loading control and the other blotted onto

nitrocellulose membranes (Bio-Rad, Hercules, CA). Membranes

were incubated in 5% nonfat milk–phosphate-buffered saline

blocking solution overnight and were incubated with rabbit

antiserum against human Prox1 (1:1,000) (Table S3). After washes,

membranes were incubated with horseradish-peroxidase-conjugated

anti-rabbit IgG (1:2,000, Amersham Biosciences, Piscataway, NJ)

and analyzed by the enhanced chemiluminescence system (Amer-

sham Biosciences).

Cell growth assays

Control and Prox1-expressing Py-4-1 clones were seeded into 96-

well plates (1,600 cells per well, n¼ 5 per clone; three independent

control and three Prox1 clones) and were incubated with or without

serum for 48 hours. The cells were then incubated in a 0.1 mg ml�1

4-methylumbelliferyl heptanoate (Sigma)/phosphate-buffered saline

solution for 1 hour at 37 1C as described (Detmar et al., 1992). After

washing, the fluorescence intensity—that corresponds to the number

of viable cells (Detmar et al., 1992)—was measured using a Victor2

Fluorometer (PerkinElmer, Boston, MA). The assay was repeated

twice with comparable results.

In vitro monolayer wounding assay

Three independent control (PC1, PC2, PC12) and three independent

Prox1-expressing Py-4-1 clones (PP8, PP11, PP17) were seeded into

24-well plates at a density of 2� 105 cells per well (n¼ 8 per clone).

After reaching confluence, cell monolayers were wounded with

a 200ml pipette tip, washed with phosphate-buffered saline

2804 Journal of Investigative Dermatology (2008), Volume 128

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

(Gibco-Invitrogen, Paisley, UK), and incubated in serum-reduced

medium (2% fetal bovine serum) for 24 hours. Identical wounded

areas were imaged at time point 0 and 24 hours with a phase-

contrast microscope, and the absolute open area (not covered by

cells) was quantified using Photoshop CS3 (Adobe, San Jose, CA).

The percentage of open area after 24 hours, as compared with

0 hours was determined with Excel 2003 (Microsoft, Redmond, WA).

The assay was performed thrice with comparable results.

Tumorigenesis assays and morphologic evaluation

The parental and the stably transfected EOMA and Py4-1 cells were

injected intradermally on each side of the back skin of 8-week-old

female 129P3 and B6D2F1 mice, respectively (Jackson Laboratory,

Bar Harbor, ME). To improve in vivo growth conditions, cells were

mixed with growth factor-reduced Matrigel Matrix (BD Biosciences,

San Jose, CA) at a 1:5 ratio. We injected a total of 14 independent

clones for the evaluation of tumorigenesis and morphology; each

independent cell clone was tested in at least 3–5 mice resulting in a

tally of at least 42 mice. Six control clones were selected from both

EOMA and Py4-1 cell lines, which were transfected with the empty

vector (pcDNA3); these six control clones (PC1, PC2, PC3, EC25,

EC27, and EC29) grew as control tumors. Eight Prox1-expressing cell

clones from both EOMA and Py4-1 cell lines (PP8, PP11, PP12,

PP16, PP21, EP38, EP44 and EP55), which were transfected with the

Prox-1 expression vector (pcDNA/hProx-1), grew as Prox1 tumors.

Necropsy and histological analysis were performed on all tumor-

bearing mice to detect possible organ or lymph node metastases. All

tumors were harvested 14 days after injection and were formalin-

fixed for histological analyses. Two pathologists (S.S.D. and M.C.M.)

analyzed tumor invasion and tumor cell differentiation, while

blinded to the identity of cell clones. The presence of tumor cells

in adjacent skeletal muscle defined tumor invasion, and tumor cell

differentiation was defined by the presence of at least five

pleomorphic cells with highly atypical nuclei per � 10 field. All

animal studies were approved by the Massachusetts General

Hospital Subcommittee on Research Animal Care.

Vascular perfusions

Mice bearing EOMA- and Py-4-1-derived tumors were subjected to

lectin vascular perfusions as described previously (Thurston et al.,

1998). Briefly, anesthetized mice received 200ml of 1 mg ml�1 FITC-

conjugated L. esculentum lectin (Vector Laboratories, Burlingame,

CA) into the retro-orbital sinus. After 2 minutes, the mice were

perfused with 60 ml of a 1% paraformaldehyde solution and then

with 60 ml phosphate-buffered saline for 60 seconds. After the

embedding of tumors and kidneys (positive control for perfusion) in

optimal cutting temperature compound (Miles, Elkhart, IN), 5 mm

cryostat sections were fixed in 4% paraformaldehyde at room

temperature for 20 minutes and were stained with Hoechst

bisbenzimide.

Immunostaining and morphometric vessel analysis

Differential immunofluorescence analyses were performed on 5-mm

cryostat sections, using a rabbit polyclonal antibody against murine

LYVE-1 (kindly provided by Dr David Jackson) and a rat mAb against

murine CD31 (Table S3), followed by secondary antibodies, as

described previously (Dadras et al., 2003). Additional immunohisto-

chemical analyses were performed using antibodies against AFP,

von Willebrand factor, podoplanin, and Prox1 (Table S3). For

specificity controls, the primary antibodies were omitted. Cell nuclei

were counterstained with Hoechst bisbenzimide or hematoxylin.

Computer-assisted morphometric analyses of tumor-associated

lymphatic and blood vessel density and size were performed in a

total of 13 tumors (control, n¼ 6; and Prox1 expressing, n¼ 7), using

the IP-Lab software (Scanalytics, Fairfax, VA) as described previously

(Dadras et al., 2003). For each tumor section, three fields with the

highest lymphatic or blood vascular density (‘‘hot spots’’) were

evaluated at � 100 magnification. Tumor borders were determined

on serial sections, using Hoechst bisbenzimide nuclear stains at

20 mg ml�1 and hematoxylin-and-eosin (H&E) stains.

Statistical analyses

The unpaired Student’s t-test was used to determine the statistical

significance (P-value) of the mean for all vascular parameters, real-

time RT-PCR, tumor cell differentiation, and invasion assays and

in vitro monolayer wounding assay.

CONFLICT OF INTERESTThe authors state no conflict of interest.

ACKNOWLEDGMENTSWe thank L. Janes and J. Bertoncini for expert technical assistance, Dr S.I.Tomarev for providing the pcDNA3/hProx-1 cDNA, Dr D.G. Jackson for themouse anti-LYVE-1 antibody, and Drs K. Alitalo and Y. Hong for the anti-Prox-1 antibodies. This work was supported by NIH/NCI grants CA69184,CA86410, and CA92644 (M.D.), the Susan G. Komen Breast CancerFoundation (M.D.), American Cancer Society Program Project Grant 99-23901 (M.D.), Swiss National Fund grant 3100A0-108207 (M.D.), AustrianScience Foundation grant S9408-B11 (M.D.), Commission of the EuropeanCommunities grant LSHC-CT-2005-518178 (M.D.), an NIH PathologyTraining Grant (S.S.D.), and by the Cutaneous Biology Research Centerthrough the Massachusetts General Hospital/Shiseido Co. Ltd. Agreement(M.D.).

SUPPLEMENTARY MATERIAL

Table S1. Upregulated genes in Prox1-expressing Py-4-1 cells as comparedwith control-transfected cells (log2 ratios; sorted by mean values).

Table S2. Genes and primer sequences used for real-time RT-PCR.

Table S3. Antibodies used for immunostaining analyses.

Figure S1. Hemangioendotheliomas were well perfused.

Figure S2. Prox1 expression induced anaplasia but did not promote cellularproliferation or tumor growth.

Figure S3. Increased LYVE-1 protein expression in Prox1 tumor cells.

REFERENCES

Amatschek S, Kriehuber E, Bauer W, Reininger B, Meraner P, Wolpl A et al.(2007) Blood and lymphatic endothelial cell-specific differentiationprograms are stringently controlled by the tissue environment. Blood109:4777–85

Burke Z, Oliver G (2002) Prox1 is an early specific marker for the developingliver and pancreas in the mammalian foregut endoderm. Mech Dev118:147–55

Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles DM et al.(1994) Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865–9

Cueni LN, Detmar M (2006) New insights into the molecular control of thelymphatic vascular system and its role in disease. J Invest Dermatol126:2167–77

Dadras SS, North PE, Bertoncini J, Mihm MC, Detmar M (2004) Infantilehemangiomas are arrested in an early developmental vascular differ-entiation state. Mod Pathol 17:1068–79

www.jidonline.org 2805

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion

Dadras SS, Paul T, Bertoncini J, Brown LF, Muzikansky A, Jackson DG et al.(2003) Tumor lymphangiogenesis: a novel prognostic indicator forcutaneous melanoma metastasis and survival. Am J Pathol 162:1951–60

Detmar M, Tenorio S, Hettmannsperger U, Ruszczak Z, Orfanos CE (1992)Cytokine regulation of proliferation and ICAM-1 expression of humandermal microvascular endothelial cells in vitro. J Invest Dermatol98:147–53

Dubois-Stringfellow N, Kolpack-Martindale L, Bautch VL, Azizkhan RG(1994) Mice with hemangiomas induced by transgenic endothelial cells.A model for the Kasabach–Merritt syndrome. Am J Pathol 144:796–806

Enjolras O, Mulliken JB, Wassef M, Frieden IJ, Rieu PN, Burrows PE et al.(2000) Residual lesions after Kasabach–Merritt phenomenon in 41patients. J Am Acad Dermatol 42:225–35

Flanagan BP, Helwig EB (1977) Cutaneous lymphangioma. Arch Dermatol113:24–30

Harvey NL, Srinivasan RS, Dillard ME, Johnson NC, Witte MH, Boyd K et al.(2005) Lymphatic vascular defects promoted by Prox1 haploinsufficiencycause adult-onset obesity. Nat Genet 37:1072–81

Hirakawa S, Hong YK, Harvey N, Schacht V, Matsuda K, Libermann T et al.(2003) Identification of vascular lineage-specific genes by transcriptionalprofiling of isolated blood vascular and lymphatic endothelial cells. Am JPathol 162:575–86

Hoak JC, Warner ED, Cheng HF, Fry GL, Hankenson RR (1971) Hemangiomawith thrombocytopenia and microangiopathic anemia (Kasabach–Merrittsyndrome): an animal model. J Lab Clin Med 77:941–50

Hong YK, Foreman K, Shin JW, Hirakawa S, Curry CL, Sage DR et al. (2004)Lymphatic reprogramming of blood vascular endothelium by Kaposisarcoma-associated herpesvirus. Nat Genet 36:683–5

Hong Y-K, Harvey N, Noh Y-H, Schacht V, Hirakawa S, Detmar M et al.(2002) Prox1 is a master control gene in the program specifyinglymphatic endothelial cell fate. Dev Dyn 225:351–7

Laerm A, Helmbold P, Goldberg M, Dammann R, Holzhausen HJ, BallhausenWG (2007) Prospero-related homeobox 1 (PROX1) is frequentlyinactivated by genomic deletions and epigenetic silencing in carcinomasof the biliary system. J Hepatol 46:89–97

Lavado A, Oliver G (2007) Prox1 expression patterns in the developing andadult murine brain. Dev Dyn 236:518–24

Nagai H, Li Y, Hatano S, Toshihito O, Yuge M, Ito E et al. (2003) Mutationsand aberrant DNA methylation of the PROX1 gene in hematologicmalignancies. Genes Chromosomes Cancer 38:13–21

Obeso J, Weber J, Auerbach R (1990) A hemangioendothelioma-derived cellline: its use as a model for the study of endothelial cell biology. LabInvest 63:259–69

Petrova TV, Maekinen T, Maekelae TP, Saarela J, Virtanen I, Ferrell RE et al.(2002) Lymphatic endothelial reprogramming of vascular endo-thelial cells by the Prox-1 homeobox transcription factor. EMBO J 21:4593–9

Sarkar M, Mulliken JB, Kozakewich HP, Robertson RL, Burrows PE (1997)Thrombocytopenic coagulopathy (Kasabach–Merritt phenomenon) isassociated with Kaposiform hemangioendothelioma and not withcommon infantile hemangioma. Plast Reconstr Surg 100:1377–86

Shimoda M, Takahashi M, Yoshimoto T, Kono T, Ikai I, Kubo H (2006)A homeobox protein, prox1, is involved in the differentiation, prolifera-tion, and prognosis in hepatocellular carcinoma. Clin Cancer Res 12:6005–11

Sosa-Pineda B, Wigle JT, Oliver G (2000) Hepatocyte migration during liverdevelopment requires Prox1. Nat Genet 25:254–5

Streit M, Riccardi L, Velasco P, Brown LF, Hawighorst T, Bornstein P et al.(1999) Thrombospondin-2: a potent endogenous inhibitor of tumorgrowth and angiogenesis. Proc Natl Acad Sci USA 96:14888–93

Su H, Reano A, Hesse S, Viac J, Thivolet J (1990) Modulation of bullouspemphigoid antigens by gamma interferon in cultured human keratino-cytes. J Dermatol 17:16–23

Thurston G, Murphy TJ, Baluk P, Lindsey JR, McDonald DM (1998)Angiogenesis in mice with chronic airway inflammation: strain-dependent differences. Am J Pathol 153:1099–112

Wang HW, Trotter MW, Lagos D, Bourboulia D, Henderson S, Makinen Tet al. (2004) Kaposi sarcoma herpesvirus-induced cellular reprogram-ming contributes to the lymphatic endothelial gene expression in Kaposisarcoma. Nat Genet 36:687–93

Wigle JT, Chowdhury K, Gruss P, Oliver G (1999) Prox1 function is crucial formouse lens-fibre elongation. Nat Genet 21:318–22

Wigle JT, Oliver G (1999) Prox1 function is required for the development ofthe murine lymphatic system. Cell 98:769–78

Witte MH, Way DL, Witte CL, Bernas M (1997) Lymphangiogenesis:mechanisms, significance and clinical implications. EXS 79:65–112

Zukerberg LR, Nickoloff BJ, Weiss SW (1993) Kaposiform hemangioendothe-lioma of infancy and childhood. An aggressive neoplasm associated withKasabach–Merritt syndrome and lymphangiomatosis. Am J Surg Pathol17:321–8

2806 Journal of Investigative Dermatology (2008), Volume 128

SS Dadras et al.Prox-1 Promotes Hemangioendothelioma Invasion